The Notch pathway is frequently altered in head and neck squamous cell carcinomas (HNSCC); however, the clinical significance of NOTCH1 dysregulation is poorly understood. This study was designed to characterize expression of the transcriptionally active NOTCH1 intracellular domain (NICD1) in HNSCCs and evaluate its association with NOTCH1 mutation status and clinical parameters. IHC for NICD1 was performed on 79 previously sequenced archival HNSCCs with known NOTCH1 mutation status. Three distinct immunohistochemical staining patterns were identified: positive/peripheral (47%), positive/nonperipheral (34%), and negative (19%). NICD1 expression was associated with NOTCH1 mutation status (P < 0.001). Most NOTCH1–wild-type tumors were peripheral (55%), whereas mutated NOTCH1 tumors were most commonly negative (47%). Nonperipheral tumors were more likely than peripheral tumors to have extracapsular spread [adjusted odds ratio (aOR), 16.01; 95% confidence interval (CI), 1.92–133.46; P = 0.010] and poor differentiation (aOR, 5.27; 95% CI, 0.90–30.86; P = 0.066). Negative staining tumors tended to be poorly differentiated (aOR, 24.71; 95% CI, 1.53–399.33; P = 0.024) and were less likely to be human papillomavirus (HPV) positive (aOR, 0.043; 95% CI, 0.001–1.59; P = 0.087). NOTCH1 mutagenesis was significantly associated with HPV status, with NOTCH1–wild-type tumors more likely to be HPV positive than NOTCH1-mutated tumors (aOR, 19.06; 95% CI, 1.31–276.15; P = 0.031). TP53 disruptive mutations were not associated with NICD1 expression or NOTCH1 mutation. In conclusion, NICD1 is expressed in three distinct patterns in HNSCC that are significantly associated with high-risk features. These findings further support a dual role for NOTCH1 as both tumor suppressor and oncogene in HNSCC. Further research is necessary to clarify the role of NOTCH1 in HNSCC and understand the clinical and therapeutic implications therein. Cancer Prev Res; 8(4); 287–95. ©2015 AACR.

Head and neck squamous cell carcinoma (HNSCC) is the seventh most common malignancy worldwide (1). Tobacco, alcohol, and human papillomavirus (HPV) are responsible for the majority of HNSCCs (2, 3). Genetic alterations in HNSCC are highly heterogeneous, and distinct for HPV-negative and HPV-positive tumors (4–6). The diversity of genetic alterations and tumor-suppressor predominance in HNSCC has underscored the importance of identifying molecular targets for tailored therapeutic regimens specific to the unique characteristics of individual tumors (7, 8).

Recently, NOTCH1 was identified as a frequently mutated gene in HNSCC, with 10% to 15% prevalence of inactivating mutations (4, 5, 9, 10). The NOTCH1 protein is one of four Notch transmembrane signaling protein paralogs with key roles in the regulation of cell differentiation, proliferation, and survival (11, 12). NOTCH1 is activated in a juxtacrine fashion when bound by ligands on neighboring cells. Following ligand binding, stepwise proteolytic cleavage releases the effector domain of the NOTCH1 protein, the Notch1 intracellular domain (NICD1), for translocation to the nucleus. The NICD1 binds transcriptional coactivators and initiates transcription of various target genes involved in cell differentiation and proliferation. The downstream effects of NOTCH1 activation are highly context dependent, and vary with cell lineage, pathology, and stage of differentiation (11, 12). In normal keratinocytes, the cell type from which HNSCCs are derived, NICD1 signaling promotes cell differentiation (13).

NOTCH1 acts as a tumor suppressor or as an oncogene in hematopoietic and solid organ malignancies, depending on the cancer type (11). In HNSCC, whole-exome sequencing revealed loss-of-function mutations consistent with a tumor-suppressor role (4, 5). Although subsequent studies confirmed the NOTCH1 inactivating mutations (9, 10) and demonstrated NOTCH1 tumor-suppressor activity in oral squamous cell carcinoma cell lines (9), NOTCH1 dysregulation appears to be more complex than simple loss-of-function (10). Indeed, approximately one third of HNSCCs displayed evidence of increased NOTCH1 pathway activation as compared with normal mucosa (10).

Studies of NOTCH1 protein expression in HNSCC are similarly conflicting. Both NOTCH1 over- and underexpression have been observed in tumors compared with normal tissue (10, 14). Increased NOTCH1 expression by IHC has been correlated with poor prognosis (15, 16), and high-risk clinical features, including cervical lymph node metastasis (15, 17), advanced stage (15), higher histologic grade (15), greater depth of invasion (17), and cisplatin resistance (18, 19). These findings appear to be at odds with the putative tumor-suppressor role of this protein. To our knowledge, IHC studies to date have only evaluated full-length NOTCH1, which is not transcriptionally active. Here, we explored the expression patterns of the transcriptionally active NICD1 in HNSCC tumor samples, and their association with NOTCH1 mutation status and clinicopathologic parameters.

Subjects

This study was approved by the Johns Hopkins Hospital Institutional Review Board (Protocol NA_00036235) and informed consent was obtained. Patients treated for HNSCC at the Johns Hopkins Hospital from 1995 to 2010 and for whom tumor whole-exome sequencing data were available were eligible for analysis. Whole-exome sequencing methods and sequencing data for all of the specimens included in this study were previously reported (4). Retrospective medical record abstraction was performed to determine clinicopathologic variables of interest.

HPV tumor status

HPV status for oropharyngeal tumors was based upon p16 IHC and/or DNA in situ hybridization (ISH) results, as available clinically (20) and/or from previous report (4). ISH for high-risk HPV DNA results were available for all 29 oropharyngeal cases and p16 IHC was available for 23 of 29 cases.

NICD1 immunohistochemistry

Paraffin-embedded archival tumor tissue was used to prepare slides with standard 4-μm tissue sections for eligible tumors with sufficient tissue available. Specimens were stained with anti-NICD1 rabbit monoclonal antibody (clone D3B8, catalog #4147, Cell Signaling Technology) using a previously described IHC protocol (21) with slight modifications as follows: slides were immuno-stained on the Ultra Benchmark autostainer, applying 64 minutes of heat-induced epitope retrieval (CC1 buffer) and 44 minutes of antibody incubation (room temperature) followed by an amplification step. The reaction was then developed with ultra-view detection (Ventana-Roche Medical Systems).

Stained specimens were reviewed and categorized into patterns of staining by a pathologist with expertise in tumors of the head and neck (J.A. Bishop). Images were captured using an Olympus BX41 microscope, Olympus DP71 camera and Olympus cellSens Standard software. Cell line–derived mouse xenografts from the HaCaT cell line (22) with known wild-type Notch1 expression (23) and normal human tonsil tissue were used for positive controls. Xenografts from the SCC47-E545K cell line (9) with a known NOTCH1 deletion and lack of NOTCH1 expression were used for negative controls. Cell lines were authenticated on August 18, 2014 (SCC47) or January 9, 2015 (HaCaT), using a short tandem repeat (STR) analysis kit, Identifiler (Applied Biosystems), as directed at the Johns Hopkins Genetic Resources Core Facility. SCC47 was obtained from the University of Michigan (Ann Arbor, MI) and modified to introduce a PI3KA E545K activating mutation to facilitate in vivo tumor growth (24). SCC47 STR was matched with published cell line genotyping (25). HaCaT was purchased from CLS Cell Lines Service GMbH (Eppelheim, Germany). Mice were 6- to 8-week-old, 20 to 22 g Hsd:Athymic Nude-FOXn1nu females purchased from Harlan Laboratories, Inc.

Characterization of TP53 mutations

TP53 mutation data were obtained from whole-exome sequencing as previously described (4). Potential functional significance of the mutations was assessed and categorized as “disruptive” or “nondisruptive” as previously described (26).

Analysis

Descriptive variables were summarized with frequencies and proportions for categorical variables, and medians and interquartile ranges for continuous variables. NICD1 IHC staining pattern, NOTCH1 mutation status, clinicopathologic characteristics, and TP53 disruptive mutation status were considered categorical variables and compared using χ2 tests. Clinicopathologic characteristics were also considered as binary outcome variables and analyzed using logistic regression. Odds ratios (ORs) were reported with 95% confidence intervals (CIs). Two tailed P values less than 0.05 were considered statistically significant. Data analysis was performed using STATA 11.2 (2012).

Study population

Tumors were available for 79 of 105 (75.2%) previously sequenced HNSCCs (4). Patients with available archival specimens were more likely to be female (P = 0.044), have higher T stage tumors (P = 0.028), and harbor a NOTCH1 mutation (P = 0.038) as compared with patients with unavailable tissue. There were no differences in terms of age, race, tobacco and alcohol use, tumor site, HPV status, differentiation, and extracapsular spread (ECS; Supplementary Table S1).

Tumor sites included oropharynx (N = 23, 29%), oral cavity (N = 38, 48%), larynx (N = 10, 13%), hypopharynx (N = 7, 9%), and an unknown primary (N = 1, 1%). Most tumors were incident disease (N = 57, 72%) and had not received prior radiation (N = 62, 78%).

NICD1 IHC patterns

Normal tonsil tissue was used as a control to determine expected NICD1 staining in nonpathologic tissue. Nuclear NICD1 staining was observed in the suprabasal layer of the stratified squamous epithelium, both on the tonsil surface and in the crypts (Fig. 1A and B) in 5 of 5 (100%) controls, which was consistent with previously published results (21). In tumor samples, 64 (81%) stained positive for NICD1 staining. Three distinct patterns of NICD1 IHC staining were observed (Fig. 1C–H). The positive, peripheral pattern consisted of nuclear staining only at the tumor periphery, sparing the outermost cell layer. This was the most common (N = 37, 47%) pattern of staining. The positive, nonperipheral pattern exhibited nuclear staining of tumor cells, but in a more diffuse distribution. The nonperipheral pattern was observed in 34% of samples (N = 27). Nuclear staining was absent in negative pattern tumors (N = 15, 19%).

Figure 1.

NICD1 immunohistochemical staining in normal and tumor tissue. A and B, normal tonsil tissue with nuclear staining in suprabasal epithelial layer. C and D, tumor tissue with nuclear staining in peripheral pattern, sparing outermost layer. E and F, tumor tissue with nuclear staining in nonperipheral pattern. G and H, tumor tissue with negative staining in tumor cells. Magnification: B, ×10; A, C to H, ×20.

Figure 1.

NICD1 immunohistochemical staining in normal and tumor tissue. A and B, normal tonsil tissue with nuclear staining in suprabasal epithelial layer. C and D, tumor tissue with nuclear staining in peripheral pattern, sparing outermost layer. E and F, tumor tissue with nuclear staining in nonperipheral pattern. G and H, tumor tissue with negative staining in tumor cells. Magnification: B, ×10; A, C to H, ×20.

Close modal

Association of NICD1 IHC pattern with NOTCH1 mutation status

NOTCH1 mutations were previously described in 17 (22%) tumors in this study and were predicted to be predominantly inactivating (Table 1 and Fig. 2; ref. 4). Most wild-type NOTCH1 tumors had positive staining (55/62, 89%), and the majority of these demonstrated a peripheral pattern (34/55, 62%; Table 2). Approximately half of the mutated NOTCH1 tumors had a negative pattern (8/17, 47%), including 6 of the 9 (67%) tumors with truncating mutations. Of the 9 tumors with mutated NOTCH1 that stained positive, the majority (6/9, 67%) were in a nonperipheral pattern (Table 1 and Table 2). Overall, NICD1 IHC staining patterns differed significantly by NOTCH1 mutation status (P < 0.001).

Figure 2.

Schematic representation of NOTCH1 mutations. A, depiction of NOTCH1 mutations within corresponding Notch1 exons. B, depiction of Notch1 protein domains corresponding to exons. The high number of mutations that are likely inactivating, for example, truncating and/or located N-terminal to the transmembrane domain, has been described as evidence for a tumor-suppressor role of NOTCH1 in HNSCC (4). LNR, Lin12/Notch repeat; HD, heterodimerization; TM, transmembrane; RAM, RBP-jkappa-associated module; TAD, transcriptional activation domain; PEST, proline (P), glutamic acid (E), serine (S), and threonine (T)-rich domain.

Figure 2.

Schematic representation of NOTCH1 mutations. A, depiction of NOTCH1 mutations within corresponding Notch1 exons. B, depiction of Notch1 protein domains corresponding to exons. The high number of mutations that are likely inactivating, for example, truncating and/or located N-terminal to the transmembrane domain, has been described as evidence for a tumor-suppressor role of NOTCH1 in HNSCC (4). LNR, Lin12/Notch repeat; HD, heterodimerization; TM, transmembrane; RAM, RBP-jkappa-associated module; TAD, transcriptional activation domain; PEST, proline (P), glutamic acid (E), serine (S), and threonine (T)-rich domain.

Close modal
Table 1.

NOTCH1 mutations in individual tumors and corresponding NICD1 IHC staining patterns

Tumor sample IDMutation typeNucleotide (cDNA)Amino acid (protein)Domain of Notch1 proteinExon #NICD1 IHC pattern
HN12PT Nonsense c.5529G>A p.W1843X RAM 30 Nonperipheral 
HN14PT Missense c.1171C>T p.P391S EGF-like domain (10) Peripheral 
HN102PT Missense c.1348G>A p.E450K EGF-like domain (11) Nonperipheral 
HN105PT Missense c.2434G>T p.G812W EGF-like domain (21) 15 Nonperipheral 
 Indel c.2436_2455delGGGT ACAAGTGCAACTGCC fs EGF-like domain (21) 15  
HN107PT Nonsense c.1205C>A p.S402X EGF-like domain (10) Negative 
HN115PT Indel c.1932_1931delGT fs EGF-like domain (17) 12 Peripheral 
 Missense c.4019G>C p.G1340A EGF-like domain (34) 25  
HN117PT Missense c.928G>A p.G310R EGF-like domain (8) Negative 
 Missense c.1366T>C p.C456R EGF-like domain (12)  
HN130PT Nonsense c.2845G>T p.E949X EGF-like domain (25) 18 Negative 
HN139PT Nonsense c.1662C>A p.C554X EGF-like domain (14) 10 Negative 
HN142PT Missense c.1058G> p.R353H EGF-like domain (9) Nonperipheral 
 Missense c.6032T>G p.M2011R Ankyrin repeats Ank_4 32  
HN183PT Nonsense c.5872C>T p.Q1958X Ankyrin repeats Ank_3 31 Negative 
HN194PT Indel c.4665delC fs LNR-3 26 Negative 
HN208PT Missense c.1093C> p.R365C EGF-like domain (9) Nonperipheral 
 Missense c.3838C>T p.R1280C EGF-like domain (33) 23  
HN227PT Missense c.1093C> p.R365C EGF-like domain (9) Nonperipheral 
HN245PT Indel c.1130delT fs EGF-like domain (10) Negative 
HN251PT Missense c.3876C>G p.F1292L EGF-like domain (33) 23 Negative 
HN255PT Missense c.6115G>T p.V2039L Ankyrin repeats Ank_5 33 Peripheral 
 Missense c.6116T>A p.V2039E Ankyrin repeats Ank_5 33  
Tumor sample IDMutation typeNucleotide (cDNA)Amino acid (protein)Domain of Notch1 proteinExon #NICD1 IHC pattern
HN12PT Nonsense c.5529G>A p.W1843X RAM 30 Nonperipheral 
HN14PT Missense c.1171C>T p.P391S EGF-like domain (10) Peripheral 
HN102PT Missense c.1348G>A p.E450K EGF-like domain (11) Nonperipheral 
HN105PT Missense c.2434G>T p.G812W EGF-like domain (21) 15 Nonperipheral 
 Indel c.2436_2455delGGGT ACAAGTGCAACTGCC fs EGF-like domain (21) 15  
HN107PT Nonsense c.1205C>A p.S402X EGF-like domain (10) Negative 
HN115PT Indel c.1932_1931delGT fs EGF-like domain (17) 12 Peripheral 
 Missense c.4019G>C p.G1340A EGF-like domain (34) 25  
HN117PT Missense c.928G>A p.G310R EGF-like domain (8) Negative 
 Missense c.1366T>C p.C456R EGF-like domain (12)  
HN130PT Nonsense c.2845G>T p.E949X EGF-like domain (25) 18 Negative 
HN139PT Nonsense c.1662C>A p.C554X EGF-like domain (14) 10 Negative 
HN142PT Missense c.1058G> p.R353H EGF-like domain (9) Nonperipheral 
 Missense c.6032T>G p.M2011R Ankyrin repeats Ank_4 32  
HN183PT Nonsense c.5872C>T p.Q1958X Ankyrin repeats Ank_3 31 Negative 
HN194PT Indel c.4665delC fs LNR-3 26 Negative 
HN208PT Missense c.1093C> p.R365C EGF-like domain (9) Nonperipheral 
 Missense c.3838C>T p.R1280C EGF-like domain (33) 23  
HN227PT Missense c.1093C> p.R365C EGF-like domain (9) Nonperipheral 
HN245PT Indel c.1130delT fs EGF-like domain (10) Negative 
HN251PT Missense c.3876C>G p.F1292L EGF-like domain (33) 23 Negative 
HN255PT Missense c.6115G>T p.V2039L Ankyrin repeats Ank_5 33 Peripheral 
 Missense c.6116T>A p.V2039E Ankyrin repeats Ank_5 33  

Abbreviations: fs, frameshift; Indel, insertion or deletion; LNR, Lin12/Notch repeat; RAM, RBP-jkappa-associated module.

Table 2.

NOTCH1 mutation compared with NICD1 IHC staining pattern

NICD1 IHC, pattern of staining
Positive, peripheralPositive, nonperipheralNegativeTotal
NOTCH1 mutationN (%)N (%)N (%)N (%)
 Wild-type 34 (55) 21 (34) 7 (11) 62 (78) 
 Missense 2 (25) 4 (50) 2 (25) 8 (10) 
 Truncating 0 (0) 1 (14) 6 (86) 7 (9) 
 Missense and truncating 1 (50) 1 (50) 0 (0) 2 (3) 
Total 37 (47) 27 (34) 15 (19) 79 (100) 
    P < 0.001 
 NICD1 IHC, positive vs. negative 
 Positive Negative Total  
NOTCH1 status N (%) N (%) N (%)  
 Wild-type 55 (89) 7 (11) 62 (78)  
 Mutated 9 (53) 8 (47) 17 (22)  
Total 64 (81) 15 (19) 79 (100)  
   P = 0.002  
NICD1 IHC, pattern of staining
Positive, peripheralPositive, nonperipheralNegativeTotal
NOTCH1 mutationN (%)N (%)N (%)N (%)
 Wild-type 34 (55) 21 (34) 7 (11) 62 (78) 
 Missense 2 (25) 4 (50) 2 (25) 8 (10) 
 Truncating 0 (0) 1 (14) 6 (86) 7 (9) 
 Missense and truncating 1 (50) 1 (50) 0 (0) 2 (3) 
Total 37 (47) 27 (34) 15 (19) 79 (100) 
    P < 0.001 
 NICD1 IHC, positive vs. negative 
 Positive Negative Total  
NOTCH1 status N (%) N (%) N (%)  
 Wild-type 55 (89) 7 (11) 62 (78)  
 Mutated 9 (53) 8 (47) 17 (22)  
Total 64 (81) 15 (19) 79 (100)  
   P = 0.002  

Clinicopathologic characteristics and TP53 disruptive mutations compared by NICD1 IHC pattern

Clinicopathologic characteristics were compared by NICD1 IHC staining pattern (Table 3). NICD1 IHC staining pattern was significantly different by HPV tumor status (P = 0.024). The majority of positive staining tumors were HPV positive (19/22, 86%). Among the positive staining tumors, similar proportions of peripheral (10/20, 50%) and nonperipheral (9/11, 45%) pattern tumors were HPV positive. By contrast, only 3 of 22 (14%) positive staining tumors were HPV negative.

Table 3.

Clinicopathologic characteristics associated with NICD1 IHC staining pattern

NICD1 IHC staining pattern
Positive, peripheralPositive, nonperipheralNegativeTotal
Clinicopathologic characteristicsN (%)N (%)N (%)N (%)P
 37 (47) 27 (34) 15 (19) 79 (100)  
Age, y     0.34 
 ≤55 18 (55) 11 (33) 4 (12) 33 (42)  
 >55 19 (41) 16 (35) 11 (24) 46 (58)  
Sex     0.14 
 Male 24 (44) 22 (41) 8 (15) 54 (68)  
 Female 13 (52) 5 (20) 7 (28) 25 (32)  
Race     0.77 
 White 31 (46) 22 (33) 14 (21) 67 (88)  
 Other 5 (56) 3 (33) 1 (11) 9 (12)  
Tobacco     0.42 
 No 14 (58) 7 (29) 3 (12) 24 (32)  
 Yes 22 (42) 20 (38) 10 (19) 52 (68)  
Alcohol     0.19 
 No 18 (58) 8 (26) 5 (16) 31 (47)  
 Yes 13 (47) 16 (46) 6 (17) 35 (53)  
Site     0.10 
 Oropharynx 8 (35) 12 (52) 3 (13) 23 (29)  
 Other 29 (52) 15 (27) 12 (21) 56 (71)  
HPV status     0.024 
 Negative 1 (17) 2 (33) 3 (50) 6 (23)  
 Positive 10 (50) 9 (45) 1 (5) 20 (77)  
T Stage     0.69 
 T1-2 18 (44) 14 (34) 9 (22) 41 (59)  
 T3-T4 14 (48) 11 (38) 4 (14) 29 (41)  
N Stage     0.54 
 N0 13 (46) 8 (29) 7 (25) 28 (39)  
 N1-N2 19 (44) 17 (40) 7 (16) 43 (61)  
Overall stage     0.29 
 Stage < 4 13 (48) 7 (26) 7 (26) 27 (38)  
 Stage 4 20 (45) 18 (41) 6 (14) 44 (62)  
Differentiation     0.012 
 Moderate or well 29 (56) 15 (29) 8 (15) 52 (74)  
 Poor 3 (17) 8 (44) 7 (39) 18 (26)  
Angiolymphatic invasion     0.30 
 No 8 (47) 6 (35) 3 (18) 17 (47)  
 Yes 7 (47) 11 (58) 1 (5) 19 (53)  
Perineural invasion     0.57 
 No 7 (41) 9 (53) 1 (6) 17 (50)  
 Yes 6 (35) 8 (47) 3 (18) 17 (50)  
ECS     0.034 
 No 9 (64) 2 (14) 3 (21) 14 (42)  
 Yes 5 (36) 11 (58) 3 (16) 19 (58)  
Locoregional recurrence     0.88 
 No 22 (45) 17 (35) 10 (20) 49 (62)  
 Yes 15 (50) 10 (33) 5 (17) 30 (38)  
Distant recurrence     0.27 
 No 34 (50) 21 (31) 13 (19) 68 (86)  
 Yes 3 (27) 6 (55) 2 (18) 11 (14)  
TP53 disruptive mutation     0.90 
 No 27 (46) 21 (36) 11 (19) 59 (75)  
 Yes 10 (50) 6 (30) 4 (20) 20 (25)  
NICD1 IHC staining pattern
Positive, peripheralPositive, nonperipheralNegativeTotal
Clinicopathologic characteristicsN (%)N (%)N (%)N (%)P
 37 (47) 27 (34) 15 (19) 79 (100)  
Age, y     0.34 
 ≤55 18 (55) 11 (33) 4 (12) 33 (42)  
 >55 19 (41) 16 (35) 11 (24) 46 (58)  
Sex     0.14 
 Male 24 (44) 22 (41) 8 (15) 54 (68)  
 Female 13 (52) 5 (20) 7 (28) 25 (32)  
Race     0.77 
 White 31 (46) 22 (33) 14 (21) 67 (88)  
 Other 5 (56) 3 (33) 1 (11) 9 (12)  
Tobacco     0.42 
 No 14 (58) 7 (29) 3 (12) 24 (32)  
 Yes 22 (42) 20 (38) 10 (19) 52 (68)  
Alcohol     0.19 
 No 18 (58) 8 (26) 5 (16) 31 (47)  
 Yes 13 (47) 16 (46) 6 (17) 35 (53)  
Site     0.10 
 Oropharynx 8 (35) 12 (52) 3 (13) 23 (29)  
 Other 29 (52) 15 (27) 12 (21) 56 (71)  
HPV status     0.024 
 Negative 1 (17) 2 (33) 3 (50) 6 (23)  
 Positive 10 (50) 9 (45) 1 (5) 20 (77)  
T Stage     0.69 
 T1-2 18 (44) 14 (34) 9 (22) 41 (59)  
 T3-T4 14 (48) 11 (38) 4 (14) 29 (41)  
N Stage     0.54 
 N0 13 (46) 8 (29) 7 (25) 28 (39)  
 N1-N2 19 (44) 17 (40) 7 (16) 43 (61)  
Overall stage     0.29 
 Stage < 4 13 (48) 7 (26) 7 (26) 27 (38)  
 Stage 4 20 (45) 18 (41) 6 (14) 44 (62)  
Differentiation     0.012 
 Moderate or well 29 (56) 15 (29) 8 (15) 52 (74)  
 Poor 3 (17) 8 (44) 7 (39) 18 (26)  
Angiolymphatic invasion     0.30 
 No 8 (47) 6 (35) 3 (18) 17 (47)  
 Yes 7 (47) 11 (58) 1 (5) 19 (53)  
Perineural invasion     0.57 
 No 7 (41) 9 (53) 1 (6) 17 (50)  
 Yes 6 (35) 8 (47) 3 (18) 17 (50)  
ECS     0.034 
 No 9 (64) 2 (14) 3 (21) 14 (42)  
 Yes 5 (36) 11 (58) 3 (16) 19 (58)  
Locoregional recurrence     0.88 
 No 22 (45) 17 (35) 10 (20) 49 (62)  
 Yes 15 (50) 10 (33) 5 (17) 30 (38)  
Distant recurrence     0.27 
 No 34 (50) 21 (31) 13 (19) 68 (86)  
 Yes 3 (27) 6 (55) 2 (18) 11 (14)  
TP53 disruptive mutation     0.90 
 No 27 (46) 21 (36) 11 (19) 59 (75)  
 Yes 10 (50) 6 (30) 4 (20) 20 (25)  

NICD1 staining pattern was also associated with tumor differentiation (P = 0.012). The majority of poorly differentiated tumors were nonperipheral (44%) or negative pattern (39%). Nonperipheral pattern was associated with a 6-fold increase in odds of poor differentiation, as compared with a peripheral pattern (OR, 6.16; 95% CI, 1.18–22.57). Negative pattern was also associated with increased odds of poor differentiation (OR, 8.46; 95% CI, 1.75–40.81; P = 0.029). These associations remained even after adjustment for HPV tumor status and overall stage of disease (adjusted odds ratio [aOR], 5.27; 95% CI, 0.90–20.86; P = 0.066 for nonperipheral tumors; and aOR, 24.71; 95% CI, 1.53–399.33; P = 0.024 for negative tumors as compared with peripheral tumors).

Finally, NICD1 staining pattern was associated with ECS of lymph node metastases (P = 0.034), and the predominant pattern among tumors with ECS was nonperipheral (58%). As compared with tumors with a peripheral pattern, tumors with a nonperipheral pattern were significantly more likely to have ECS (OR, 9.90; 95% CI, 1.50–65.55; P = 0.017). This association remained robust after adjustment for HPV tumor status and overall stage of disease (aOR, 16.01; 95% CI, 1.92–133.46; P = 0.010). Negative staining pattern, conversely, was not associated with ECS (OR, 1.80; 95% CI, 0.25–12.88; P = 0.56).

TP53 is frequently mutated in HNSCC; therefore, the relationship between disruptive TP53 mutations and NICD1 IHC pattern was explored (Table 3). Disruptive TP53 mutation was not associated with NICD1 IHC staining pattern (P = 0.90), and the proportion of disruptive TP53 mutations was similar for each NICD1 IHC pattern.

Clinicopathologic characteristics and NICD1 IHC pattern in NOTCH1–wild-type tumors

NOTCH1 mutation status was associated with NICD1 IHC staining pattern (Table 2); therefore, clinicopathologic characteristics were compared by NICD1 IHC pattern for NOTCH1–wild-type (WT) tumors only (Supplementary Table S2). Among NOTCH1-WT tumors, oropharyngeal site was significantly associated with IHC pattern (P = 0.0089). All oropharyngeal tumors stained positively (18/18, 100%) and most had a nonperipheral pattern (11/18, 61%). The majority (90%) of oropharyngeal tumors with NOTCH1-WT were HPV positive. There were limited HPV-negative tumors with NOTCH1-WT; therefore, analysis of IHC pattern by HPV tumor status was not feasible.

When restricting to NOTCH-WT tumors, nonperipheral NICD1 IHC pattern was associated with ECS (OR, 14.40; 95% CI, 1.32–157.45; P = 0.029) and poor differentiation (OR, 4.73; 95% CI, 0.98–22.71; P = 0.052).

TP53 disruptive mutation status was not significantly associated with NICD1 IHC patterns among NOTCH1-WT tumors (P = 0.54, Supplementary Table S2).

Clinicopathologic characteristics associated with NOTCH1 mutation status

Clinicopathologic characteristics were then compared by NOTCH1 mutation status. Clinicopathologic characteristics and the proportion of disruptive TP53 mutations were largely similar for Notch1–wild-type and mutated tumors (Table 4), except for HPV tumor status, which was significantly associated with NOTCH1 mutation status (P = 0.006). NOTCH1–wild-type tumors were more commonly HPV-positive than NOTCH1-mutated tumors both in univariate analysis (OR, 13.33; 95% CI, 1.59–111.47; P = 0.017) and after adjustment for overall stage and gender (aOR, 19.06; 95% CI, 1.31–276.15; P = 0.031).

Table 4.

Clinicopathologic characteristics associated with NOTCH1 mutation

NOTCH1 mutation status
Wild-typeMutatedTotal
Clinicopathologic characteristicsN (%)N (%)N (%)p-value
 87 (83) 18 (17) 105 (100)  
Age    0.42 
 ≤55 years 38 (86) 6 (14) 44 (42)  
 >55 years 49 (80) 12 (20) 61 (58)  
Sex    0.061 
 Male 67 (87) 10 (13) 77 (73)  
 Female 20 (71) 8 (29) 28 (27)  
Race    0.91 
 White 73 (82) 16 (18) 89 (88)  
 Other 10 (83) 2 (17) 12 (12)  
Tobacco    0.83 
 No 27 (84) 5 (16) 32 (32)  
 Yes 57 (83) 12 (17) 69 (68)  
Alcohol    0.32 
 No 38 (86) 6 (14) 44 (52)  
 Yes 32 (78) 9 (22) 41 (48)  
Site    0.77 
 Oropharynx 26 (81) 6 (19) 32 (30)  
 Other 61 (84) 12 (16) 73 (70)  
HPV status    0.006 
 Negative 3 (43) 4 (57) 7 (24)  
 Positive 20 (91) 2 (9) 22 (76)  
T Stage    0.43 
 T1-2 52 (85) 9 (15) 61 (65)  
 T3-T4 26 (79) 7 (21) 33 (35)  
N Stage    0.18 
 N0 28 (76) 9 (24) 37 (39)  
 N1-N2 51 (86) 8 (14) 59 (61)  
Overall stage    0.33 
 Stage<4 36 (88) 5 (12) 41 (42)  
 Stage 4 45 (80) 11 (20) 56 (58)  
Differentiation    0.23 
 Moderate or well 53 (84) 10 (16) 63 (71)  
 Poor 19 (73) 7 (27) 26 (29)  
Angiolymphatic invasion    0.58 
 No 16 (80) 4 (20) 20 (48)  
 Yes 19 (86) 3 (14) 22 (52)  
Perineural invasion    0.52 
 No 20 (83) 4 (17) 24 (55)  
 Yes 18 (90) 2 (10) 20 (45)  
ECS    0.98 
 No 15 (88) 2 (12) 17 (40)  
 Yes 22 (88) 3 (12) 25 (60)  
Locoregional recurrence    0.86 
 No 55 (83) 11 (17) 66 (63)  
 Yes 32 (82) 7 (18) 39 (37)  
Distant recurrence    0.59 
 No 73 (82) 16 (18) 89 (85)  
 Yes 14 (88) 2 (12) 16 (15)  
TP53 disruptive mutation    0.66 
 No 67 (84) 13 (16) 80 (76)  
 Yes 20 (80) 5 (20) 25 (24)  
NOTCH1 mutation status
Wild-typeMutatedTotal
Clinicopathologic characteristicsN (%)N (%)N (%)p-value
 87 (83) 18 (17) 105 (100)  
Age    0.42 
 ≤55 years 38 (86) 6 (14) 44 (42)  
 >55 years 49 (80) 12 (20) 61 (58)  
Sex    0.061 
 Male 67 (87) 10 (13) 77 (73)  
 Female 20 (71) 8 (29) 28 (27)  
Race    0.91 
 White 73 (82) 16 (18) 89 (88)  
 Other 10 (83) 2 (17) 12 (12)  
Tobacco    0.83 
 No 27 (84) 5 (16) 32 (32)  
 Yes 57 (83) 12 (17) 69 (68)  
Alcohol    0.32 
 No 38 (86) 6 (14) 44 (52)  
 Yes 32 (78) 9 (22) 41 (48)  
Site    0.77 
 Oropharynx 26 (81) 6 (19) 32 (30)  
 Other 61 (84) 12 (16) 73 (70)  
HPV status    0.006 
 Negative 3 (43) 4 (57) 7 (24)  
 Positive 20 (91) 2 (9) 22 (76)  
T Stage    0.43 
 T1-2 52 (85) 9 (15) 61 (65)  
 T3-T4 26 (79) 7 (21) 33 (35)  
N Stage    0.18 
 N0 28 (76) 9 (24) 37 (39)  
 N1-N2 51 (86) 8 (14) 59 (61)  
Overall stage    0.33 
 Stage<4 36 (88) 5 (12) 41 (42)  
 Stage 4 45 (80) 11 (20) 56 (58)  
Differentiation    0.23 
 Moderate or well 53 (84) 10 (16) 63 (71)  
 Poor 19 (73) 7 (27) 26 (29)  
Angiolymphatic invasion    0.58 
 No 16 (80) 4 (20) 20 (48)  
 Yes 19 (86) 3 (14) 22 (52)  
Perineural invasion    0.52 
 No 20 (83) 4 (17) 24 (55)  
 Yes 18 (90) 2 (10) 20 (45)  
ECS    0.98 
 No 15 (88) 2 (12) 17 (40)  
 Yes 22 (88) 3 (12) 25 (60)  
Locoregional recurrence    0.86 
 No 55 (83) 11 (17) 66 (63)  
 Yes 32 (82) 7 (18) 39 (37)  
Distant recurrence    0.59 
 No 73 (82) 16 (18) 89 (85)  
 Yes 14 (88) 2 (12) 16 (15)  
TP53 disruptive mutation    0.66 
 No 67 (84) 13 (16) 80 (76)  
 Yes 20 (80) 5 (20) 25 (24)  

The Notch signaling pathway is a critical component of normal keratinocyte development, promoting differentiation and cell-cycle withdrawal. Here, we demonstrate that expression of the transcriptionally active NICD1 in HNSCC occurs in three distinct immunohistochemical patterns. In addition, NICD1 expression pattern is associated with NOTCH1 mutation and the high-risk clinical features of ECS and poor differentiation.

Three distinct NICD1 expression patterns

The three NICD1 expression patterns observed by IHC were (i) positive, peripheral; (ii) positive, nonperipheral; and (iii) negative. The peripheral pattern was most common (n = 37, 47%) and resembled the suprabasal nuclear expression of NICD1 observed in normal human tonsil, oropharyngeal mucosa, and skin (21). Therefore, we considered the peripheral pattern to be the “normal” pattern, with retained features of NICD1 expression in nonpathologic keratinocyte differentiation. The peripheral pattern was also significantly more common in the more differentiated tumors, substantiating this hypothesis.

In the nonperipheral pattern, NICD1 expression appeared disorganized, with more diffuse staining among tumor cells and no distinct peripheral pattern. Tumors with nonperipheral NICD1 pattern were more likely to have the high-risk clinical features of poor differentiation and ECS compared with peripheral staining tumors. These findings may be consistent with Notch pathway escape from normal regulatory mechanisms, widespread NOTCH1 activation, and potentially the promotion of an epithelial–mesenchymal transition phenotype that has been associated with NOTCH1 activation in other malignancies (27–29). Although it is not possible to conclude that NOTCH1 is activated in this subset of tumors without examining its downstream targets, we speculate that NOTCH1 may behave as an oncogene in tumors with nonperipheral staining pattern.

In negative staining tumors, the loss of NICD1 expression and the association with poor differentiation suggest a tumor-suppressor role for NOTCH1, as previously suspected from sequencing studies (4, 5). Over half of the negative staining tumors (8/15, 53%) contained putative inactivating (4) NOTCH1 mutations, and most of these (6/8, 75%) were truncating. Negative tumors were less differentiated than peripheral tumors, but unlike nonperipheral tumors, did not have a greater likelihood of ECS. Loss of NICD1 expression is likely tumorigenic due to disruption of the established Notch1 tumor-suppressive functions in keratinocytes, such as promotion of terminal differentiation and inhibition of cell proliferation (13, 30, 31).

The immunohistochemical pattern of NICD1 staining in HSNCC has not been previously reported. Importantly, clinically significant high-risk markers, ECS, and poor differentiation, were associated with NICD1 IHC pattern but not with NOTCH1 mutation status, which may indicate a role for aberrant Notch signaling in HNSCC independent of NOTCH1 mutation.

The importance of NICD1 expression pattern is perhaps not surprising because canonical Notch signaling is mediated by cell-to-cell interactions, and is, therefore, dependent on tissue architecture (31, 32). The Notch pathway has been shown in other malignancies to interact with various components of the tumor microenvironment, including endothelial cells, stroma, and secreted molecules (31, 33–35). The significant association of nonperipheral NICD1 expression with high-risk features in our study implies that dysregulation of NOTCH1-mediated communication of HNSCC tumors cells with the surrounding microenvironment may be a determinant of disease progression.

Dual role of NOTCH1 in HNSCC

A dual role for NOTCH1 in HNSCC as both a tumor suppressor and oncogene would be consistent with the heterogeneous and context-specific actions of NOTCH1 in normal development and in other cancers. In normal development, Notch signaling promotes a variety of competing cell-cycle regulatory decisions, including terminal differentiation, proliferation and apoptosis, depending on the tissue, developmental stage and other parameters (12, 36). In human malignancies, NOTCH1 behaves as an oncogene in T-ALL (37, 38) and several solid tumors, but as a tumor suppressor in other cancers (32, 39). In fact, there is increasing support for a dual oncogenic and tumor-suppressor role for Notch signaling within the same solid tumor type in several malignancies, including breast, pancreatic, esophageal, and non-small cell lung cancer (32, 40).

Specific to HNSCC, a dual role for NOTCH1 may help to explain apparent incongruities in studies reported thus far. Initially, whole-exome sequencing revealed inactivating NOTCH1 mutations in 10% to 15% of HNSCCs, indicative of a tumor-suppressor role (4, 5). An integrative genomic analysis then reported that NICD1 expression in HNSCC cell lines with inactivating NOTCH1 mutations resulted in decreased cell growth and reduced tumor size in mice, again consistent with NOTCH1 functioning as a tumor suppressor (9). However, a subsequent comprehensive characterization of NOTCH1 in HNSCC demonstrated NOTCH1 pathway activation in a subset of tumors, with increased copy number or gene expression of NOTCH1 ligands such as JAG1 and JAG2. Furthermore, nearly one third of tumors (14/44) exhibited increased expression of NOTCH1 downstream target genes HES1 and/or HEY1 (10), suggesting an oncogenic rather than tumor-suppressor mechanism. Our findings introduce the possibility that the characteristics of Notch dysregulation may in fact differ from tumor to tumor among HNSCCs, with NOTCH1 functioning as a tumor suppressor in a subset of tumors and as an oncogene in another subset, while retaining normal function in the remainder. This paradigm would aid in interpreting the apparent inconsistencies of existing literature.

NICD1 is dysregulated independent of NOTCH1 mutation

Although NICD1 expression was significantly associated with NOTCH1 mutation status, there was an evident discordance. Abnormal (nonperipheral or negative) NICD1 IHC patterns were observed in 45% of wild-type NOTCH1 tumors and a normal (peripheral) pattern was seen in 18% of tumors with mutated NOTCH1. There are several explanations for this discrepancy, including heterogeneous functional consequences of NOTCH1 mutation dependent upon location within the NOTCH1 protein, potential sampling error in some tumors, and additional mechanisms of Notch pathway alterations independent of gene mutation. The latter is likely the driving factor behind our observations given that when analysis was limited to wild-type NOTCH1 tumors, the associations with ECS and differentiation noted in the whole cohort remained robust. There are numerous modalities of Notch signaling perturbations in other diseases, such as chromosomal translocation (38), abnormal activation by Notch ligands such as JAG1 and JAG2 (10, 27, 41), epigenetic modifications (42), and altered transcriptional coactivation (43). Downregulation or inhibition of NOTCH1 cleavage by gamma-secretase, required for release of NICD1, is another possible mechanism to explain loss of NICD1 expression in tumors with wild-type NOTCH1. Pathway-level Notch signaling aberrations in HNSCC have not yet been fully elucidated, but our results support their importance in a significant subset of tumors.

Therapeutic and clinical implications

The findings reported in this study may have therapeutic implications. If indeed Notch pathway dysregulation in HNSCC is bimodal, there may be an opportunity to tailor Notch-targeted therapy based on biomarkers of oncogenic or tumor-suppressor NOTCH1 activity in specific tumors. Several Notch pathway–specific drugs are currently under study for the treatment of solid tumors, including both inhibitors such as gamma-secretase inhibitors, and activators such as valproic acid (11).

The association of nonperipheral staining tumors with ECS and poor differentiation may be consistent with prior studies correlating increased NOTCH1 expression with poor prognostic parameters in HNSCC such as decreased survival (15, 16), cervical lymph node metastasis (15, 17), advanced stage (15), higher histologic grade (15), and cisplatin resistance (18, 19). Although these studies did not specifically evaluate transcriptionally active NOTCH1, that is, the NICD1, it is conceivable that increased NOTCH1 expression as reported corresponds to the nonperipheral pattern in our study, with its more diffuse NICD1 expression. Additional larger studies are necessary to determine the prognostic significance of NICD1 expression pattern.

HPV status and NOTCH1

The association of HPV-negative tumor status with both negative NICD1 expression and presence of NOTCH1 mutation has not been previously reported to our knowledge. The significance of this finding is unclear. In cervical cancer cell line studies, NOTCH1 interacts with HPV oncoproteins E6 and E7 in a complex manner: NOTCH1 has been shown to inhibit E6/E7 expression and induce growth arrest (44, 45), but E6/E7 have also been reported to upregulate NOTCH1 expression and lead to cell transformation (46). The association observed in our study may be a result of NOTCH1–E6/E7 interaction, but may also be a reflection of fewer genetic alterations in HPV-positive versus HPV-negative tumors (4, 5).

Disruptive TP53 mutations and NOTCH1

TP53 is the most commonly mutated gene in HNSCC and considerable cross-talk is observed between p53 and Notch signaling in keratinocytes (47–50); however, in our study there was no significant correlation between TP53 disruptive mutations and either NICD1 expression or NOTCH1 mutation status. This may indicate that Notch dysregulation is TP53 independent in HNSCCs, although it is difficult to draw conclusions in this relatively small sample size.

Limitations

The findings in this study are largely descriptive, and are, therefore, limited to hypothesis generation with regard to mechanistic reasons for and molecular consequences of the distinct NICD1 expression patterns. Without characterization of upstream regulators and downstream targets associated with the NICD1 expression patterns described here it is not possible to draw conclusions regarding the significance of these patterns in HNSCC. In addition, among the eligible archival tumors specimens, there were significant differences in those with available tissue for NICD1 IHC staining compared with those without tissue available, including higher T stage and higher likelihood of both having a NOTCH1 mutation and being from a female patient. The increased amount of tissue in higher T stage tumors likely improved availability. However, the higher prevalence of NOTCH1 mutation and greater female representation in this group are unexplained, and may have affected the results reported here.

Notch signaling alterations occur in a subset of HNSCCs but are poorly understood. The findings reported in this study indicate that there are three distinct patterns of NICD1 expression in HNSCC, and that these patterns are significantly associated with the presence or absence of clinically high-risk features. Although this may be consistent with a dual tumor suppressor and oncogene role for NOTCH1 in HNSCC, further research is necessary to confirm these findings, fully characterize Notch signaling aberrations in HNSCC, and elucidate the clinical importance and therapeutic implications therein.

No potential conflicts of interest were disclosed.

The content is solely the responsibility of the authors and does not necessarily represent the official view of the NIH.

Conception and design: E.M. Rettig, C.H. Chung, J.D. Howard, N. Agrawal, C. Fakhry

Development of methodology: E.M. Rettig, C.H. Chung, J.D. Howard, R. Sharma, W. Koch, N. Agrawal, C. Fakhry

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): E.M. Rettig, C.H. Chung, J.D. Howard, R.J. Li, D. Sidransky, W. Koch, N. Agrawal, C. Fakhry

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): E.M. Rettig, C.H. Chung, J.A. Bishop, R.J. Li, C. Douville, R. Karchin, E. Izumchenko, D. Sidransky, W. Koch, J. Califano, N. Agrawal, C. Fakhry

Writing, review, and/or revision of the manuscript: E.M. Rettig, C.H. Chung, J.A. Bishop, J.D. Howard, C. Douville, E. Izumchenko, D. Sidransky, W. Koch, J. Califano, N. Agrawal, C. Fakhry

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): C.H. Chung, E. Izumchenko, C. Fakhry

Study supervision: J.D. Howard, N. Agrawal, C. Fakhry

The research reported in this article was supported by the National Institute of Dental and Craniofacial Research (NIDCR) and NIH Specialized Program of Research Excellence grant P50DE019032 (to E.M. Rettig, C.H. Chung, J.A. Bishop, E. Izumchenko, D. Sidransky, W. Koch, J. Califano, N. Agrawal, and C. Fakhry), NIDCR grant 2T32DC000027-26 (to E.M. Rettig), and the Oral Cancer Foundation (C. Fakhry).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Ferlay
J
,
Shin
HR
,
Bray
F
,
Forman
D
,
Mathers
C
,
Parkin
DM
. 
Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008
.
Int J Cancer
2010
;
127
:
2893
917
.
2.
Gillison
ML
,
D'Souza
G
,
Westra
W
,
Sugar
E
,
Xiao
W
,
Begum
S
, et al
Distinct risk factor profiles for human papillomavirus type 16-positive and human papillomavirus type 16-negative head and neck cancers
.
J Natl Cancer Inst
2008
;
100
:
407
20
.
3.
Forastiere
A
,
Koch
W
,
Trotti
A
,
Sidransky
D
. 
Head and neck cancer
.
N Engl J Med
2001
;
345
:
1890
900
.
4.
Agrawal
N
,
Frederick
MJ
,
Pickering
CR
,
Bettegowda
C
,
Chang
K
,
Li
RJ
, et al
Exome sequencing of head and neck squamous cell carcinoma reveals inactivating mutations in NOTCH1
.
Science
2011
;
333
:
1154
7
.
5.
Stransky
N
,
Egloff
AM
,
Tward
AD
,
Kostic
AD
,
Cibulskis
K
,
Sivachenko
A
, et al
The mutational landscape of head and neck squamous cell carcinoma
.
Science
2011
;
333
:
1157
60
.
6.
Seiwert
TY
,
Zuo
Z
,
Keck
MK
,
Khattri
A
,
Pedamallu
CS
,
Stricker
TP
, et al
Integrative and comparative genomic analysis of HPV-positive and HPV-negative head and neck squamous cell carcinomas
.
Clin Cancer Res
2015
;
21
:
632
41
.
7.
Loyo
M
,
Li
RJ
,
Bettegowda
C
,
Pickering
CR
,
Frederick
MJ
,
Myers
JN
, et al
Lessons learned from next-generation sequencing in head and neck cancer
.
Head Neck
2013
;
35
:
454
63
.
8.
Leemans
CR
,
Braakhuis
BJ
,
Brakenhoff
RH
. 
The molecular biology of head and neck cancer
.
Nat Rev Cancer
2011
;
11
:
9
22
.
9.
Pickering
CR
,
Zhang
J
,
Yoo
SY
,
Bengtsson
L
,
Moorthy
S
,
Neskey
DM
, et al
Integrative genomic characterization of oral squamous cell carcinoma identifies frequent somatic drivers
.
Cancer Discov
2013
;
3
:
770
81
.
10.
Sun
W
,
Gaykalova
DA
,
Ochs
MF
,
Mambo
E
,
Arnaoutakis
D
,
Liu
Y
, et al
Activation of the NOTCH pathway in head and neck cancer
.
Cancer Res
2014
;
74
:
1091
104
.
11.
Egloff
AM
,
Grandis
JR
. 
Molecular pathways: context-dependent approaches to Notch targeting as cancer therapy
.
Clin Cancer Res
2012
;
18
:
5188
95
.
12.
Louvi
A
,
Artavanis-Tsakonas
S
. 
Notch and disease: a growing field
.
Semin Cell Dev Biol
2012
;
23
:
473
80
.
13.
Dotto
GP
. 
Notch tumor suppressor function
.
Oncogene
2008
;
27
:
5115
23
.
14.
Sakamoto
K
,
Fujii
T
,
Kawachi
H
,
Miki
Y
,
Omura
K
,
Morita
K
, et al
Reduction of NOTCH1 expression pertains to maturation abnormalities of keratinocytes in squamous neoplasms
.
Lab Invest
2012
;
92
:
688
702
.
15.
Li
D
,
Dong
P
,
Wu
C
,
Cao
P
,
Zhou
L
. 
Notch1 overexpression associates with poor prognosis in human laryngeal squamous cell carcinoma
.
Ann Otol Rhinol Laryngol
2014
;
123
:
705
10
.
16.
Lin
JT
,
Chen
MK
,
Yeh
KT
,
Chang
CS
,
Chang
TH
,
Lin
CY
, et al
Association of high levels of Jagged-1 and Notch-1 expression with poor prognosis in head and neck cancer
.
Ann Surg Oncol
2010
;
17
:
2976
83
.
17.
Joo
YH
,
Jung
CK
,
Kim
MS
,
Sun
DI
. 
Relationship between vascular endothelial growth factor and Notch1 expression and lymphatic metastasis in tongue cancer
.
Otolaryngol Head Neck Surg
2009
;
140
:
512
8
.
18.
Gu
F
,
Ma
Y
,
Zhang
Z
,
Zhao
J
,
Kobayashi
H
,
Zhang
L
, et al
Expression of Stat3 and Notch1 is associated with cisplatin resistance in head and neck squamous cell carcinoma
.
Oncol Rep
2010
;
23
:
671
6
.
19.
Zhang
ZP
,
Sun
YL
,
Fu
L
,
Gu
F
,
Zhang
L
,
Hao
XS
. 
Correlation of Notch1 expression and activation to cisplatin-sensitivity of head and neck squamous cell carcinoma
.
Ai Zheng
2009
;
28
:
100
3
.
20.
Singhi
AD
,
Westra
WH
. 
Comparison of human papillomavirus in situ hybridization and p16 immunohistochemistry in the detection of human papillomavirus-associated head and neck cancer based on a prospective clinical experience
.
Cancer
2010
;
116
:
2166
73
.
21.
Kluk
MJ
,
Ashworth
T
,
Wang
H
,
Knoechel
B
,
Mason
EF
,
Morgan
EA
, et al
Gauging NOTCH1 activation in cancer using immunohistochemistry
.
PLoS ONE
2013
;
8
:
e67306
.
22.
Boukamp
P
,
Stanbridge
EJ
,
Foo
DY
,
Cerutti
PA
,
Fusenig
NE
. 
c-Ha-ras oncogene expression in immortalized human keratinocytes (HaCaT) alters growth potential in vivo but lacks correlation with malignancy
.
Cancer Res
1990
;
50
:
2840
7
.
23.
Fertig
EJ
,
Ren
Q
,
Cheng
H
,
Hatakeyama
H
,
Dicker
AP
,
Rodeck
U
, et al
Gene expression signatures modulated by epidermal growth factor receptor activation and their relationship to cetuximab resistance in head and neck squamous cell carcinoma
.
BMC Genomics
2012
;
13
:
160
.
24.
Qiu
W
,
Schonleben
F
,
Li
X
,
Ho
DJ
,
Close
LG
,
Manolidis
S
, et al
PIK3CA mutations in head and neck squamous cell carcinoma
.
Clin Cancer Res
2006
;
12
:
1441
6
.
25.
Brenner
JC
,
Graham
MP
,
Kumar
B
,
Saunders
LM
,
Kupfer
R
,
Lyons
RH
, et al
Genotyping of 73 UM-SCC head and neck squamous cell carcinoma cell lines
.
Head Neck
2010
;
32
:
417
26
.
26.
Masica
DL
,
Li
S
,
Douville
C
,
Manola
J
,
Ferris
RL
,
Burtness
B
, et al
Predicting survival in head and neck squamous-cell carcinoma from TP53 mutation
.
Hum Genet
. 
2014
Aug 10. [Epub ahead of print]
.
27.
Leong
KG
,
Niessen
K
,
Kulic
I
,
Raouf
A
,
Eaves
C
,
Pollet
I
, et al
Jagged1-mediated Notch activation induces epithelial-to-mesenchymal transition through Slug-induced repression of E-cadherin
.
J Exp Med
2007
;
204
:
2935
48
.
28.
Bao
B
,
Wang
Z
,
Ali
S
,
Kong
D
,
Li
Y
,
Ahmad
A
, et al
Notch-1 induces epithelial-mesenchymal transition consistent with cancer stem cell phenotype in pancreatic cancer cells
.
Cancer Lett
2011
;
307
:
26
36
.
29.
Santagata
S
,
Demichelis
F
,
Riva
A
,
Varambally
S
,
Hofer
MD
,
Kutok
JL
, et al
JAGGED1 expression is associated with prostate cancer metastasis and recurrence
.
Cancer Res
2004
;
64
:
6854
7
.
30.
Nicolas
M
,
Wolfer
A
,
Raj
K
,
Kummer
JA
,
Mill
P
,
van Noort
M
, et al
Notch1 functions as a tumor suppressor in mouse skin
.
Nat Genet
2003
;
33
:
416
21
.
31.
Demehri
S
,
Turkoz
A
,
Kopan
R
. 
Epidermal Notch1 loss promotes skin tumorigenesis by impacting the stromal microenvironment
.
Cancer Cell
2009
;
16
:
55
66
.
32.
Ranganathan
P
,
Weaver
KL
,
Capobianco
AJ
. 
Notch signalling in solid tumours: a little bit of everything but not all the time
.
Nat Rev Cancer
2011
;
11
:
338
51
.
33.
Howard
JD
,
Moriarty
WF
,
Park
J
,
Riedy
K
,
Panova
IP
,
Chung
CH
, et al
Notch signaling mediates melanoma-endothelial cell communication and melanoma cell migration
.
Pigment Cell Melanoma Res
2013
;
26
:
697
707
.
34.
Charles
N
,
Ozawa
T
,
Squatrito
M
,
Bleau
AM
,
Brennan
CW
,
Hambardzumyan
D
, et al
Perivascular nitric oxide activates notch signaling and promotes stem-like character in PDGF-induced glioma cells
.
Cell Stem Cell
2010
;
6
:
141
52
.
35.
Lu
J
,
Ye
X
,
Fan
F
,
Xia
L
,
Bhattacharya
R
,
Bellister
S
, et al
Endothelial cells promote the colorectal cancer stem cell phenotype through a soluble form of Jagged-1
.
Cancer Cell
2013
;
23
:
171
85
.
36.
Aster
JC
,
Blacklow
SC
. 
Targeting the Notch pathway: twists and turns on the road to rational therapeutics
.
J Clin Oncol
2012
;
30
:
2418
20
.
37.
Weng
AP
,
Ferrando
AA
,
Lee
W
,
Morris
JPt
,
Silverman
LB
,
Sanchez-Irizarry
C
, et al
Activating mutations of NOTCH1 in human T cell acute lymphoblastic leukemia
.
Science
2004
;
306
:
269
71
.
38.
Ellisen
LW
,
Bird
J
,
West
DC
,
Soreng
AL
,
Reynolds
TC
,
Smith
SD
, et al
TAN-1, the human homolog of the Drosophila notch gene, is broken by chromosomal translocations in T lymphoblastic neoplasms
.
Cell
1991
;
66
:
649
61
.
39.
Klinakis
A
,
Lobry
C
,
Abdel-Wahab
O
,
Oh
P
,
Haeno
H
,
Buonamici
S
, et al
A novel tumour-suppressor function for the Notch pathway in myeloid leukaemia
.
Nature
2011
;
473
:
230
3
.
40.
Kagawa
S
,
Natsuizaka
M
,
Whelan
KA
,
Facompre
N
,
Naganuma
S
,
Ohashi
S
, et al
Cellular senescence checkpoint function determines differential Notch1-dependent oncogenic and tumor-suppressor activities
.
Oncogene
. 
2014
Jun 16. [Epub ahead of print]
.
41.
Zeng
Q
,
Li
S
,
Chepeha
DB
,
Giordano
TJ
,
Li
J
,
Zhang
H
, et al
Crosstalk between tumor and endothelial cells promotes tumor angiogenesis by MAPK activation of Notch signaling
.
Cancer Cell
2005
;
8
:
13
23
.
42.
Felician
G
,
Collesi
C
,
Lusic
M
,
Martinelli
V
,
Dal Ferro
M
,
Zentilin
L
, et al
Epigenetic modification at Notch responsive promoters blunts efficacy of inducing Notch pathway reactivation after myocardial infarction
.
Circ Res
2014
;
115
:
636
49
.
43.
Wu
L
,
Griffin
JD
. 
Modulation of Notch signaling by mastermind-like (MAML) transcriptional co-activators and their involvement in tumorigenesis
.
Semin Cancer Biol
2004
;
14
:
348
56
.
44.
Talora
C
,
Sgroi
DC
,
Crum
CP
,
Dotto
GP
. 
Specific down-modulation of Notch1 signaling in cervical cancer cells is required for sustained HPV-E6/E7 expression and late steps of malignant transformation
.
Genes Dev
2002
;
16
:
2252
63
.
45.
Talora
C
,
Cialfi
S
,
Segatto
O
,
Morrone
S
,
Kim Choi
J
,
Frati
L
, et al
Constitutively active Notch1 induces growth arrest of HPV-positive cervical cancer cells via separate signaling pathways
.
Exp Cell Res
2005
;
305
:
343
54
.
46.
Weijzen
S
,
Zlobin
A
,
Braid
M
,
Miele
L
,
Kast
WM
. 
HPV16 E6 and E7 oncoproteins regulate Notch-1 expression and cooperate to induce transformation
.
J Cell Physiol
2003
;
194
:
356
62
.
47.
Yugawa
T
,
Handa
K
,
Narisawa-Saito
M
,
Ohno
S
,
Fujita
M
,
Kiyono
T
. 
Regulation of Notch1 gene expression by p53 in epithelial cells
.
Mol Cell Biol
2007
;
27
:
3732
42
.
48.
Lefort
K
,
Mandinova
A
,
Ostano
P
,
Kolev
V
,
Calpini
V
,
Kolfschoten
I
, et al
Notch1 is a p53 target gene involved in human keratinocyte tumor suppression through negative regulation of ROCK1/2 and MRCKalpha kinases
.
Genes Dev
2007
;
21
:
562
77
.
49.
Duan
L
,
Yao
J
,
Wu
X
,
Fan
M
. 
Growth suppression induced by Notch1 activation involves Wnt-beta-catenin down-regulation in human tongue carcinoma cells
.
Biol Cell
2006
;
98
:
479
90
.
50.
Dotto
GP
. 
Crosstalk of Notch with p53 and p63 in cancer growth control
.
Nat Rev Cancer
2009
;
9
:
587
95
.