Skip to main content
  • AACR Journals
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

AACR logo

  • Register
  • Log in
  • My Cart
Advertisement

Main menu

  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
    • Reviewing
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Focus on Radiation Oncology
      • Novel Combinations
      • Reviews
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citation
    • Author/Keyword
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

  • AACR Journals
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

User menu

  • Register
  • Log in
  • My Cart

Search

  • Advanced search
Molecular Cancer Therapeutics
Molecular Cancer Therapeutics
  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
    • Reviewing
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Focus on Radiation Oncology
      • Novel Combinations
      • Reviews
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citation
    • Author/Keyword
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

Small Molecule Therapeutics

LCL161, a SMAC-mimetic, Preferentially Radiosensitizes Human Papillomavirus–negative Head and Neck Squamous Cell Carcinoma

Linlin Yang, Bhavna Kumar, Changxian Shen, Songzhu Zhao, Dukagjin Blakaj, Tianyun Li, Mitchell Romito, Theodoros N. Teknos and Terence M. Williams
Linlin Yang
The Ohio State University Medical Center, Arthur G. James Comprehensive Cancer Center and Richard J. Solove Research Institute, Columbus, Ohio.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Bhavna Kumar
The Ohio State University Medical Center, Arthur G. James Comprehensive Cancer Center and Richard J. Solove Research Institute, Columbus, Ohio.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Changxian Shen
The Ohio State University Medical Center, Arthur G. James Comprehensive Cancer Center and Richard J. Solove Research Institute, Columbus, Ohio.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Songzhu Zhao
The Ohio State University Medical Center, Arthur G. James Comprehensive Cancer Center and Richard J. Solove Research Institute, Columbus, Ohio.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Dukagjin Blakaj
The Ohio State University Medical Center, Arthur G. James Comprehensive Cancer Center and Richard J. Solove Research Institute, Columbus, Ohio.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Tianyun Li
The Ohio State University Medical Center, Arthur G. James Comprehensive Cancer Center and Richard J. Solove Research Institute, Columbus, Ohio.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Mitchell Romito
The Ohio State University Medical Center, Arthur G. James Comprehensive Cancer Center and Richard J. Solove Research Institute, Columbus, Ohio.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Theodoros N. Teknos
The Ohio State University Medical Center, Arthur G. James Comprehensive Cancer Center and Richard J. Solove Research Institute, Columbus, Ohio.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Terence M. Williams
The Ohio State University Medical Center, Arthur G. James Comprehensive Cancer Center and Richard J. Solove Research Institute, Columbus, Ohio.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: terence.williams@osumc.edu
DOI: 10.1158/1535-7163.MCT-18-1157 Published June 2019
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Targeting inhibitor of apoptosis proteins (IAP) with second mitochondria-derived activator of caspase (SMAC) mimetics may promote cancer cell death. We tested whether cIAP1 predicts poor prognosis in head and neck squamous cell carcinoma (HNSCC) and whether a novel Smac-mimetic, LCL161, could radiosensitize human papillomavirus–positive (HPV+) and -negative (HPV−) HNSCC. The association of BIRC2 (encoding cIAP1) mRNA level with HPV status in HNSCC was analyzed using The Cancer Genome Atlas (TCGA) database. cIAP1 was assessed by IHC on an HNSCC tissue microarray (TMA, n = 84) followed by correlation analysis with HPV status and patient outcomes. Human cell culture and animal models of HNSCC were used to analyze the outcome and molecular characteristics following radiotherapy in combination with LCL161. cIAP1 expression is increased in HPV− compared with HPV+HNSCC tumors in the TCGA database. In our TMA, cIAP1 was overexpressed in HNSCC compared with normal tissues (P = 0.0003) and associated with a poor overall survival (P = 0.0402). cIAP1 levels were higher in HPV− than that in HPV+HNSCC tumors (P = 0.004) and patients with cIAP1+/HPV− HNSCC had the worst survival. LCL161 effectively radiosensitized HPV− HNSCC cells, which was accompanied with enhanced apoptosis, but not HPV+ HNSCC cells. Importantly, LCL161 in combination with radiotherapy led to dramatic tumor regression of HPV− HNSCC tumor xenografts, accompanied by cIAP1 degradation and apoptosis activation. These results reveal that cIAP1 is a prognostic and a potential therapeutic biomarker for HNSCC, and targeting cIAP1 with LCL161 preferentially radiosensitizes HPV− HNSCC, providing justification for clinical testing of LCL161 in combination with radiation for patients with HPV− HNSCC.

This article is featured in Highlights of This Issue, p. 1023

Introduction

Head and neck squamous cell carcinoma (HNSCC) is the sixth most common cancer and accounts for 600,000 new cancer cases per year in the world (1, 2). Common treatments include surgery, radiotherapy, chemotherapy, targeted therapy, and more recently immunotherapy. Although improvements in these treatments have greatly benefited patients with HNSCC, the 5-year survival rate of patients with HNSCC remains low at about 50% (1). HNSCC is clinically classified as human papillomavirus–positive (HPV+) and -negative (HPV−) subgroups, with different genetic alteration patterns and clinical outcomes associated with them (3). HPV− HNSCC represents a distinct clinicopathologic entity and typically has a worse prognosis than patients with HPV+ HNSCC (4–7). In comparison with HPV+ HNSCC, patients with HPV− HNSCC are more resistant to radiotherapy. It has been proposed that p53 dysfunction resulting from TP53 and CDKN2A mutations and uncontrolled activity of EGFR/PI3K/AKT signalling may contribute to the radioresistance of HPV− HNSCC (8–10). Indeed, targeting EGFR with cetuximab significantly improved the outcome of HNSCC when compared with radiotherapy alone in a large randomized phase III trial; however, HPV status was not determined for patients on this trial (11). However, the most recent randomized phase III clinical trial has shown that cetuximab does not improve outcomes when used in combination with cisplatin and radiotherapy (12, 13). Because radioresistance is a significant challenge for patients with HNSCC, particularly HPV− patients (14), it is of high importance to elucidate the precise mechanism of radioresistance, which will engender novel strategies to overcome radioresistance of HPV− patients.

Apoptosis is a tightly regulated multi-step cell suicide program that is critical for the development and homeostasis of multicellular organisms (15). Evasion of apoptosis is a characteristic feature of human cancer cells and represents an important basis of resistance to current treatment approaches, including radiation (16, 17). It has been widely accepted that reversal of cancer cell apoptosis evasion is a pivotal strategy for cancer therapy (18, 19). Inhibitor of apoptosis proteins (IAP) originally discovered in Baculoviral genomes by Miller and colleagues in 1993, comprise a family of antiapoptotic proteins that promote prosurvival signalling pathways and prevent activation of apoptosis by interfering with the activation of caspases (20, 21). Overexpression of IAPs frequently occurs in various human cancers, including esophageal carcinoma (22), cervical cancer (23), and pancreatic cancer (24), and correlates with tumor progression, treatment failure, and poor prognosis (25–27), making IAPs important targets for therapeutic intervention. Endogenously, the role of IAPs in preventing apoptosis is inhibited by the second mitochondria-derived activator of caspase (SMAC), a mitochondria protein that is released to the cytoplasm upon induction of apoptosis (28, 29). SMAC (also called DIABLO) physically interacts with the conserved Baculovirus IAP repeat (BIR) domains thereby preventing the apoptosis inhibition functions of IAPs. Accordingly, several SMAC mimetics have been designed to prevent IAPs inhibitory action on caspases to promote apoptosis. The SMAC-mimetic, LCL161, is a monovalent SMAC-mimetic, which binds IAPs with high affinity and initiates the destruction of cIAP1 and cIAP2 (encoded by BIRC2 and BIRC3, respectively) and prevention of caspase inhibition by XIAP (30, 31). LCL161 has shown safety and efficacy in a phase I study of advanced solid tumors (32). Furthermore, a preclinical study has shown good antitumor activity of LCL161 in human hepatocellular carcinoma (33). LCL161 has also been shown to sensitize esophageal carcinoma to radiotherapy (34). In addition, SMAC mimetics SM-164 and birinapant were reported to sensitize HNSCC to radiotherapy (35, 36). Dual inhibition of p53 and Rb by E6 and E7, respectively, in HPV+ HNSCC cells may lead to loss of G1 checkpoint, but the presence of wild-type p53 has been proposed to contribute to the sensitivity of HPV+ HNSCC cells to radiotherapy (37). In contrast, TP53 and CDKN2A mutations that are commonly found in HPV− HNSCC cells may not only result in loss of G1 phase checkpoint, but also apoptosis evasion in response to DNA damage. We hypothesize that HPV− HNSCC cells might rely on attenuated apoptosis for survival and be more susceptible to radiotherapy following reactivation of apoptosis by a potent SMAC-mimetic, LCL161.

In this study, we compared the expression of cIAP1 between HPV− HNSCC and HPV+ HNSCC in The Cancer Genome Atlas (TCGA) database, cell lines, and tissue microarray (TMA), and evaluated the radiosensitizing potential of LCL161 in in vitro and in vivo models of HPV− and HPV+ HNSCC. We revealed that cIAP1 is a prognostic and therapeutic biomarker for HPV− HNSCC and targeting cIAP1 with LCL161 preferentially radiosensitizes HPV− HNSCC. Our findings may provide a novel strategy for the management of patients with HPV− HNSCC.

Materials and Methods

Cell culture, chemicals, antibodies, and ELISA

HNSCC cell line UD-SCC-2 was a gift from Henning Bier (University of Dusseldorf, Dusseldorf, Germany, 2009); UM-SCC-47, UM-SCC-1, UM-SCC-11B, and UM-SCC-74A were gifts from Thomas Carey (University of Michigan, Ann Arbor, MI, 2009); 93VU147T was gift from Jim Rocco (Ohio State University, Columbus, OH, 2015); UPC1:SCC090 was gift from Susanne M. Gollin (University of Pittsburgh, Pittsburg, PA, 2009); Normal Oral Epithelial cells (NOE) was gift from Quintin Pan (University Hospitals Cleveland Medical Center, Cleveland, OH, 2009); and Cal27 and FaDu were purchased from ATCC. Except for UPCI:SCC090 and NOE cells, cell lines were maintained in DMEM (Life Technologies Inc.) containing 10% (v/v) heat-inactivated FBS (Life Technologies Inc.) and 1% (v/v) non-essential amino acid (NEAA; Life Technologies Inc.). UPCI:SCC090 cells were cultured in MEM (Life Technologies Inc.) with 10% FBS (v/v) and 1% NEAA (v/v). NOE cells were maintained in a serum-free keratinocyte growth medium (Life Technologies Inc.). All cell lines were routinely cultured at humidified atmosphere in an incubator with 5% CO2 at 37°C. Typically cells were kept in culture for minimum two passages prior and maximum 20 passages when the experiments were performed. The identity of all cell lines was confirmed by STR genotyping (Identifier Kit, Applied Biosystems). For the detection of Mycoplasma in cell culture, the Universal Mycoplasma Detection Kit (ATCC) was used. LCL161 was synthesized and provided by Novartis Pharma AG. Pan-caspase inhibitor, Z-VAD-FMK, was purchased from Selleck Chemicals LLC. Anti- c-IAP1, c-IAP2, XIAP, Survivin, Smac, Bcl-xL, Bcl-2, caspase 3, cleaved caspase 3, caspase 7, cleaved caspase 7, caspase 8, cleaved caspase 8, caspase 9, cleaved caspase 9, PARP, FADD, c-FLIP, p65/RELA, phospho-p65, TNFα, gamma-H2A.X, and GAPDH antibodies were purchased from Cell Signalling Technology. TNFα ELISA kit (#ELH-TNFa-1) was purchased from RayBiotech Life and experiments performed as per the manufacturer's instructions. Recombinant human TNFα (catalog no. 210-TA-005) was purchased from R&D Biosystems.

Immunoblotting

Immunoblotting was performed as described previously (38). Briefly, total proteins were extracted from cells using RIPA buffer (Thermo Fisher Scientific) supplemented with 1× phosphatase inhibitors (PhosSTOP, Roche) and protease inhibitors (cOmplete, Roche) followed by protein quantification with Dc Protein Assay Kit (Bio-Rad). Equal amounts of protein were resolved by SDS-PAGE and transferred onto nitrocellulose membranes, followed by incubation with primary antibodies (dilution of 1:500–1,000) at 4°C overnight. After washing with TBS-Tween, membranes were incubated with horseradish peroxidase–conjugated secondary antibodies (dilution of 1:2,500–5,000) at room temperature for 1 hour, followed by signal detection with enhanced chemiluminescence reagent (Immobilon, Millipore).

Study population, TMA, IHC, and scoring

A TMA was constructed from human oropharyngeal squamous cell carcinoma tissue specimens, which were obtained from the surgical resection of the patients at The Ohio State University James Cancer Hospital and Solove Research Institute from 2002 to 2009 (39). All patients underwent surgical resection as a first-line of therapy with a curative intent. This was followed by no additional treatment or adjuvant chemotherapy and/or radiotherapy as needed. The Ohio State University Institutional Review Board approved a retrospective analysis study of these specimens and a waiver of HIPAA authorization was obtained. Patient characteristics including HPV status are shown in Supplementary Table S1. TMA slides were stained to assess the tumor expression of cIAP1 using IHC as described previously (40). Stained slides were interpreted by a pathologist who was blinded to treatment outcome at the time of review. Tumor cells were scored for staining proportion (0%–100%) and intensity (0, none; 1, low; 2, moderate; 3, high). An H-score was generated by multiplying the stain proportion scores with stain intensity to obtain values between 0–300.

WST-1 assay and IC50 determination

WST-1 cytotoxicity assay was performed according to the manufacturer's instructions (Roche). Briefly, cells were seeded in 96-well plates in 5–6 replicates at a density of 2,000 cells per well in 100 μL medium. The next day, the cells were treated with LCL161 at various concentrations. After 72 hours, WST-1 cell proliferation reagent was added and incubated at 37°C for 4 hours, and absorbance was measured at 490 nm. IC50 was determined using the nonlinear four parameter regression function in GraphPad Prism.

Radiation clonogenic assay

Exponentially growing cells were harvested to generate single-cell suspensions and seeded into 60- or 100-mm tissue culture dishes in triplicate at predetermined densities. Twenty-four hours later, cells were irradiated using a RS-2000 Biological Irradiator (Rad Source Technologies) for different doses, followed by incubation at 37°C for 10–14 days. Colonies were then fixed with methanol/acetic and stained with 1% crystal violet. The number of colony containing at least 50 cells was counted using a dissecting microscope (Leica Microsystems, Inc.). The surviving fractions and dose-enhancement ratios (DER) at 2 Gy were calculated. For LCL161 or Z-VAD-FMK treatment, cells were pretreated with either drug or vehicle (DMSO) for 2 hours prior to radiation, and incubated for additional 24 hours post radiation. The cells were then washed and cultured in fresh media for the remainder of the experiments. Experiments were repeated at multiples for independent times.

Detection of cell apoptosis

Cells were seeded into 6-well plates at a density of 3 × 105 cells per well in 2 mL medium for 16 hours. The cells were treated with the indicated drugs and ionizing radiation, and cultured for 48 hours. Cell apoptosis was assessed by Annexin V-FITC (Invitrogen) and propidium iodide (PI; Sigma-Aldrich) staining coupled with flow cytometry analysis. For cell-cycle distribution analysis, cells were fixed in 70% ethanol at −20°C and stained with DNA staining solution containing PI and RNaseA (Sigma-Aldrich) overnight. All data were acquired on LSRII Cytometry (BD Biosciences) and each sample was assessed using a collection of 10,000 events, followed by analysis using FlowJo Software (FlowJo).

Animal experiments

All animal studies were conducted under our protocol approved by The Ohio State University Institutional Animal Care and Usage Committee. Six- to 8-week-old athymic nude mice (Taconic Farms Inc.) were housed in a pathogen-free facility. A total of 5 × 106 Cal27 or FaDu cells were injected subcutaneously into the left flank of each mouse. Mice were randomized to treatment regimens when the tumor size reached 150 mm3 (∼10 days after inoculation). As reported previously (41), LCL161 was formulated for oral gavage by dissolving in 0.1 N HCL and diluting with sodium acetate buffer (100 mmol/L, pH 4.63) to produce a solution with pH 4.3–4.6. LCL161 (50 mg/kg, oral gavage) and radiation (6 Gy) were given once a day for 5 continuous days over 1 week. Radiation was delivered directly to the tumor with the rest of the animal body shielded. For combination treatment, LCL161 was given to the mouse 2–3 hours before radiation exposure. The tumor growth was monitored three times a week. The average tumor volumes of at least 10 tumors in each group were calculated according to the formula [(Length × Width2)/2].

Statistical analysis

Data are presented as the mean ± SEM for clonogenic survival and tumor growth experiments. Statistical comparisons were made between the control and experimental groups using the unpaired two-tailed Student t test. Cox proportional hazards models were used to assess univariate associations of cIAP1 expression as predictors for death. Unadjusted HR and 95% confidence intervals (CI) were reported. cIAP1 expression was grouped into two categories: positive (cIAP1 H-score ≥ 100) and negative (cIAP1 H-score < 100). Kaplan–Meier survival curves were plotted by cIAP1 status or by cIAP1 and HPV dual markers. Log-rank test was used to compare survival curves with the Šídák multiple-comparison adjustment. A multivariate Cox proportional hazards model including tumor stage, node stage, gender, age, and cIAP1 expression was built to estimate adjusted HRs. cIAP1 expression was compared between HPV+ and HPV− using Wilcoxon two-sample test. A P < 0.05 was considered as statistically significant. All analyses were conducted in SAS, version 9.3 (SAS Institute).

Results

cIAP1 expression is upregulated in HPV− HNSCC

Overexpression of IAPs has been detected in numerous cancers and found to be associated with the resistance of cancer cells to radiotherapy (42). To explore the association between the expression of IAPs and HPV status in HNSCC, we compared the mRNA expression of BIRC2 (cIAP1), BIRC3 (cIAP2), XIAP, and SURVIVIN, well-characterized members of the IAP family, and other apoptosis related genes, between the 36 HPV+ (13%) and 243 HPV− (87%) HNSCC tumors with mRNA expression available in TCGA database. These tumor samples are derived from primary, previously untreated tumors. Between HPV+ and HPV− HNSCC tumors in this cohort, BIRC2 and SURVIVIN mRNA levels were significantly upregulated in HPV− HNSCC, whereas there were no apparent differences in XIAP mRNA level; BIRC3 mRNA levels were elevated in HPV+ tumors but not statistically significant (Fig. 1A). We also noted significantly higher expression of FADD and RELA/p65 in HPV− HNSCC. We next determined the protein expression of these IAPs by immunoblotting in the well-characterized HPV+ HNSCC cell lines UD-SCC-2, UM-SCC-47, and UPCI:SCC090, as well as the HPV− HNSCC cell lines Cal27, UM-SCC-1, and UM-SCC-74A. Consistent with the significant upregulation of BIRC2 and SURVIVIN mRNA in HPV− HNSCC from the TCGA database, we found cIAP1 and Survivin protein levels were also elevated in HPV− HNSCC cell lines when compared with that in HPV+ HNSCC cell lines (Fig. 1B). We also noted higher levels of XIAP, suggesting that multiple IAP family members are upregulated in HPV− HNSCC. Finally, we noted several other proteins were also elevated in HPV− HNSCC cells, such as RELA/p65, TNFα, FADD, and BCL-xL, which paralleled the trends or significant differences observed with mRNA expression in TCGA data.

Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1.

Differential expression of IAPs between HPV− and HPV+ HNSCC. A, The mRNA expression of BIRC2 (cIAP1), BIRC3 (cIAP2), XIAP, and SURVIVIN between HPV− and HPV+ HNSCC of HNSCC cohort in TCGA database was compared using t test. The mRNA expression data were derived from RNA-seq and converted to z scores. *, P < 0.05; **, P < 0.001. B, Exponentially growing HPV− and HPV+ HNSCC cells of the indicated cell lines were subjected to immunoblotting analyses of cIAP1, cIAP2, XIAP, Survivin, SMAC/DIABLO, and other NF-kB and apoptosis pathway components. GAPDH shown as loading control.

cIAP1 overexpression is associated with poor overall survival in HNSCC and is associated with HPV-negative status

To confirm the upregulation of cIAP1 in HPV− HNSCC, we evaluated the protein expression pattern of cIAP1 by IHC in an institutional TMA, which was constructed from 84 surgically treated HNSCC samples and 72 paired normal tissues (39). The 84 HNSCC cases were composed of 46 HPV+ and 38 HPV− HNSCC (Supplementary Table S1). Representative IHC images of low and high cIAP1 protein staining are shown in Fig. 2A, and validation of the antibody in Supplementary Fig. S1. We graded the cores for intensity and percent of cells staining positive and derived an “H-score” for each tumor sample. Wilcoxon signed rank sum test was used to assess the difference between cIAP1 tumor H-score versus normal H-score. We found cIAP1 protein levels in tumor cells were significantly higher than that in normal cells (P = 0.0003; Fig. 2B). When cIAP1 H-score increased by 10 units, the HR increased by 3.7% (HR, 1.037; 95% CI, 1.001–1.074; P = 0.0422). Kaplan–Meier survival plot showed that patients having cIAP1-positive expression had worse overall survival than patients having negative cIAP1 expression (log-rank P = 0.0322; Fig. 2C). Similar to TCGA data analysis, HPV− tumors had significantly higher expression of cIAP1 protein as compared with HPV+ tumors (Wilcoxon two-sample test, P = 0.004; Fig. 2D). In multiple comparisons, cIAP1-positive/HPV-negative (cIAP1+/HPV−) group had the worst survival rate compared with cIAP1−/HPV+ (P = 0.0004) or cIAP1+/HPV+ (log-rank P = 0.0046; Fig. 2E). Finally, we performed multivariate analysis with common clinical covariates, including gender, T stage, N stage, and age. After adjusting for other covariates, cIAP1 expression remained a significant predictor of overall survival. Notably, when the cIAP1 H-score increased by 10 units, the HR for death increased by 3.9% (HR, 1.039; 95% CI, 1.002–1.078; P = 0.0402; Table 1).

Figure 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 2.

cIAP1 protein expression is associated with poor overall survival and elevated in HPV− HNSCC. A, Representative IHC images of HNSCC tumor cores negative (left) and positive (right) for cIAP1 expression in the TMA. B, cIAP1 protein expression in tumor versus paired normal tissues. C, Kaplan–Meier survival plot by cIAP1 status. D, cIAP1 expression in HPV− versus HPV+ patients. E, Kaplan–Meier survival plot by cIAP1 status and HPV status (*, P < 0.05; **, P < 0.001).

View this table:
  • View inline
  • View popup
Table 1.

Overall survival multivariate analysis

HPV− HNSCC cells are preferentially radiosensitized by LCL161

The higher expression of cIAP1 in HPV− HNSCCs and the poor survival of cIAP1+/HPV− HNSCC subgroup implies that HPV− HNSCC cells may depend on cIAP1 for survival. LCL161 is a small molecule Smac-mimetic that binds to cIAP1 and cIAP2 with high affinity, initiating their destruction, and also binds to XIAP and inhibits XIAP activity. Previous studies have shown LCL161 potently induces the degradation of cIAP1 (34, 41). This led us to hypothesize that LCL161 may be, in general, more effective on HPV− than HPV+ cells with regard to radiosensitization. To this end, we first compared the radiosensitivity of HPV− cell lines (Cal27 and UM-SCC-1) with that of HPV+ cell lines (UD-SCC-2 and UM-SCC-47) using standard radiation clonogenic assay. Consistent with the previous observations that HPV− HNSCC cells generally display higher resistance to radiation than HPV+ cells (37), we found that HPV− cell lines were relatively resistant to radiation whereas HPV+ cell lines were relatively sensitive (Supplementary Fig. S2A).

We next investigated the cytotoxic effect of LCL161 as a single agent on HNSCC cell lines through WST1 assay. We observed that all HNSCC cell lines were highly resistant to LCL161 as a single agent with the IC50 values ranging from 32 to 95 μmol/L (Supplementary Fig. S2B), suggesting LCL161 as monotherapy may not be effective in HNSCC. However, HPV− cells showed significantly lower IC50 than HPV+ cells, suggesting increased sensitivity to cIAP1 inhibition (P = 0.006; Supplementary Fig. S2C). We further determined the doses and timing of LCL161 to reduce IAPs in HNSCC cells. LCL161 treatment for 2 hours led to a dose-dependent decrease of both cIAP1 and cIAP2 expression, but did not decrease XIAP expression in HPV− Cal27 and UM-SCC-1 cells, consistent with its known effects on XIAP enzymatic activity (not expression; Fig. 3A). LCL161 at 100 nmol/L induced potent downregulation of cIAP1 and cIAP2 in all tested HNSCC cell lines and was chosen for subsequent experiments. We next treated HPV− UM-SCC-1 and Cal27 cells with 100 nmol/L LCL161 for different time periods and found LCL161 treatment resulted in rapid decrease of cIAP1 and cIAP2 expression as early as 30 minutes and sustained after 24 hours (Fig. 3B). We determined whether LCL161 treatment for 2 hours could alter the expression and activity of important tumor cell proliferation and survival pathways. We did not detect any significant acute changes in the phosphorylation levels of STAT3, AKT, and ERK in HNSCC cells (Supplementary Fig. S3), suggesting that LCL161-mediated inhibition of IAPs does not alter the acute activation of the JAK/STAT, PI3K/AKT, and MEK/MAPK signaling pathways in HNSCC cells.

Figure 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 3.

LCL161 sensitizes HPV− HNSCC cells to radiation. A, The indicated HNSCC cell lines were treated with increasing concentrations of LCL161 for 2 hours. Total proteins were extracted for immunoblotting of cIAP1, cIAP2, XIAP, and Survivin with GAPDH as loading control. B, UM-SSC-1 and Cal27 cells were treated with 100 nmol/L LCL161 for different time points and subjected to immunoblotting for the indicated proteins. C, Schedule of combined treatment in clonogenic assay. Cells were seeded in 6-well plates for 24 hours, followed by exposure to irradiation (IR) of increasing dose. LCL161 (100 nmol/L) was added to the culture media 2 hours prior to irradiation, and removed 24 hours after radiation. The cells were cultured for 10–14 days to determine colony formation. Representative radiation clonogenic assay results of HPV− Cal27 (D) and HPV+ UD-SCC-2 (E) are shown. F, DERs at 2 Gy were compared between various types of cancer using radiation clonogenic assay. Multiple independent experiments were performed (**, P < 0.001).

To explore the potential of LCL161 as a radiosensitizer, cells were treated with different doses of radiation in the presence or absence of 100 nmol/L LCL161, followed by clonogenic assay (treatment schema shown in Fig. 3C). With 2 hours of treatment prior to radiation, LCL161 significantly induced radiosensitization of HPV− cells (representative Cal27 cells shown in Fig. 3D) with an average DER of 1.31 (Fig. 3F), but not HPV+ cells (representative UD-SCC-2 cells shown in Fig. 3E) with an average DER of 1.01 (Fig. 3F). To determine whether pretreatment of cells with LCLC161 is critical for radiosensitization, we also compared treatment with an alternate treatment schedule of radiation first, followed by LCL161 treatment 2 hours later. The results showed that LCL161 also radiosensitized Cal27 cells in both treatment schedules (Supplementary Fig. S4). For the remaining in vitro experiments, LCL161 was administered prior to radiation. To determine whether the radiosensitizing effect of LCL161 is tumor type–specific, we treated many additional types of cancer cells with radiation in combination with 100 nmol/L LCL161 followed by clonogenic assays, and found all other cancer cell lines except for HPV− HNSCC cells, were essentially resistant to LCL161 radiosensitization with a mean DER of 1.1 or less (Fig. 3F).

LCL161 enhances radiation-induced apoptosis of HPV− HNSCC cells

Impaired apoptosis is one of the mechanisms for radiation resistance in cancer cells. To assess whether LCL161-mediated inhibition of IAPs enhances radiation-induced apoptosis, 2 HPV− and 2 HPV+ HNSCC cell lines, as well as NOE were treated with 4 Gy radiation alone or in combination with 100 nmol/L LCL161 for 24 hours (including pretreatment for 2 hours), and subjected to analysis of cell death by PI flow cytometry and apoptosis by Annexin V staining. In all cell lines, LCL161 treatment alone did not lead to an increase of cells in the sub-G1 fraction, indicating that LCL161 monotherapy was not cytotoxic to both tumor and normal cells (Fig. 4A). On the other hand, radiation monotherapy did induce a mild increase in sub-G1 fraction in the four tumor cell lines. However, when compared with radiation alone, the combined treatment of radiation with LCL161 significantly increased the percentage of sub-G1 cells from 2.2% and 2.24%, to 7.98% and 6.5% in the 2 HPV− cell lines Cal27 and UM-SCC-1, respectively. In sharp contrast, LCL161 did not significantly increase the radiation-induced sub-G1 fraction in either HPV+ cell lines or NOE cells (Fig. 4A). Furthermore, Annexin V/PI staining coupled with flow cytometry analysis demonstrated that the percentage of Annexin V–positive cells was significantly enhanced by the combination treatment (7.54%) compared with radiation alone (4.39%) in HPV− Cal27 cells; however, there was no difference of Annexin V–positive cells in HPV+ UM-SCC-47 and normal NOE cells treated with radiation alone or radiation plus LCL161 (Fig. 4B).

Figure 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 4.

LCL161 enhances radiation-induced apoptosis in HPV− but not HPV+ HNSCC and normal oral epithelial cell lines. Cells were treated with 4 Gy radiation, 100 nmol/L LCL161 alone, or in combination for 24 hours, followed by flow cytometry analysis of sub-G1 populations (A) and Annexin V–positive cells (B). C, Cells were treated as in A and subjected to immunoblotting for the indicated proteins. D, Cal27 cells were treated with 4 Gy radiation and 100 nmol/L LCL161 in the presence or absence of 10 μmol/L pan-caspase inhibitor-z-VAD-fmk for 24 hours. Total proteins were extracted for immunoblotting of the indicated proteins. E, Cal27 cells were treated with 4 Gy radiation, 100 nmol/L LCL161, 4 Gy radiation + 100 nmol/L CL161 in the presence or absence of 10 μmol/L z-VAD-fmk for 24 hours. Cell apoptosis was assessed by Annexin V staining coupled with flow cytometry analysis. F, Effect of z-VAD-fmk on LCL161 radiosensitization was assessed by clonogenic assay. Cal27 cells were treated with LCL161 alone (2 hours prior to radiation) or in combination with z-VAD-fmk (10 mmol/L). For all assays, multiple independent experiments were performed (*, P < 0.05; **, P < 0.001).

Our above results imply that LCL161 may preferentially enhance radiation-induced apoptosis and cell death in HPV− HNSCC to a greater extent than HPV+ HNSCC and normal NOE cells. To further support this conclusion, caspase activation was assessed by immunoblotting. Treatment with 100 nmol/L LCL161 for 24 hours led to marked degradation of cIAP1 in all the four HNSCC cell lines and NOE cells, whereas radiation alone had minimal effect on cIAP1 protein expression (Fig. 4C). Consistent with the cell death and apoptosis assays (Fig. 4A and B), LCL161 monotherapy caused marginal cleavage of caspase-3, 7, 8, 9, and PARP in all five cell lines, whereas radiation alone had moderate caspase cleavage in HPV+ and HPV− cells but not NOE cells. Compared with radiation alone, the combined treatment of LCL161 and radiation resulted in increased cleavage of caspase-3, 7, 8, 9, and PARP only in HPV− cells, but not HPV+ and NOE cells, suggesting potentiation of apoptotic pathway activation with the combination (Fig. 4C). Also noted was persistence of the DNA damage marker phosphorylated-H2A.X (Ser139) in the combination treated HPV− cells, suggestive of heightened DNA damage at 24 hours after the combination, which was not observed in the HPV+ or NOE cells.

To confirm that caspase activation contributes to LCL161-mediated radiosensitization of HPV− HNSCC cells in vitro, Cal27 cells were treated for 24 hours with 100 nmol/L LCL161 + 4 Gy irradiation in the presence or absence of 10 μmol/L z-VAD-fmk, a pan-caspase inhibitor, followed by detection of cleavage of caspase-3, 8, and PARP by immunoblotting, and apoptosis by Annexin V staining. z-VAD-fmk suppressed the cleavage of caspase-3, 8, and PARP (Fig. 4D), and abolished the increase of Annexin V–positive cells by LCL161 in combination with radiation (Fig. 4E). Consistent with these findings, radiation clonogenic assay showed that LCL161-mediated radiosensitization of Cal27 cells was blocked by z-VAD-fmk (Fig. 4F). Collectively, these data indicate that LCL161 enhances radiosensitivity of HPV− HNSCC cells through caspase-mediated apoptosis.

Because of the important role of TNFα in the tumor microenvironment, as well as in the recent study with birinapant and radiation (36), we assessed TNFα levels in our cell lines and indeed found upregulation of TNFα in our HPV− cells compared with HPV+ cells (Fig. 1B). Next, we assessed whether the various treatments could result in further activation of TNFα expression in HPV− cell lines and xenograft tumors 24–48 hours after treatment, but did not find significant upregulation of TNFα expression after treatment with LCL161, radiation, or the combination (Supplementary Fig. S5A and S5B). Interestingly, we did detect heightened levels of TNFα in the medium of tumor cells by ELISA after 48 hours of treatment with LCL161 + radiation in Cal27 cells, but not UD-SCC-2 cells (Supplementary Fig. S5C). Because TNFα (and other death receptor ligands like TRIAL or Fas ligand) could contribute to heightened apoptotic-mediated tumor cell death during this process, we investigated whether supplementing TNFα could increase rates of apoptosis in HPV− tumor cells. Indeed, treatment with TNFα led to significantly increased rates of radiation-induced apoptosis in Cal27 cells, but only slightly increased rates of apoptosis further when LCL161 was added to this combination (Supplementary Fig. S5D).

LCL161 radiosensitizes HPV− HNSCC cells in mouse tumor xenografts

To translate our findings to preclinical investigation, we evaluated LCL161 radiosensitization of HPV− HNSCC in nude mice xenografts. LCL161 was delivered by oral gavage with a dose of 50 mg/kg as described previously (43). When tumors derived from HNSCC HPV− Cal27 and FaDu cell lines reached approximately 100–150 mm3, the mice were randomized to treatment with vehicle, LCL161 alone, 6 Gy radiation alone, or the combination of LCL161 + 6 Gy, all for 5 consecutive days (Fig. 5A). Consistent with in vitro data, LCL161 monotherapy did not alter tumor growth, whereas radiation treatment resulted in partial tumor growth delay. However, LCL161 in combination with radiation treatment led to remarkable and sustained regression of both Cal27 (Fig. 5B) and FaDu (Fig. 5C) xenografts, with the majority of Cal27 tumors showing no evidence of tumor recurrence (survival curves shown in Supplementary Fig. S6). The treatment was tolerated well, although mice in the combination treatment cohort lost weight during the first week (no more than 10% or less of starting body weight), but fully recovered within 1–2 weeks after the last treatment (Supplementary Fig. S7). Immunoblotting of tumor lysates several hours after day 3 of treatment in each of the groups demonstrated degradation of cIAP1 by LCL161 alone and in combination with radiation, confirming pharmacodynamic inhibition by LCL161. Consistent with the cell culture results, radiation alone did not induce degradation of cIAP1 and cleavage of caspase-3, 7, 8, 9, and PARP. In sharp contrast, combination treatment of LCL161 with radiation resulted in potent cleavage of caspase-3, 7, 8, 9, and PARP, implicating activation of apoptosis as a major mechanism for tumor growth regression and sustained response (Fig. 5D).

Figure 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 5.

LCL161 radiosensitization of HPV− HNSCC in in vivo xenograft tumor models. A, Schematic schedule of combined in vivo treatment. A total of 5 × 106 Cal27 or FaDu cells were inoculated subcutaneously in the left flank of nude mice. Indicated treatments were started on 14 days after inoculation when tumor size reached 100–150 mm3, followed by tumor growth measurement (mean ± SEM, n = 10). The change of the average tumor volume of each group was shown for Cal27 (B) and FaDu (C) xenografts models. D, Cal27 tumor lysates were made from two representative tumors per group 3 hours after the indicated treatment on day 3. LCL161 eliminated cIAP1 and enhanced radiation-induced apoptosis as assessed by immunoblotting of cleaved caspase-3, 7, 8, 9, and PARP (**, P < 0.001).

Discussion

In this study, we found that cIAP1 (BIRC2) expression is significantly higher in HNSCC tumor cells than normal tissue cells, and increased cIAP1 is significantly associated with a poor overall survival of patients with HNSCC. For the first time, to our knowledge, we demonstrated that HPV− HNSCC has significantly higher expression of cIAP1 as compared with HPV+ HNSCC and that patients with cIAP1+/HPV− HNSCC have reduced survival. Most importantly, we showed that targeting IAPs with LCL161 preferentially sensitizes HPV− HNSCC cells to radiotherapy both in in vitro and mouse xenograft models, which is accompanied by degradation of cIAP1 and enhanced apoptosis. The effects of the combination of LCL161 and radiation appear to demonstrate tumor selectivity, as minimal induction of apoptotic response was observed in the normal oral epithelial (nontransformed) cell line.

The HPV status of HNSCC has an important impact on the prognosis and response of patients to treatments. It has been well documented that patients with HPV+ HNSCC have a better prognosis than patients with HPV− HNSCC (4–7); moreover, patients with HPV− HNSCC are resistant to radiotherapy (37). However, the exact underlying molecular mechanisms are largely unknown. The TCGA study show that HPV− HNSCC has a characteristic coamplification of 11q22 (containing BIRC2 and YAP1) and 11q13 (containing CCND1, FADD, and CTTN), suggesting an important role for cIAP1 in HPV− HNSCC. Indeed, data from TCGA suggests approximately 30% of HNSCC harbors chromosome 11q13/22 amplifications and overexpression of FADD, with or without BIRC2/3 genes that encode cIAP1/2 (36). In addition, 86% of HPV− HNSCC harbor TP53 mutations (3). In this study, using a TMA, we found that HPV− HNSCC has significantly higher cIAP1 protein expression as compared with HPV+ HNSCC, and patients with cIAP1+/HPV− HNSCC have the lowest survival. cIAP1 is a potent inhibitor of apoptosis and p53 promotes apoptosis by stimulating the expression of proapoptosis genes including PUMA and BAX, while suppressing the expression of antiapoptosis genes such as BCL-2 and BCL-XL (44). It is therefore possible that simultaneous BIRC2 overexpression and TP53 mutation may contribute in part to the resistance of HPV− HNSCC to therapies which rely on activation of apoptosis.

Radiation is commonly used to treat patients with HNSCC, both in the curative and palliative settings. However, radioresistance, which contributes to local recurrence, is currently a challenge for the management of patients with HNSCC, particularly patients with HPV− HNSCC. The only proven radiosensitizer in HNSCC that was shown to improve overall survival in combination with radiation was cetuximab, targeting EGFR, in the RTOG 0522 randomized trial (45). Current ongoing trials are exploring various other radiosensitizers, including cytochlor/tetrahydrouridine (NCT00077051), olaparib (PARP inhibitor, NCT02229656), PS-341/bortezomib (proteasome inhibitor, NCT00011778), and nimorazole (hypoxia modifying drug, NCT01880359), but results have yet to be reported. Our findings support that an IAP antagonist may be an effective radiosensitizer for HNSCC. Our finding that overexpression and negative correlation of cIAP1 with the survival of patients with HPV− HNSCC led us to explore the potential of targeting cIAP1 with SMAC-mimetic in the sensitization of HPV− HNSCC to radiotherapy. LCL161 is an orally bioavailable, cell permeable small-molecule mimetic of SMAC developed by Novartis Pharmaceuticals. LCL161 promotes the degradation of certain IAP family members (i.e., cIAP1 and cIAP2) and disengages IAPs from caspases (e.g., XIAP), thereby sensitizing cells to apoptosis-inducing treatments (46, 47). Certainly, although we found that LCL161 promotes a rapid and potent degradation of cIAP1 in HNSCC cells at nanomolar concentrations, we cannot exclude the possibility that the effects we observed are not all dependent on cIAP1 (or cIAP2), because LCL161 potently inhibits XIAP activity by forcing disengagement of XIAP from caspases. Further evaluation into the role of XIAP in LCL161 response is warranted, but cIAP1 may be a more effective pharmacodynamic biomarker because degradation may be more readily measurable than caspase disengagement.

LCL161 has been shown to exert antitumor activity against multiple cancers including myeloma, glioblastoma, and sarcoma. In agreement with the finding from a preclinical study in pediatric tumors (41), the IC50 values of LCL161 on HNSCC cells are at range of 32–95 μmol/L, indicating LCL161 may not have therapeutic potential as a single treatment for HNSCC. With regards to a role for LCL161 in combination therapy, several SMAC mimetics have been reported to sensitize cancer cells to radiation, including SM164 in HNSCC (35) and breast cancer (48), Debio 1143 in HNSCC (49), ASTX660 in HNSCC (50), and JP-1201 in colorectal cancer (51). Likewise, a more recent study showed that SMAC-mimetic compound, birinapant, sensitizes HNSCC with FADD and BIRC2 amplification (36). It is interesting to note that tumors with FADD and BIRC2 amplification are generally HPV− tumors. In our study, we demonstrated that LCL161 enhances the radiosensitization of HNSCC cell lines with the greatest effect in HPV− cells. In preclinical modeling, we demonstrated that oral administration of LCL161 synergizes with radiation to induce remarkable and virtually complete regression of HPV− HNSCC tumour xenografts. Such dramatic findings of SMAC-mimetic in combination with radiation using xenografts were likewise noted in the study with birinapant and radiation (36). Consistent with previous reports showing that SMAC mimetics radiosensitize cancer cells through activation of caspases and enhanced induction of apoptosis (35, 48, 49), we revealed that the combination treatment of LCL161 and radiation activates caspases and induces apoptosis of HPV− HNSCC cells both in in vitro and in tumour xenografts. Our findings suggest that cIAP1+/HPV− might serve as a predictive biomarker for patients with HNSCC who would benefit most from the combination of SMAC-mimetic and radiotherapy.

Aside from radiotherapy, IAP antagonists have also been shown to sensitize various cancers including pancreatic, lung, colon, and acute leukemia to chemotherapy such as gemcitabine, doxorubicin, cisplatin, and irinotecan (19). Clinically, LCL161 has been tested in combination with paclitaxel chemotherapy for triple-negative breast cancer and found to result in higher pathologic response rates compared with chemotherapy alone for patients with a TNFα gene expression signature (52). Similar to radiotherapy, one of the main mechanisms by which many chemotherapy drugs act is to induce DNA damage. Thus, it will be interesting to investigate whether LCL161 sensitizes HPV− HNSCC to DNA-damaging based chemotherapy. In addition, the pharmacology of SMAC-mimetic sensitization of cancer cells to radiotherapy and chemotherapy is complex. It has been reported that IAP antagonists can initiate NF-kB activation, as well as autocrine and/or paracrine TNF signalling to induce cell apoptosis (53). Of note, IAPs are emerging as modulators of both innate and adaptive immunity during pathogen recognition and inflammasome activation (54). A weakness of our study is that the mouse modeling was performed in an immunocompromised mouse model, with lack of proper T-cell–mediated cell killing. Because checkpoint inhibitors such as anti-PD-1 antibodies (which activate T-cell infiltration and T-cell–mediated immunogenic tumor cell death) are increasingly being used in the clinic, it will be important in the future to explore the modulation of immune response by LCL161 in response to radiotherapy, chemotherapy, and immune checkpoint blockade based therapy. Certainly, there is increasing data that radiation may help to activate an immune response by enhanced tumor neoantigen release and cross-presentation of tumor antigens, leading to activation of tumor-specific T cells (55). As proof of principle, a recent study found that another IAP antagonist (ASTX660) enhances antitumor immunity when combined with PD-1 inhibition and radiation in an immunocompetent model of HNSCC (50).

In conclusion, higher cIAP1 expression is correlated with HPV− status and is associated with a poor prognosis of HNSCC. Targeting IAPs with LCL161 preferentially sensitizes HPV− HNSCC to radiotherapy over HPV+ HSNSCC and normal epithelial cells. Our study could have implications for the use of cIAP1 expression as a surrogate biomarker in future clinical trials combing LCL161 and radiation. Finally, our preclinical data demonstrate the susceptibility of HPV− HNSCC tumors to SMAC-mimetics in combination with radiotherapy, and supports initiation of clinical trials with LCL161 in combination with radiation, particularly in patients with HPV− HNSCC tumors.

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

Disclaimer

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

Authors' Contributions

Conception and design: L. Yang, B. Kumar, C. Shen, T.N. Teknos, T.M. Williams

Development of methodology: L. Yang, B. Kumar, C. Shen, T.M. Williams

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): L. Yang, B. Kumar, T. Li, M. Romito, T.M. Williams

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): S. Zhao, M. Romito, T.N. Teknos, T.M. Williams

Writing, review, and/or revision of the manuscript: L. Yang, C. Shen, S. Zhao, D. Blakaj, M. Romito, T.M. Williams

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): L. Yang, B. Kumar, T.N. Teknos, T.M. Williams

Study supervision: T.M. Williams

Acknowledgments

Research reported in this article was supported by The Ohio State University Comprehensive Cancer Center (OSU-CCC) and the NIH under grant no. P30 CA016058, as well as NIH grant no. R01 CA198128 (to T.M. Williams).

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.

Footnotes

  • Note: Supplementary data for this article are available at Molecular Cancer Therapeutics Online (http://mct.aacrjournals.org/).

  • Prior presentation: Presented in part at the 2016 American Association for Cancer Research (AACR) Annual Meeting, New Orleans, LA.

  • Mol Cancer Ther 2019;18:1025–35

  • Received October 8, 2018.
  • Revision received January 23, 2019.
  • Accepted April 4, 2019.
  • Published first April 23, 2019.
  • ©2019 American Association for Cancer Research.

References

  1. 1.↵
    1. Siegel R,
    2. Ma J,
    3. Zou Z,
    4. Jemal A
    . Cancer statistics, 2014. CA Cancer J Clin 2014;64:9–29.
    OpenUrlCrossRefPubMed
  2. 2.↵
    1. Leemans CR,
    2. Braakhuis BJ,
    3. Brakenhoff RH
    . The molecular biology of head and neck cancer. Nat Rev Cancer 2011;11:9–22.
    OpenUrlCrossRefPubMed
  3. 3.↵
    Cancer Genome Atlas Network. Comprehensive genomic characterization of head and neck squamous cell carcinomas. Nature 2015;517:576–82.
    OpenUrlCrossRefPubMed
  4. 4.↵
    1. Chung CH,
    2. Gillison ML
    . Human papillomavirus in head and neck cancer: its role in pathogenesis and clinical implications. Clin Cancer Res 2009;15:6758–62.
    OpenUrlAbstract/FREE Full Text
  5. 5.↵
    1. Marur S,
    2. D'Souza G,
    3. Westra WH,
    4. Forastiere AA
    . HPV-associated head and neck cancer: a virus-related cancer epidemic. Lancet Oncol 2010;11:781–9.
    OpenUrlCrossRefPubMed
  6. 6.↵
    1. Chaturvedi AK
    . Epidemiology and clinical aspects of HPV in head and neck cancers. Head Neck Pathol 2012;6:S16–24.
    OpenUrlPubMed
  7. 7.↵
    1. Ang KK,
    2. Harris J,
    3. Wheeler R,
    4. Weber R,
    5. Rosenthal DI,
    6. Nguyen-Tan PF,
    7. et al.
    Human papillomavirus and survival of patients with oropharyngeal cancer. N Engl J Med 2010;363:24–35.
    OpenUrlCrossRefPubMed
  8. 8.↵
    1. Skinner HD,
    2. Sandulache VC,
    3. Ow TJ,
    4. Meyn RE,
    5. Yordy JS,
    6. Beadle BM,
    7. et al.
    TP53 disruptive mutations lead to head and neck cancer treatment failure through inhibition of radiation-induced senescence. Clin Cancer Res 2012;18:290–300.
    OpenUrlAbstract/FREE Full Text
  9. 9.↵
    1. Jedlinski A,
    2. Ansell A,
    3. Johansson AC,
    4. Roberg K
    . EGFR status and EGFR ligand expression influence the treatment response of head and neck cancer cell lines. J Oral Pathol Med 2013;42:26–36.
    OpenUrlCrossRefPubMed
  10. 10.↵
    1. Bussink J,
    2. van der Kogel AJ,
    3. Kaanders JH
    . Activation of the PI3-K/AKT pathway and implications for radioresistance mechanisms in head and neck cancer. Lancet Oncol 2008;9:288–96.
    OpenUrlCrossRefPubMed
  11. 11.↵
    1. Bonner JA,
    2. Harari PM,
    3. Giralt J,
    4. Azarnia N,
    5. Shin DM,
    6. Cohen RB,
    7. et al.
    Radiotherapy plus cetuximab for squamous-cell carcinoma of the head and neck. N Engl J Med 2006;354:567–78.
    OpenUrlCrossRefPubMed
  12. 12.↵
    1. Ang KK,
    2. Zhang Q,
    3. Rosenthal DI,
    4. Nguyen-Tan PF,
    5. Sherman EJ,
    6. Weber RS,
    7. et al.
    Randomized phase III trial of concurrent accelerated radiation plus cisplatin with or without cetuximab for stage III to IV head and neck carcinoma: RTOG 0522. J Clin Oncol 2014;32:2940–50.
    OpenUrlAbstract/FREE Full Text
  13. 13.↵
    1. Yamamoto VN,
    2. Thylur DS,
    3. Bauschard M,
    4. Schmale I,
    5. Sinha UK
    . Overcoming radioresistance in head and neck squamous cell carcinoma. Oral Oncol 2016;63:44–51.
    OpenUrl
  14. 14.↵
    1. Perri F,
    2. Pacelli R,
    3. Della Vittoria Scarpati G,
    4. Cella L,
    5. Giuliano M,
    6. Caponigro F,
    7. et al.
    Radioresistance in head and neck squamous cell carcinoma: biological bases and therapeutic implications. Head Neck 2015;37:763–70.
    OpenUrlCrossRefPubMed
  15. 15.↵
    1. Cotter TG
    . Apoptosis and cancer: the genesis of a research field. Nat Rev Cancer 2009;9:501–7.
    OpenUrlCrossRefPubMed
  16. 16.↵
    1. Fulda S
    . Tumor resistance to apoptosis. Int J Cancer 2009;124:511–5.
    OpenUrlCrossRefPubMed
  17. 17.↵
    1. Hanahan D,
    2. Weinberg RA
    . Hallmarks of cancer: the next generation. Cell 2011;144:646–74.
    OpenUrlCrossRefPubMed
  18. 18.↵
    1. Ashkenazi A,
    2. Fairbrother WJ,
    3. Leverson JD,
    4. Souers AJ
    . From basic apoptosis discoveries to advanced selective BCL-2 family inhibitors. Nat Rev Drug Discov 2017;16:273–84.
    OpenUrl
  19. 19.↵
    1. Fulda S,
    2. Vucic D
    . Targeting IAP proteins for therapeutic intervention in cancer. Nat Rev Drug Discov 2012;11:109–24.
    OpenUrlCrossRefPubMed
  20. 20.↵
    1. Crook NE,
    2. Clem RJ,
    3. Miller LK
    . An apoptosis-inhibiting baculovirus gene with a zinc finger-like motif. J Virol 1993;67:2168–74.
    OpenUrlAbstract/FREE Full Text
  21. 21.↵
    1. Salvesen GS,
    2. Duckett CS
    . IAP proteins: blocking the road to death's door. Nat Rev Mol Cell Biol 2002;3:401–10.
    OpenUrlCrossRefPubMed
  22. 22.↵
    1. Imoto I,
    2. Yang ZQ,
    3. Pimkhaokham A,
    4. Tsuda H,
    5. Shimada Y,
    6. Imamura M,
    7. et al.
    Identification of cIAP1 as a candidate target gene within an amplicon at 11q22 in esophageal squamous cell carcinomas. Cancer Res 2001;61:6629–34.
    OpenUrlAbstract/FREE Full Text
  23. 23.↵
    1. Imoto I,
    2. Tsuda H,
    3. Hirasawa A,
    4. Miura M,
    5. Sakamoto M,
    6. Hirohashi S,
    7. et al.
    Expression of cIAP1, a target for 11q22 amplification, correlates with resistance of cervical cancers to radiotherapy. Cancer Res 2002;62:4860–6.
    OpenUrlAbstract/FREE Full Text
  24. 24.↵
    1. Bashyam MD,
    2. Bair R,
    3. Kim YH,
    4. Wang P,
    5. Hernandez-Boussard T,
    6. Karikari CA,
    7. et al.
    Array-based comparative genomic hybridization identifies localized DNA amplifications and homozygous deletions in pancreatic cancer. Neoplasia 2005;7:556–62.
    OpenUrlCrossRefPubMed
  25. 25.↵
    1. Tamm I,
    2. Kornblau SM,
    3. Segall H,
    4. Krajewski S,
    5. Welsh K,
    6. Kitada S,
    7. et al.
    Expression and prognostic significance of IAP-family genes in human cancers and myeloid leukemias. Clin Cancer Res 2000;6:1796–803.
    OpenUrlAbstract/FREE Full Text
  26. 26.↵
    1. Carter BZ,
    2. Kornblau SM,
    3. Tsao T,
    4. Wang RY,
    5. Schober WD,
    6. Milella M,
    7. et al.
    Caspase-independent cell death in AML: caspase inhibition in vitro with pan-caspase inhibitors or in vivo by XIAP or Survivin does not affect cell survival or prognosis. Blood 2003;102:4179–86.
    OpenUrlAbstract/FREE Full Text
  27. 27.↵
    1. Tamm I,
    2. Richter S,
    3. Scholz F,
    4. Schmelz K,
    5. Oltersdorf D,
    6. Karawajew L,
    7. et al.
    XIAP expression correlates with monocytic differentiation in adult de novo AML: impact on prognosis. Hematol J 2004;5:489–95.
    OpenUrlCrossRefPubMed
  28. 28.↵
    1. Du C,
    2. Fang M,
    3. Li Y,
    4. Li L,
    5. Wang X
    . Smac, a mitochondrial protein that promotes cytochrome c-dependent caspase activation by eliminating IAP inhibition. Cell 2000;102:33–42.
    OpenUrlCrossRefPubMed
  29. 29.↵
    1. Verhagen AM,
    2. Ekert PG,
    3. Pakusch M,
    4. Silke J,
    5. Connolly LM,
    6. Reid GE,
    7. et al.
    Identification of DIABLO, a mammalian protein that promotes apoptosis by binding to and antagonizing IAP proteins. Cell 2000;102:43–53.
    OpenUrlCrossRefPubMed
  30. 30.↵
    1. Weisberg E,
    2. Ray A,
    3. Barrett R,
    4. Nelson E,
    5. Christie AL,
    6. Porter D,
    7. et al.
    Smac mimetics: implications for enhancement of targeted therapies in leukemia. Leukemia 2010;24:2100–9.
    OpenUrlCrossRefPubMed
  31. 31.↵
    1. Chen KF,
    2. Lin JP,
    3. Shiau CW,
    4. Tai WT,
    5. Liu CY,
    6. Yu HC,
    7. et al.
    Inhibition of Bcl-2 improves effect of LCL161, a SMAC mimetic, in hepatocellular carcinoma cells. Biochem Pharmacol 2012;84:268–77.
    OpenUrlPubMed
  32. 32.↵
    1. Infante JR,
    2. Dees EC,
    3. Olszanski AJ,
    4. Dhuria SV,
    5. Sen S,
    6. Cameron S,
    7. et al.
    Phase I dose-escalation study of LCL161, an oral inhibitor of apoptosis proteins inhibitor, in patients with advanced solid tumors. J Clin Oncol 2014;32:3103–10.
    OpenUrlAbstract/FREE Full Text
  33. 33.↵
    1. Tian A,
    2. Wilson GS,
    3. Lie S,
    4. Wu G,
    5. Hu Z,
    6. Hebbard L,
    7. et al.
    Synergistic effects of IAP inhibitor LCL161 and paclitaxel on hepatocellular carcinoma cells. Cancer Lett 2014;351:232–41.
    OpenUrlCrossRefPubMed
  34. 34.↵
    1. Qin Q,
    2. Zuo Y,
    3. Yang X,
    4. Lu J,
    5. Zhan L,
    6. Xu L,
    7. et al.
    Smac mimetic compound LCL161 sensitizes esophageal carcinoma cells to radiotherapy by inhibiting the expression of inhibitor of apoptosis protein. Tumour Biol 2014;35:2565–74.
    OpenUrlPubMed
  35. 35.↵
    1. Yang J,
    2. McEachern D,
    3. Li W,
    4. Davis MA,
    5. Li H,
    6. Morgan MA,
    7. et al.
    Radiosensitization of head and neck squamous cell carcinoma by a SMAC-mimetic compound, SM-164, requires activation of caspases. Mol Cancer Ther 2011;10:658–69.
    OpenUrlAbstract/FREE Full Text
  36. 36.↵
    1. Eytan DF,
    2. Snow GE,
    3. Carlson S,
    4. Derakhshan A,
    5. Saleh A,
    6. Schiltz S,
    7. et al.
    SMAC mimetic birinapant plus radiation eradicates human head and neck cancers with genomic amplifications of Cell Death Genes FADD and BIRC2. Cancer Res 2016;76:5442–54.
    OpenUrlAbstract/FREE Full Text
  37. 37.↵
    1. Kimple RJ,
    2. Smith MA,
    3. Blitzer GC,
    4. Torres AD,
    5. Martin JA,
    6. Yang RZ,
    7. et al.
    Enhanced radiation sensitivity in HPV-positive head and neck cancer. Cancer Res 2013;73:4791–800.
    OpenUrlCrossRefPubMed
  38. 38.↵
    1. Williams TM,
    2. Flecha AR,
    3. Keller P,
    4. Ram A,
    5. Karnak D,
    6. Galban S,
    7. et al.
    Cotargeting MAPK and PI3K signaling with concurrent radiotherapy as a strategy for the treatment of pancreatic cancer. Mol Cancer Ther 2012;11:1193–202.
    OpenUrlAbstract/FREE Full Text
  39. 39.↵
    1. Kumar B,
    2. Brown NV,
    3. Swanson BJ,
    4. Schmitt AC,
    5. Old M,
    6. Ozer E,
    7. et al.
    High expression of myoferlin is associated with poor outcome in oropharyngeal squamous cell carcinoma patients and is inversely associated with HPV-status. Oncotarget 2016;7:18665–77.
    OpenUrl
  40. 40.↵
    1. Kumar B,
    2. Cordell KG,
    3. Lee JS,
    4. Worden FP,
    5. Prince ME,
    6. Tran HH,
    7. et al.
    EGFR, p16, HPV Titer, Bcl-xL and p53, sex, and smoking as indicators of response to therapy and survival in oropharyngeal cancer. J Clin Oncol 2008;26:3128–37.
    OpenUrlAbstract/FREE Full Text
  41. 41.↵
    1. Houghton PJ,
    2. Kang MH,
    3. Reynolds CP,
    4. Morton CL,
    5. Kolb EA,
    6. Gorlick R,
    7. et al.
    Initial testing (stage 1) of LCL161, a SMAC mimetic, by the Pediatric Preclinical Testing Program. Pediatr Blood Cancer 2012;58:636–9.
    OpenUrlCrossRefPubMed
  42. 42.↵
    1. Fulda S
    . Targeting IAP proteins in combination with radiotherapy. Radiat Oncol 2015;10:105.
    OpenUrl
  43. 43.↵
    1. Beug ST,
    2. Tang VA,
    3. LaCasse EC,
    4. Cheung HH,
    5. Beauregard CE,
    6. Brun J,
    7. et al.
    Smac mimetics and innate immune stimuli synergize to promote tumor death. Nat Biotechnol 2014;32:182–90.
    OpenUrlCrossRefPubMed
  44. 44.↵
    1. Vazquez A,
    2. Bond EE,
    3. Levine AJ,
    4. Bond GL
    . The genetics of the p53 pathway, apoptosis and cancer therapy. Nat Rev Drug Discov 2008;7:979–87.
    OpenUrlCrossRefPubMed
  45. 45.↵
    1. Bonner JA,
    2. Harari PM,
    3. Giralt J,
    4. Cohen RB,
    5. Jones CU,
    6. Sur RK,
    7. et al.
    Radiotherapy plus cetuximab for locoregionally advanced head and neck cancer: 5-year survival data from a phase 3 randomised trial, and relation between cetuximab-induced rash and survival. Lancet Oncol 2010;11:21–8.
    OpenUrlCrossRefPubMed
  46. 46.↵
    1. Varfolomeev E,
    2. Alicke B,
    3. Elliott JM,
    4. Zobel K,
    5. West K,
    6. Wong H,
    7. et al.
    X chromosome-linked inhibitor of apoptosis regulates cell death induction by proapoptotic receptor agonists. J Biol Chem 2009;284:34553–60.
    OpenUrlAbstract/FREE Full Text
  47. 47.↵
    1. Petersen SL,
    2. Wang L,
    3. Yalcin-Chin A,
    4. Li L,
    5. Peyton M,
    6. Minna J,
    7. et al.
    Autocrine TNFα signaling renders human cancer cells susceptible to Smac-mimetic-induced apoptosis. Cancer Cell 2007;12:445–56.
    OpenUrlCrossRefPubMed
  48. 48.↵
    1. Yang D,
    2. Zhao Y,
    3. Li AY,
    4. Wang S,
    5. Wang G,
    6. Sun Y
    . Smac-mimetic compound SM-164 induces radiosensitization in breast cancer cells through activation of caspases and induction of apoptosis. Breast Cancer Res Treat 2012;133:189–99.
    OpenUrlCrossRefPubMed
  49. 49.↵
    1. Matzinger O,
    2. Viertl D,
    3. Tsoutsou P,
    4. Kadi L,
    5. Rigotti S,
    6. Zanna C,
    7. et al.
    The radiosensitizing activity of the SMAC-mimetic, Debio 1143, is TNFα-mediated in head and neck squamous cell carcinoma. Radiother Oncol 2015;116:495–503.
    OpenUrl
  50. 50.↵
    1. Xiao R,
    2. Allen CT,
    3. Tran L,
    4. Patel P,
    5. Park SJ,
    6. Chen Z,
    7. et al.
    Antagonist of cIAP1/2 and XIAP enhances anti-tumor immunity when combined with radiation and PD-1 blockade in a syngeneic model of head and neck cancer. Oncoimmunology 2018;7:e1471440.
    OpenUrl
  51. 51.↵
    1. Huerta S,
    2. Gao X,
    3. Livingston EH,
    4. Kapur P,
    5. Sun H,
    6. Anthony T
    . In vitro and in vivo radiosensitization of colorectal cancer HT-29 cells by the smac mimetic JP-1201. Surgery 2010;148:346–53.
    OpenUrlCrossRefPubMed
  52. 52.↵
    1. Bardia A,
    2. Parton M,
    3. Kummel S,
    4. Estevez LG,
    5. Huang CS,
    6. Cortes J,
    7. et al.
    Paclitaxel with inhibitor of apoptosis antagonist, LCL161, for localized triple-negative breast cancer, prospectively stratified by gene signature in a biomarker-driven neoadjuvant trial. J Clin Oncol 2018;36:3126–33.
    OpenUrl
  53. 53.↵
    1. Stadel D,
    2. Cristofanon S,
    3. Abhari BA,
    4. Deshayes K,
    5. Zobel K,
    6. Vucic D,
    7. et al.
    Requirement of nuclear factor kappaB for Smac mimetic-mediated sensitization of pancreatic carcinoma cells for gemcitabine-induced apoptosis. Neoplasia 2011;13:1162–70.
    OpenUrlCrossRefPubMed
  54. 54.↵
    1. Beug ST,
    2. Cheung HH,
    3. LaCasse EC,
    4. Korneluk RG
    . Modulation of immune signalling by inhibitors of apoptosis. Trends Immunol 2012;33:535–45.
    OpenUrlCrossRefPubMed
  55. 55.↵
    1. Demaria S,
    2. Golden EB,
    3. Formenti SC
    . Role of local radiation therapy in cancer immunotherapy. JAMA Oncol 2015;1:1325–32.
    OpenUrl
View Abstract
PreviousNext
Back to top
Molecular Cancer Therapeutics: 18 (6)
June 2019
Volume 18, Issue 6
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Editorial Board (PDF)

Sign up for alerts

View this article with LENS

Open full page PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for sharing this Molecular Cancer Therapeutics article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
LCL161, a SMAC-mimetic, Preferentially Radiosensitizes Human Papillomavirus–negative Head and Neck Squamous Cell Carcinoma
(Your Name) has forwarded a page to you from Molecular Cancer Therapeutics
(Your Name) thought you would be interested in this article in Molecular Cancer Therapeutics.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
LCL161, a SMAC-mimetic, Preferentially Radiosensitizes Human Papillomavirus–negative Head and Neck Squamous Cell Carcinoma
Linlin Yang, Bhavna Kumar, Changxian Shen, Songzhu Zhao, Dukagjin Blakaj, Tianyun Li, Mitchell Romito, Theodoros N. Teknos and Terence M. Williams
Mol Cancer Ther June 1 2019 (18) (6) 1025-1035; DOI: 10.1158/1535-7163.MCT-18-1157

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
LCL161, a SMAC-mimetic, Preferentially Radiosensitizes Human Papillomavirus–negative Head and Neck Squamous Cell Carcinoma
Linlin Yang, Bhavna Kumar, Changxian Shen, Songzhu Zhao, Dukagjin Blakaj, Tianyun Li, Mitchell Romito, Theodoros N. Teknos and Terence M. Williams
Mol Cancer Ther June 1 2019 (18) (6) 1025-1035; DOI: 10.1158/1535-7163.MCT-18-1157
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Introduction
    • Materials and Methods
    • Results
    • Discussion
    • Disclosure of Potential Conflicts of Interest
    • Disclaimer
    • Authors' Contributions
    • Acknowledgments
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF
Advertisement

Related Articles

Cited By...

More in this TOC Section

  • Sertraline Targets Serine/Glycine Synthesis Enzyme SHMT
  • Combination BETi/MEKi Treatments in Ovarian Cancer
  • Discovery of a Novel Anticancer Therapeutic
Show more Small Molecule Therapeutics
  • Home
  • Alerts
  • Feedback
  • Privacy Policy
Facebook  Twitter  LinkedIn  YouTube  RSS

Articles

  • Online First
  • Current Issue
  • Past Issues
  • Meeting Abstracts

Info for

  • Authors
  • Subscribers
  • Advertisers
  • Librarians

About MCT

  • About the Journal
  • Editorial Board
  • Permissions
  • Submit a Manuscript
AACR logo

Copyright © 2021 by the American Association for Cancer Research.

Molecular Cancer Therapeutics
eISSN: 1538-8514
ISSN: 1535-7163

Advertisement