Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 Sep 1.
Published in final edited form as: Clin Cancer Res. 2011 Jul 12;17(17):5755–5764. doi: 10.1158/1078-0432.CCR-11-0861

Early tumor progression associated with enhanced EGFR signaling with bortezomib, cetuximab, and radiotherapy for head and neck cancer

Athanassios Argiris 1, Austin G Duffy 2, Shivaani Kummar 2, Nicole L Simone 3, Yoshio Arai 4, Seungwon W Kim 1, Susan F Rudy 5, Vishnu R Kannabiran 5, Xinping Yang 5, Minyoung Jang 5, Zhong Chen 5, Nanette Suksta 5, Theresa Cooley-Zgela 3, Susmita G Ramanand 6, Aarif Ahsan 6, Mukesh K Nyati 6, John J Wright 7, Carter Van Waes 3,5
PMCID: PMC3368806  NIHMSID: NIHMS311443  PMID: 21750205

Abstract

PURPOSE

A phase I clinical trial and molecular correlative studies were performed to evaluate preclinical evidence for combinatorial activity of proteasome inhibitor bortezomib, epidermal growth factor receptor (EGFR) inhibitor cetuximab, and radiation therapy.

EXPERIMENTAL DESIGN

Patients with radiotherapy-naïve stage IV or recurrent squamous cell carcinoma of the head and neck (SCCHN) were studied. Escalating doses of bortezomib (0.7, 1.0 and 1.3 mg/m2) were given intravenously twice weekly on days 1, 4, 8, 11, every 21 days, with weekly cetuximab beginning 1 week prior and concurrently with intensity modulated radiotherapy (IMRT), delivered in 2Gy fractions to 70-74 Gy. Molecular effects were examined in serial serum and SCCHN tumor specimens, and the cell line UMSCC-1.

RESULTS

Seven patients were accrued before the study was terminated when 5/6 previously untreated patients with favorable prognosis oropharyngeal SCCHN progressed within 1 year (PFS =4.8 months; 95% CI, 2.6-6.9). Three patients each received bortezomib 0.7 or 1.0 mg/m2, without dose-limiting toxicities; 1 patient treated at 1.3 mg/m2 was taken off study due to recurring cetuximab infusion reaction and progressive disease. Expected grade 3 toxicities included radiation mucositis (n=4), dermatitis (n=1), and rash (n=1). SCCHN-related cytokines increased in serial serum specimens of patients developing progressive disease (P=0.029). Bortezomib antagonized cetuximab- and radiation-induced cytotoxicity, degradation of EGFR, and enhanced prosurvival signal pathway activation in SCCHN tumor biopsies and UMSCC-1.

CONCLUSIONS

Combining bortezomib with cetuximab and radiation therapy demonstrated unexpected early progression, evidence for EGFR stabilization, increased prosurvival signaling and SCCHN cytokine expression, warranting avoidance of this combination.

Keywords: bortezomib, cetuximab, epidermal growth factor receptor, radiotherapy, head and neck cancer

Introduction

Epidermal growth factor receptor (EGFR) is upregulated in many cancers, including approximately 90% of squamous cell carcinomas of the head and neck (SCCHN), where it is associated with decreased patient survival.1,2 Cetuximab (ERBITUX™) is a humanized chimera of C225 that is FDA-approved for use in combination with radiation for treatment of SCCHN. A phase III clinical trial showed that the addition of cetuximab to radiotherapy (RT) results in an approximately 10% improvement in survival over RT alone in patients with locally advanced SCCHN, particularly those of the oropharynx.3 EGFR is implicated in cellular transformation, cell-cycle progression, DNA repair, prosurvival signal pathway activation, and angiogenesis. 4-8 Inhibition of EGFR by anti-EGFR monoclonal antibody C225 has been shown to block pathways leading to inhibition of tumorigenesis and sensitization of EGFR driven tumors. Resistance of remaining SCCHN to EGFR inhibitors has been attributed to EGFR overexpression, mutations, or EGFR-independent mechanisms that co-activate multiple signal pathways important for cancer cell survival.1,2, 9-13

Several prosurvival pathways have been reported to be variably activated by EGFR and other signals in SCCHN, including the Mitogen-Activated Protein Kinases (MAPKs), AKT, Nuclear Factor-kappa B (NF-κB), and Signal Transducer and Transcription (STAT)-3 pathways.8-11 Among these, studies using SCCHN cell lines have revealed that aberrant signaling by cytokine and other growth factor pathways mediate EGFR-independent activation of NF-κB.9,12 NF-κB is a key family of signal-activated transcription factors that affect prosurvival gene activation, the malignant phenotype, and prognosis.12 Bortezomib (VELCADE™, PS-341) is an inhibitor of the 26S proteasome, a macromolecular complex important in degradation of proteins, including Inhibitor-κBs, that can block activation of NF-κBs.14 In preclinical and phase I studies, bortezomib was shown to inhibit NF-κB activation and have cytotoxic, anti-angiogenic, and radiosensitizing activity in SCCHN and other tumors. 15-18 However, in combination with re-irradiation, bortezomib showed limited clinical activity, and lacked the ability to inhibit activated components of the EGFR-inducible MAPK and STAT3 pathways.18-19 Together, preclinical and clinical results suggested that EGFR inhibitor-dependent signal pathways, and NF-κB proteasome-dependent pathways, are independently activated and contribute to the malignant phenotype and clinical response of SCCHN. 8,9, 19 Combined treatment with either of these agents individually with radiation, or with proteasome and EGFR inhibitors, had cytotoxic activity in preclinical and/or early phase clinical studies.1, 2, 16, 17, 20-24

We conducted a phase I study to examine the effects of combination of bortezomib-proteasome and cetuximab-EGFR inhibition with intensity modulated radiation therapy (IMRT) in patients with advanced SCCHN. The primary objectives included evaluation for the toxicities and the maximum tolerated dose (MTD) of this combination. Secondary objectives included clinical response, progression-free and overall survival. Correlative studies evaluated the effects of combined bortezomib and cetuximab to inhibit activation of the EGFR, MAPK, AKT, STAT3 and NF-κB signal pathways, tumor cell survival, and levels of pro-inflammatory and angiogenic cytokines regulated by these pathways and detectable in serum of patients with SCCHN.

Methods

Patient selection

Protocol NCI-7893 was conducted at the National Institute of Health and the University of Pittsburgh, after obtaining approval by the respective Institutional Review Boards and informed consent. Eligibility criteria included age≥18 years; pathologically confirmed SCCHN or poorly/undifferentiated carcinoma of any head/neck site except the nasopharynx; previously untreated stage IV disease, residual disease or regional recurrence, without or with distant metastatic disease <3cm; ECOG performance status 0-1; adequate organ function; recovery from any prior surgery or chemotherapy including prior cisplatin >3 months; and no prior systemic EGFR inhibitors, bortezomib, head and neck radiation, uncontrolled intercurrent illness; or grade ≥2 peripheral sensory neuropathy.

Treatment Plan and Patient Assessments

The schema for the treatment plan and correlative studies is shown in Fig. 1. A standard 3+3 dose escalation design (3 subjects without, or up to 6 subjects after a dose limiting toxicity per dose level) was planned. Bortezomib (0.7, 1.0 and 1.3 mg/m2) was given intravenously twice weekly on days 1, 4, 8, 11, every 21 days. To obtain serum and optional tumor biopsies with the drug combination without and with radiation, bortezomib and cetuximab 400 mg/m2 were started 1 week prior to combining bortezomib and weekly cetuximab 250 mg/m2 with intensity modulated radiation therapy. Tumor received 2Gy per fraction once daily 5 days per week to 70-74Gy. Regions of intermediate and low risk were received 60-64 and 50 Gy, respectively. Bortezomib (Millennium Pharmaceuticals Inc, Cambridge, MA) was provided through a Clinical Trials Agreement, Cancer Therapeutics Evaluation Program, National Cancer Institute (NCI).

Figure 1. Dosing Schema NCI 7893 Bortezomib with weekly Cetuximab and IMRT.

Figure 1

Patients were given escalating doses of Bortezomib (0.7, 1.0 and 1.3 mg/m2), twice weekly by IV on days 1, 4, 8 and 11 every 3 weeks. Bortezomib and cetuximab (400mg/m2 loading dose, bold arrow) were started during week 1, followed by bortezomib and weekly cetuximab (250 mg/m2, non-bold arrows) concurrent with IMRT 2Gy/day 5 days per week to 70-74 Gy. Serum was collected as indicated for SCCHN-related cytokines and optional tumor biopsies were obtained prior and during the first cycle of treatment as indicated.

Baseline evaluation included history, physical exam, standard laboratory tests, and CT or CT-PET imaging of the head, neck and chest obtained within 2 weeks of treatment. During treatment, patients underwent weekly physical exam, toxicity evaluation, CBC and blood chemistries. Toxicities were assessed by NCI common terminology criteria for adverse events (http://ctep.cancer.gov/protocoldevelopment/electronic_applications/docs/ctcaev3.pdf). Collection of serum was planned pretreatment and during each cycle, and optional tumor biopsies were planned pretreatment and week 1 and 2 of cycle 1 as in Fig. 1. Tumor measurements were performed at baseline, 2 and 5 months after completion of radiotherapy. Response and progression was evaluated using RECIST criteria.25

Study Endpoints

The primary endpoints included dose limiting toxicities (DLTs), other toxicities, and the maximum tolerated dose (MTD) of bortezomib for this combination regimen. Patients were evaluable for toxicity if they received one cycle of therapy, or if they had DLT during the first cycle. Evaluation for DLT included the period on drug/radiation treatment plus 4 weeks of follow-up. DLTs were defined as CTCAE 3.0 grade 4 toxicities for: in-field stomatitis/mucositis, dermatitis or dysphagia lasting >5 days; rash; nausea/vomiting despite appropriate antiemetic therapy; absolute neutropenia <500/μL for more than 7 days, or neutropenic fever; thrombocytopenia; and recurrent grade 4 hematologic toxicities following delay or dose modification. Other DLTs included grade 3 or recurrent grade 2 neuropathy despite dose delay or modifications; all other grade 3 or higher toxicities, except grade 3 fatigue; infection without grade 4 neutropenia; in-field toxicities and nausea/vomiting as above; weight loss; dehydration; creatinine; hypotension; anorexia; pain; and any grade hypomagnesemia, hypokalemia or hyponatremia. DLT also included treatment delay due to toxicity of more than 3 weeks, except cetuximab infusion reactions ≥ Gr3, for which study removal and replacement were planned. Toxicities attributable (possible, probable or definite) to the study treatment were used for determination of DLT and MTD.

Secondary clinical endpoints included objective response, progression-free survival and overall survival. Secondary correlative endpoints included pre- to post-treatment changes in a set of serum and tumor biomarkers below.

Serum Cytokine and Growth Factor Assays

Concentrations of serum cytokine and growth factors were determined as described previously.18, 26 Peripheral blood sample collection was planned within 2 weeks of initiation of treatment, and then after initiation of study drugs on days 1, 5 and 12 of the first cycle. Thereafter, optional blood for serum could be collected on day 1, 5 and 12 of the second and third cycles (weeks 5 and 8) of bortezomib; following completion of RT at 8 weeks; and up to 3, 6, 12, 15, 18, 21 and 24 months. See Supplemental Methods for details.

Correlative studies of SCCHN tumor and cell line UMSCC-1

Serial SCCHN tumor biopsies were obtained pretreatment, on day 5 after induction with bortezomib and cetuximab, and on day 12 after combination with IMRT from one patient (#7) who consented to optional biopsies. To confirm and elucidate the mechanism of results obtained, HNSCC cell line UM-SCC1 was treated with bortezomib, cetuximab and/or radiation. Methods for immunoblotting of SCCHN tumor and UMSCC-113,17 and clonogenic survival assays17 for correlative studies were described previously, and as modified in Supplementary Methods.

Statistics

Using the standard 3+3 design, dose escalation is based on a ≤33% true rate of DLT in 3 patients, and MTD on a ≤16% true rate if the cohort is expanded to 6 subjects for a DLT.18 Based on previous studies linking increasing cytokine levels with progressive disease,26 an exploratory comparison of the progression free survival between patients whose early cytokine changes after initiating treatment tended to increase vs. those whose values tended to decrease or remain steady was done using an exact log-rank test. For clonogenic survival assays, the difference in the surviving fraction after combination of drug treatments and 2 Gy irradiation was compared, as the surviving fraction in cell lines has been reported to correlate with the radiocurability of the corresponding human tumors in vivo.27

Results

Patient characteristics, treatment and response

Subjects (Table 1) included 6 previously untreated patients with stage IV and 1 patient who presented after incomplete staging and surgical neck dissection with recurrent tonsil and neck SCCHN; 6 had oropharyngeal and 1 had a laryngeal primary. Treatment delivery (Supplemental Table 1) was completed in the first 6 patients, although bortezomib dose reduction was required in one dose level 2 patient for thrombocytopenia. Patient 7 (1.3 mg/m2 dose level) was taken off study after 8 doses of bortezomib and 6 doses of cetuximab for recurrent grade 2 cetuximab infusion reactions and progressive disease (PD). There were no DLTs and a MTD for combination of bortezomib with cetuximab and radiation was not reached before the study was ended. Clinical outcomes (Table 2) precipitated termination of the study after 5 out of 6 of the previously untreated patients exhibited progression within one year. Only 3 of 7 patients achieved a complete response (CR) within 2 to 5 months (patients 1,4,6). Of these, patient 4 developed a solitary pulmonary metastasis at 11 months. Three patients had PD during (patient 7) or within 5 months of treatment (patients 3, 5). Overall, 3 had local-regional and 2 had pulmonary PD. Local-regional failures occurred within the 70Gy treatment region.

Table 1. Patient and tumor characteristics.

Patient
number
Age
(years)
Gender Stage/
resectability
Primary site HPV & p16
status
Smoking
history
Alcohol
history
1 52 Male Original
TxN2aM0,
Recurrence
tonsil, neck
resectable
Tonsil N/A Former
35pk/yrs
No
2 48 Male T3N2cM0
resectable
Base of
tongue
HPV+ Current
68pk/yrs
No
3 58 Male T1N2aM0
resectable
Base of
Tongue
HPV+ Never Yes
4 62 Male T4N2cM0
resectable
Base of
Tongue
HPV − / p 16+ Former
weekly
pipe/cigar
Yes
5 61 Male T3N2cM0
unresectable
Base of
Tongue
HPV+ Former
48pk/yrs
Yes
6 50 Female T3N2cM0
resectable
Larynx HPV− Former
35pk/yrs
Yes
7 54 Female T1N2aM0
resectable
Tonsil HPV+/p16+ Current
30pk/yrs
Yes

Abbreviations: TNM, Tumor, Node, Metastasis; Pk/yrs=cigarette pack years; N/A, not available Patient 1 presented with incomplete TxN2aM0 staging and treatment consisting of neck dissection only, after which he sought care with recurrent tonsil and neck disease

Table 2. Treatment and outcomes.

Patient
number
Bortezomib
dose level
(per m2)
Best
objective
response
Disease
progression
Progressionfree
interval
(months)
Site of
progression
Salvage therapy Overall
survival
(months)
Survival
status
1 0.7 mg/m2 CR No 14 None None 14 NED
2 0.7 mg/m2 PD Yes 6 Distant None 8 DOD
3 0.7 mg/m2 PD Yes 5 Regional Neck surgery 17 NED
4 1 mg/m2 CR Yes 11 Distant Lobectomy 23 NED
5 1 mg/m2 PD Yes 5 Local None 17 DOD
6 1 mg/m2 CR No 24 None None 24 NED
7 1.3 mg/m2 PD Yes 1 Regional Chemoradiotherapy 18 NED

Best response by CT/PET at 2 or 5 months post-treatment: CR, complete response; PD, progressive Disease; Clinical Progression-Free and Overall Survival as of last visit or 12/1/10; NED, no evidence of disease; DOD, dead of disease

The median Progression Free Survival (PFS) was only 4.8 months (95% CI, 2.6-6.9), including early recurrence in 5/6 with previously untreated oropharynx cancer, which compared unfavorably with a median PFS of 17 months reported for cetuximab and radiation at that site.3 While stratification for tumor site and testing for human papillomavirus (HPV) or p16 status were not incorporated in design of this phase I study, of six patients with oropharyngeal primary site lesions, patients 2, 3 and 5 were reportedly HPV+ prior to study entry, and specimens from PD lesions in patients 4 (lung) and 5 (oropharynx) tested p16+ at the site of progressive disease, consistent with HPV origin. Of the 5 with PD, patients 2, 4, 5, and 7 had other co-factors considered to increase risk of recurrence (T stage≥3 or unresectable; N stage≥2; and current or former smoking ≥10 pack/years). Early detection of PD and salvage therapy by parotidectomy (patient 3), lobectomy (patient 5), or cisplatin concurrent with remaining radiation (patient 7) achieved disease-free status in 3/5 recurrent patients, who together have a median overall survival of 18 months at last follow-up. The patient with a laryngeal primary and CR remains disease-free after 24 months.

Toxicities

Toxicities (Table 3) included expected grade 3 toxicities for the treatment combination, such as mucositis (4), dysphagia (3), xerostomia (1), and dermatitis (1); cetuximab-associated acneiform rash (1); and bortezomib-associated peripheral neuropathy (1). One Grade 3 infection occurred in a patient without neutropenia.

Table 3. Worst toxicities (n=7).

Grade 0 Grade 1 Grade 2 Grade 3 Grade 4
Neutropenia 6 1 0 0 0
Anemia 3 3 1 0 0
Thrombocytopenia 5 0 2 0 0
Lymphopenia 4 0 0 1 2
Infection with normal or grade1/2 neutrophils 5 0 0 2 0
Infection, other 5 0 2 0 0
Mucositis/stomatisis (clinical exam) 0 0 3 4 0
Dysphagia 0 0 3 4 0
Radiation dermatitis 0 1 2 4 0
Rash – Acneiform 4 0 2 1 0
Rash – Desquamation 5 1 1 0 0
Dermatology-Other 5 1 1 0 0
Allergic reaction/hypersensitivity (drug fever) 6 0 1 0 0
Motor and Sensory Neuropathy 5 0 1 1 0
Muscle weakness, generalized, whole body 6 1 0 0 0
Diarrhea 5 2 0 0 0
Constipation 4 1 2 0 0
Anorexia 4 1 2 0 0
Nausea 2 3 2 0 0
Vomiting 3 3 1 0 0
Weight loss 4 1 2 0 0
Elevated Alkaline Phosphatase 5 2 0 0 0
Elevated iver Transaminases, AST 5 2 0 0 0
Elevated Liver Transaminases, ALT 2 5 0 0 0
Fever without neutropenia 4 3 0 0 0
Gastrointestinal - Other 4 1 2 0 0
Hiccoughs 6 1 0 0 0
Allergic rhinitis 5 1 1 0 0
Fatigue/Asthenia 0 1 6 0 0
Cytokine Release Syndrome 6 0 1 0 0
Diaphoresis 6 1 0 0 0
Dysarthria/Voice Changes 6 0 1 0 0
Dysgeusia/Taste Changes 4 1 2 0 0
Dyspepsia 6 0 1 0 0
Edema 6 1 0 0 0
Hyperbilirubinemia 6 1 0 0 0
Hypermagnesemia 4 3 0 0 0
Potassium, serum-high 6 1 0 0 0
Hypoalbuminemia 4 1 2 0 0
Hypocalcemia 5 1 1 0 0
Hypomagnesemia 5 1 1 0 0
Hyponatremia 4 3 0 0 0
Hypophosphatemia 5 1 0 1 0
Hypotension 5 0 2 0 0
Insomnia 6 1 0 0 0
Pain, all types 0 0 4 3 0
Rigors, Chills 5 1 1 0 0
Sino-nasal reactions 4 2 1 0 0
Skin breakdown/Decubitus Ulcer 6 0 1 0 0
Xerosis/ Dry Skin 3 3 1 0 0
Xerostomia/Dry Mouth 3 3 1 0 0

Correlative Studies of Serum Cytokines

Previously, a pre-treatment increase in multiple tumor-related cytokine and angiogenic growth factors was detected in patients with SCCHN.26 Based on the rationale that this set of cytokines were co-regulated by NF-κB, the predictive value of coordinate changes in 3 or more of these cytokines was evaluated. Longitudinal increases in 3 or more of these factors was associated with decreased response and survival in patients with oropharyngeal SCCHN. Consistent with previous findings, increases in 3 or more cytokines occurred in 3 patients (2,3, 5, and 7) who developed PD, and increases in 2 or fewer cytokines, or decline, in 3 patients with CRs (1,4,6) (Fig. 2). Although based on fewer patients, there was evidence suggesting that those patients whose initial cytokine profile was generally associated with increasing values after starting treatment were more likely to have shorter progression free survival than those whose cytokine levels tended to decline with greater PFS (P=0.029 by exact two-tailed log-rank test). The patient showing the greatest increase in all 4 cytokines during cycle 1 was treated with the highest dose of bortezomib (#7, 1.3mg/m2), and developed PD in the neck by week 5 while on treatment.

Figure 2. Longitudinal changes in serum cytokine levels in seven patients.

Figure 2

Based on the rationale of co-regulation of 5 serum cytokines by NF-κB, the predictive value of coordinate changes in 3 or more of these cytokines was evaluated, as previously (23). Cytokine concentrations (pg/ml) are presented as mean ± standard deviation of replicates on a log scale vs. days since beginning treatment. Pre-Tx = pre-treatment. Longitudinal increase is slope of 3 or more cytokines previously associated with poor response and survival in patients with oropharyngeal carcinoma23 was seen in patients 2, 3, 5, and 7, and associated with decreased progression free survival (P=0.029, log-rank test).

Correlative studies of markers of prosurvival signal, transcription factors and apoptosis

We examined the pharmacodynamic effects of bortezomib and cetuximab on EGFR, downstream signal, and apoptosis markers we previously validated for SCCHN in multiple studies.8, 10, 11, 15, 17-20 Only patient 7 consented to optional serial biopsies of SCCHN primary tumor, which were obtained pretreatment, on day 5 after induction with bortezomib 1.3 mg/m2 (days 1, 4) and cetuximab 400 mg/m2(day 1), and on day 12, after combination with IMRT. Fig. 3A shows that by day 5, combination of bortezomib and cetuximab enhanced, rather than inhibited, phosphorylated and total EGFR, pERK1/2, and NF-κB p65 subunit. By day 12, with addition of IMRT, further enhancement of phosphorylated and total EGFR, pAKT, STAT3 and NF-κB p65 was observed. Increase in cleaved PARP as an indicator of cytotoxicity was only detected after initiation of IMRT (day 12).

Figure 3. Combined bortezomib, cetuximab and radiation enhances co-activation of EGFR and multiple prosurvival pathways in SCCHN tumor biopsies, along with clonogenic survival in line UMSCC-1.

Figure 3

A. Tumor biopsies were obtained from patient #7 before and on day 5 after initiating combined treatment with bortezomib and cetuximab, and on day 12 after addition of IMRT. Protein extracts from tumor specimens were subjected to SDS-PAGE and Western blots were performed for activated EGFR and signal phospho- and total proteins shown. Combined bortezomib and cetuximab treatment increased phosphorylation and total EGFR, and phosphorylation of downstream prosurvival signal kinases and transcription factors, including p-AKT, p-ERK1/2, p-STAT3 and p-NF-κB p65. B. Clonogenic survival assays. UMSCC-1 cells were treated with bortezomib, cetuximab, and RT alone or in combinations as indicated. Clonogenic assays were performed, and surviving fractions are presented. C. Western blots were performed for activated EGFR and signal phospho- and total proteins shown. Combined bortezomib and cetuximab treatment with RT resulted in stabilization of EGFR, and phosphorylated EGFR and downstream prosurvival signal kinases and transcription factors, including p-AKT, p-ERK1/2, p-STAT3.

Molecular effects in SCCHN in vitro

To further determine how these effects observed in tumor specimens were related to the activity of the individual or combination of agents, SCCHN cell line UMSCC-1 was treated as indicated, and effects were examined by clonogenic survival assay, and western blot for EGFR and downstream signaling components (Fig. 3B, C). Combination of C225 or bortezomib and radiation reduced clonogenic survival (Fig. 3B). However, combination of cetuximab and bortezomib when combined with radiation, reduced the overall effect of treatment to a level intermediate between that observed with either C225 or bortezomib with radiation, and the control (Fig. 3B). Reduction of survival was accompanied by reduction in EGFR and pEGFR (Fig. 3C). Inhibition of one or more downstream signal mediators including pAKT, pERK, and pSTAT3 was often observed with C225, or combination of C225 and radiation, but bortezomib attenuated these effects (Fig. 3C). These findings may explain the reduced efficacy of C225 and radiation when combined with bortezomib, which can inhibit proteasome activity, and possibly, C225-induced EGFR degradation.

Discussion

The combination of bortezomib, cetuximab, and IMRT was tolerated with supportive care, but resulted in a median PFS of only 4.8 months. These poor efficacy results included 5/6 previously untreated patients with HPV and/or p16 positive oropharyngeal carcinomas, which compared unfavorably to results of 17.1 months for oropharynx site tumors reported for cetuximab and radiotherapy.3 This group has been associated with favorable prognosis in additional studies, even though unfavorable characteristics (advanced stage and history of heavy smoking) which can influence outcome, were also present.28 While it is possible these adverse risk factors contributed to the unexpectedly low response and early recurrence in the small cohort in the present study, translational studies provided additional evidence for an adverse interaction of the combination of bortezomib, cetuximab and radiation. Greater than expected EGFR and cell survival signaling, and angiogenesis factor expression by SCCHN was observed. Together, the clinical and molecular findings caution against further clinical investigation of this combination of agents.

The clinical results of this study were initially surprising after early preclinical and clinical studies provided evidence that combined treatment with either of these agents individually with radiation, or a combination of proteasome and EGFR inhibitors, potentiated cytotoxic activity.1, 2, 16, 17, 20-24 However, evidence emerging from one of our laboratories concurrent with this trial indicated that proteasome inhibitors could potentially antagonize chemotherapy or radiation induced EGFR degradation, and anti-proliferative and cytotoxic effects (M. Nyati, unpublished observations).27 Consistent with this possibility, analysis of serial tumor biopsies from the patient who developed PD on-treatment revealed increased EGFR and prosurvival signaling instead of EGFR degradation and attenuation of prosurvival signaling previously reported with cetuximab or cetuximab and radiation.27 Further studies in the UMSCC-1 cell line showed that combination of C225 or bortezomib with radiation reduced clonogenic survival consistent with previous preclinical studies (Fig. 3B), but combination of cetuximab and bortezomib with radiation, reduced the overall effect of treatment to a level intermediate between that observed with either drug with radiation, and the control (Fig. 3B). Bortezomib also attenuated the effects of cetuximab- and radiation-induced EGFR degradation, and inhibition of prosurvival signaling in UMSCC-1 (Fig. 3C).

Since recent evidence suggests EGFR is degraded by the ubiqutin-proteasome system,30,31 it appears likely that proteasome inhibition by bortezomib could attenuate the cytotoxic effects of cetuximab and radiation by protecting EGFR from degradation. Further, recent reports demonstrate proteasome inhibitor-induced activation of EGFR as well as EGFR-independent mechanisms can induce MAPK, AKT and STAT3 prosurvival pathways, as observed here.9,19, 32-34 Additionally, while proteasome inhibitors radiosensitized cancer cells and smaller xenograft tumors in experimental models,15-17 they may enhance radioprotection of SCC tumor cells under hypoxic conditions,35 such as occur in large SCCHN in advanced stage patients. Cytokines and angiogenesis factors expressed by SCCHN in response to prosurvival26 and hypoxia signals36 were detected in serum of all 7 patients pretreatment. Concentrations of 3 or more serum cytokine and angiogenic growth factors previously shown to increase with poor response and survival in patients with oropharyngeal SCCHN,26 increased in the 3 patients with early PD. We and others have shown that these cytokines may be produced in other patient tumors and cell lines by SCCHN epithelial and stromal cells.37,38 We have further shown that cytokines such as IL-8 may be induced in SCCHN lines by bortezomib through activation of MAPK signaling and transcription factor AP-1. 32 Thus, proteasome inhibitor and EGFR induced expression of IL-8., VEGF and HGF could enhance angiogenesis, tumorigenesis and metastasis. 32, 37, 38

Together, the stabilization or enhancement of EGFR-mediated survival signaling and angiogenesis factor expression may help further explain the suboptimal efficacy of the combination of these drugs with radiation. These observations suggest that cetuximab and radiation have multiple effects on cancer besides DNA repair, and that combination studies should be pursued with caution. Both drug-drug and drug-radiation interactions affecting diverse mechanisms may need to be considered when developing therapeutic regimens.

The first in human phase I study of bortezomib in combination with re-irradiation for recurrent SCCHN was also recently concluded at NIH. While bortezomib inhibited proteasome, NF-κB p65 subunit, and prosurvival genes,18 clinical activity of bortezomib plus reirradiation was limited.19 PRs were seen in 5/10 patients receiving lower doses and bortezomib treatment breaks, while PD occurred in patients receiving a continuous schedule or higher doses of bortezomib with re-irradiation. The limited clinical activity observed was also associated with lack of inhibition of EGFR-activated ERK or STAT3 pathways, as well as other non-canonical NF-κB/REL family members, which may also contribute to cell survival.19 A recent phase I study of bortezomib plus cetuximab in treatment refractory patients with tumors expressing EGFR yielded SD but no PRs or CRs in 5/6 with SCCHN or lung cancer.24 Altogether, the results of these studies demonstrate that bortezomib in combination with cetuximab or re-irradiation results in incomplete clinical and molecular responses in SCCHN.

Recently completed phase II studies of bortezomib with other chemotherapies for recurrent SCCHN also showed evidence of limited combinatorial activity or possible chemoprotection.39,40 One of these studies showed that the response rate was lower than expected for docetaxel alone, and PD was associated with an increased NF-κB and EGFR gene profile.39 Another phase I trial evaluated bortezomib in combination with weekly cisplatin 30 mg/m2 and RT for advanced SCCHN.41 Twenty-seven patients with previously untreated local-regionally advanced (10 patients) or recurrent/previously irradiated (17 patients) SCCHN were studied. Only 8 patients (30%) were without PD at a median 7.3 month follow-up. Interestingly, there is now also evidence that proteasome inhibition may antagonize chemotherapy-mediated EGFR degradation and cytotoxicity as well. Gemcitabine or cisplatin chemotherapy cytotoxicity was shown to involve ubiquitination and proteasome-dependent EGFR degradation.30, 42

In conclusion, the present and other clinical and mechanistic studies suggest that bortezomib may have limited clinical efficacy, and in some instances, lower than expected activity due to antagonism, when combined with cetuximab and other cytotoxic therapies of known activity in SCCHN. Proteasome inhibitor-mediated activation of EGFR-dependent and independent MAPK, AKT or STAT3 prosurvival signaling may be countered by combination with ERK, JNK and AKT inhibitors. 31, 32 However, as learned here and other recent trials cited above, further study of proteasome inhibitors in combination with other targeted therapies should be considered only with caution after testing in appropriate non-HPV and HPV+ HNSCC xenograft models appropriate to the patient population to be studied. These results also provide several insights important in avoiding or reducing the impact of unfavorable outcomes in the future. Despite the inherent challenges and limitations in preclinical modeling of the combination and sequencing of multiple therapies to be used in clinical trials, accurate modeling is important to identify potential interactions and mechanisms that could result in unfavorable clinical outcomes. Close monitoring is important for early recognition of unfavorable outcomes for provision of additional therapy, early stoppage of the study, and reporting. Obtaining paired pre and on-treatment specimens for correlative studies can support the identification of possible underlying mechanisms.

Supplementary Material

1

Translational Relevance.

Epidermal growth factor receptor (EGFR) inhibitor cetuximab and radiotherapy is approved for squamous cell carcinoma of the head and neck (SCCHN), but the added benefit is limited to a subset of patients. EGFR inhibitors attenuate signaling via Mitogen Activated Protein Kinases (MAPKs) and Signal Transduction and Transcription Factor 3 (STAT3), while proteasome inhibitors block activation of Nuclear Factor-κB, another signal-activated transcription factor important in survival of SCCHN. Combined treatment with proteasome and EGFR inhibitors, or these agents individually with radiation demonstrated cytotoxic activity in preclinical and/or clinical studies. In this phase I trial, combining bortezomib with cetuximab and radiation therapy demonstrated unexpectedly short progression free survival that led to early study termination. There was evidence that bortezomib antagonized cetuximab- and radiation-induced degradation of EGFR, enhanced prosurvival signal pathway activation, and cell survival. Further clinical studies of proteasome inhibitors in combination with other therapies in SCCHN should be undertaken with caution.

Acknowledgements

The assistance of Dr. Seth Steinberg, NCI, in statistical analysis of the serum cytokine data, and Drs. Barbara Burtness, Fox Chase Cancer Center, and Arlene Forastiere, MD, ITA Partners, for helpful critique is gratefully appreciated.

Research funding was provided through a Clinical Trials Agreement between NCI and Milliennium Pharmaceuticals to CVW, and Millennium Pharmaceuticals to AA.

Supported by NIH Projects ZIADC-000016, U01 CA099168-01, Head and Neck Cancer SPORE Grant No. P50 CA097190-06 from the National Cancer Institute, Millennium Pharmaceuticals, and a Clinical Trials Agreement between NCI CTEP and Millennium Pharmaceuticals.

Footnotes

Authors’ Disclosure of Potential Conflicts of Interest

Presented in part at the 46th Annual Meeting of the American Society of Clinical Oncology, Chicago, IL, June 4-8, 2010.

References

  • 1.Rubin Grandis J, Melhem MF, Gooding WE, et al. Levels of TGF-alpha and EGFR protein in head and neck squamous cell carcinoma and patient survival. J Natl Cancer Inst. 1998;90:824–832. doi: 10.1093/jnci/90.11.824. [DOI] [PubMed] [Google Scholar]
  • 2.Ang KK, Berkey BA, Tu X, et al. Impact of epidermal growth factor receptor expression on survival and pattern of relapse in patients with advanced head and neck carcinoma. Cancer Res. 2002;62:7350–6. [PubMed] [Google Scholar]
  • 3.Bonner JA, Harari PM, Giralt J, et al. Radiotherapy plus cetuximab for squamous-cell carcinoma of the head and neck. N Engl J Med. 2006;354:567–78. doi: 10.1056/NEJMoa053422. [DOI] [PubMed] [Google Scholar]
  • 4.Huang SM, Harari PM. Modulation of radiation response after epidermal growth factor receptor blockade in squamous cell carcinomas: inhibition of damage repair, cell cycle kinetics, and tumor angiogenesis. Clin Cancer Res. 2000;6:2166–74. [PubMed] [Google Scholar]
  • 5.Harari PM, Huang SM. Head and neck cancer as a clinical model for molecular targeting of therapy: Combining EGFR blockade with radiation. Int J Rad Oncol Biol Phys. 2001;49:427–33. doi: 10.1016/s0360-3016(00)01488-7. [DOI] [PubMed] [Google Scholar]
  • 6.Dittmann K, Mayer C, Rodemann HP. Inhibition of radiation-induced EGFR nuclear import by C225 (Cetuximab) suppresses DNA-PK activity. Radiotherapy and Oncology. 2005;76:157–61. doi: 10.1016/j.radonc.2005.06.022. [DOI] [PubMed] [Google Scholar]
  • 7.Kriegs M, Kasten-Pisula U, Rieckmann T, et al. The epidermal growth factor receptor modulates DNA double-strand break repair by regulating non-homologous end-joining. DNA Repair. 2000;9:889–97. doi: 10.1016/j.dnarep.2010.05.005. [DOI] [PubMed] [Google Scholar]
  • 8.Bancroft CC, Chen Z, Yeh J, Sunwoo JB, et al. Effects of pharmacologic antagonists of epidermal growth factor receptor, PI3K and MEK signal kinases on NF-kappaB and AP-1 activation and IL-8 and VEGF expression in human head and neck squamous cell carcinoma lines. Int J Cancer. 2002;99:538–48. doi: 10.1002/ijc.10398. [DOI] [PubMed] [Google Scholar]
  • 9.Lee TL, Yeh J, Van Waes C, Chen Z. Epigenetic modification of SOCS-1 differentially regulates STAT3 activation in response to interleukin-6 receptor and epidermal growth factor receptor signaling through JAK and/or MEK in head and neck squamous cell carcinomas. Mol Cancer Ther. 2006;5:8–19. doi: 10.1158/1535-7163.MCT-05-0069. [DOI] [PubMed] [Google Scholar]
  • 10.Pernas FG, Allen CT, Winters ME, et al. Proteomic signatures of epidermal growth factor receptor and survival signal pathways correspond to gefitinib sensitivity in head and neck cancer. Clin Cancer Res. 2009;15:2361–72. doi: 10.1158/1078-0432.CCR-08-1011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Van Waes C, Allen CT, Citrin D, et al. Molecular and clinical responses in a pilot study of gefitinib with paclitaxel and radiation in locally advanced head-and-neck cancer. Int J Radiat Oncol Biol Phys. 2010;77:447–54. doi: 10.1016/j.ijrobp.2009.05.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Allen CT, Ricker JL, Chen Z, et al. Role of activated nuclear factor-kappaB in the pathogenesis and therapy of squamous cell carcinoma of the head and neck. Head Neck. 2007;29:959–71. doi: 10.1002/hed.20615. [DOI] [PubMed] [Google Scholar]
  • 13.Cohen J, Chen Z, Lu SL, et al. Attenuated transforming growth factor beta signaling promotes nuclear factor-kappaB activation in head and neck cancer. Cancer Res. 2009;69:3415–24. doi: 10.1158/0008-5472.CAN-08-3704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Adams J, Kauffman M. Development of the proteasome inhibitor Velcade (Bortezomib) Cancer Invest. 2004;22:304–311. doi: 10.1081/cnv-120030218. [DOI] [PubMed] [Google Scholar]
  • 15.Sunwoo JB, Chen Z, Dong G, et al. Novel proteasome inhibitor PS-341 inhibits activation of nuclear factor-kappa B, cell survival, tumor growth, and angiogenesis in squamous cell carcinoma. Clin Cancer Res. 2001;7:1419–1428. [PubMed] [Google Scholar]
  • 16.Pervan M, Pajonk F, Sun JR, et al. Molecular pathways that modify tumor radiation response. Am J Clin Oncol. 2001;24:481–485. doi: 10.1097/00000421-200110000-00013. [DOI] [PubMed] [Google Scholar]
  • 17.Van Waes C, Sunwoo JB, DeGraff W, et al. Radiosensitization and proteasome inhibition. In: Adams J, editor. Proteasome Inhibitors in Cancer Therapy. Humana Press; Totowa, NJ: 2004. pp. 123–130. [Google Scholar]
  • 18.Van Waes C, Chang AA, Lebowitz PF, et al. Inhibition of nuclear factor-kappaB and target genes during combined therapy with proteasome inhibitor bortezomib and reirradiation in patients with recurrent head-and-neck squamous cell carcinoma. Int J Radiat Oncol Biol Phys. 2005;63:1400–1412. doi: 10.1016/j.ijrobp.2005.05.007. [DOI] [PubMed] [Google Scholar]
  • 19.Allen C, Saigal K, Nottingham L, et al. Bortezomib-induced apoptosis with limited clinical response is accompanied by inhibition of canonical but not alternative nuclear factor-{kappa}B subunits in head and neck cancer. Clin Cancer Res. 2008;14:4175–4185. doi: 10.1158/1078-0432.CCR-07-4470. [DOI] [PubMed] [Google Scholar]
  • 20.Morris JC, Citrin D, Nottingham L, et al. Phase I study of proteasome inhibitor bortezomib (B) concurrent with re-irradiation therapy (re-RT) for recurrent squamous cell carcinoma of the head and neck (SCCHN) J Clin Oncol. 2010;28:15S–2603. [Google Scholar]
  • 21.Lorch JH, Thomas TO, Schmoll HJ. Bortezomib Inhibits Cell-Cell Adhesion and Cell Migration and Enhances Epidermal Growth Factor Receptor Inhibitor-Induced Cell Death in Squamous Cell Cancer. Cancer Research. 2007;67:727–734. doi: 10.1158/0008-5472.CAN-06-2162. [DOI] [PubMed] [Google Scholar]
  • 22.Wagenblast J, Baghi M, Arnoldner C, et al. Cetuximab enhances the efficacy of bortezomib in squamous cell carcinoma cell lines. J Cancer Res Clin Oncol. 2009;135:387–393. doi: 10.1007/s00432-008-0477-0. [DOI] [PubMed] [Google Scholar]
  • 23.Cascone T, Morelli MP, Morgillo F, et al. Synergistic anti-proliferative and pro-apoptotic activity of combined therapy with bortezomib, a proteasome inhibitor, with anti-epidermal growth factor receptor (EGFR) drugs in human cancer cells. J Cell Physiol. 2008;216:698–707. doi: 10.1002/jcp.21444. [DOI] [PubMed] [Google Scholar]
  • 24.Dudek AZ, Lesniewski-Kmak K, Shehadeh NJ, et al. Phase I study of bortezomib and cetuximab in patients with solid tumours expressing epidermal growth factor receptor. Br J Cancer. 2009;100:1379–1384. doi: 10.1038/sj.bjc.6605043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Therasse P, Arbuck SG, Eisenhauer EA, et al. New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada. J Natl Cancer Inst. 2000;92:205–216. doi: 10.1093/jnci/92.3.205. [DOI] [PubMed] [Google Scholar]
  • 26.Allen C, Duffy S, Teknos T, et al. Nuclear factor-kappaB-related serum factors as longitudinal biomarkers of response and survival in advanced oropharyngeal carcinoma. Clin Cancer Res. 2007;13:3182–3190. doi: 10.1158/1078-0432.CCR-06-3047. [DOI] [PubMed] [Google Scholar]
  • 27.Fertil B, Malaise EP. Inherent cellular radiosensitivity as a basic concept for human tumor radiotherapy. Int J Radiat Oncol Biol Phys. 1981;7:621–629. doi: 10.1016/0360-3016(81)90377-1. [DOI] [PubMed] [Google Scholar]
  • 28.Ang KK, Harris J, Wheeler R, et al. Human papillomavirus and survival of patients with oropharyngeal cancer. N Engl J Med. 2010;363:24–35. doi: 10.1056/NEJMoa0912217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Feng FY, Lopez CA, Normolle DP, et al. Effect of epidermal growth factor receptor inhibitor class in the treatment of head and neck cancer with concurrent radiochemotherapy in vivo. Clin Cancer Res. 2007;13:2512–18. doi: 10.1158/1078-0432.CCR-06-2582. [DOI] [PubMed] [Google Scholar]
  • 30.Feng FY, Varambally S, Tomlins SA, et al. Role of epidermal growth factor receptor degradation in gemcitabine-mediated cytotoxicity. Oncogene. 2007;26:3431–9. doi: 10.1038/sj.onc.1210129. [DOI] [PubMed] [Google Scholar]
  • 31.Kesarwala AH, Samrakandi MM, Piwnica-Worms D. Proteasome inhibition blocks ligand-induced dynamic processing and internalization of epidermal growth factor receptor via altered receptor ubiquitination and phosphorylation. Cancer Res. 2009;69:976–83. doi: 10.1158/0008-5472.CAN-08-2938. [DOI] [PubMed] [Google Scholar]
  • 32.Chen Z, Ricker JL, Malhotra PS, Nottingham L. Differential bortezomib sensitivity in head and neck cancer lines corresponds to proteasome, nuclear factor-kappaB and activator protein-1 related mechanisms. Mol Cancer Ther. 2008;7:1949–60. doi: 10.1158/1535-7163.MCT-07-2046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Sloss CM, Wang F, Liu R, et al. Proteasome inhibition activates epidermal growth factor receptor (EGFR) and EGFR-independent mitogenic kinase signaling pathways in pancreatic cancer cells. Clin Cancer Res. 2008;14:5116–23. doi: 10.1158/1078-0432.CCR-07-4506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Li C, Zang Y, Sen M, Leeman-Neill RJ, et al. Bortezomib up-regulates activated signal transducer and activator of transcription-3 and synergizes with inhibitors of signal transducer and activator of transcription-3 to promote head and neck squamous cell carcinoma cell death. Mol Cancer Ther. 2009;8:2211–20. doi: 10.1158/1535-7163.MCT-09-0327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Pajonk F, Grumann T, McBride WH. The proteasome inhibitor MG-132 protects hypoxic SiHa cervical carcinoma cells after cyclic hypoxia/reoxygenation from ionizing radiation. Neoplasia. 2006;8:1037–41. doi: 10.1593/neo.06634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Byers LA, Holsinger FC, Kies MS, et al. Serum signature of hypoxia-regulated factors is associated with progression after induction therapy in head and neck squamous cell cancer. Mol Cancer Ther. 2010;9:1755–63. doi: 10.1158/1535-7163.MCT-09-1047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Chen Z, Malhotra PS, Thomas GR, et al. Expression of proinflammatory and proangiogenic cytokines in patients with head and neck cancer. Clin Cancer Res. 1999;5:1369–79. [PubMed] [Google Scholar]
  • 38.Dong G, Chen Z, Li ZY, Yeh NT, Bancroft CC, Van Waes C. Hepatocyte growth factor/scatter factor-induced activation of MEK and PI3K signal pathways contributes to expression of proangiogenic cytokines interleukin-8 and vascular endothelial growth factor in head and neck squamous cell carcinoma. Cancer Res. 2001;61:5911–8. [PubMed] [Google Scholar]
  • 39.Chung CH, Aulino J, Muldowney NJ, et al. Nuclear factor-kappa B pathway and response in a phase II trial of bortezomib and docetaxel in patients with recurrent and/or metastatic head and neck squamous cell carcinoma. Ann Oncol. 2010;21:864–70. doi: 10.1093/annonc/mdp390. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Gilbert J, Aulino J, Muldowney NJ, et al. Phase II randomized trial of bortezomib (B) plus irinotecan (I) or B with addition of I at progression in recurrent (R) or metastatic (M) squamous cell carcinoma of the head and neck (SCCHN) (E1304): A trial of the Eastern Cooperative Oncology Group. J Clin Oncol. 2009;27(suppl):15s. abstr 6020. [Google Scholar]
  • 41.Axelrod RS, et al. Mature results of a phase I trial of bortezomib, cisplatin, and radiotherapy for advanced or recurrent head and neck cancer. J Clin Oncol. 2010;28(suppl):15s. abstr 5560. [Google Scholar]
  • 42.Ahsan A, Hiniker SM, Ramanand SG, et al. Role of Epidermal Growth Factor Receptor Degradation in Cisplatin-Induced Cytotoxicity in Head and Neck Cancer. Cancer Res. 2010;70:2862–2869. doi: 10.1158/0008-5472.CAN-09-4294. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

1

RESOURCES