Purpose: Anemia is associated with poor tumor control. It was previously observed that accelerated radiotherapy combined with carbogen breathing and nicotinamide (ARCON) can correct this adverse outcome in patients with head and neck cancer. The purpose of this study was to validate this observation based on data from a randomized trial.

Experimental Design: Of 345 patients with cT2-4 laryngeal cancer, 174 were randomly assigned to accelerated radiotherapy and 171 to ARCON. Hemoglobin levels, measured before treatment, were defined as low when <7.5 mmol/L for women and <8.5 mmol/L for men. The hypoxia marker pimonidazole was used to assess the oxygenation status in tumor biopsies. Data were analyzed 2 years after inclusion of the last patient.

Results: Pretreatment hemoglobin levels were available and below normal in 27 of 173 (16%) accelerated radiotherapy and 27 of 167 (16%) ARCON patients. In patients with normal pretreatment, hemoglobin levels treatment with ARCON had no significant effect on 5-year loco-regional control (LRC, 79% versus 75%; P = 0.44) and disease-free survival (DFS, 75% vs. 70%; P = 0.46) compared with accelerated radiotherapy. However, in patients with low pretreatment, hemoglobin levels ARCON significantly improved 5-year LRC (79% vs. 53%; P = 0.03) and DFS (68% vs. 45%; P = 0.04). In multivariate analysis including other prognostic factors, pretreatment hemoglobin remained prognostic for LRC and DFS in the accelerated radiotherapy treatment arm. No correlation between pretreatment hemoglobin levels and pimonidazole uptake was observed.

Conclusion: Results from the randomized phase III trial support previous observations that ARCON has the potential to correct the poor outcome of cancer patients with anemia (ClinicalTrials.gov number, NCT00147732). Clin Cancer Res; 20(5); 1345–54. ©2014 AACR.

Translational Relevance

Anemia is a strong prognostic indicator of poor disease control and survival in patients presenting with head and neck cancer. Randomized trials failed to demonstrate outcome improvement with transfusions, and erythropoietin was even counterproductive. In contrast, elimination of the adverse impact of anemia was observed in patients with head and neck cancer participating in a nonrandomized phase II trial when accelerated radiotherapy was combined with carbogen breathing and nicotinamide (ARCON), a hypoxia-modifying regimen. Data from the current phase III trial demonstrate that the poor recurrence-free survival, observed in patients with locally advanced laryngeal cancers presenting with anemia and treated with accelerated radiotherapy, can be corrected by an ARCON regimen. Shorter oxygen diffusion distances in the tumor, higher levels of free oxygen in plasma, and reduced blood viscosity can explain the effect of ARCON in patients with anemia. The potential of ARCON should be further explored in a prospective randomized trial with primary focus on patients with anemia.

Up to 40% of patients with solid tumors undergoing radiotherapy have anemia at presentation (1). A wealth of data indicates that low pretreatment hemoglobin levels are a strong prognostic indicator of poor disease control and survival (2–4). To overcome the adverse impact of low pretreatment hemoglobin levels, correction strategies have been applied, such as administering erythropoietin and packed cell transfusions. However, randomized trials failed to demonstrate the effectiveness of these approaches in patients with head and neck or breast cancer, and erythropoietin was even counterproductive in some studies (3, 5–7).

Disappearance of the adverse impact of anemia was observed in a nonrandomized phase II trial when accelerated radiotherapy was combined with carbogen breathing and nicotinamide (ARCON; ref. 8). This strategy counteracts tumor cell repopulation and hypoxic radioresistance (9). Carbogen breathing (98% O2 + 2% CO2) is used to decrease diffusion-limited hypoxia whereas nicotinamide, a vasoactive agent, is added to limit hypoxia caused by intermittent reduction of blood flow (9). The same regimen has subsequently been studied in a randomized phase III trial, comparing ARCON for locally advanced laryngeal cancers. Although local control is not improved, it was demonstrated that ARCON results in a benefit of 8% in regional control and excellent larynx preservation rates without increase of toxicity (10, 11). Translational research using the hypoxia marker pimonidazole demonstrated that proper patient selection based on tumor biology is the key to the success of this approach.

Based on the results of the preceding phase II study the hypothesis was generated that ARCON can abolish the adverse impact of anemia (8). The purpose of this study is to find support for this observation in the data from the randomized phase III study. This article reports on the impact of ARCON on the prognosis of a cohort of patients with laryngeal cancer presenting with pretreatment anemia.

Study design and eligibility

This was an open-label, randomized phase III trial comparing accelerated radiotherapy with ARCON in patients with cT2-4 laryngeal cancer. The trial (ClinicalTrials.gov NCT00147732) was conducted under the auspices of the Dutch Head and Neck Cancer Group in 7 centers in the Netherlands and in the United Kingdom (Supplementary Table S1). Eligibility criteria are provided in the Supplementary Table S2.

Approval for the study was obtained from the Radboud University Nijmegen Medical Centre Research Ethics Committee with ratification from each center. Written informed consent was obtained before randomization.

Randomization

Patients were centrally randomized by phone at the Integraal Kankercentrum Oost trials office. Treatment arm assignments (accelerated radiotherapy vs. ARCON) were stratified for tumor site (glottic vs. supraglottic) and institution. A dynamic allocation method was used to avoid imbalance of treatment assignment within an institution. Randomization took place after all study investigations and no longer than 4 weeks before the anticipated start of treatment.

Procedure

A radiation dose of 44 Gy in 22 daily fractions of 2 Gy was prescribed to the primary tumor and neck nodes followed by a boost dose of 24 Gy in twice daily fractions of 2 Gy to the primary tumor and involved lymph nodes. Because a decrease in the radiation tolerance was observed for cartilage and spinal cord in earlier studies with hypoxic sensitization, the total dose to the arytenoid cartilage and the spinal cord in the ARCON arm was limited to 64 and 40 Gy, respectively (12, 13).

Patients allocated to the ARCON arm received carbogen (98% O2 + 2% CO2, 4 minutes before and during radiotherapy) and nicotinamide (60 mg/kg, 1–1.5 hours before fractions) concurrently with radiotherapy. Details of the procedure are described previously (10, 11).

All patients participating in this study were treated in academic hospitals with accreditation in head and neck oncology by the Dutch Cooperative Head and Neck Oncology Group and institution-wide quality assurance programs.

Hypoxia marker analysis

After additional informed consent, patients received pimonidazole (Hypoxyprobe-1; Natural Pharmacia International) intravenously (500 mg/m2) 2 hours before biopsy taking. Biopsies were snap frozen in liquid nitrogen, immunohistochemically stained, and semi-automatically analyzed (14). Of each biopsy, one complete section was analyzed for the hypoxic fraction, that is, the tumor area positive for pimonidazole relative to the total tumor area.

Monitoring during treatment and follow-up evaluations

Before and weekly during treatment hemoglobin, hematocrit, creatinine, and urea levels were obtained. Normal hemoglobin levels were defined as 7.5 to 10 mmol/L for women and 8.5 to 11 mmol/L for men (women, 12–16 g/dL; men, 13.6–17.7 g/dL). Follow-up visits took place every 2, 3, and 4 months during the first, second, and third year, respectively, then every 6 months for another 2 years. The larynx was assessed by fiberoptic or indirect laryngoscopy. Regional control was assessed by palpation of the neck. When tumor recurrence was suspected, imaging (computed tomography scan or MRI) was performed to document the extent of the disease and biopsies were taken for pathologic confirmation.

Endpoints and statistics

Survival endpoints used were loco-regional control (LRC), metastasis-free survival (MFS), disease-free survival (DFS), and overall survival (OS) at 5 years from randomization. All intervals were calculated from the date of randomization and censored after 60 months or at last follow-up. LRC was defined as freedom of first recurrence at the primary tumor site and complete and persistent disappearance of the pathologic lymph nodes after radiotherapy, not including salvage procedures. MFS was defined as the time from randomization to distant metastasis. DFS was defined as the time to local or regional recurrence, or distant metastasis. OS was defined as time to death.

The primary endpoint of the randomized trial was local control. In the accelerated radiotherapy arm, a local control rate of 60% at 2 years after completion of radiotherapy was expected. An improvement by 15%, resulting in a local control rate of 75% at 2 years, was assumed for the ARCON arm. To detect this difference of 15% with a significance level of 0.05 and a power of 0.80 (2-sided log-rank test), 156 patients were required in each treatment group. To account for a drop out percentage of 10%, an additional 16 patients were added for each group.

Statistical analyses were performed using SPSS 19.0.0. Mann–Whitney U and χ2 tests were used at a 2-sided significance level of 0.05. Endpoints were evaluated by the Kaplan–Meier method based on intent-to-treat policy and compared with log-rank testing. A multivariate Cox proportional hazards analysis, with stepwise backward elimination of variables at P > 0.1, was used for both patient groups and included N-classification (N+ vs. N0), performance status (0 vs. 1), and treatment arm (ARCON vs. accelerated radiotherapy). An interaction between treatment arm and pretreatment hemoglobin level was assessed using Cox regression.

Role of the funding source

The study sponsors had no role in the study design, data collection, data analysis, data interpretation, or writing of the report. The corresponding author had full access to all of the data and the final responsibility to submit for publication.

Patient characteristics and protocol compliance

Between April 2001 and February 2008, 345 patients were randomized to either accelerated radiotherapy (n = 174) or ARCON (n = 171; Fig. 1). The median follow-up time was 44 (range 18–103), 55, and 60 months for the whole group and for patients still alive receiving accelerated radiotherapy and ARCON, respectively. Patient demographics and clinical tumor characteristics were well balanced without significant differences within the whole study population and the group of patients presenting with anemia (Table 1). Patients presenting with anemia were older (P = 0.01), had a poorer performance status (P < 0.01), more N2 stages (P < 0.01), and higher stage grouping (P = 0.02) compared with the whole group of patients. Compliance to radiotherapy, carbogen breathing, and nicotinamide intake was high and comparable between both groups. Detailed information is listed in the Supplementary Table S3.

Figure 1.

CONSORT diagram.

Figure 1.

CONSORT diagram.

Close modal
Table 1.

Demographics and clinical characteristics for all patients (N = 345) and patients presenting with anemia (N = 54)

All patientsPatients with anemia
CharacteristicsAccelerated radiotherapy (N = 174)ARCON (N = 171)PAccelerated radiotherapy (N = 27)ARCON (N = 27)P
Age, y   0.56a   0.48a 
 Median 60 61  63 64  
 Range 38–88 41–84  48–76 43–80  
Sex, number (%)   0.16b   0.10b 
 Male 136 (78) 142 (83)  22 (81) 26 (96)  
 Female 38 (22) 29 (17)  5 (19) 1 (4)  
Performance status, number (%)   0.54b   0.43b 
 0 140 (80) 137 (81)  16 (59) 17 (63)  
 1 34 (20) 34 (19)  11 (41) 10 (37)  
Site of the primary tumor, number (%)   0.49b   0.50b 
 Supraglottic 100 (57) 97 (56)  17 (63) 17 (63)  
 Glottic 74 (43) 74 (44)  10 (37) 10 (37)  
AJCC stage groupings, number (%)   0.35b   0.07b 
 Stage II 53 (31) 46 (27)  9 (33) 4 (15)  
 Stage III 65 (37) 77 (45)  4 (15) 11 (41)  
 Stage IV 56 (32) 48 (28)  14 (52) 12 (44)  
T-stage, number (%)   0.20b   0.12b 
 T2 67 (38) 55 (32)  13 (48) 6 (22)  
 T3 80 (46) 95 (56)  11 (41) 15 (56)  
 T4 27 (16) 21 (12)  3 (11) 6 (22)  
N-stage, number (%)   0.58b   0.29b 
 N0 117 (67) 116 (68)  15 (56) 17 (63)  
 N1 20 (12) 23 (13)  0 (0) 2 (7)  
 N2a 4 (2) 7 (4)  2 (7) 3 (11)  
 N2b 10 (6) 5 (3)  2 (7) 0 (0)  
 N2c 23 (13) 20 (12)  8 (30) 5 (19)  
 N3 0 (0) 0 (0)  0 (0) 0 (0)  
Pretreatment hemoglobin, number (%)   0.49b    
 Normal 146/173 (84) 140/167 (84)  0/27 (0) 0/27 (0)  
 Low 27/173 (16) 27/167 (16)  27/27 (100) 27/27 (100)  
 Male, mmol/L       
  Median 9.2 9.1  8.0 8.1  
  Range 6.1–14.5 6.1–13.4  6.1–8.4 6.1–8.4  
 Female, mmol/L       
  Median 8.4 8.3  7.3 7.2  
  Range 5.8–11.0 7.2–9.9  5.8–7.4 7.2–7.2  
All patientsPatients with anemia
CharacteristicsAccelerated radiotherapy (N = 174)ARCON (N = 171)PAccelerated radiotherapy (N = 27)ARCON (N = 27)P
Age, y   0.56a   0.48a 
 Median 60 61  63 64  
 Range 38–88 41–84  48–76 43–80  
Sex, number (%)   0.16b   0.10b 
 Male 136 (78) 142 (83)  22 (81) 26 (96)  
 Female 38 (22) 29 (17)  5 (19) 1 (4)  
Performance status, number (%)   0.54b   0.43b 
 0 140 (80) 137 (81)  16 (59) 17 (63)  
 1 34 (20) 34 (19)  11 (41) 10 (37)  
Site of the primary tumor, number (%)   0.49b   0.50b 
 Supraglottic 100 (57) 97 (56)  17 (63) 17 (63)  
 Glottic 74 (43) 74 (44)  10 (37) 10 (37)  
AJCC stage groupings, number (%)   0.35b   0.07b 
 Stage II 53 (31) 46 (27)  9 (33) 4 (15)  
 Stage III 65 (37) 77 (45)  4 (15) 11 (41)  
 Stage IV 56 (32) 48 (28)  14 (52) 12 (44)  
T-stage, number (%)   0.20b   0.12b 
 T2 67 (38) 55 (32)  13 (48) 6 (22)  
 T3 80 (46) 95 (56)  11 (41) 15 (56)  
 T4 27 (16) 21 (12)  3 (11) 6 (22)  
N-stage, number (%)   0.58b   0.29b 
 N0 117 (67) 116 (68)  15 (56) 17 (63)  
 N1 20 (12) 23 (13)  0 (0) 2 (7)  
 N2a 4 (2) 7 (4)  2 (7) 3 (11)  
 N2b 10 (6) 5 (3)  2 (7) 0 (0)  
 N2c 23 (13) 20 (12)  8 (30) 5 (19)  
 N3 0 (0) 0 (0)  0 (0) 0 (0)  
Pretreatment hemoglobin, number (%)   0.49b    
 Normal 146/173 (84) 140/167 (84)  0/27 (0) 0/27 (0)  
 Low 27/173 (16) 27/167 (16)  27/27 (100) 27/27 (100)  
 Male, mmol/L       
  Median 9.2 9.1  8.0 8.1  
  Range 6.1–14.5 6.1–13.4  6.1–8.4 6.1–8.4  
 Female, mmol/L       
  Median 8.4 8.3  7.3 7.2  
  Range 5.8–11.0 7.2–9.9  5.8–7.4 7.2–7.2  

aMann–Whitney U test.

bχ2 test.

Hemoglobin levels

Pretreatment hemoglobin levels were available and below normal in 27 of 173 (16%) accelerated radiotherapy and 27 of 167 (16%) ARCON patients (Table 1). Four of them (accelerated radiotherapy, n = 3; ARCON, n = 1) received a transfusion, given as 2 units of packed red blood cells. A correlation was observed between low hemoglobin at presentation and lower performance status (P < 0.01), and a trend was observed between low hemoglobin and higher N-status (P = 0.06).

LRC and survival

In patients with normal pretreatment hemoglobin levels treatment with ARCON had no significant effect on 5-year LRC (79% vs. 75%, P = 0.44) and DFS (75% vs. 70%, P = 0.46) compared with accelerated radiotherapy. However, in patients with low pretreatment hemoglobin levels ARCON treatment significantly improved 5-year LRC (79% vs. 53%, P = 0.03) and DFS (68% vs. 45%, P = 0.04; Fig. 2A and B). No significant benefit of ARCON was observed for MFS (normal hemoglobin: ARCON vs. accelerated radiotherapy: 91% vs. 89%, P = 0.56; low hemoglobin: ARCON vs. accelerated radiotherapy 87% vs. 69%, P = 0.10; Fig. 3A). Patients presenting with low hemoglobin levels had a worse 5-year OS regardless of the treatment regimen (normal hemoglobin: ARCON vs. accelerated radiotherapy: 65% vs. 65%, P = 0.93; low hemoglobin: ARCON vs. accelerated radiotherapy 46% vs. 28%, P = 0.97; Fig. 3B).

Figure 2.

A and B, LRC (A) and DFS (B) for patients with low and normal pretreatment hemoglobin (Hb) levels, treated by accelerated radiotherapy (AR) or ARCON.

Figure 2.

A and B, LRC (A) and DFS (B) for patients with low and normal pretreatment hemoglobin (Hb) levels, treated by accelerated radiotherapy (AR) or ARCON.

Close modal
Figure 3.

A and B, MFS (A) and OS (B) for patients with low and normal pretreatment hemoglobin (Hb) levels, treated by accelerated radiotherapy (AR) or ARCON.

Figure 3.

A and B, MFS (A) and OS (B) for patients with low and normal pretreatment hemoglobin (Hb) levels, treated by accelerated radiotherapy (AR) or ARCON.

Close modal

Prognostic factors for tumor control

The impact of various common prognostic factors and hemoglobin levels on LRC, MFS, DFS, and OS is summarized in Table 2. On multivariate analysis after correction for N-stage and hemoglobin level, ARCON treatment remained an independent prognostic factor for LRC (P = 0.04) and DFS (P = 0.09) in patients with anemia only. A significant interaction between treatment effect and hemoglobin level was found for LRC (P = 0.02) and DFS (P = 0.05).

Table 2.

Univariate and multivariate analysis per hemoglobin levels

Normal hemoglobin (N = 286)Low hemoglobin (N = 54)Normal hemoglobin (N = 286)Low hemoglobin (N = 54)
HR (95% CI)PHR (95% CI)PHR (95% CI)PHR (95% CI)P
  LRC DFS 
Univariate parameter 
 Age >60 vs. ≤60 0.94 (0.57–1.55) 0.81 0.47 (0.18–1.22) 0.12 0.90 (0.57–1.43) 0.65 0.55 (0.23–1.30) 0.17 
 Sex Female vs. male 0.30 (0.12–0.76) 0.01 1.31 (0.30–5.77) 0.72 0.42 (0.20–0.87) 0.02 1.06 (0.25–4.59) 0.94 
 Site Supraglottic vs. glottic 0.81 (0.49–1.33) 0.40 1.45 (0.51–4.13) 0.48 1.02 (0.64–1.62) 0.93 1.59 (0.61–4.09) 0.34 
 T-classification T2 vs. T3-4 0.85 (0.52–1.42) 0.54 0.99 (0.37–2.67) 0.98 0.93 (0.58–1.50) 0.77 0.92 (0.38–2.22) 0.85 
 N-classification N+ vs. N0 1.85 (1.12–3.06) 0.02 2.91 (1.10–7.68) 0.03 2.20 (1.39–3.48) <0.01 2.98 (1.25–7.12) 0.01 
 Performance status 0 vs. 1 0.58 (0.25–1.34) 0.20 1.68 (0.65–4.35) 0.29 0.86 (0.44–1.69) 0.67 1.39 (0.58–3.30) 0.46 
 Treatment ARCON vs. accelerated radiotherapy 0.82 (0.49–1.35) 0.43 0.31 (0.11–0.89) 0.03 0.84 (0.53–1.33) 0.45 0.44 (0.18–1.05) 0.06 
Multivariate analysis 
 N-classification N+ vs. N0 1.86 (1.12–3.06) 0.02 3.17 (1.19–8.42) 0.02 2.20 (1.39–3.48) <0.01 3.56 (1.49–8.50) <0.01 
 Performance status 0 vs. 1 0.60 (0.26–1.39) 0.23 1.91 (0.72–5.06) 0.19 0.91 (0.46–1.77) 0.77 1.44 (0.61–3.42) 0.41 
 Treatment ARCON vs. accelerated radiotherapy 0.80 (0.48–1.33) 0.39 0.33 (0.11–0.93) 0.04 0.82 (0.51–1.30) 0.40 0.47 (0.20–1.13) 0.09 
  MFS OS 
Univariate parameter 
 Age >60 vs. ≤60 0.63 (0.28–1.44) 0.28 0.61 (0.17–2.16) 0.44 1.54 (1.03–2.30) 0.04 1.08 (0.48–2.43) 0.86 
 Sex Female vs. male 0.70 (0.24–2.04) 0.51 1.07 (0.14–8.50) 0.95 0.74 (0.44–1.24) 0.25 1.84 (0.54–6.20) 0.33 
 Site Supraglottic vs. glottic 1.95 (0.81–4.71) 0.14 1.26 (0.32–4.85) 0.74 1.18 (0.79–1.76) 0.41 0.82 (0.38–1.76) 0.61 
 T-classification T2 vs. T3-4 1.47 (0.61–3.54) 0.40 0.86 (0.24–3.07) 0.82 1.29 (0.85–1.97) 0.23 1.24 (0.57–2.68) 0.59 
 N-classification N+ vs. N0 4.80 (2.06–11.22) <0.01 2.82 (0.79–10.06) 0.11 1.66 (1.12–2.48) 0.01 1.81 (0.87–3.77) 0.11 
 Performance status 0 vs. 1 1.51 (0.56–4.05) 0.41 1.78 (0.51–6.14) 0.37 1.99 (1.25–3.16) <0.01 1.44 (0.67–3.09) 0.35 
 Treatment ARCON vs. accelerated radiotherapy 0.78 (0.34–1.76) 0.55 0.35 (0.09–1.33) 0.12 1.02 (.69–1.51) 0.93 0.99 (0.47–2.05) 0.97 
Multivariate analysis 
 N-classification N+ vs. N0 4.80 (2.06–11.22) <0.01 3.31 (0.95–11.5) 0.06 1.17 (1.03–1.33) 0.02 1.22 (0.99–1.50) 0.06 
 Performance status 0 vs. 1 1.78 (0.66–4.78) 0.26 1.63 (0.49–5.39) 0.43 2.04 (1.29–3.25) <0.01 1.56 (0.72–3.37) 0.26 
 Treatment ARCON vs. accelerated radiotherapy 0.74 (0.33–1.66) 0.46 0.41 (0.11–1.55) 0.19 1.05 (0.70–1.55) 0.83 1.18 (0.56–2.47) 0.67 
Normal hemoglobin (N = 286)Low hemoglobin (N = 54)Normal hemoglobin (N = 286)Low hemoglobin (N = 54)
HR (95% CI)PHR (95% CI)PHR (95% CI)PHR (95% CI)P
  LRC DFS 
Univariate parameter 
 Age >60 vs. ≤60 0.94 (0.57–1.55) 0.81 0.47 (0.18–1.22) 0.12 0.90 (0.57–1.43) 0.65 0.55 (0.23–1.30) 0.17 
 Sex Female vs. male 0.30 (0.12–0.76) 0.01 1.31 (0.30–5.77) 0.72 0.42 (0.20–0.87) 0.02 1.06 (0.25–4.59) 0.94 
 Site Supraglottic vs. glottic 0.81 (0.49–1.33) 0.40 1.45 (0.51–4.13) 0.48 1.02 (0.64–1.62) 0.93 1.59 (0.61–4.09) 0.34 
 T-classification T2 vs. T3-4 0.85 (0.52–1.42) 0.54 0.99 (0.37–2.67) 0.98 0.93 (0.58–1.50) 0.77 0.92 (0.38–2.22) 0.85 
 N-classification N+ vs. N0 1.85 (1.12–3.06) 0.02 2.91 (1.10–7.68) 0.03 2.20 (1.39–3.48) <0.01 2.98 (1.25–7.12) 0.01 
 Performance status 0 vs. 1 0.58 (0.25–1.34) 0.20 1.68 (0.65–4.35) 0.29 0.86 (0.44–1.69) 0.67 1.39 (0.58–3.30) 0.46 
 Treatment ARCON vs. accelerated radiotherapy 0.82 (0.49–1.35) 0.43 0.31 (0.11–0.89) 0.03 0.84 (0.53–1.33) 0.45 0.44 (0.18–1.05) 0.06 
Multivariate analysis 
 N-classification N+ vs. N0 1.86 (1.12–3.06) 0.02 3.17 (1.19–8.42) 0.02 2.20 (1.39–3.48) <0.01 3.56 (1.49–8.50) <0.01 
 Performance status 0 vs. 1 0.60 (0.26–1.39) 0.23 1.91 (0.72–5.06) 0.19 0.91 (0.46–1.77) 0.77 1.44 (0.61–3.42) 0.41 
 Treatment ARCON vs. accelerated radiotherapy 0.80 (0.48–1.33) 0.39 0.33 (0.11–0.93) 0.04 0.82 (0.51–1.30) 0.40 0.47 (0.20–1.13) 0.09 
  MFS OS 
Univariate parameter 
 Age >60 vs. ≤60 0.63 (0.28–1.44) 0.28 0.61 (0.17–2.16) 0.44 1.54 (1.03–2.30) 0.04 1.08 (0.48–2.43) 0.86 
 Sex Female vs. male 0.70 (0.24–2.04) 0.51 1.07 (0.14–8.50) 0.95 0.74 (0.44–1.24) 0.25 1.84 (0.54–6.20) 0.33 
 Site Supraglottic vs. glottic 1.95 (0.81–4.71) 0.14 1.26 (0.32–4.85) 0.74 1.18 (0.79–1.76) 0.41 0.82 (0.38–1.76) 0.61 
 T-classification T2 vs. T3-4 1.47 (0.61–3.54) 0.40 0.86 (0.24–3.07) 0.82 1.29 (0.85–1.97) 0.23 1.24 (0.57–2.68) 0.59 
 N-classification N+ vs. N0 4.80 (2.06–11.22) <0.01 2.82 (0.79–10.06) 0.11 1.66 (1.12–2.48) 0.01 1.81 (0.87–3.77) 0.11 
 Performance status 0 vs. 1 1.51 (0.56–4.05) 0.41 1.78 (0.51–6.14) 0.37 1.99 (1.25–3.16) <0.01 1.44 (0.67–3.09) 0.35 
 Treatment ARCON vs. accelerated radiotherapy 0.78 (0.34–1.76) 0.55 0.35 (0.09–1.33) 0.12 1.02 (.69–1.51) 0.93 0.99 (0.47–2.05) 0.97 
Multivariate analysis 
 N-classification N+ vs. N0 4.80 (2.06–11.22) <0.01 3.31 (0.95–11.5) 0.06 1.17 (1.03–1.33) 0.02 1.22 (0.99–1.50) 0.06 
 Performance status 0 vs. 1 1.78 (0.66–4.78) 0.26 1.63 (0.49–5.39) 0.43 2.04 (1.29–3.25) <0.01 1.56 (0.72–3.37) 0.26 
 Treatment ARCON vs. accelerated radiotherapy 0.74 (0.33–1.66) 0.46 0.41 (0.11–1.55) 0.19 1.05 (0.70–1.55) 0.83 1.18 (0.56–2.47) 0.67 

Hypoxia marker study

Tumor biopsies of 79 patients were available for pimonidazole staining. Characteristics were well balanced between patients receiving accelerated radiotherapy and ARCON (Table 3). However, the group of patients participating in the hypoxia marker study differed in some aspects from the entire study population: patients involved were more frequently female (P < 0.01) and presented with higher tumor stage (P < 0.01).

Table 3.

Demographics and clinical characteristics of patients (N = 79) participating in the hypoxia marker study

CharacteristicsAccelerated radiotherapy (N = 38)ARCON (N = 41)P
Age, y   0.41a 
 Median 61 61  
 Range 38–81 46–83  
Sex, number (%)   0.99b 
 Male 26 (68) 28 (68)  
 Female 12 (32) 13 (32)  
Performance status, number (%)   0.71b 
 0 29 (76) 34 (83)  
 1 9 (24) 7 (17)  
Site of the primary tumor, number (%)   0.99b 
 Supraglottic 25 (66) 27 (66)  
 Glottic 13 (34) 14 (34)  
T-stage, number (%)   0.16b 
 T2 11 (29) 6 (15)  
 T3 20 (53) 30 (73)  
 T4 7 (18) 5 (12)  
N-stage, number (%)   0.24b 
 N0 17 (45) 20 (49)  
 N1 10 (26) 5 (12)  
 N2a 3 (8) 1 (2)  
 N2b 1 (3) 4 (10)  
 N2c 7 (18) 11 (27)  
 N3 0 (0) 0 (0)  
Pretreatment hemoglobin, number (%)   0.27b 
 Normal 31 (82) 37 (90)  
 Low 7 (18) 4 (10)  
 Male, mmol/L    
  Median 9.0 9.1  
  Range 7.7–10.3 6.8–10.4  
 Female, mmol/L    
  Median 7.9 8.2  
  Range 7.4–9.3 7.5–9.9  
CharacteristicsAccelerated radiotherapy (N = 38)ARCON (N = 41)P
Age, y   0.41a 
 Median 61 61  
 Range 38–81 46–83  
Sex, number (%)   0.99b 
 Male 26 (68) 28 (68)  
 Female 12 (32) 13 (32)  
Performance status, number (%)   0.71b 
 0 29 (76) 34 (83)  
 1 9 (24) 7 (17)  
Site of the primary tumor, number (%)   0.99b 
 Supraglottic 25 (66) 27 (66)  
 Glottic 13 (34) 14 (34)  
T-stage, number (%)   0.16b 
 T2 11 (29) 6 (15)  
 T3 20 (53) 30 (73)  
 T4 7 (18) 5 (12)  
N-stage, number (%)   0.24b 
 N0 17 (45) 20 (49)  
 N1 10 (26) 5 (12)  
 N2a 3 (8) 1 (2)  
 N2b 1 (3) 4 (10)  
 N2c 7 (18) 11 (27)  
 N3 0 (0) 0 (0)  
Pretreatment hemoglobin, number (%)   0.27b 
 Normal 31 (82) 37 (90)  
 Low 7 (18) 4 (10)  
 Male, mmol/L    
  Median 9.0 9.1  
  Range 7.7–10.3 6.8–10.4  
 Female, mmol/L    
  Median 7.9 8.2  
  Range 7.4–9.3 7.5–9.9  

aMann–Whitney U test.

bχ2 test.

The hypoxic fraction, as defined by pimonidazole staining, varied from 0% to 19.4% with a median value of 1.6%. Eleven of the 79 patients had hemoglobin levels below normal at diagnosis. No correlation was observed between pretreatment hemoglobin levels and hypoxic fraction (P = 0.11).

It has been demonstrated by numerous reports that anemia in patients with cancer of the cervix, head and neck, bladder, breast, and lung is associated with poor outcome (2–4). This is also found in the current study for patients with T2-4 laryngeal cancer treated with radiotherapy alone. However, this impaired outcome in patients with anemia is no longer observed when carbogen and nicotinamide are added to radiotherapy, supporting the results of a previous phase II ARCON trial in head and neck cancer (8).

Attempts have been made to improve outcome of cancer patients with anemia using erythropoietin or red blood cell transfusions. A Cochrane review based on 5 randomized controlled trials found strong indications that for patients with head and neck cancer, the addition of erythropoietin to radiotherapy negatively affects patient outcome in terms of loco-regional progression-free survival and OS (7). Suggested explanations for the lack of benefit of erythropoietin include presence of erythropoietin receptors on the tumor cell membranes stimulating tumor growth, and a decrease of tissue oxygenation because of increased viscosity when hemoglobin concentrations become too high (15–17). Patients with head and neck cancer with low hemoglobin levels treated in the DAHANCA 5 (radiotherapy and nimorazole vs. radiotherapy and placebo) and DAHANCA 7 (conventional radiotherapy vs. accelerated radiotherapy) studies were subrandomized to receive red blood cell transfusions before and during radiotherapy (3). Also with this approach, the increased hemoglobin level was not able to improve tumor control or survival. Stimulation of inflammatory and immunosuppressive pathways was proposed as a possible factor involved (18). Randomized trials exploring the role of chemotherapy in addition to radiotherapy for patients presenting with anemia are lacking. However, prospective and retrospective studies indicate that the poor prognosis of patients presenting with anemia cannot be improved by a chemoradiotherapy approach (19, 20).

The “reduced cord radius” model described by Hirst and colleagues could explain the success of ARCON in patients with anemia (21). The term “tumor cord” describes the functional unit of a blood vessel and its dependent tumor cell volume*. The model assumes that the ability of cancer cells to survive at a distance from blood vessels is dependent on the local supply and diffusion distance of oxygen and nutrients from each vessel. Histologic examination of tumors in animals exposed to low oxygen tension for several days has shown that the thickness of the tumor cords is less than in animals breathing normal air (22). It has been proposed that anemia causes a reduced cord radius by the same mechanism. Restoration of hemoglobin levels by blood transfusion in animals with anemia produced a markedly increased tumor radiosensitivity supposedly by improved oxygenation of tumor cells in the peripheral zones of the cords. However, this effect was only transient and was lost within 24 hours (23). Tumor cords that are chronically exposed to higher oxygen levels, will adapt and begin to proliferate more actively and will once again outgrow their oxygen supply. In contrast, daily carbogen breathing immediately followed by radiotherapy will not cause this adaptive response because the oxygenation increase is too short and only for the duration of the radiation treatment (10–15 minutes). The status quo of reduced cord radius and, consequently, shorter oxygen diffusion distances in combination with higher levels of free oxygen in plasma explains how ARCON can exploit adaptive mechanisms in patients with anemia. In addition, because the effect of carbogen relies on oxygen transportation by the plasma, the reduced blood viscosity and consequential increased flow through the tumor microvasculature will further benefit the patients with anemia. Finally, other compensatory mechanisms such as a shift in the oxygen–hemoglobin dissociation relationship may contribute as well.

*Although the typical corded structure is histologically recognizable in some tumors and tumor types, the cord radius model has a broader application. “Cord radius” is the equivalence of the more generally applicable and easier to recognize “distance from blood vessel to necrosis”. However, for readability we use the term “tumor cord” here.

Although the effect of hyperbaric oxygen in patients with head and neck cancer has recently been demonstrated in a systematic review and meta-analysis, prospective data testing hyperbaric oxygen in relation to anemia are lacking (24). However, a retrospective analysis of patients with carcinoma of the uterine cervix treated with radiotherapy in hyperbaric oxygen also revealed a marked improvement in local tumor control in patients with severe anemia before radiotherapy (25). Additional support comes from a preclinical study demonstrating that hyperbaric oxygen was successful in overcoming the increased radioresistance associated with anemia in mouse mammary adenocarcinomas (26). These observations are of interest because several mechanisms of action of ARCON discussed above also apply to hyperbaric oxygen.

Another issue is why patients with anemia have an inferior outcome compared with patients without anemia in the first place. It is generally assumed that this is primarily a consequence of the impaired tumor oxygenation resulting in a more aggressive and treatment resistant tumor phenotype (15, 27). Indeed, clinical and preclinical studies do provide evidence for a correlation between hemoglobin level and tumor hypoxia measured by polarographic pO2 electrodes (15, 16, 28, 29). In particular, this association is present in patients with severe anemia [hemoglobin < 11.0 g/dL (<6.9 mmol/L)], whereas it is much weaker in patients with mild anemia or low normal hemoglobin levels. One study did not demonstrate such correlation, but it should be noted that in that study there were hardly any patients with severe anemia (30). This was also the case in the current study: in the subgroup of patients, participating in the hypoxia marker side study, no correlation was found between tumor hypoxia as measured by pimonidazole binding and hemoglobin level but there was only one patient with a hemoglobin level below 11.0 g/dL. Only 79 of 345 patients took part in this side study. This is mainly because of a late amendment to the initial protocol and the limited number of participating centers to the side study. However, we argue that it is not the absolute amount of hypoxia but the distribution as function of distance to the vessels that is relevant for the response to ARCON.

Tumor hypoxia may be one reason for the poor prognosis of patients with anemia but most likely not the only one. A typical molecular feature of malignancies is the switch in tumor cell metabolism from oxidative to glycolytic pathways (31, 32). Anaerobic glycolysis provides cells with a growth advantage in the tumor microenvironment and promotes metastasis formation. With decreased blood viscosity and increased plasma flow in patients with anemic there will be greater availability of glucose, fueling the process of malignant progression. This can explain the observation of an association between anemia and the trend toward a higher N-stage (P = 0.06) in this study. Apart from a causative factor, anemia can also be an epiphenomenon of aggressive tumor behavior. Activation of the immune and inflammatory system by the malignant disease produces cytokines, including interferons, TNF, and interleukin-1 (33). These cytokines inhibit erythropoiesis, affect the life span of erythrocytes and impair iron metabolism.

Despite reduction of tumor recurrence, no benefit of ARCON was observed on OS in patients with anemia. The correlation between low hemoglobin levels and a poorer performance status (P < 0.01), observed in this study, suggests that associated comorbidity in patients with low hemoglobin levels at diagnosis may affect survival independent of tumor control. Supportive evidence for this comes from the GORTEC 94-01 trial comparing radiotherapy alone with concomitant radiochemotherapy in advanced-stage oropharynx carcinoma. This study showed that anemia was associated with a higher probability of death caused by intercurrent disease (20). In our cohort of patients presenting with anemia, approximately 40% exhibit significant comorbidity. It is obvious that the survival impact of comorbidity cannot be influenced by ARCON or any other cancer treatment (34).

In conclusion, the poor prognosis of patients with laryngeal cancer with pretreatment anemia is no longer observed when radiotherapy is combined with carbogen breathing and nicotinamide. This observation of improved outcome in patients with anemia supports an earlier proposed hypothesis (8). Reduced oxygen diffusion distances in the tumor and improved oxygen transportation by the plasma because of reduced blood viscosity can explain the effectiveness of ARCON in patients with anemia. The potential of ARCON should be further explored in a large prospective randomized trial with focus on patients presenting with anemia.

No potential conflicts of interest were disclosed.

Conception and design: C.H. Terhaard, J.H. Kaanders

Development of methodology: C.H. Terhaard, J. Bussink, J.H. Kaanders

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): G.O. Janssens, C.H. Terhaard, P.A. Doornaert, H.P. Bijl, P. van den Ende, A. Chin, R.P. Takes, R. de Bree, I.J. Hoogsteen, J.H. Kaanders

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): G.O. Janssens, S.E. Rademakers, I.J. Hoogsteen, P.N. Span, J.H. Kaanders

Writing, review, and/or revision of the manuscript: G.O. Janssens, S.E. Rademakers, C.H. Terhaard, P.A. Doornaert, H.P. Bijl, A. Chin, R.P. Takes, R. de Bree, J. Bussink, P.N. Span, J.H. Kaanders

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): G.O. Janssens, C.H. Terhaard, I.J. Hoogsteen

Study supervision: C.H. Terhaard, J.H. Kaanders

This work was supported by the Dutch Cancer Society (KWF) Research Fund No. CKTO-2000-09.

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

1.
Harrison
L
,
Shasha
D
,
Shiaova
L
,
White
C
,
Ramdeen
B
,
Portenoy
R
. 
Prevalence of anemia in cancer patients undergoing radiation therapy
.
Semin Oncol
2001
;
28
:
54
9
.
2.
Grau
C
,
Overgaard
J
. 
Significance of hemoglobin concentration for treatment outcome
. In:
Molls
M
,
Vaupel
P
, editors. 
Blood perfusion and microenvironment of human tumors
.
Berlin/Heidelberg/New York
:
Springer-Verlag
; 
1998
.
p.
101
12
.
3.
Hoff
CM
,
Lassen
P
,
Eriksen
JG
,
Hansen
HS
,
Specht
L
,
Overgaard
M
, et al
Does transfusion improve the outcome for HNSCC patients treated with radiotherapy?—Results from the randomized DAHANCA 5 and 7 trials
.
Acta Oncol
2011
;
50
:
1006
14
.
4.
Henke
M
,
Sindlinger
F
,
Ikenberg
H
,
Gerds
T
,
Schumacher
M
. 
Blood hemoglobin level and treatment outcome of early breast cancer
.
Strahlenther Onkol
2004
;
180
:
45
51
.
5.
Henke
M
,
Laszig
R
,
Rübe
C
,
Schäfer
U
,
Haase
KD
,
Schilcher
B
, et al
Erythropoietin to treat head and neck cancer patients with anemia undergoing radiotherapy: randomized, double-blind, placebo-controlled trial
.
Lancet
2003
;
362
:
1255
60
.
6.
Leyland-Jones
B
. 
Breast cancer trial with erythropoietin terminated unexpectedly
.
Lancet Oncol
2003
;
4
:
459
60
.
7.
Lambin
P
,
Ramaekers
BL
,
van Mastrigt
GA
,
van den Ende
P
,
de Jong
J
,
De Ruysscher
DK
, et al
Erythropoietin as an adjuvant treatment with (chemo) radiation therapy for head and neck cancer (review)
.
Cochrane Database Syst Rev
2009
;
3
:
1
30
.
8.
Hoogsteen
IJ
,
Pop
LA
,
Marres
HA
,
Merkx
MA
,
van den Hoogen
FJ
,
van der Kogel
AJ
, et al
Oxygen-modifying treatment with ARCON reduces the prognostic significance of hemoglobin in squamous cell carcinoma of the head and neck
.
Int J Radiat Biol Phys
2006
;
64
:
83
9
.
9.
Kaanders
JH
,
Bussink
J
,
van der Kogel
AJ
. 
ARCON: a novel biology-based approach in radiotherapy
.
Lancet Oncol
2002
;
3
:
728
37
.
10.
Janssens
GO
,
Terhaard
CH
,
Doornaert
PA
,
Bijl
HP
,
van den Ende
P
,
Chin
A
, et al
Acute toxicity profile and compliance to ARCON for clinical stage T2-4 laryngeal cancer: results of a phase III randomized trial
.
Int J Radiat Oncol Biol Phys
2012
;
82
:
532
8
.
11.
Janssens
GO
,
Rademakers
SE
,
Terhaard
CH
,
Doornaert
PA
,
Bijl
HP
,
van den Ende
P
, et al
Accelerated radiotherapy with carbogen and nicotinamide for laryngeal cancer: Results of a phase III randomized trial
.
J Clin Oncol
2012
;
30
:
1777
82
.
12.
Henk
JM
,
Kunkler
PB
,
Smith
CW
. 
Radiotherapy and hyperbaric oxygen in head and neck cancer. Final report of first controlled clinical trial
.
Lancet
1977
;
2
:
101
3
.
13.
Haustermans
K
,
van der Kogel
AJ
,
Vanacker
B
,
van der Schueren
E
. 
Influence of combined use of nicotinamide and carbogen on rat spinal cord radiation tolerance
.
Radiother Oncol
1994
;
31
:
123
8
.
14.
Hoogsteen
IJ
,
Lok
J
,
Marres
HA
,
Takes
RP
,
Rijken
PF
,
van der Kogel
AJ
, et al
Hypoxia in larynx carcinomas assessed by pimonidazole binding and the value of CA-IX and vascularity as surrogate markers of hypoxia
.
Eur J Cancer
2009
;
45
:
2906
14
.
15.
Vaupel
P
. 
Hypoxia and aggressive tumor phenotype: implications for therapy and prognosis
.
Oncologist
2008
;
13
:
21
6
.
16.
Becker
A
,
Stadler
P
,
Lavey
RS
,
Hänsgen
G
,
Kuhnt
T
,
Lautenschlaäger
C
, et al
Severe anemia is associated with poor tumor oxygenation in head and neck squamous cell carcinomas
.
Int J Radiat Oncol Biol Phys
2000
;
46
:
459
66
.
17.
Henke
M
,
Mattern
D
,
Pepe
M
,
Bezay
C
,
Weissenberger
C
,
Werner
M
, et al
Do erythropoietin receptors on cancer cells explain unexpected clinical findings?
J Clin Oncol
2006
;
24
:
4708
13
.
18.
Varlotto
J
,
Stevenson
MA
. 
Anemia, tumor hypoxemia, and the cancer patient
.
Int J Radiat Oncol Biol Phys
2005
;
63
:
25
36
.
19.
Prosnitz
RG
,
Yao
B
,
Farrell
CL
,
Clough
R
,
Brizel
DM
. 
Pretreatment anemia is correlated with reduced effectiveness of radiation and concurrent chemotherapy in advanced head and neck cancer
.
Int J Radiat Oncol Biol Phys
2005
;
61
:
1087
95
.
20.
Denis
F
,
Garaud
P
,
Bardet
E
,
Alfonsi
M
,
Sire
C
,
Germain
T
, et al
Final results of the 94-01 French head and neck oncology and radiotherapy group randomized trial comparing radiotherapy alone with concomitant radiochemotherapy in advanced-stage oropharynx carcinoma
.
J Clin Oncol
2004
;
22
:
69
76
.
21.
Hirst
DG
. 
Anemia: a problem or an opportunity in radiotherapy?
Int J Radiat Biol Phys
1986
;
12
:
2009
17
.
22.
Tannock
IF
. 
Effects of pO2 on cell proliferation kinetics
. In:
Bond
VP
,
Suit
HD
,
Marcial
V
, editors. 
Time and dose relationships in radiation biology as applied to radiotherapy
.
Upton, Brookhaven National Laboratory
; 
1970
.
p.
215
24
.
23.
Hirst
DG
,
Wood
PJ
. 
The adaptive response of mouse tumors to anemia and retransfusion
.
Int J Radiat Biol Rel Stud Phys Chem Med
1987
;
51
:
597
609
.
24.
Overgaard
J
. 
Hypoxic modification of radiotherapy in squamous cell carcinoma of the head and neck: a systematic review and meta-analysis
.
Radiother Oncol
2011
;
100
:
22
32
.
25.
Dische
S
,
Anderson
PJ
,
Sealy
R
,
Watson
ER
. 
Carcinoma of the cervix—anaemia, radiotherapy and hyperbaric oxygen
.
Br J Radiol
1983
;
56
:
251
5
.
26.
McCormack
M
,
Nias
AH
,
Smith
E
. 
Chronic anaemia, hyperbaric oxygen and tumour radiosensitivity
.
Br J Radiol
1990
;
63
:
752
9
.
27.
Harrison
L
,
Blackwell
K
. 
Hypoxia and anemia: factors in decreased sensitivity to radiation therapy and chemotherapy?
Oncologist
2004
;
9
:
31
40
.
28.
Kelleher
DK
,
Matthiensen Thews
O
,
Vaupel
P
. 
Blood flow, oxygenation, and bioenergetic status of tumors after erythropoietin treatment in normal and anemic rats
.
Cancer Res
1996
;
56
:
4728
34
.
29.
Nordsmark
M
,
Bentzen
SM
,
Rudat
V
,
Brizel
D
,
Lartigau
E
,
Stadler
P
, et al
Prognostic value of tumor oxygenation in 397 head and neck tumors after primary radiation therapy: an international multi-center study
.
Radiother Oncol
2005
;
77
:
18
24
.
30.
Rudat
V
,
Vanselow
B
,
Wollensack
P
,
Bettscheider
C
,
Osman-Ahmet
S
,
Eble
MJ
, et al
Repeatability and prognostic impact of the pretreatment pO2 histography in patients with advanced head and neck cancer
.
Radiother Oncol
2000
;
57
:
31
7
.
31.
Kim
JW
,
Dang
CV
. 
Cancer's molecular sweet tooth and the Warburg effect
.
Cancer Res
2006
;
66
:
8927
30
.
32.
Meijer
TW
,
Kaanders
JH
,
Span
PN
,
Bussink
J
. 
Targeting hypoxia, HIF-1 and tumor glucose metabolism to improve radiotherapy efficacy
.
Clin Cancer Res
2012
;
18
:
5585
94
.
33.
Bron
D
,
Meuleman
N
,
Mascaux
C
. 
Biological basis of anemia
.
Semin Oncol
2001
;
28
:
1
6
.
34.
Forastiere
AA
,
Goepfert
H
,
Maor
M
,
Pajak
TF
,
Weber
R
,
Morrison
W
, et al
Concurrent chemotherapy and radiotherapy for organ preservation in advanced laryngeal cancer
.
N Engl J Med
2003
;
349
:
2091
8
.