Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2009 Jun 1.
Published in final edited form as: Semin Oncol. 2008 Jun;35(3):236–250. doi: 10.1053/j.seminoncol.2008.03.003

NEW DEVELOPMENTS IN RADIATION THERAPY FOR HEAD AND NECK CANCER: INTENSITY MODULATED RADIATION THERAPY AND HYPOXIA TARGETING

Nancy Y Lee 1, Quynh-Thu Le 2,*
PMCID: PMC2494523  NIHMSID: NIHMS56290  PMID: 18544439

Abstract

Intensity modulated radiation therapy (IMRT) has revolutionized radiation treatment for head and neck cancers (HNC). When compared to the traditional techniques, IMRT has the unique ability to minimize the dose delivered to normal tissues without compromising tumor coverage. As a result, side effects from high dose radiation have decreased and patient quality of life has improved. In addition to toxicity reduction, excellent clinical outcomes have been reported for IMRT. The first part of this review will focus on clinical results of IMRT for HNC.

Tumor hypoxia or the condition of low oxygen is a key factor for tumor progression and treatment resistance. Hypoxia develops in solid tumors due to aberrant blood vessel formation, fluctuation in blood flow and increasing oxygen demands for tumor growth. Because hypoxic tumor cells are more resistant to ionizing radiation, hypoxia has been a focus of clinical research in radiation therapy for half a decade. Interest for targeting tumor hypoxia have waxed and waned as promising treatments emerged from the laboratory, only to fail in the clinics. However, with the development of new technologies, the prospect of targeting tumor hypoxia is more tangible. The second half of the review will focus on approaches for assessing tumor hypoxia and on the strategies for targeting this important microenvironmental factor in HNC.

INTENSITY MODULATED RADIOTHERAPY (IMRT)

Overview of IMRT

IMRT is a refinement of three-dimensional conformal radiotherapy (3DCRT). It utilizes a computerized treatment planning system along with a sophisticated delivery machineries to tailor the radiation dose to the tumor target.1,2 By subdividing a broad radiation beam into smaller pencil beams and by varying the intensities of these pencil beams, a conformal dose distribution is generated. Tumor coverage is improved, particularly in situations where the disease lies in close proximity to critical normal tissues. Given its highly conformal dose distribution, several advantages are noted when compared to conventional RT techniques.35 First, by decreasing the dose to the normal tissues, RT-related toxicities are minimized. Second, by conforming the doses to the irregularly shaped tumor, dose escalation is possible with IMRT, which can potentially lead to improvement in local control. Third, for each daily fraction, IMRT can give a higher dose to the gross tumor volume (GTV), resulting in a more effective biologic dose.6

Although IMRT has several advantages in comparison with conventional RT, one must not overlook several important issues. First, because of the varying degrees of intensities in the pencil beams, a greater target dose inhomogeneity is noted for IMRT. “Hot spots” are found within the GTV where nerves and blood vessels reside that can potentially result in unwarranted complications.7 Efforts should be made by the treating physician and treatment planner to minimize these “hot spots.” Fortunately, no higher rates of late effects have been reported to date. In fact, published reports have shown outstanding clinical results, particularly with decreased xerostomia, leading to improved quality of life.811 However, longer follow up is needed to validate these findings. Second, due to the sharp dose fall-off gradient between the target and the surrounding normal tissues, accurate target delineation is absolutely essential.1214 Inaccurate coverage of the target volume may lead to marginal misses since the treatment planning system will not treat areas that are not drawn on the computed tomography (CT) slices. Precise target delineation should be a multidisciplinary effort, integrating all clinical and radiographic findings. Available tools such as fusion of magnetic resonance imaging (MRI) and/or positron emission tomography (PET) to the treatment planning CT can be performed to improve the accuracy of target delineation.13

A more challenging question is what constitutes the clinical target volume (CTV) in order to encompass all potential microscopic spread. Several papers examining the precise definition of cervical nodal levels have been published and can serve as guidelines to help the radiation oncologists to delineate the CTV.5,15 Efforts by HNC experts from the Radiation Therapy Oncology Group (RTOG), European Organization for the Research and Treatment of Cancer (EORTC), and the Danish Head and Neck Cancer (DAHANCA) cooperative groups have generated a consensus guideline for the non-surgically violated node negative neck.15 This consensus can be downloaded from the RTOG website (www.rtog.org). However, the primary focus of these atlases is on the delineation of the nodal levels for the uninvolved, non-surgically violated neck. Such a consensus is also necessary for the node positive disease and the postoperative neck, which was recently published by a core group from the first consensus conference.5 Essentially, patients with gross nodes or those with extracapsular extension should have larger CTV than those who did not and the entire surgical bed should be included.

Treatment Planning and Radiation Delivery

IMRT can be divided into forward-planning (FP) and inverse-planning (IP).16 FP requires an experienced treatment planner who through multiple trials and errors designs the treatment fields for a given tumor.17 The planning process begins with the planner determining the beam directions, shapes, beam weighting. A desired dose distribution then results after multiple iterations and dose calculations by the treatment planner. FP can be used to treat HNC.18 However, to maximize the full potential of IMRT for complex tumors, IP is required. IP starts with the treatment planner and physician setting desired clinical objectives for the GTV, CTV, and the nearby normal tissues. With clinical objectives specified mathematically, a computerized algorithm determines the beam parameters that will yield the desired dose distribution for the target. The computer will undergo thousands of iterations to find the best solution. This cannot be achieved without a reliable and sophisticated planning system. Several commercial and noncommercial treatment planning systems are available and they are, in general, comparable.1,19

With regards to radiation delivery, different methods of beam intensity modulation are available.2,16 In the past, wedge filters and/or compensators have been used. Recently, a more sophisticated delivery system utilizes the multileaf collimator (MLC), situated in the aperture of the linear accelerator, to delivery radiation in a “step-and shoot fashion.” The MLC steps to a predetermined configuration and delivers the pencil beam. The MLC then steps to a different configuration and shoots another pencil beam. MLC system can also function in a “sliding window” fashion to deliver the radiation through the constant movements of different shapes of the MLC across the target.20 Another treatment delivery system is called Tomotherapy where radiation is delivered in a constant rotation of the gantry (linear accelerator head) using a concept similar to spiral CT scan.21 In general, these delivery systems are comparable to one another.22

Clinical results

There are increasing reports published from different institutions on the use of IMRT for HNC.23,24 Most reports focused on the decreased rates of xerostomia in HNC with IMRT.23,25 Besides sparing the parotid glands, IMRT can also spare the oral cavity, which houses several minor salivary glands. Investigators have also used IMRT to spare the constrictor muscles in attempt to minimize swallowing difficulties as a result of intense RT treatments.26 In addition, reduction of mucositis have also been reported, although this is based on retrospective reviews.27 Several centers also reported excellent locoregional control. Most of these series reported on their experience in treating a mixture of different sites within the head and neck.25,28 Experience from Stanford University on 69 head and neck cancer patients has shown a remarkable locoregional control of 92% with a median follow-up of 25 months.25 Sites included were oropharynx, oral cavity, larynx, hypopharynx, and unknown primary. In addition, in most series, definitive IMRT treated patients are reported along with post-operative treated patients.12,13 Although the results are promising, longer follow-up is needed to validate these findings, particularly with regards to the locoregional control and late complication. One interesting finding is the shift of the failure pattern with the predominant failures being distant rather than local. This changing pattern of failure can also be attributed to the use of concurrent chemotherapy with RT. It is beyond the scope of this article to discuss in detail these findings. Below, the authors present the most updated clinical results, broken down by the disease site.

Nasopharynx

Due to the multiple critical normal tissues surrounding the nasopharynx, such as the optic structures, brain stem, and salivary glands, IMRT is especially useful in treating these tumors. The earliest published dose comparison study on IMRT with conventional RT was reported by Xia et al. from University of California-San Francisco Medical Center (UCSF).3 Superior tumor coverage was shown with IMRT when compared to all other conventional techniques. In addition, the dose delivered to the normal tissue was decreased with IMRT. Other dosimetric analyses corroborated these findings.29,30 Since then, several centers have reported decreased rates of xerostomia in nasopharyngeal cancer (NPC) patients treated with IMRT.11,3133 There is even phase III evidence which confirms the advantage of IMRT in improving xerostomia compared to conventional RT. Besides the dosimetric advantages, several centers also reported excellent early clinical outcomes in NPC for IMRT.34

The most mature IMRT clinical data came from UCSF.11 The 4-year local progression-free and regional progression-free rates for 67 loco-regional advanced NPC patients were 97% and 98%, respectively. An update with more patients (n=118), continued to show excellent locoregional control.35 Several centers from Hong Kong also have shown similar findings.31,32 A recent experience from MSKCC reported a 91% locoregional control rate for IMRT treated NPC with a median follow-up of 35 months.33 (Table 1) The RTOG completed a phase II trial of IMRT with or without chemotherapy for non-stage IVC NPC. Preliminary data showed decreased xerostomia when compared to historical RTOG trials where conventional RT was used. An example of a NPC patient treated with IMRT is shown in figure 1, showing excellent conformation of the isodose lines to the tumor while sparing the optic chiasm and the brain stem.

Table 1.

Results of IMRT for nasopharyngeal carcinomas

Author N Stage F/U (month) Local Control
Lee14 67 T1–T4 33 97%
Wolden36 74 T1–T4 35 91%
Kwong 37 33 T1 24 100%
Kam34 63 T1–T4 29 92%
Figure 1.

Figure 1

An example of a T4N0 nasopharyngeal carcinoma treated with IMRT.

With the integration of IMRT and chemotherapy, centers are reporting a changing pattern of failure for NPC. Although the locoregional control is in excess of 90%, the distant metastases rates can be as high as 30%. Present regimens of cisplatin-based chemotherapy may not be effective in preventing distant metastases. Increased vascular endothelial growth factor-A (VEGF-A) has been associated with poor prognosis in HNC.36 One study showed significant elevation of serum VEGF in patients with metastastic NPC.37 In another series, VEGF overexpression was noted in 67% of the NPC patients and elevated VEGF expression in Epstein Barr Virus (EBV) positive tumors was associated with more recurrence, advance nodal stage, and lower overall survival.38 Therefore, the RTOG is currently conducting a phase II trial adding bevacizumab to IMRT and chemotherapy for locoregionally advanced NPC to address this issue.

Oropharynx

Although locoregional control rates for oropharyngeal (OP) cancer patients treated with non-IMRT techniques are excellent, both parotid glands are almost always included in the RT portal. As a result, patients suffer from permanent xerostomia, which has a negative impact on nutrition, dentition, communication and emotional well-being. With parotid-sparing IMRT, patient quality of life has improved when compared to conventional RT.39

Dose sculpting and parotid sparing raise a general concern for possible tumor misses with IMRT. However, no clinical reports to date have shown any compromise in local control for OP cancers treated with IMRT. With a median follow-up of 32 months, Eisbruch et al. reported a 3-year locoregional control rate of 94% in 80 OP cancer patients.40 Over 90% of the patients had stage III or IV disease. Chao et al reported a local control rate of 87% using IMRT in 74 patients with OP cancer.41 In this report, 46% had T3/T4 tumors while 76% of the patients had stage III/IV disease. Other centers also reported excellent locoregional control rates. De Arruda et al noted a 100% tumor control rate at a median follow-up of 24 months. An update with a median follow-up of 31 months continues to show excellent local control in this patient cohort.27 (Table 2) A multi-institutional RTOG study, H-0022, using IMRT for early-stage OP cancer has completed accrual. The protocol tests the transportability of IMRT to multiple institutions. Preliminary results have been presented in the American Society of Therapeutic Radiation Oncology (ASTRO) that showed low IMRT violation rates and reduced xerostomia when compared to historical controls. Figure 2 shows an example of an IMRT plan for an OP cancer.

Table 2.

Results of IMRT for oropharyngeal carcinomas

Author N Stage F/U (month) Loco-regional Control
Chao46 74 I–IV 33 87%
Eisbruch45 80 I–IV 32 94%
De Arruda61 50 I–IV 24 92%
Figure 2.

Figure 2

An example of a T3N0 base of tongue cancer treated with IMRT.

Larynx & Hypopharynx

There are scanty reports on the use of IMRT for laryngeal and hypopharyngeal tumors. Perhaps because these tumors are situated inferior to the parotid glands, using IMRT for parotid sparing are of less importance when compared tumors situated higher in the head. However, IMRT can still offer several advantages in the management of laryngeal and hypopharyngeal cancers. These tumors often present in close proximity to the spinal cord. In order to respect the spinal cord tolerance, conventional treatments require photon/electron matching which results in dose uncertainty at the match line. By using IMRT, a concave dose distribution can be achieved which allows improved tumor coverage while sparing the spinal cord. In addition, significant external contour changes from the head to the low neck can result in dose inhomogeneity and cause unwarranted hot and cold spots within the tumor. IMRT can minimize dose inhomogeneity without the need of tissue compensators. Lastly, for tumors that have a high risk of retropharyngeal nodal involvement, i.e. node positive hypopharyngeal tumors, where target delineation extends to the skull base, IMRT can spare the parotid glands where it would be difficult with conventional techniques.10,42 (Figure 3)

Figure 3.

Figure 3

An example of a T3N1 laryngeal carcinoma treated with IMRT.

With these theoretical advantages, recent studies have reported promising results for IMRT for laryngeal and hypopharyngeal carcinomas. At a median follow-up of 2.2 years, the 2-year locoregional progression-free survival was 84% and the laryngectomy-free survival was 89% for patients treated with IMRT and concurrent chemotherapy.10 These results were comparable to those seen for the best arm of the intergroup larynx preservation trial.43 In addition, no grade 2 xerostomia was observed in this study. In another report by the Swiss group where IMRT was used to treat hypopharyngeal cancer, 86% of the 29 patients received concurrent chemotherapy and 60% had locally advanced disease.42 With a short mean follow-up of 16 months, the 2-year local control and disease-free survival estimates were both 90%.

Paranasal Sinuses (PNS)

Due to the proximity of multiple surrounding critical tissues, IMRT is of particular clinical interest in the treatment of PNS tumors. Numerous treatment-planning studies have demonstrated the superior dose distribution in favor of IMRT for these tumors in comparison to 3DCRT.4,44,45 Early clinical results suggest that the theoretical advantages associated IMRT do indeed translate into clinical benefits.46,47 Since tumor control is highly dependent on appropriate target volume delineation, which is often difficult for PNS tumors in the post-operative settings, integration of pre-operative imaging studies and inputs from HN surgeons and pathologists are critical for accurate target contouring.

Claus et al. at the Ghent University Hospital has reported on the use of IMRT for 44 patients with PNS cancers.48 In general, the prescription dose to the gross disease was 70 Gy, with a maximum dose constraint of 60 Gy to the optic structures. Although the follow-up is relatively short, none of the patients in whom optic pathway was spared had developed severely dry eyes. Locoregional control was excellent for T1-3 tumors but poor for T4 tumors. Other centers have also reported encouraging toxicity reduction when compared to historical controls where the complication rates can be as high as 35%. Chen et al. reported a longitudinal study of 127 patients treated with RT from 1960 to 2005 at UCSF. The incidence of grade 3 and 4 late ocular toxicity among patients treated with conventional RT, 3D CRT, and IMRT was 20%, 9%, and 0%, respectively.49 In another series by Hoppe et al, none of the patients who underwent IMRT developed Grade 3–4 late ocular complication.46 As more centers gain experience with IMRT, it is anticipated that the late complication rates will continue to decrease. The North American Skull Base Society is currently investigating the role of high dose IMRT and concomitant cisplatin chemotherapy with or without surgery for advanced PNS cancers. The protocol is under development with the goal to determine the feasibility of non-surgical therapy for these tumors in a multi-institution setting.

Thyroid

Tumors arising from the thyroid gland are primarily treated by surgical resection and the role of external beam RT is largely restricted to the post-operative setting.54 Due to the proximity of the tumor bed to the spinal cord, it has been very difficult to deliver adequate doses without exceeding spinal cord tolerance. Dosimetric studies from different centers have shown improvements in target coverage while reducing the spinal cord dose with IMRT.50,51

Clinically, IMRT was found to be effective and feasible in a small series by Rosenbluth et al, reporting on 20 patients treated with IMRT between 2001 and 2004.52 All patients had non-anaplastic thyroid cancer and many had recurrent or T4N1 tumor. The authors noted 2 local failures and reported a 2-year local progression-free rate of 85%. The 2-year overall survival was 60%. Acute grade 3 toxicities included 7 mucositis, 3 pharyngitis, 2 dermatitis and 2 laryngitis. No significant radiation-related late effects were reported at the time of their analysis. Though impressive, longer follow-up and larger studies are needed to validate these results.

Re-irradiation

Re-irradiation using conventional techniques is limited by the tolerance of surrounding critical structures. Current recommendation, based on the RTOG re-irradiation trials, limits the combined total spinal cord dose (both initial and retreatment) to 50 Gy. Therefore, a major benefit of IMRT over conventional RT techniques is to provide superior coverage for the recurrent tumor while minimizing the dose delivered to the adjacent normal tissues.53 There are few clinical reports on the use of IMRT to retreat recurrent HNC. Lu et al. reported on their experience of IMRT in treating recurrent NPC.54 Acute toxicity of the skin, mucosa, and salivary glands was acceptable. Tumor necrosis was noted in 14 patients (29%) toward the end of IMRT. With a median follow-up of 9 months, the locoregional control rate was 100%. Another study included all head and neck sites showed that IMRT was associated with better locoregional progression-free survival than conventional RT on multivariate analysis. In this study, IMRT was intricately related to the ability to deliver higher RT doses, which was associated with better overall survival.55 Although promising, longer follow-up, larger and more uniform studies are required to fully assess the clinical utility of IMRT in the recurrent settings.

TUMOR HYPOXIA IN HEAD AND NECK CANCERS

Techniques to measure tumor hypoxia and their clinical significance in HNSCC

Currently, there exist several approaches for detecting tumor hypoxia in HNC. At a recent hypoxia workshop, convened the National Cancer Institute (NCI), the consensus among the experts is that “there is not, and probably will never be, a single clear ‘gold standard’ for in vivo hypoxia measurement”.56 The workshop report also presented a comprehensive review of different approaches for measuring tumor hypoxia. Briefly, techniques for measuring tumor oxygen can be categorized into 2 groups: direct and indirect. Their advantages and disadvantages are summarized in Table 3.

Table 3.

Different techniques for assessing tumor hypoxia

Method Examples Measure Spatial resolution Advantages Disadvantages
PO2
Histography
Eppendorf electrode
OxyLite fiber optic probe
pO2 0.5 mm
(thousands of cells)
  • Direct

  • Rapid real-time measurements

  • Validated in human tumors

  • Invasive

  • Tumor inaccessibility

  • Pressure dependence

  • Inter-observer variability

  • Readings affected by necrosis

  • No spatial information

Exogenous
Markers
EF5
Pimonidazole
Chronic hypoxia 1.0 um
(single cell)
  • Highly sensitive

  • Reproducible

  • Require drug injection

  • Require extra biopsies

Endogenous hypoxia marker HIF-1
CA-IX
Glut-1
Biologic hypoxia 1.0 um
(single cell)
  • Apply to archival tissues

  • No drug injection

  • No extra biopsies

  • Less hypoxia specific

  • Variability in staining & interpretation

Secreted markers OPN
VEGF
Biologic hypoxia N/A
  • Non invasive

  • Inexpensive

  • No biopsy or drug injection

  • Serial measurements

  • Less hypoxia specific

  • Less tumor specific

  • Specimen processing critical

  • Antibody specificity

PET-based hypoxia imaging 18F-MISO
18F-FAZA
18F-EF5
18FETNIM
60CuATSM
24I-IAZGP
Chronic hypoxia 2–10 mm
  • Spatial resolution

  • Serial measurements

  • RT directed targeting

  • Reproducible

  • Requires dedicated equipments (tracer generation & imaging)

  • Expensive

  • Radiation exposure

  • Tracer synthesis expertise

Direct oxygen measurements in tissues

Direct approaches can be applied to tissue (needle electrodes, fiberoptic probes) or blood (measurements or imaging of oxyhemoglobin saturation and oxygen diffusion). Polarographic needle electrodes (pO2 histograph, Eppendorf, Hamburg, Germany) provided the first convincing evidence that hypoxia existed in human solid tumors.57,58 The sensing electrode, mounted on the tip of a needle, is advanced via a step motor through the tissue, taking rapid measurements (1.4 s) to avoid spurious readings from pressure artifacts. A histogram of oxygen tensions (pO2) can then be obtained from multiple sampling points along different tracks. Normal tissues typically show a Gaussian pO2 distribution with the median value between 40–60 mm Hg; whereas tumors invariably show lower pO2 measurements. Several studies have showed that low tumoral pO2, defined by either the median value or the hypoxic fraction (% readings below 2.5 or 5 mm Hg), correlated with treatment outcomes in HNC patients treated with RT or chemoradiotherapy.5961 One study also found that tumor pO2 prediected for pathologically persistent neck nodes in patients undergoing a neck dissection for clinical N2-3 necks after chemoradiation treatment.62 Pooled data in 397 HNC patients provided definitive evidence that tumor pO2 is an independent predictor for survival.63 Although the microelectrode technique directly measures tumor pO2, it suffers from several drawbacks that make it difficult for general use. These include invasiveness, tumor inaccessibility, pressure dependence, inter-observer variability, failure to distinguish necrosis from hypoxia, and the lack of spatial information. Despite these limitations, it is the most studied approach for assessing hypoxia to date.64

Indirect approach – Injectable markers

Indirect approaches use injectable molecular reporters of oxygen as the endpoints. Injectable reporters include 2-nitroimidazole compounds such as misonidazole, pimonidazole (1-(2-nitro-1-imidazolyl)-3-N-piperidino-2-propanolol)65, and EF5 (nitroimidazole[2-(2-nitro-1H-imidazol-1-yl)-N-(2,2,3,3,3-pentaflouropropyl) acetamide).66 These compounds form stable adducts with intracellular macromolecules only in hypoxic regions (< 10 mm Hg).67 Detection of these adducts with antibodies can provide information on the relative oxygenation at cellular resolution.68,69 In general, 2-nitroimidazole markers stain for areas of chronic hypoxia and more sensitive at severe hypoxic conditions than the microelectrode.69,70 This approach is limited by the requirement for exogenous drug administration, additional biopsies for staining and quantification of staining. Presently there is minimal clinical data regarding the prognostic significance of these agents in HNC. In a small study evaluating pimonidazole, microvessel density count and carbonic anhydrase IX (CA IX) binding in 42 HNC, pimonidazole staining was more pronounced at distance > 100 υm from blood vessels than CA IX, suggesting that it is more specific for chronic hypoxia.71 High pimonidazole staining correlated with a higher risk locoregional relapse in patients treated with RT alone but not in those treated with RT plus carbogen and nicotinamide, which were used to modulate tumor hypoxia.

These exogenous markers can also be labeled with 18F and employed as special tracers for hypoxia imaging using PET or SPECT imaging approaches. Hypoxia imaging is discussed in details in the “Novel Imaging Approach” section by Drs Krohn and Yueh.

Indirect approach – Endogenous markers

Endogenous molecular markers for tumor hypoxia represent proteins and genes whose expressions are induced by hypoxic exposure. One of the most studied oxygen response pathways is that mediated by the hypoxia inducible factor-1 (HIF-1), which regulates genes that are involved in cell metabolism, angiogenesis, invasion, metastasis and apoptosis. HIF-1 and several of its downstream targets including Glut-1 (glucose transporter-1), CA IX and VEGF have been widely investigated as prognostic markers in HNC with mixed results (Table 4). In general, elevated expression of these markers portends poorer outcomes in patients treated with non-surgical therapies. The advantage of endogenous markers is that levels of these proteins can be assessed on archival materials, thereby allowing rapid correlation to treatment outcomes. In addition it requires neither the injection of foreign material nor any additional invasive procedure beyond that a biopsy at diagnosis. A significant drawback to these approaches is that these proteins can be regulated by factors other than hypoxia. For example, HIF-1α expression can be influenced by several non-hypoxic stimuli including nitric oxide, cytokines, trophic stimuli and oncogenes.7275 Comparison of the staining patterns between endogenous and injectable markers showed that the former, in general, stained more diffusely and closer to the blood vessels than the latter, suggesting other modes of induction and activation at a wider range of oxygen concentration.71,76 To circumvent this dilemma, suggestions have been made to combine several endogenous markers together to improve hypoxia specificity. For example, gene expression analysis has been used to generate a hypoxia gene signature or a hypoxia metagene to predict treatment outcomes in several solid tumors, including HNC.77,78 Using gene expression profiling of 59 HNC, Winter et al generated a hypoxia metagene by identifying genes whose expression clustered with 10 known hypoxia regulated genes.78 They found that this metagene was able to predict recurrence-free survival in an independent HNC data set as well as overall survival in another breast cancer series. We have also used a combination of gene expression and proteomic analyses to identify novel hypoxia induced proteins. After confirming their hypoxic inducibility in cell lines and animal models, we investigated their utility in combination with CA IX to predict outcomes by staining a HNC tissue array from with known tumor pO2. These studies resulted in a panel of 4 hypoxia markers (CA IX, Lysyl oxidase (LOX), Galectin-1 and Ephrin A1) that can be used to predict treatment outcomes in terms of cancer-specific survival. 79 (Figure 4) These endogenous hypoxia signatures, though promising, need to be validated in larger independent datasets before they can be used in the clinical settings.

Table 4.

Significance of endogenous markers for hypoxia in head and neck cancers

Author Marker # Pts Tumor site Treatment Respond LRC Survival Associated parameters
Aebersold126 HIF-1α 98 Oropharynx RT or CRT Yes Yes Yes Grade (inverse)
Koukourakis127 HIF-1α, HIF-2α 75 H&N CRT Yes Yes Yes T-stage, MVD, VEGF
Beasley128 HIF-1α 79 H&N Surgery No* Necrosis
Hui129 HIF-1α, CA IX 90 Nasopharynx RT or CRT Yes** CA IX, VEGF
Beasley130 CA IX 79 H&N Surgery MVD, necrosis
Koukourakis131 CA IX 75 H&N CRT Yes Yes Yes MVD, necrosis
Kaanders71 CA IX 43 H&N RT +/− ARCON No No No Pimonidazole staining
Winter132 HIF-1α, HIF-2α, CA IX 140 H&N Surgery Yes***
Koukourakis133 HIF-2α, CA IX 198 H&N RT **** Yes Yes Higher T-stage

Yr: year; Pt: patients; H&N: head and neck; RT: radiotherapy; CRT: chemoradiotherapy; ARCON: Carbogen and nicotidamide; LRC: locoregional control; MVD: microvessel density count.

*

Improved disease-free and overall survival with HIF-1α over expression

**

The combination of HIF-1α and CA IX positive staining (hypoxic profile) was associated with worse progression-free survival. Positive individual marker staining did not correlated with survival.

***

Improved disease-specific survival but not overall survival with HIF-1α & HIF-2α expression, but not with CA IX

****

RT: Conventional fraction versus continuous hyperfractionated accelerated radiation therapy (CHART)

Figure 4.

Figure 4

Cancer-specific survival by hypoxia marker score comprised of Galectin-1, Ephrin A1, Lysyl Oxidase, CA IX cytoplasmic and CA IX membrane staining, where a score of 1 was assigned to strong staining for each marker and a score of 0 to negative and week staining. This has been adjusted for age and hemoglobin levels, 2 other significant factors on univariate analysis.

Our laboratory has focused on identifying secreted markers of hypoxia that can be rapidly and inexpensively measured in the blood. Two markers that have been tested clinically with mixed results are VEGF and osteopontin (OPN). Although circulating VEGF levels were elevated in cancer patients80,81 and in those with acute hypoxia such as obstructive apnea82, the relationship between tumor hypoxia and systemic VEGF levels is unclear. Dunst et al found that serum VEGF levels independently correlated with hypoxic tumor subvolume in 56 HNC patients.83 However, it also correlated with total tumor volume, hemoglobin level and platelet counts. They did not report on the clinical significance of serum VEGF levels in terms of treatment outcomes. In contrast, we did not find a direct relationship between plasma VEGF and tumor pO2 in 48 HNC patients in our study (unpublished observations). We did however found a small but significant relationship between OPN level and tumor pO2 in our patient cohort.84 This was confirmed by Nordsmark et al.85 In addition, plasma OPN was an independent and significant predictor for treatment outcomes in these patients and another independent group of HNC patients.86 These results were confirmed by the DAHANCA group in a larger cohort of HNC patients treated with radiation therapy +/− nimorazole, a hypoxic cell radiosensititizer.87 Intriguingly, only patients with high pretreatment circulating OPN levels benefited from nimorazole whereas those with low-intermediate levels did not, suggesting that OPN may be use to select patients for hypoxia targeting. Further validation of this marker is ongoing in another set of HNC patients treated with or without Tirapazamine (TPZ), a hypoxic cell cytotoxin.

Hypoxia Targeted Therapy

Since the 1950s, enormous efforts have been devoted to develop strategies to target hypoxia either directly or indirectly. These include techniques to enhance oxygen tissue delivery, generation hypoxic cell radiosensitizers and hypoxic cell cytotoxin, treatment with high linear-energy transfer (LET) radiation that is less oxygen dependence and, more recently, development of inhibitors to signaling pathways that are activated by hypoxia. Although most of these strategies have not achieved general acceptance, a meta-analysis of trials using hypoxic cell sensitizers or hyperbaric oxygen showed a small but statistically significant benefit in terms of locoregional control and survival.88 In this part of the paper, we will touch on some past strategies and future approaches for exploiting hypoxia.

Past Strategies

The most straightforward strategy to overcome hypoxia is to administer oxygen at pressure higher than room air (usually 3 atmospheres), i.e. hyperbaric oxygen treatment. Although one study showed promising results in HNC patients, the results have been mixed.89,90 In retrospect, this strategy only affects chronically hypoxic cells but is not expected to change acute hypoxia. Although a meta-analysis suggests that the use of hyperbaric oxygen breathing during RT can improve local control by 10%, it has not gained general acceptance for clinical use due to inconsistent response, safety issues and high cost for implementation.88

A widely investigated hypoxia targeted strategy is to use electron-affinic drugs (nitroimidazoles) to sensitize tumors to radiation. Xenograft studies showed significant radiosensitization with nitroimidazole compounds in tumors without enhancing normal tissue toxicity.91,92 For 2 decades, nitroimidazole compounds had been extensively studied by RTOG and DAHANCA groups in HNC as an adjunct to RT with mixed results.9397 Most trials reported disappointing outcomes except for one large study. In this phase III study (DAHANCA 5-85), the addition of nimorazole to RT resulted in improved locoregional control (49% versus 33%, p = 0.002) and cancer-related survival (52% versus 41%) when compared to the placebo arm in patients with supraglottic larynx and pharynx cancers.96 The main draw back of these compounds is the neurotoxicity associated with multiple doses.98,99 This toxicity limits the total amount of drugs that can be administered to achieve maximal efficacy.

Present Strategies

Erythropoietin manipulation

It is hypothesized that increase oxygen delivery via modifications of hemoglobin levels can attenuate tumor hypoxia. Henke et al performed a phase III randomized study of RT +/− erythropoietin (EPO) in HNC. Unfortunately, patients who received EPO had worse locoregional progression-free survival.100 This could be explained partially by the quadratic regression in the correlation between tumor pO2 and hemoglobin levels: at levels ≤ 14 g/dL, increasing hemoglobin directly correlates with increasing pO2; however, at levels above 14 g/dL, an inverse correlation is noted with lower pO2 associated with increasing hemoglobin, presumably from venostasis.63 More recently, the presence of EPO receptors in tumor cells has also been implicated with worse outcomes in patients receiving EPO on this study.101

Vascular normalization

Increased vascular leakage from immature tumor blood vessels can result in higher interstitial fluid pressure, thereby, worsening tumor hypoxia and impeding effective tumoral drug delivery. Jain et al. popularized the concept of normalization of tumor vasculature through antiangiogenic therapy such as bevacizumab.102 This concept was supported by clinical data in colorectal, breast and brain cancers, where treatment with bevacizumab, paclitaxel (a chemotherapy with antiangiogenic properties) or AZD2171 (a VEGF receptor tyrosine kinase inhibitor), respectively, was shown to reduce tumor interstitial pressure.103 103105 This concept has yet to be tested in HNC.

ARCON (Accelerated Radiotherapy with CarbOgen and Nicotinamide)

Another promising approach to target hypoxia in HNC is the combined use of the nicotinamide vasodilator and carbogen breathing to increase tumor pO2. ARCON has produced a 3-year local control rate in excess of 80% for advanced stage T3-4 laryngeal and oropharyngeal cancers.106 Presently, a phase III clinical trial testing the efficacy of ARCON in laryngeal cancers is ongoing in Europe and will address the effectiveness of this strategy in HNC.106

Hypoxic cell cytotoxin

An important strategy to exploit tumor hypoxia is the use of bioredutive agents that can selectively kill hypoxic cells. The first bioreductive drug used in clinical trials is mitomycin-C. Pooled data from 2 randomized trials in HNC suggested that the addition of mitomycin-C to RT resulted in statistically significant improvement in locoregional control and cause-specific survival.107 Another study comparing conventional fractionated RT to the Vienna Continuous Hyperfractionated Accelerated RT regimen (V-CHART) or to V-CHART plus mitomycin-C showed that the best survival and locoregional control rates were observed for the V-CHART and mitomycin-C group.108,109 Although promising, mitomycin-C toxicity limits the frequency of drug delivery, making it unlikely to be the ideal drug for exploiting tumor hypoxia.

A widely studied hypoxic cell cytotoxin is Tirapazamine (TPZ). In a randomized phase II trial (TROG), the combination of TPZ, cisplatin and RT was found to be superior to 5FU, cisplatin and RT for locally advanced HNC.110 In contrast, we found that the addition of TPZ to an aggressive regimen of induction and concurrent cisplatin and 5FU with RT did not result in improved outcomes in a small randomized phase II study.111 A large multi-institutional phase III trial testing the benefit of adding TPZ to concurrent RT and cisplatin has been completed and the results are pending.

TPZ has several limitations including poor drug diffusion through hypoxic tissues and become activate under less stringent hypoxia, a feature that can cause normal tissue toxicity in poorly oxygenated organs. Therefore, there are strong interests in developing novel hypoxic cell cytotoxins with more specific antitumor activity. Dinitrobenzamide mustards (DNBMs) belong to a new and highly potent class of hypoxic cytotoxins discovered by the Auckland University group. These compounds have improved properties over TPZ, including activation by more stringent hypoxia and a substantial bystander killing effect. A lead compound of the DNBM family, PR-104 is being studied in phase I-II clinical trials for several solid tumors.112,113

Future directions: Targeting HIF-1

Since HIF-1 is a key transcriptional factor for several hypoxia induced genes, there is a strong interest in developing specific HIF-1 inhibitors. The rationales for targeting HIF-1 have been eloquently outlined in a review by Giaccia et al.114 Although targeting HIF-1 is a conceptually attractive, it’s highly challenging; therefore, blocking upstream signaling pathways leading to HIF-1 inactivation or downstream effector functions may represent a more practical strategy. For a thorough review of emerging HIF-1 inhibitors, please refer to Melillo et al.115 To date, inhibitors of several upstream signaling pathways of HIF-1 have either been approved by the FDA for clinical use or are being investigated in clinical trials. Examples of these include mTOR inhibitors (Rapamycin and Temsirolimus) that can suppress mTOR-dependent HIF-1 translation and epidermal growth factor receptor (EGFR) inhibitors (Gefitinib, Erlotinib) or antibodies (Cetuximab, Panitumumab) that inhibit HIF-1 induction by EGFR-dependent pathways.115 Another approach is to inhibit HIF protein accumulation. Chemicals in this class include Topotecan116, 2-methoxyestradiol (2ME2)117, Hsp90 inhibitors (17-N-Allylamino-17-demethoxy geldanamycin [17-AAG] and 17-dimethylaminoethylamino-17-demethoxygeldanamycin [17-DMAG])118, Histone deacetylase (HDAC) inhibitors (Vorinostat)119, YC-1 (3-(5′-hydroxymethyl-2′-furyl)-1-benzylindazole) a soluble guanylate cyclase activator120, PX478121, Thioredoxin inhibitors (PX-12)122, and flavopiridol, a cyclin dependent kinase inhibitor.123 Although most of these compounds have been identified as anticancer drugs by their other activities, all negatively affect HIF-1 protein levels. One of such drug, topotecan, is being tested in a clinical trial at the NCI to specifically assess its HIF-1 inhibitory function in patients with advanced tumors, refractory to standard therapies.115 A crucial feature of this trial is that the enrolled population is intentionally enriched for tumors with elevated HIF-1 protein expression. Inhibitors that directly target HIF-1 transactivating activity are slowly emerging from high throughput screenings.115 These include echinomycin and chetomin, both of which are currently being evaluated only in animal models.

An interesting proposal to target HIF-1 that has yet to be tested in patients is to take advantage of its involvement in tumor metabolism, which is different from that of normal tissues. Several of HIF-1 target genes are key regulators of tumor metabolism. One of such genes is pyruvate dehydrogenase kinase 1 (PDK1), which limits the amounts of pyruvate entering into the citric acid cycle, thereby decreasing mitochondrial oxygen consumption. Cairn et al showed that inhibition of HIF-1 and PDK1 either genetically or pharmacologically resulted in enhanced oxygen consumption and a transient rise in tumor hypoxia.124 They theorize that pretreatment with either HIF-1 or PDK1 inhibitors would transiently decrease tumor oxygenation, which in turns would improve the effectiveness of hypoxic cell cytotoxins such as TPZ. They elegantly showed that HIF-1 inhibitors dramatically increased TPZ anticancer effectiveness when administered before but not after TPZ, and this enhancement is dependent on HIF-1 being functional. This novel strategy for hypoxia modification may circumvent the dilemma of hypoxia heterogeneity and may also alleviate the need for accurately identifying hypoxic tumors. In addition, it highlights the importance of appropriate sequencing of RT and HIF-1 inhibitors as HIF-1 blockade before RT may increase tumor hypoxia and negate RT effectiveness. This is also consistent with the data from Moeller et al who showed that HIF inhibition increased radiation effectiveness when occurred after but not before RT.125

CONCLUSIONS

Given the available dosimetric and clinical data, IMRT is an effective treatment modality for HNC. It conforms the dose to the tumor while sparing nearby normal tissues. Toxicity from high dose RT has decreased when compared to historical controls. Clinical reports have shown great promise in terms of tumor control. With advances in anatomic and functional imaging, along with more clinical experience, it is likely that IMRT will lead to further improvements in tumor control, survival and quality of life in HNC patients.

After half a decade of efforts, tumor hypoxia continues to be a therapeutic challenge in HNC. Nonetheless, the prospect of reducing its impact is looking brighter with improved ability of detecting hypoxia and better understanding of its molecular targets for therapeutic exploitation. Testing new leads from the laboratory will require clinical trials with innovative designs that incorporate serial novel non-invasive surrogate endpoints for hypoxia such as molecular makers or imaging methods.

Acknowledgments

Supported in part by 1 R01 CA118582-01 (QTL) and ASTRO Junior Investigator Award (NYL)

Financial disclosure: None

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Bibliography

  • 1.Chui CS, Spirou SV. Inverse planning algorithms for external beam radiation therapy. Medical Dosimetry. 2001;26:189–97. doi: 10.1016/s0958-3947(01)00069-3. [DOI] [PubMed] [Google Scholar]
  • 2.Chui CS, Chan MF, Yorke E, et al. Delivery of intensity-modulated radiation therapy with a conventional multileaf collimator: comparison of dynamic and segmental methods. Medical Physics. 2001;28:2441–9. doi: 10.1118/1.1418018. [DOI] [PubMed] [Google Scholar]
  • 3.Xia P, Fu KK, Wong GW, et al. Comparison of treatment plans involving intensity-modulated radiotherapy for nasopharyngeal carcinoma. [see comment] Int J Radiat Oncol Biol Phys. 2000;48:329–37. doi: 10.1016/s0360-3016(00)00585-x. [DOI] [PubMed] [Google Scholar]
  • 4.Huang D, Xia P, Akazawa P, et al. Comparison of treatment plans using intensity-modulated radiotherapy and three-dimensional conformal radiotherapy for paranasal sinus carcinoma. Int J Radiat Oncol Biol Phys. 2003;56:158–68. doi: 10.1016/s0360-3016(03)00080-4. [DOI] [PubMed] [Google Scholar]
  • 5.Gregoire V, Eisbruch A, Hamoir M, et al. Proposal for the delineation of the nodal CTV in the node-positive and the post-operative neck. Radiother Oncol. 2006;79:15–20. doi: 10.1016/j.radonc.2006.03.009. [DOI] [PubMed] [Google Scholar]
  • 6.Gregoire V. Implementation of intensity-modulated radiation therapy in head and neck tumors: what are the important data for biological optimization? Rays. 2004;29:271–4. [PubMed] [Google Scholar]
  • 7.de Neve W, Duthoy W, Claus F, et al. Dose conformation in IMRT for head and neck tumors: which solution to apply? Cancer Radiother. 2002;6 Suppl 1:32s–36s. doi: 10.1016/s1278-3218(02)00216-0. [DOI] [PubMed] [Google Scholar]
  • 8.Eisbruch A, Kim HM, Terrell JE, et al. Xerostomia and its predictors following parotid-sparing irradiation of head-and-neck cancer. Int J Radiat Oncol Biol Phys. 2001;30:695–704. doi: 10.1016/s0360-3016(01)01512-7. [DOI] [PubMed] [Google Scholar]
  • 9.Chao KS, Deasy JO, Markman J, et al. A prospective study of salivary function sparing in patients with head-and-neck cancers receiving intensity-modulated or three-dimensional radiation therapy: initial results. Int J Radiat Oncol Biol Phys. 2001;49:907–16. doi: 10.1016/s0360-3016(00)01441-3. [DOI] [PubMed] [Google Scholar]
  • 10.Lee NY, O’Meara W, Chan K, et al. Concurrent chemotherapy and intensity-modulated radiotherapy for locoregionally advanced laryngeal and hypopharyngeal cancers. Int J Radiat Oncol Biol Phys. 2007 doi: 10.1016/j.ijrobp.2007.03.013. [DOI] [PubMed] [Google Scholar]
  • 11.Lee N, Xia P, Quivey JM, et al. Intensity-modulated radiotherapy in the treatment of nasopharyngeal carcinoma: an update of the UCSF experience. [see comment] Int J Radiat Oncol Biol Phys. 2002;53:12–22. doi: 10.1016/s0360-3016(02)02724-4. [DOI] [PubMed] [Google Scholar]
  • 12.Chao KS, Ozyigit G, Tran BN, et al. Patterns of failure in patients receiving definitive and postoperative IMRT for head-and-neck cancer. Int J Radiat Oncol Biol Phys. 2003;55:312–21. doi: 10.1016/s0360-3016(02)03940-8. [DOI] [PubMed] [Google Scholar]
  • 13.Lee N, Xia P, Fischbein NJ, et al. Intensity-modulated radiation therapy for head-and-neck cancer: the UCSF experience focusing on target volume delineation. Int J Radiat Oncol Biol Phys. 2003;57:49–60. doi: 10.1016/s0360-3016(03)00405-x. [DOI] [PubMed] [Google Scholar]
  • 14.Dawson LA, Anzai Y, Marsh L, et al. Patterns of local-regional recurrence following parotid-sparing conformal and segmental intensity-modulated radiotherapy for head and neck cancer. Int J Radiat Oncol Biol Phys. 2000;46:1117–26. doi: 10.1016/s0360-3016(99)00550-7. [DOI] [PubMed] [Google Scholar]
  • 15.Gregoire V, Levendag P, Ang KK, et al. CT-based delineation of lymph node levels and related CTVs in the node-negative neck: DAHANCA, EORTC, GORTEC, NCIC, RTOG consensus guidelines. Radiother Oncol. 2003;69:227–36. doi: 10.1016/j.radonc.2003.09.011. [DOI] [PubMed] [Google Scholar]
  • 16.Intensity Modulated Radiation Therapy Collaborative Working Group. Intensity-modulated radiotherapy: current status and issues of interest. International Journal of Radiation Oncology Biology Physics. 2001;51:880–914. doi: 10.1016/s0360-3016(01)01749-7. [DOI] [PubMed] [Google Scholar]
  • 17.Xia P, Pickett B, Vigneault E, et al. Forward or inversely planned segmental multileaf collimator IMRT and sequential tomotherapy to treat multiple dominant intraprostatic lesions of prostate cancer to 90 Gy. Int J Radiat Oncol Biol Phys. 2001;51:244–54. doi: 10.1016/s0360-3016(01)01643-1. [DOI] [PubMed] [Google Scholar]
  • 18.Lee N, Akazawa C, Akazawa P, et al. A forward-planned treatment technique using multisegments in the treatment of head-and-neck cancer. Int J Radiat Oncol Biol Phys. 2004;59:584–94. doi: 10.1016/j.ijrobp.2004.02.005. [DOI] [PubMed] [Google Scholar]
  • 19.Xia P, Lee N, Liu YM, et al. A study of planning dose constraints for treatment of nasopharyngeal carcinoma using a commercial inverse treatment planning system. Int J Radiat Oncol Biol Phys. 2004;59:886–96. doi: 10.1016/j.ijrobp.2004.02.040. [DOI] [PubMed] [Google Scholar]
  • 20.Boyer AL, Yu CX. Intensity-modulated radiation therapy with dynamic multileaf collimators. Semin Radiat Oncol. 1999;9:48–59. doi: 10.1016/s1053-4296(99)80054-x. [DOI] [PubMed] [Google Scholar]
  • 21.Michalski J, Purdy JA, Gaspar L, et al. Radiation Therapy Oncology Group. Research Plan 2002-2006 Image-Guided Radiation Therapy Committee. Int J Radiat Oncol Biol Phys. 2001;51:60–5. [PubMed] [Google Scholar]
  • 22.Xia P, Verhey LJ. Delivery systems of intensity-modulated radiotherapy using conventional multileaf collimators. Medical Dosimetry. 2001;26:169–77. doi: 10.1016/s0958-3947(01)00060-7. [DOI] [PubMed] [Google Scholar]
  • 23.Lee N, Puri DR, Blanco AI, et al. Intensity-modulated radiation therapy in head and neck cancers: an update. Head Neck. 2007;29:387–400. doi: 10.1002/hed.20332. [DOI] [PubMed] [Google Scholar]
  • 24.Eisbruch A. Intensity-modulated radiotherapy of head-and-neck cancer: encouraging early results. Int J Radiat Oncol Biol Phys. 2002;53:1–3. doi: 10.1016/s0360-3016(02)02725-6. [DOI] [PubMed] [Google Scholar]
  • 25.Daly ME, Lieskovsky Y, Pawlicki T, et al. Evaluation of patterns of failure and subjective salivary function in patients treated with intensity modulated radiotherapy for head and neck squamous cell carcinoma. Head Neck. 2007;29:211–20. doi: 10.1002/hed.20505. [DOI] [PubMed] [Google Scholar]
  • 26.Eisbruch A, Schwartz M, Rasch C, et al. Dysphagia and aspiration after chemoradiotherapy for head-and-neck cancer: which anatomic structures are affected and can they be spared by IMRT? Int J Radiat Oncol Biol Phys. 2004;60:1425–39. doi: 10.1016/j.ijrobp.2004.05.050. [DOI] [PubMed] [Google Scholar]
  • 27.Lee NY, de Arruda FF, Puri DR, et al. A comparison of intensity-modulated radiation therapy and concomitant boost radiotherapy in the setting of concurrent chemotherapy for locally advanced oropharyngeal carcinoma. Int J Radiat Oncol Biol Phys. 2006;66:966–74. doi: 10.1016/j.ijrobp.2006.06.040. [DOI] [PubMed] [Google Scholar]
  • 28.Yao M, Dornfeld KJ, Buatti JM, et al. Intensity-modulated radiation treatment for head-and-neck squamous cell carcinoma--the University of Iowa experience. Int J Radiat Oncol Biol Phys. 2005;63:410–21. doi: 10.1016/j.ijrobp.2005.02.025. [DOI] [PubMed] [Google Scholar]
  • 29.Kam MK, Chau RM, Suen J, et al. Intensity-modulated radiotherapy in nasopharyngeal carcinoma: dosimetric advantage over conventional plans and feasibility of dose escalation. Int J Radiat Oncol Biol Phys. 2003;56:145–57. doi: 10.1016/s0360-3016(03)00075-0. [DOI] [PubMed] [Google Scholar]
  • 30.Hunt MA, Zelefsky MJ, Wolden S, et al. Treatment planning and delivery of intensity-modulated radiation therapy for primary nasopharynx cancer. Int J Radiat Oncol Biol Phys. 2001;49:623–32. doi: 10.1016/s0360-3016(00)01389-4. [DOI] [PubMed] [Google Scholar]
  • 31.Kwong DL, Pow EH, Sham JS, et al. Intensity-modulated radiotherapy for early-stage nasopharyngeal carcinoma: a prospective study on disease control and preservation of salivary function. Cancer. 2004;101:1584–93. doi: 10.1002/cncr.20552. [DOI] [PubMed] [Google Scholar]
  • 32.Kam MK, Teo PM, Chau RM, et al. Treatment of nasopharyngeal carcinoma with intensity-modulated radiotherapy: the Hong Kong experience. Int J Radiat Oncol Biol Phys. 2004;60:1440–50. doi: 10.1016/j.ijrobp.2004.05.022. [DOI] [PubMed] [Google Scholar]
  • 33.Wolden SL, Chen WC, Pfister DG, et al. Intensity-modulated radiation therapy (IMRT) for nasopharynx cancer: update of the Memorial Sloan-Kettering experience. Int J Radiat Oncol Biol Phys. 2006;64:57–62. doi: 10.1016/j.ijrobp.2005.03.057. [DOI] [PubMed] [Google Scholar]
  • 34.Nagata Y, Takayama K, Matsuo Y, et al. Clinical outcomes of a phase I/II study of 48 Gy of stereotactic body radiotherapy in 4 fractions for primary lung cancer using a stereotactic body frame. Int J Radiat Oncol Biol Phys. 2005;63:1427–31. doi: 10.1016/j.ijrobp.2005.05.034. [DOI] [PubMed] [Google Scholar]
  • 35.Bucci M, XIa P, Lee N, et al. Intensity Modulated Radiation Therapy for Carcinoma of the Nasopharynx: An Update of the UCSF Experience. Int J Radiat Oncol Biol Phys. 2004;60:S317–318. [Google Scholar]
  • 36.Kyzas PA, Cunha IW, Ioannidis JP. Prognostic significance of vascular endothelial growth factor immunohistochemical expression in head and neck squamous cell carcinoma: a meta-analysis. Clin Cancer Res. 2005;11:1434–40. doi: 10.1158/1078-0432.CCR-04-1870. [DOI] [PubMed] [Google Scholar]
  • 37.Qian CN, Zhang CQ, Guo X, et al. Elevation of serum vascular endothelial growth factor in male patients with metastatic nasopharyngeal carcinoma. Cancer. 2000;88:255–61. [PubMed] [Google Scholar]
  • 38.Krishna SM, James S, Balaram P. Expression of VEGF as prognosticator in primary nasopharyngeal cancer and its relation to EBV status. Virus Res. 2006;115:85–90. doi: 10.1016/j.virusres.2005.07.010. [DOI] [PubMed] [Google Scholar]
  • 39.Lin A, Kim HM, Terrell JE, et al. Quality of life after parotid-sparing IMRT for head-and-neck cancer: a prospective longitudinal study. Int J Radiat Oncol Biol Phys. 2003;57:61–70. doi: 10.1016/s0360-3016(03)00361-4. [DOI] [PubMed] [Google Scholar]
  • 40.Eisbruch A, Marsh LH, Dawson LA, et al. Recurrences near base of skull after IMRT for head-and-neck cancer: implications for target delineation in high neck and for parotid gland sparing. Int J Radiat Oncol Biol Phys. 2004;59:28–42. doi: 10.1016/j.ijrobp.2003.10.032. [DOI] [PubMed] [Google Scholar]
  • 41.Chao KS, Ozyigit G, Blanco AI, et al. Intensity-modulated radiation therapy for oropharyngeal carcinoma: impact of tumor volume. Int J Radiat Oncol Biol Phys. 2004;59:43–50. doi: 10.1016/j.ijrobp.2003.08.004. [DOI] [PubMed] [Google Scholar]
  • 42.Studer G, Lutolf UM, Davis JB, et al. IMRT in Hypopharyngeal Tumors. Strahlenther Onkol. 2006;182:331–335. doi: 10.1007/s00066-006-1556-2. [DOI] [PubMed] [Google Scholar]
  • 43.Forastiere AA, Goepfert H, Maor M, et al. Concurrent chemotherapy and radiotherapy for organ preservation in advanced laryngeal cancer. N Engl J Med. 2003;349:2091–8. doi: 10.1056/NEJMoa031317. [DOI] [PubMed] [Google Scholar]
  • 44.Combs SE, Konkel S, Schulz-Ertner D, et al. Intensity modulated radiotherapy (IMRT) in patients with carcinomas of the paranasal sinuses: clinical benefit for complex shaped target volumes. Radiat Oncol. 2006;1:23. doi: 10.1186/1748-717X-1-23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Claus F, Mijnheer B, Rasch C, et al. Report of a study on IMRT planning strategies for ethmoid sinus cancer. Strahlenther Onkol. 2002;178:572–6. doi: 10.1007/s00066-002-0999-3. [DOI] [PubMed] [Google Scholar]
  • 46.Hoppe BS, Stegman LD, Zelefsky MJ, et al. Treatment of nasal cavity and paranasal sinus cancer with modern radiotherapy techniques in the postoperative setting--the MSKCC experience. Int J Radiat Oncol Biol Phys. 2007;67:691–702. doi: 10.1016/j.ijrobp.2006.09.023. [DOI] [PubMed] [Google Scholar]
  • 47.Daly ME, Chen AM, Bucci MK, et al. Intensity-modulated radiation therapy for malignancies of the nasal cavity and paranasal sinuses. Int J Radiat Oncol Biol Phys. 2007;67:151–7. doi: 10.1016/j.ijrobp.2006.07.1389. [DOI] [PubMed] [Google Scholar]
  • 48.Claus F, Boterberg T, Ost P, et al. Short term toxicity profile for 32 sinonasal cancer patients treated with IMRT. Can we avoid dry eye syndrome? 2002;64:205–8. doi: 10.1016/s0167-8140(02)00172-x. [DOI] [PubMed] [Google Scholar]
  • 49.Chen AM, Daly ME, Bucci MK, et al. Carcinomas of the paranasal sinuses and nasal cavity treated with radiotherapy at a single institution over five decades: Are we making improvement? Int J Radiat Oncol Biol Phys. 2007 doi: 10.1016/j.ijrobp.2007.02.031. [DOI] [PubMed] [Google Scholar]
  • 50.Nutting CM, Convery DJ, Cosgrove VP, et al. Improvements in target coverage and reduced spinal cord irradiation using intensity-modulated radiotherapy (IMRT) in patients with carcinoma of the thyroid gland. Radiother Oncol. 2001;60:173–80. doi: 10.1016/s0167-8140(01)00382-6. [DOI] [PubMed] [Google Scholar]
  • 51.Posner MD, Quivey JM, Akazawa PF, et al. Dose optimization for the treatment of anaplastic thyroid carcinoma: a comparison of treatment planning techniques. Int J Radiat Oncol Biol Phys. 2000;48:475–83. doi: 10.1016/s0360-3016(00)00621-0. [DOI] [PubMed] [Google Scholar]
  • 52.Rosenbluth BD, Serrano V, Hunt MA, et al. Intensity-modulated radiation therapy for the treatment of non-anaplastic thyroid cancer. Int J Radiat Oncol Biol Phys. 2005;63:1419–1426. doi: 10.1016/j.ijrobp.2005.05.043. [DOI] [PubMed] [Google Scholar]
  • 53.Chen YJ, Kuo JV, Ramsinghani NS, et al. Intensity-modulated radiotherapy for previously irradiated, recurrent head-and-neck cancer. Med Dosim. 2002;27:171–6. doi: 10.1016/s0958-3947(02)00100-0. [DOI] [PubMed] [Google Scholar]
  • 54.Lu TX, Mai WY, Teh BS, et al. Initial experience using intensity-modulated radiotherapy for recurrent nasopharyngeal carcinoma. Int J Radiat Oncol Biol Phys. 2004;58:682–7. doi: 10.1016/S0360-3016(03)01508-6. [DOI] [PubMed] [Google Scholar]
  • 55.Lee N, Chan K, Bekelman JE, et al. Salvage re-irradiation for recurrent head and neck cancer. Int J Radiat Oncol Biol Phys. 2007;68:731–40. doi: 10.1016/j.ijrobp.2006.12.055. [DOI] [PubMed] [Google Scholar]
  • 56.Tatum JL, Kelloff GJ, Gillies RJ, et al. Hypoxia: importance in tumor biology, noninvasive measurement by imaging, and value of its measurement in the management of cancer therapy. Int J Radiat Biol. 2006;82:699–757. doi: 10.1080/09553000601002324. [DOI] [PubMed] [Google Scholar]
  • 57.Gatenby RA, Kessler HB, Rosenblum JS, et al. Oxygen distribution in squamous cell carcinoma metastases and its relationship to outcome of radiation therapy. Int J Radiat Oncol Biol Phys. 1988;14:831–8. doi: 10.1016/0360-3016(88)90002-8. [DOI] [PubMed] [Google Scholar]
  • 58.Wendling P, Manz R, Thews G, et al. Heterogeneous oxygenation of rectal carcinomas in humans: a critical parameter for preoperative irradiation? Adv Exp Med Biol. 1984;180:293–300. doi: 10.1007/978-1-4684-4895-5_28. [DOI] [PubMed] [Google Scholar]
  • 59.Brizel DM, Dodge RK, Clough RW, et al. Oxygenation of head and neck cancer: changes during radiotherapy and impact on treatment outcome. Radiother Oncol. 1999;53:113–7. doi: 10.1016/s0167-8140(99)00102-4. [DOI] [PubMed] [Google Scholar]
  • 60.Nordsmark M, Overgaard J. A confirmatory prognostic study on oxygenation status and loco-regional control in advanced head and neck squamous cell carcinoma treated by radiation therapy. Radiother Oncol. 2000;57:39–43. doi: 10.1016/s0167-8140(00)00223-1. [DOI] [PubMed] [Google Scholar]
  • 61.Rudat V, Stadler P, Becker A, et al. Predictive value of the tumor oxygenation by means of pO2 histography in patients with advanced head and neck cancer. Strahlenther Onkol. 2001;177:462–8. doi: 10.1007/pl00002427. [DOI] [PubMed] [Google Scholar]
  • 62.Brizel DM, Prosnitz RG, Hunter S, et al. Necessity for adjuvant neck dissection in setting of concurrent chemoradiation for advanced head-and-neck cancer. Int J Radiat Oncol Biol Phys. 2004;58:1418–23. doi: 10.1016/j.ijrobp.2003.09.004. [DOI] [PubMed] [Google Scholar]
  • 63.Nordsmark M, Bentzen SM, Rudat V, 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. doi: 10.1016/j.radonc.2005.06.038. [DOI] [PubMed] [Google Scholar]
  • 64.Stone HB, Brown JM, Phillips TL, et al. Oxygen in human tumors: correlations between methods of measurement and response to therapy. Radiat Res. 1993;136:422–434. [PubMed] [Google Scholar]
  • 65.Raleigh JA, Calkins-Adams DP, Rinker LH, et al. Hypoxia and vascular endothelial growth factor expression in human squamous cell carcinomas using pimonidazole as a hypoxia marker. Cancer Res. 1998;58:3765–8. [PubMed] [Google Scholar]
  • 66.Evans SM, Hahn S, Pook DR, et al. Detection of hypoxia in human squamous cell carcinoma by EF5 binding. Cancer Res. 2000;60:2018–24. [PubMed] [Google Scholar]
  • 67.Varghese AJ, Gulyas S, Mohindra JK. Hypoxia-dependent reduction of 1-(2-nitro-1-imidazolyl)-3-methoxy-2-propanol by Chinese hamster ovary cells and KHT tumor cells in vitro and in vivo. Cancer Res. 1976;36:3761–5. [PubMed] [Google Scholar]
  • 68.Ljungkvist AS, Bussink J, Kaanders JH, et al. Dynamics of tumor hypoxia measured with bioreductive hypoxic cell markers. Radiat Res. 2007;167:127–45. doi: 10.1667/rr0719.1. [DOI] [PubMed] [Google Scholar]
  • 69.Evans SM, Koch CJ. Prognostic significance of tumor oxygenation in humans. Cancer Lett. 2003;195:1–16. doi: 10.1016/s0304-3835(03)00012-0. [DOI] [PubMed] [Google Scholar]
  • 70.Raleigh JA, Chou SC, Arteel GE, et al. Comparisons among pimonidazole binding, oxygen electrode measurements, and radiation response in C3H mouse tumors. Radiat Res. 1999;151:580–9. [PubMed] [Google Scholar]
  • 71.Kaanders JH, Wijffels KI, Marres HA, et al. Pimonidazole binding and tumor vascularity predict for treatment outcome in head and neck cancer. Cancer Res. 2002;62:7066–74. [PubMed] [Google Scholar]
  • 72.Stroka DM, Burkhardt T, Desbaillets I, et al. HIF-1 is expressed in normoxic tissue and displays an organ-specific regulation under systemic hypoxia. Faseb J. 2001;15:2445–53. doi: 10.1096/fj.01-0125com. [DOI] [PubMed] [Google Scholar]
  • 73.Zundel W, Schindler C, Haas-Kogan D, et al. Loss of PTEN facilitates HIF-1-mediated gene expression. Genes Dev. 2000;14:391–6. [PMC free article] [PubMed] [Google Scholar]
  • 74.Zelzer E, Levy Y, Kahana C, et al. Insulin induces transcription of target genes through the hypoxia-inducible factor HIF-1alpha/ARNT. Embo J. 1998;17:5085–94. doi: 10.1093/emboj/17.17.5085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Zhong H, Chiles K, Feldser D, et al. Modulation of hypoxia-inducible factor 1alpha expression by the epidermal growth factor/phosphatidylinositol 3-kinase/PTEN/AKT/FRAP pathway in human prostate cancer cells: implications for tumor angiogenesis and therapeutics. Cancer Res. 2000;60:1541–5. [PubMed] [Google Scholar]
  • 76.Janssen HL, Haustermans KM, Sprong D, et al. HIF-1A, pimonidazole, and iododeoxyuridine to estimate hypoxia and perfusion in human head-and-neck tumors. Int J Radiat Oncol Biol Phys. 2002;54:1537–49. doi: 10.1016/s0360-3016(02)03935-4. [DOI] [PubMed] [Google Scholar]
  • 77.Chi JT, Wang Z, Nuyten DS, et al. Gene Expression Programs in Response to Hypoxia: Cell Type Specificity and Prognostic Significance in Human Cancers. PLoS Med. 2006;3:e47. doi: 10.1371/journal.pmed.0030047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Winter SC, Buffa FM, Silva P, et al. Relation of a hypoxia metagene derived from head and neck cancer to prognosis of multiple cancers. Cancer Res. 2007;67:3441–9. doi: 10.1158/0008-5472.CAN-06-3322. [DOI] [PubMed] [Google Scholar]
  • 79.Le QT, Kong C, Lavori PW, et al. Expression and prognostic significance of a panel of tissue hypoxia markers in head and neck squamous cell carcinomas. Int J Radiat Oncol Biol Phys. 2007 doi: 10.1016/j.ijrobp.2007.01.071. in press. [DOI] [PubMed] [Google Scholar]
  • 80.Riedel F, Gotte K, Schwalb J, et al. Serum levels of vascular endothelial growth factor in patients with head and neck cancer. Eur Arch Otorhinolaryngol. 2000;257:332–6. doi: 10.1007/s004059900208. [DOI] [PubMed] [Google Scholar]
  • 81.Salven P, Manpaa H, Orpana A, et al. Serum vascular endothelial growth factor is often elevated in disseminated cancer. Clin Cancer Res. 1997;3:647–51. [PubMed] [Google Scholar]
  • 82.Imagawa S, Yamaguchi Y, Higuchi M, et al. Levels of vascular endothelial growth factor are elevated in patients with obstructive sleep apnea--hypopnea syndrome. Blood. 2001;98:1255–7. doi: 10.1182/blood.v98.4.1255. [DOI] [PubMed] [Google Scholar]
  • 83.Dunst J, Stadler P, Becker A, et al. Tumor hypoxia and systemic levels of vascular endothelial growth factor (VEGF) in head and neck cancers. Strahlenther Onkol. 2001;177:469–73. doi: 10.1007/pl00002428. [DOI] [PubMed] [Google Scholar]
  • 84.Le QT, Sutphin PD, Raychaudhuri S, et al. Identification of osteopontin as a prognostic plasma marker for head and neck squamous cell carcinomas. Clin Cancer Res. 2003;9:59–67. [PMC free article] [PubMed] [Google Scholar]
  • 85.Nordsmark M, Eriksen JG, Gebski V, et al. Differential risk assessments from five hypoxia specific assays: The basis for biologically adapted individualized radiotherapy in advanced head and neck cancer patients. Radiother Oncol. 2007 doi: 10.1016/j.radonc.2007.04.021. [DOI] [PubMed] [Google Scholar]
  • 86.Petrik D, Lavori PW, Cao H, et al. Plasma osteopontin is an independent prognostic marker for head and neck cancers. J Clin Oncol. 2006;24:5291–7. doi: 10.1200/JCO.2006.06.8627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Overgaard J, Eriksen JG, Nordsmark M, et al. Plasma osteopontin, hypoxia, and response to the hypoxia sensitiser nimorazole in radiotherapy of head and neck cancer: results from the DAHANCA 5 randomised double-blind placebo-controlled trial. Lancet Oncol. 2005;6:757–64. doi: 10.1016/S1470-2045(05)70292-8. [DOI] [PubMed] [Google Scholar]
  • 88.Overgaard J, Horsman MR. Modification of Hypoxia-Induced Radioresistance in Tumors by the Use of Oxygen and Sensitizers. Semin Radiat Oncol. 1996;6:10–21. doi: 10.1053/SRAO0060010. [DOI] [PubMed] [Google Scholar]
  • 89.Watson ER, Halnan KE, Dische S, et al. Hyperbaric oxygen and radiotherapy: a Medical Research Council trial in carcinoma of the cervix. Br J Radiol. 1978;51:879–87. doi: 10.1259/0007-1285-51-611-879. [DOI] [PubMed] [Google Scholar]
  • 90.Dische S, Anderson PJ, Sealy R, et al. Carcinoma of the cervix--anaemia, radiotherapy and hyperbaric oxygen. Br J Radiol. 1983;56:251–5. doi: 10.1259/0007-1285-56-664-251. [DOI] [PubMed] [Google Scholar]
  • 91.Sheldon PW, Foster JL, Fowler JF. Radiosensitization of C3H mouse mammary tumours by a 2-nitroimidazole drug. Br J Cancer. 1974;30:560–5. doi: 10.1038/bjc.1974.235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Brown JM. Selective radiosensitization of the hypoxic cells of mouse tumors with the nitroimidazoles metronidazole and Ro 7-0582. Radiat Res. 1975;64:633–47. [PubMed] [Google Scholar]
  • 93.Lee DJ, Pajak TF, Stetz J, et al. A phase I/II study of the hypoxic cell sensitizer misonidazole as an adjunct to high fractional dose radiotherapy in patients with unresectable squamous cell carcinoma of the head and neck: a RTOG randomized study (#79-04) Int J Radiat Oncol Biol Phys. 1989;16:465–70. doi: 10.1016/0360-3016(89)90343-x. [DOI] [PubMed] [Google Scholar]
  • 94.Lee DJ, Cosmatos D, Marcial VA, et al. Results of an RTOG phase III trial (RTOG 85-27) comparing radiotherapy plus etanidazole with radiotherapy alone for locally advanced head and neck carcinomas. Int J Radiat Oncol Biol Phys. 1995;32:567–76. doi: 10.1016/0360-3016(95)00150-W. [DOI] [PubMed] [Google Scholar]
  • 95.Overgaard J, Hansen HS, Anderson AP, et al. Misonidazole combined with split course radiotherapy in the treatment of invasive carcinoma of larynx and pharynx: report from the DAHANCA study. Int J Radiat Oncol Biol Phys. 1989;16:1065–1068. doi: 10.1016/0360-3016(89)90917-6. [DOI] [PubMed] [Google Scholar]
  • 96.Overgaard J, Hansen HS, Overgaard M, et al. A randomized double-blind phase III study of nimorazole as a hypoxic radiosensitizer of primary radiotherapy in supraglottic larynx and pharynx carcinoma. Results of the Danish Head and Neck Cancer Study (DAHANCA) Protocol 5–85. Radiother Oncol. 1998;46:135–46. doi: 10.1016/s0167-8140(97)00220-x. [DOI] [PubMed] [Google Scholar]
  • 97.Wasserman TH, Lee DJ, Cosmatos D, et al. Clinical trials with etanidazole (SR-2508) by the Radiation Therapy Oncology Group (RTOG) Radiother Oncol. 1991;20 Suppl 1:129–35. doi: 10.1016/0167-8140(91)90200-z. [DOI] [PubMed] [Google Scholar]
  • 98.Dische S, Saunders MI, Lee ME, et al. Clinical testing of the radiosensitizer Ro 07-0582: experience with multiple doses. Br J Cancer. 1977;35:567–79. doi: 10.1038/bjc.1977.90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Coleman CN, Wasserman TH, Urtasun RC, et al. Phase I trial of the hypoxic cell radiosensitizer SR-2508: the results of the five to six week drug schedule. Int J Radiat Oncol Biol Phys. 1986;12:1105–8. doi: 10.1016/0360-3016(86)90236-1. [DOI] [PubMed] [Google Scholar]
  • 100.Henke M, Laszig R, Rube C, et al. Erythropoietin to treat head and neck cancer patients with anaemia undergoing radiotherapy: randomised, double-blind, placebo-controlled trial. Lancet. 2003;362:1255–60. doi: 10.1016/S0140-6736(03)14567-9. [DOI] [PubMed] [Google Scholar]
  • 101.Henke M, Mattern D, Pepe M, et al. Do erythropoietin receptors on cancer cells explain unexpected clinical findings? J Clin Oncol. 2006;24:4708–13. doi: 10.1200/JCO.2006.06.2737. [DOI] [PubMed] [Google Scholar]
  • 102.Jain RK. Normalization of tumor vasculature: an emerging concept in antiangiogenic therapy. Science. 2005;307:58–62. doi: 10.1126/science.1104819. [DOI] [PubMed] [Google Scholar]
  • 103.Willett CG, Boucher Y, di Tomaso E, et al. Direct evidence that the VEGF-specific antibody bevacizumab has antivascular effects in human rectal cancer. Nat Med. 2004;10:145–7. doi: 10.1038/nm988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Taghian AG, Abi-Raad R, Assaad SI, et al. Paclitaxel decreases the interstitial fluid pressure and improves oxygenation in breast cancers in patients treated with neoadjuvant chemotherapy: clinical implications. J Clin Oncol. 2005;23:1951–61. doi: 10.1200/JCO.2005.08.119. [DOI] [PubMed] [Google Scholar]
  • 105.Batchelor TT, Sorensen AG, di Tomaso E, et al. AZD2171, a pan-VEGF receptor tyrosine kinase inhibitor, normalizes tumor vasculature and alleviates edema in glioblastoma patients. Cancer Cell. 2007;11:83–95. doi: 10.1016/j.ccr.2006.11.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Kaanders JH, Pop LA, Marres HA, et al. ARCON: experience in 215 patients with advanced head-and-neck cancer. Int J Radiat Oncol Biol Phys. 2002;52:769–78. doi: 10.1016/s0360-3016(01)02678-5. [DOI] [PubMed] [Google Scholar]
  • 107.Haffty BG, Son YH, Papac R, et al. Chemotherapy as an adjunct to radiation in the treatment of squamous cell carcinoma of the head and neck: results of the Yale Mitomycin Randomized Trials. J Clin Oncol. 1997;15:268–76. doi: 10.1200/JCO.1997.15.1.268. [DOI] [PubMed] [Google Scholar]
  • 108.Dobrowsky W, Naude J, Widder J, et al. Continuous hyperfractionated accelerated radiotherapy with/without mitomycin C in head and neck cancer. Int J Radiat Oncol Biol Phys. 1998;42:803–6. doi: 10.1016/s0360-3016(98)00321-6. [DOI] [PubMed] [Google Scholar]
  • 109.Dobrowsky W, Naude J. Continuous hyperfractionated accelerated radiotherapy with/without mitomycin C in head and neck cancers. Radiother Oncol. 2000;57:119–24. doi: 10.1016/s0167-8140(00)00233-4. [DOI] [PubMed] [Google Scholar]
  • 110.Rischin D, Peters L, Fisher R, et al. Tirapazamine, Cisplatin, and Radiation versus Fluorouracil, Cisplatin, and Radiation in patients with locally advanced head and neck cancer: a randomized phase II trial of the Trans-Tasman Radiation Oncology Group (TROG 98.02) J Clin Oncol. 2005;23:79–87. doi: 10.1200/JCO.2005.01.072. [DOI] [PubMed] [Google Scholar]
  • 111.Le QT, Taira A, Budenz S, et al. Mature results from a randomized Phase II trial of cisplatin plus 5-fluorouracil and radiotherapy with or without tirapazamine in patients with resectable Stage IV head and neck squamous cell carcinomas. Cancer. 2006;106:1940–9. doi: 10.1002/cncr.21785. [DOI] [PubMed] [Google Scholar]
  • 112.Wilson WR, Hicks KO, Pullen SM, et al. Bystander effects of bioreductive drugs: potential for exploiting pathological tumor hypoxia with dinitrobenzamide mustards. Radiat Res. 2007;167:625–36. doi: 10.1667/RR0807.1. [DOI] [PubMed] [Google Scholar]
  • 113.Patterson AV, Ferry DM, Edmunds SJ, et al. Mechanism of action and preclinical antitumor activity of the novel hypoxia-activated DNA crosslinking agent PR-104. Clin Can Res. 2007 doi: 10.1158/1078-0432.CCR-07-0478. in press. [DOI] [PubMed] [Google Scholar]
  • 114.Giaccia A, Siim BG, Johnson RS. HIF-1 as a target for drug development. Nat Rev Drug Discov. 2003;2:803–11. doi: 10.1038/nrd1199. [DOI] [PubMed] [Google Scholar]
  • 115.Melillo G. Targeting hypoxia cell signaling for cancer therapy. Cancer Metastasis Rev. 2007 doi: 10.1007/s10555-007-9059-x. [DOI] [PubMed] [Google Scholar]
  • 116.Rapisarda A, Uranchimeg B, Scudiero DA, et al. Identification of small molecule inhibitors of hypoxia-inducible factor 1 transcriptional activation pathway. Cancer Res. 2002;62:4316–24. [PubMed] [Google Scholar]
  • 117.Mabjeesh NJ, Escuin D, LaVallee TM, et al. 2ME2 inhibits tumor growth and angiogenesis by disrupting microtubules and dysregulating HIF. Cancer Cell. 2003;3:363–75. doi: 10.1016/s1535-6108(03)00077-1. [DOI] [PubMed] [Google Scholar]
  • 118.Mabjeesh NJ, Post DE, Willard MT, et al. Geldanamycin induces degradation of hypoxia-inducible factor 1alpha protein via the proteosome pathway in prostate cancer cells. Cancer Res. 2002;62:2478–82. [PubMed] [Google Scholar]
  • 119.Kong X, Lin Z, Liang D, et al. Histone deacetylase inhibitors induce VHL and ubiquitin-independent proteasomal degradation of hypoxia-inducible factor 1alpha. Mol Cell Biol. 2006;26:2019–28. doi: 10.1128/MCB.26.6.2019-2028.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Yeo EJ, Chun YS, Cho YS, et al. YC-1: a potential anticancer drug targeting hypoxia-inducible factor 1. J Natl Cancer Inst. 2003;95:516–25. doi: 10.1093/jnci/95.7.516. [DOI] [PubMed] [Google Scholar]
  • 121.Welsh S, Williams R, Kirkpatrick L, et al. Antitumor activity and pharmacodynamic properties of PX-478, an inhibitor of hypoxia-inducible factor-1alpha. Mol Cancer Ther. 2004;3:233–44. [PubMed] [Google Scholar]
  • 122.Welsh S, Williams R, Birmingham A, et al. The Thioredoxin Redox Inhibitors 1-Methylpropyl 2-Imidazolyl Disulfide and Pleurotin Inhibit Hypoxia-induced Factor 1α and Vascular Endothelial Growth Factor Formation. Mol Cancer Ther. 2003;2:235–243. [PubMed] [Google Scholar]
  • 123.Melillo G, Sausville EA, Cloud K, et al. Flavopiridol, a protein kinase inhibitor, down-regulates hypoxic induction of vascular endothelial growth factor expression in human monocytes. Cancer Res. 1999;59:5433–7. [PubMed] [Google Scholar]
  • 124.Cairns RA, Papandreou I, Sutphin PD, et al. Metabolic targeting of hypoxia and HIF1 in solid tumors can enhance cytotoxic chemotherapy. Proc Natl Acad Sci U S A. 2007;104:9445–50. doi: 10.1073/pnas.0611662104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Moeller BJ, Cao Y, Li CY, et al. Radiation activates HIF-1 to regulate vascular radiosensitivity in tumors: role of reoxygenation, free radicals, and stress granules. Cancer Cell. 2004;5:429–41. doi: 10.1016/s1535-6108(04)00115-1. [DOI] [PubMed] [Google Scholar]
  • 126.Aebersold DM, Burri P, Beer KT, et al. Expression of hypoxia-inducible factor-1alpha: a novel predictive and prognostic parameter in the radiotherapy of oropharyngeal cancer. Cancer Res. 2001;61:2911–6. [PubMed] [Google Scholar]
  • 127.Koukourakis MI, Giatromanolaki A, Sivridis E, et al. Hypoxia-inducible factor (HIF1A and HIF2A), angiogenesis, and chemoradiotherapy outcome of squamous cell head-and-neck cancer. Int J Radiat Oncol Biol Phys. 2002;53:1192–202. doi: 10.1016/s0360-3016(02)02848-1. [DOI] [PubMed] [Google Scholar]
  • 128.Beasley NJ, Leek R, Alam M, et al. Hypoxia-inducible factors HIF-1alpha and HIF-2alpha in head and neck cancer: relationship to tumor biology and treatment outcome in surgically resected patients. Cancer Res. 2002;62:2493–7. [PubMed] [Google Scholar]
  • 129.Hui EP, Chan AT, Pezzella F, et al. Coexpression of hypoxia-inducible factors 1alpha and 2alpha, carbonic anhydrase IX, and vascular endothelial growth factor in nasopharyngeal carcinoma and relationship to survival. Clin Cancer Res. 2002;8:2595–604. [PubMed] [Google Scholar]
  • 130.Beasley NJ, Wykoff CC, Watson PH, et al. Carbonic anhydrase IX, an endogenous hypoxia marker, expression in head and neck squamous cell carcinoma and its relationship to hypoxia, necrosis, and microvessel density. Cancer Res. 2001;61:5262–7. [PubMed] [Google Scholar]
  • 131.Koukourakis MI, Giatromanolaki A, Sivridis E, et al. Hypoxia-regulated carbonic anhydrase-9 (CA9) relates to poor vascularization and resistance of squamous cell head and neck cancer to chemoradiotherapy. Clin Cancer Res. 2001;7:3399–403. [PubMed] [Google Scholar]
  • 132.Winter SC, Shah KA, Han C, et al. The relation between hypoxia-inducible factor (HIF)-1alpha and HIF-2alpha expression with anemia and outcome in surgically treated head and neck cancer. Cancer. 2006;107:757–66. doi: 10.1002/cncr.21983. [DOI] [PubMed] [Google Scholar]
  • 133.Koukourakis MI, Bentzen SM, Giatromanolaki A, et al. Endogenous markers of two separate hypoxia response pathways (hypoxia inducible factor 2 alpha and carbonic anhydrase 9) are associated with radiotherapy failure in head and neck cancer patients recruited in the CHART randomized trial. J Clin Oncol. 2006;24:727–35. doi: 10.1200/JCO.2005.02.7474. [DOI] [PubMed] [Google Scholar]

RESOURCES