Онкологический центр лечения перитонеального карциноза

Специализированный центр лечения канцероматоза брюшины.
Хирургическая онкология. Регионарная химиотерапия.

Head and Neck Cancer

Arlene Forastiere, M.D., Wayne Koch, M.D., Andrew Trotti, M.D., and David Sidransky, M.D.


Since the subject was last reviewed in the Journal, there have been important advances in the treatment of locally advanced head and neck squamous-cell carcinoma. These new approaches incorporate chemotherapy into initial curative treatment to achieve organ preservation and to improve survival. Moreover, progress in the elucidation of the molecular genetic changes that lead to the development of these tumors should soon bring novel diagnostic and therapeutic procedures into clinical practice. This review will highlight these important advances in the treatment of patients with head and neck cancer (Table 1) and emphasize the ways in which molecular biology is likely to affect the development of future therapies.

07t1

Table 1. Head and Neck Cancer in the United States, 1975–1998.
 
Molecular Progression of Head and Neck Cancer
Neoplasms arise clonally from transformed cells that have undergone specific genetic alterations in proto-oncogenes or tumor-suppressor genes (Table 2). Tabulation of the critical genetic changes in each step of the progression of head and neck cancer from preneoplastic lesions to invasive cancer has allowed the delineation of a model of molecular progression. Loss of chromosomal region 9p21 is the most common of all genetic changes and occurs early in the progression of these tumors. The main effect of this loss is the inactivation of the p16 gene, an inhibitor of cyclin-dependent kinase (CDK) that is important in regulating the cell cycle. This early inactivation is consistent with the finding that keratinocytes in culture often lose p16 function and thus escape senescence. Approximately half of all head and neck cancers contain a mutation of the p53 gene located at 17p13. The loss of p53 function due to a mutation results in a progression from preinvasive to invasive lesions and increases the likelihood of further genetic progression. Amplification of the oncogene cyclin D1, which constitutively activates cell-cycle progression, is seen in about a third of all tumors and is usually associated with invasive disease. Tumor-suppressor genes have not been isolated or characterized for most of the regions that are commonly lost in these tumors.

Table 2. Common Oncogene Alterations Identified in Primary Head and Neck Squamous-Cell Carcinoma.
 
The model of the molecular progression of head and neck squamous-cell carcinoma demonstrates that tissue with a normal or benign appearance (i.e., minimal dysplasia) can also contain clonal genetic changes (Figure 1). Several studies of head and neck cancer suggest that early genetic changes do not necessarily correlate with observable changes in morphology. Thus, testing for these genetic alterations or ploidy in early lesions may identify patients who are at the greatest risk for progression and lead to more definitive therapy.

Figure 1. Clinical, Pathological, and Molecular Progression of Oral Cancer.
Panel A shows a typical clinical presentation of oral cancer. Benign squamous hyperplasia can often appear similar to normal mucosa. Novel molecular approaches have yielded considerable understanding of the field-cancerization hypothesis originally proposed by Slaughter and colleagues. In most patients, cellular repopulation in geographically distinct areas gives rise to multiple clinical lesions. Although these lesions may have different histopathological patterns, as shown, they are often clonally related, arising from the same cell. The progression from normal-appearing mucosa to invasive cancer is depicted in Panel B. Normal-appearing mucosa already harbors early genetic changes (Panel C), often with loss of 9p21 and inactivation of p16. Further clinical progression through dysplasia is associated with further genetic changes. Carcinoma in situ often harbors most of the genetic changes described in invasive carcinoma. LOH denotes loss of heterozygosity. (Courtesy of Joseph A. Califano.)
 
Some patients present with cervical nodal metastases from clinically occult primary mucosal lesions. Many of those occult primary tumors are later found within the lymphoepithelium of Waldeyer's ring, especially in the tonsils and the base of the tongue. One recent study demonstrated that clonal genetic changes could be detected in specimens from directed biopsies in patients with this presentation. In more than half the patients, at least one pathologically benign biopsy specimen demonstrated a pattern of genetic alterations identical to that present in the cells from the cervical lymph-node metastases. These data support the notion that pathologically benign mucosa harbors patches of clonal cells that can give rise to metastatic lesions of squamous-cell carcinoma.
Molecular Epidemiology
Squamous-cell carcinoma of the head and neck is a heterogeneous disease with distinct patterns of presentation and behavior. More than 50 percent of these cancers arising in the oropharynx, particularly in the palatine tonsils and the base of the tongue, contain oncogenic human papillomavirus (HPV) DNA. In a recent study, patients who were HPV-positive were less likely to have tumors that harbored a p53 mutation. These patients had better overall survival and disease-specific survival than HPV-negative patients. These results confirm a large body of earlier literature and suggest that HPV-positive oropharyngeal tumors compose a distinct clinical and pathological disease entity caused at least in part by HPV (Figure 2).

Figure 2. Human Papillomavirus (HPV) in Oropharyngeal Cancers.
Recent studies confirm that oropharyngeal tumors are often HPV-positive and compose a distinct clinical and pathologic disease entity. In Panel A, a typical large tonsillar lesion (arrows) is shown. Panel B shows the typical basaloid appearance often seen in HPV-positive tumors. In Panel C, the same tissue section was subjected to in situ hybridization with an HPV-E7–specific probe. The dark brown spots indicate the presence of HPV DNA in virtually all the neoplastic cells. (Courtesy of Wayne M. Koch and William H. Westra.)
 
Similarly, the association of head and neck squamous-cell carcinoma with alcohol and tobacco consumption has been clear for many years. Molecular data provide evidence that the carcinogens found in these substances have a causal role, in that the prevalence and spectrum of p53 mutations are significantly greater in cancers in patients who smoke and drink alcohol than in those in patients who abstain from these substances. Laryngeal, hypopharyngeal, and floor-of-mouth cancers rarely develop in patients who do not smoke, whereas the lateral border of the tongue is a common site for head and neck cancer in this nonsmoking group. Nonsmokers are more likely to present with head and neck squamous-cell carcinoma at a relatively young or relatively old age as compared with their smoking counterparts.
Patients with primary cancers who often smoke and drink heavily may present with multiple precancerous and cancerous lesions, so-called field cancerization, throughout the upper aerodigestive tract. Second cancers develop in nonsmokers and smokers with similar frequency, but patients with second primary head and neck squamous-cell carcinomas are much more likely to have abused tobacco and alcohol. Molecular evidence indicates that some, if not all, multiple primary cancers within the upper aerodigestive tract derive from a common clonal progenitor cell that undergoes a common early molecular alteration (Figure 1). This phenomenon has driven pioneering efforts to prevent recurrences through the use of retinoids and related compounds during the past two decades. Novel biomarkers, including the retinoic acid receptor, are promising as indicators of biologic response, but the morphologic regression of these lesions does not necessarily correlate with the abrogation of genetic changes.
Diagnosis
Studies have shown that clonal genetic alterations can be identified in blood and other bodily fluids. With the use of a panel of 21 microsatellite markers, clonal genetic changes (loss of heterozygosity, microsatellite instability, or both) were detected in 80 percent of saliva samples from patients with head and neck cancer. DNA from these exfoliated cell samples harbors genetic changes identical to those observed in the primary tumors, whereas samples from smokers without cancer as well as from other controls were negative according to this type of molecular analysis. Thus, a noninvasive test for specific DNA-sequence variants in saliva may someday be useful in identifying either early lesions or patients with cancer.
Clonal genetic changes identical to those found in primary head and neck squamous-cell carcinomas have been identified in circulating plasma or serum; these changes may be useful as a means of monitoring patients with head and neck cancer. For example, Epstein–Barr virus (EBV) has been stongly linked to the development of nasopharyngeal carcinoma. EBV DNA has been found in the serum of patients with nasopharyngeal carcinoma and HPV in a subgroup of patients with other head and neck cancers.The results of quantitative analysis of EBV DNA correlated precisely with disease status and were therefore useful for monitoring and tracking patients. Some tumors do not harbor viral markers but do display promoter hypermethylation, a mechanism for inactivating tumor-suppressor genes; this methylation was detected in the saliva and serum of patients with head and neck cancer and precisely matched the methylation patterns seen in the primary tumors. A panel of such molecular markers could be used to detect early cancer, to detect minimal residual disease, and to help in the monitoring of patients for early recurrence.
Staging
The staging of head and neck squamous-cell carcinoma has changed very little in the past decade. Traditional staging methods are weak, and several other systems, including the rating of symptom severity and coexisting conditions by means of the standard tumor–node–metastasis staging method, have been proposed. Computed tomography and magnetic resonance imaging have been universally accepted as vital tools for clinical staging, and newer imaging approaches, including positron-emission tomography and the identification of sentinel nodes by means of lymphoscintigraphy, show promise.
The concept of molecular staging has also been introduced. Genetic alterations can be used to detect rare cancer cells in samples with normal histologic appearance, including lymph nodes and tissue margins at the periphery of the tumor. The molecular analysis of tissue from the margins of head and neck squamous-cell carcinoma has been shown to predict the likelihood of tumor recurrence and to allow determination of the relation between an index lesion of head and neck squamous-cell carcinoma and subsequent lesions. Information derived from these molecular studies may assist clinicians in determining whether to treat a second lesion aggressively with curative intent or to take a palliative approach in the case of distant metastatic disease.
Surgical Innovation
The focus of surgical innovation during the past decade has been on the conservation of organ function and more effective means of reconstruction.
Larynx
In the management of cancer of the larynx, new surgical procedures have extended the amount of laryngeal tissue that can be resected while preserving the ability of the larynx to protect against aspiration and to produce sound. In supracricoid laryngectomy, nearly the entire larynx is removed, and only the arytenoids and the cricoid cartilage are preserved. These structures, together with the intact base of the tongue, permit the recovery of a breathy but naturally produced voice, an intact airway, and safe deglutition in many cases. Studies of quality of life are needed in order to make direct comparisons between the functional outcomes of these surgical approaches and those of nonsurgical approaches that preserve the larynx.
Neck Dissection
The debilitating effect of neck dissection on shoulder and neck function is diminished with less radical procedures such as modified comprehensive neck dissection and selective dissection. Selective neck dissection, in which only the anatomical regions that are most likely to contain involved nodes are removed, has been used with increasing frequency. The nodal status in patients with head and neck squamous-cell carcinoma has been established with the use of radionucleotide-tagged technetium injected into the vicinity of a primary mucosal lesion in order to identify the first-echelon lymph nodes that drain that region and permit selective resection.
Reconstruction
The reconstruction of tissue to correct defects caused by surgery has been improved with the use of tissue whose characteristics are similar to those of the native organ. For example, radial forearm skin provides enough pliable tissue to reline defects of the oral cavity and pharynx in a manner that does not greatly impede the ability of the remaining musculature to perform articulatory and gustatory functions. Reconstruction for mandibular defects may now be reliably performed with flaps taken from the iliac crest or fibula, with excellent aesthetic and functional results. Oral alimentation often can be resumed after partial or total glossectomy in patients in whom the tongue bulk was replaced with the use of microvascular flaps from the rectus abdominus muscles.
Radiotherapy
Altered Fractionation
There are two different approaches to parceling a dose of radiation over time — a procedure known as fractionation; these approaches are hyperfractionation and accelerated fractionation. In hyperfractionation, small-dose fractions are delivered two to three times per day in order to take advantage of the more limited damage to normal tissue that is associated with smaller fractions. This approach has often been combined with an escalation of the total dose by 10 to 15 percent over that given with standard fractionation. The accelerated fractionation approach aims to deliver treatment at a rate of accumulation of the weekly dose that is 20 to 50 percent faster than that of standard fractionation; this acceleration substantially shortens the overall duration of treatment in order to reduce the potential for tumor repopulation that may occur between fractions.
A number of phase 3 trials testing both of these approaches have now documented a 10 to 15 percent improvement in local and regional tumor control in patients with middle- and advanced-stage disease without a significant change in overall survival. Although gains in tumor control have been moderate, these trials have confirmed important radiobiologic principles. Selected schedules of both hyperfractionation and accelerated fractionation have now been adopted as treatment options at many large cancer centers and in the community at large.
Radiation Sensitizers
The presence of oxygen is critical to the ability of radiation to damage DNA and kill cancer cells. Since most head and neck tumors are both acutely and chronically hypoxic, they are relatively resistant to the effects of radiation. Hypoxic-cell radiation sensitizers, such as misonidazole and etanidazole, have been tried but in general do not enhance tumor control. Nimorazole was tested in a randomized trial by the Danish Head and Neck Cancer Study Group and led to improved local and regional control in supraglottic and pharyngeal cancers. The bulk of such trials, however, have been disappointing because they have been unable to demonstrate improvements that translate into improved survival. Sensitizers activated by hypoxia are currently being tested. Other approaches under investigation are the use of sensitizing chemotherapy or growth factors that augment the availability of oxygen in hemoglobin (unpublished data).
Postoperative Radiotherapy
The most common approach to the management of advanced head and neck cancer has traditionally been resection and postoperative radiotherapy, but survival and rates of local recurrence have been disappointing. A number of novel approaches are being tried, including the use of risk profiles to define specific target populations at high risk for recurrence and altered fractionation of postoperative radiotherapy. Another strategy to reduce clonal repopulation in the postoperative setting is the use of sensitizing chemotherapy concurrently with radiation.
Repeated Irradiation
A previously held dogma that normal tissues can only tolerate a specific maximal lifetime dose of radiation has been challenged by studies in which second doses of radiation of up to twice the expected tolerance of normal tissue have been given with a surprisingly low incidence of serious toxic effects. These clinical data support laboratory investigations that have demonstrated that normal tissue may recover in time, suggesting that it may be possible to give additional doses of radiation in many types of tissue. This approach is currently under investigation by multiple clinical-trial groups.
Treatment of Patients with Recurrent and Metastatic Disease
The goals of treatment for patients for whom cure is no longer an option include prolonging life with various cytotoxic regimens that may achieve measurable reductions in tumor and providing palliation through control of symptoms. Many patients with locally recurrent or metastatic squamous-cell carcinoma of the head and neck have substantial coexisting disease as a consequence of age and tobacco and alcohol abuse, which makes the use of cytotoxic drugs problematic. Tumor recurrence at the primary site in the neck or the base of the skull may result in cranial-nerve dysfunction, pain, impaired speech and swallowing, and airway compromise; the provision of pain control, symptom management, and psychosocial services is integral to optimal palliative care for patients with recurrent or metastatic head and neck cancer.
The cytotoxic drugs most commonly used to treat head and neck cancer are methotrexate, cisplatin, carboplatin, fluorouracil, paclitaxel, and docetaxel. The rate of response, defined as a decrease of at least 50 percent in the dimensions of measurable tumors for a duration of at least one month, is influenced by the amount of previous treatment, the tumor burden, and the patient's performance status. In general, 15 to 30 percent of patients have brief responses to these single agents.
The combination of cisplatin and fluorouracil given by continuous intravenous infusion is the mainstay of treatment. Prospective, multicenter, randomized trials in patients with recurrent disease have shown significantly higher response rates in patients treated with this combination than in those treated with single agents, but there have been no differences in the median duration of survival or the overall survival rates. Randomized trials directly comparing newer regimens, such as paclitaxel or docetaxel plus cisplatin or carboplatin, with the standard cisplatin-and-fluorouracil regimen are in progress.
Novel Therapies
New agents, including agents that inhibit signal transduction, cell-cycle traversal, programmed cell death, the regulation of transcription, matrix invasion, and angiogenesis, are in development. Therapeutic agents targeted specifically to patients with head and neck cancer include the family of tyrosine kinase inhibitors — in particular, the epidermal growth factor–receptor antagonists, CDK (cyclin-dependent kinase) inhibitors, and replication-competent adenoviruses. More than 90 percent of squamous-cell head and neck cancers overexpress the epidermal growth factor receptor, a transmembrane glycoprotein encoded by c-erbB. Transforming growth factor , epidermal growth factor, and other growth factors bind to the extracellular domain of the epidermal growth factor receptor, stimulating tumor growth through autocrine and paracrine pathways. Thus, monoclonal antibodies directed against epidermal growth factor receptor could block ligand binding and inhibit cell proliferation. The chimeric IgG antibody C225, which has binding affinity equal to that of the natural ligand, can effectively block the effect of epidermal growth factor and transforming growth factor in patients. Enhanced cytotoxicity has been observed when C225 is used in combination with a number of conventional cytotoxic therapies, including cisplatin and paclitaxel. When used in combination with radiotherapy, C225 also enhances the effect of killing tumor cells by inhibiting tumor-cell repopulation. A number of small-molecule drugs that block the epidermal growth factor receptor are in phase 1 and 2 testing.
CDKs phosphorylate key substances that regulate the transitions between cell-cycle phases. Cyclin D is commonly overexpressed in head and neck cancer, and p16, the endogenous inhibitor of CDK4, is commonly deleted or not transcribed. In preclinical testing, the CDK inhibitor flavopiridol repressed the transcription of cyclin D, induced cell-cycle arrest at the transitions between the G2 and M phases and between the G1 and S phases, and induced p53-independent apoptosis. Clinical trials with flavopiridol in head and neck cancer and other solid tumors are in progress. When administered in preclinical studies, another CDK inhibitor, CCI-779 (a structural analogue of the macrocyclic lactone sirolimus, previously called rapamycin), decreased the kinase activity of the CDK4–cyclin D complex in a p53-independent fashion.
Replication-competent adenoviruses have been developed that selectively replicate in and cause lysis of cells deficient in p53 tumor-suppressor activity. An example is ONYX-015, which is constructed with a deletion of the E1b protein and will not proliferate in normal cells but has the capacity to replicate in tumor cells that are deficient in p53. Phase 1 and 2 clinical trials of the intralesional injection of ONYX-015 alone or in combination with systemic chemotherapy suggest that it is effective and that the responses at injected tumor sites are durable.
Combination Therapy
Three strategies for combining chemotherapy with surgery and radiotherapy are induction chemotherapy, which consists of several courses of chemotherapy before surgery or radiotherapy; the concurrent administration of chemotherapy and radiotherapy; and adjuvant chemotherapy administered after the patient has been rendered free of disease.
Cancers of the Larynx and Hypopharynx
More than a decade ago, induction chemotherapy followed by radiotherapy became a standard treatment option for patients with advanced cancer of the larynx who wanted to preserve their voice. This practice was based on the Veterans Affairs Laryngeal Cancer Study Group trial published in the Journal in 1991. More than 300 patients with stage III or IV cancer of the larynx were randomly assigned to receive induction chemotherapy with cisplatin and fluorouracil followed by radiotherapy (in patients with a response to the chemotherapy) or total laryngectomy and postoperative radiation. In the chemotherapy group, surgery was reserved for patients with no response to the induction chemotherapy and for those with persistent or recurrent disease after radiotherapy. A functioning larynx was preserved in 62 percent of surviving patients treated with chemotherapy, and survival in the chemotherapy group was not significantly different from that in the surgery group. A similarly designed trial in Europe involving patients with cancer of the hypopharynx showed equivalent survival in the surgery group and the induction-chemotherapy group and a rate of larynx preservation of 42 percent. These trials established induction therapy with cisplatin and fluorouracil followed by radiotherapy as an alternative standard of care to preserve the larynx when total laryngectomy would otherwise have been required.
The precise role of chemotherapy, however, could not be determined from these studies, which led to a follow-up trial (R91-11) conducted by the Head and Neck Intergroup in the United States. This trial compared induction chemotherapy, radiotherapy alone, and radiotherapy with concurrent cisplatin. There was a significant advantage of radiotherapy with concurrent cisplatin treatment, with 88 percent of patients preserving their larynx at two years, as compared with 74 percent of those treated with induction chemotherapy (P=0.005) and 69 percent of those treated with radiotherapy alone. These results represent a reduction of nearly 50 percent in the rate of laryngectomy with concurrent treatment as compared with the previous standard of care, and the study has established this approach as the preferred alternative to surgery for patients who desire larynx preservation. Because concurrent treatment is associated with more severe toxic effects, however, it is appropriate to recommend radiotherapy alone for patients with a poor performance status or a limited psychosocial support system.
Cancer of the Oropharynx
The majority of oropharyngeal cancers originate from the base of the tongue and the tonsils, and more than 90 percent are squamous-cell carcinomas. Randomized, controlled trials in patients with locally advanced oropharyngeal cancer have evaluated platinum-based chemotherapy given concurrently with radiotherapy, delivered according to either a standard or an altered fractionation schedule. These studies have demonstrated that such concurrent chemotherapy results in significant improvements in local control, relapse-free survival, and overall survival as compared with radiotherapy alone (Table 3).

Table 3. Randomized Trials of Concurrent Multiagent Chemotherapy and Radiotherapy as Compared with Radiotherapy Alone in Stage III or IV Disease.
 
Calais and colleagues reported a randomized trial involving 222 patients with stage III or IV cancer of the oropharynx who were treated with either radiotherapy alone (with standard fractionation) or the same radiotherapy delivered concurrently with three courses of chemotherapy with carboplatin and fluorouracil. This study and others demonstrating an improvement in survival with concurrent chemoradiotherapy report differences in local and regional control of 20 to 30 percentage points, which are sufficiently large to alter the likelihood of survival.
These data support the use of concurrent chemotherapy and radiotherapy as the standard of care for the treatment of stage III and IV cancers of the oropharynx when nonsurgical treatment is planned. No randomized trials have been performed comparing the combination of chemotherapy and radiotherapy with surgery. Moreover, these complex treatments have substantial toxicity and are therefore not suitable for all patients. A good performance status, a strong psychosocial support system, and an experienced team of providers are essential for the successful completion of treatment.
Locally Advanced, Unresectable Disease
Patients whose disease cannot be resected with an expectation of achieving tumor-free margins have until recently been treated with radiotherapy alone (Table 3). All of the trials comparing concurrent chemotherapy and radiotherapy with radiotherapy alone have demonstrated statistically significant differences in local and regional control and disease-free survival.
Concurrent chemoradiotherapy is superior to radiotherapy alone and should be the accepted standard of care for patients with locally advanced, unresectable squamous-cell carcinoma of all sites in the head and neck. We make this recommendation with the caveat that patients must have adequate performance status and the support necessary for dealing with the effects of this more complex and toxic therapy. Otherwise, radiotherapy alone is a more appropriate form of care.
Nasopharyngeal Carcinoma
Poorly differentiated or undifferentiated nasopharyngeal carcinoma differs from squamous-cell carcinoma of other sites in the head and neck. The rates of local control are high, but distant recurrence is common and is often the cause of death. Autopsy series document distant metastases in 87 percent of patients, mostly to bone, followed in order of frequency by lung, liver, and extraregional nodes. Patients with bulky regional nodes, bilateral involvement, or nodes low in the neck are at greatest risk.
Important advances in treatment have emerged from randomized trials conducted over the past decade. Concurrent chemoradiotherapy has proved superior to the previous standard of care of radiotherapy alone for the treatment of patients with T3, T4, or N1, N2, or N3 disease. Strong positive results in terms of survival — strong enough to cause the trial to be stopped after the first interim analysis — were reported by the Head and Neck Intergroup in its comparison of radiotherapy alone with concurrent cisplatin and radiotherapy followed by three courses of adjuvant cisplatin and fluorouracil. These results led to a change in the standard of care to concurrent treatment. The applicability of these trial results to ethnic groups in which the disease is endemic, such as Asians, is unclear.
Postoperative Adjuvant Chemotherapy
The use of postoperative adjuvant chemotherapy for patients who are at high risk for local and regional recurrence and death from cancer is under evaluation in randomized, controlled trials in both the United States and Europe. Patients with two or more positive regional nodes, extracapsular extension of disease, or positive margins of resection are known to be in a high-risk category. Data from Radiation Therapy Oncology Group trials have shown that the rate of local and regional recurrence at three years is 14 percent for those with none of these risk factors, 27 percent for those with two or more positive nodes or extracapsular extension of tumor, and 49 percent for those with a positive margin with or without other risk factors. The corresponding median durations of survival are 5.6 years, 2.6 years, and 1.5 years. There is no role for adjuvant systemic therapies in patients who are at low risk.
Toxicity of Chemotherapy
The recent revision and expansion of the Common Toxicity Criteria by the National Cancer Institute have improved the standardization of toxicity grading for radiation, chemotherapy, and surgery. Data on speech and swallowing function are now routinely incorporated into clinical trials. Validated psychometric tools for measuring the effects of the tumor burden and the treatment on the quality of life now enable investigators to measure these effects from the patient's perspective. Mucositis is recognized as a major impediment to further intensification of treatment for head and neck cancer.
Another debilitating long-term side effect of radiation to the head and neck region is xerostomia; amifostine given daily by intravenous infusion concurrently with postoperative radiotherapy can reduce xerostomia. Pilocarpine used after radiation has been shown to reduce the sensation of oral dryness but has had no detectable effect on salivary flow. Ongoing studies incorporating amifostine into chemoradiotherapy regimens and trials in which pilocarpine is being given concurrently with radiation are attempts to optimize the effect of these agents. There are additional studies under way to address the long-term side effects as well as the acute toxic effects of combination therapy to the head and neck region.
Summary
Head and neck cancer remains a disfiguring disease associated with a high mortality rate. Abstinence from smoking and alcohol is the best approach to prevent its occurrence. New molecular methods of detection may help clinicians diagnose tumors earlier and may provide more accurate staging systems and means of monitoring patients with this type of cancer. Aggressive surgical resection is the cornerstone of therapy, with increasing roles for both radiation and chemotherapy, especially for organ preservation.



Supported by a grant (RO1 DE12588) from the National Institute of Dental and Craniofacial Research and a Lung Spore grant (P50 CA58184).

Source Information
From the Department of Otolaryngology–Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore.
Address reprint requests to Dr. Sidransky at the Department of Otolaryngology–Head and Neck Surgery, Johns Hopkins University School of Medicine, 818 Ross Research Bldg., 720 Rutland Ave., Baltimore, MD 21205-2196, or at Этот адрес электронной почты защищён от спам-ботов. У вас должен быть включен JavaScript для просмотра..
References

  1. Vokes EE, Weichselbaum RR, Lippman SM, Hong WK. Head and neck cancer. N Engl J Med 1993;328:184-194.
  2. Canto MT, Devesa SS. Oral cavity and pharynx cancer incidence rates in the United States 1975-1998. Oral Oncol (in press).
  3. Hibi K, Trink B, Patturajan M, et al. AIS is an oncogene amplified in squamous cell carcinoma. Proc Natl Acad Sci U S A 2000;97:5462-5467.
  4. Mills AA, Zheng B, Wang X-J, Vogel H, Roop DR, Bradley A. p63 Is a p53 homologue required for limb and epidermal morphogenesis. Nature 2001;398:708-713.
  5. Fearon ER, Vogelstein B. A genetic model of colorectal tumorigenesis. Cell 1990;61:759-767.
  6. Califano J, van der Riet P, Westra W, et al. Genetic progression model for head and neck cancer: implications for field cancerization. Cancer Res 1996;56:2488-2492.
  7. van der Riet P, Nawroz H, Hruban RH, et al. Frequent loss of chromosome 9p21-22 early in head and neck cancer progression. Cancer Res 1994;54:1156-1158.
  8. Cairns P, Polascik TJ, Eby Y, et al. Frequency of homozygous deletion at p16/CDKN2 in primary human tumours. Nat Genet 1995;11:210-212.
  9. Kamb A, Gruis NA, Weaver-Feldhaus J, et al. A cell cycle regulator potentially involved in genesis of many tumor types. Science 1994;264:436-440.
  10. Reed A, Califano J, Cairns P, et al. High frequency of p16 (CDKN2/MTS-1/INK4A) inactivation in head and neck squamous cell carcinoma. Cancer Res 1996;56:3630-3633.
  11. Papadimitrakopoulou V, Izzo J, Lippman SM, et al. Frequent inactivation of p16INK4a in oral premalignant lesions. Oncogene 1997;14:1799-1803.
  12. Munro J, Stott FJ, Vousden KH, Peters G, Parkinson EK. Role of the alternative INK4A proteins in human keratinocyte senescence: evidence for the specific inactivation of p16INK4A upon immortalization. Cancer Res 1999;59:2516-2521.
  13. Hollstein M, Sidransky D, Vogelstein B, Harris CC. p53 Mutation in human cancers. Science 1991;253:49-53.
  14. Somers K, Merrick MA, Lopez ME, Incognito LS, Schechter GL, Casey G. Frequent p53 mutations in head and neck cancer. Cancer Res 1992;52:5997-6000.
  15. Boyle JO, Hakim J, Koch W, et al. The incidence of p53 mutations increases with progression of head and neck cancer. Cancer Res 1993;53:4477-4480.
  16. Hartwell LH, Kastan MB. Cell cycle control and cancer. Science 1994;266:1821-1828.
  17. Koch WM, Brennan JA, Zahurak M, et al. p53 Mutation and locoregional treatment failure in head and neck squamous cell carcinoma. J Natl Cancer Inst 1996;88:1580-1586.
  18. Berenson JR, Yang J, Mickel RA. Frequent amplification of the bcl-1 locus in head and neck squamous cell carcinomas. Oncogene 1989;4:1111-1116.
  19. Callender T, el-Naggar AK, Lee MS, Frankenthaler R, Luna MA, Batsakis JG. PRAD-1 (CCND1)/cyclin D1 oncogene amplification in primary head and neck squamous cell carcinoma. Cancer 1994;74:152-158.
  20. Jares P, Fernandez PL, Campo E, et al. PRAD-1/cyclin D1 gene amplification correlates with messenger RNA overexpression and tumor progression in human laryngeal carcinomas. Cancer Res 1994;54:4813-4817.
  21. Nawroz H, van der Riet P, Hruban RH, Koch WM, Ruppert JM, Sidransky D. Allelotype of head and neck squamous cell carcinoma. Cancer Res 1994;54:1152-1155.
  22. Ah-See KW, Cooke TG, Pickford IR, Soutar D, Balmain A. An allelotype of squamous carcinoma of the head and neck using microsatellite markers. Cancer Res 1994;54:1617-1621.
  23. Slaughter DP, Southwick HW, Smejkal W. "Field cancerization" in oral stratified squamous epithelium: clinical implications of multicentric origin. Cancer 1953;6:963-968.
  24. Rosin MP, Cheng X, Poh C, et al. Use of allelic loss to predict malignant risk for low-grade oral epithelial dysplasia. Clin Cancer Res 2000;6:357-362.
  25. Partridge M, Pateromichelakis S, Phillips E, Emilion GG, A'Hern RP, Langdon JD. A case-control study confirms that microsatellite assay can identify patients at risk of developing oral squamous cell carcinoma within a field of cancerization. Cancer Res 2000;60:3893-3898.
  26. Sudbø J, Kildal W, Risberg B, Koppang HS, Danielsen HE, Reith A. DNA content as a prognostic marker in patients with oral leukoplakia. N Engl J Med 2001;344:1270-1278.
  27. Califano J, Westra WH, Koch W, et al. Unknown primary head and neck squamous cell carcinoma: molecular identification of the site of origin. J Natl Cancer Inst 1999;91:599-604.
  28. Gillison ML, Koch WM, Capone RB, et al. Evidence for a causal association between human papillomavirus and a subset of head and neck cancers. J Natl Cancer Inst 2000;92:709-720.
  29. Brennan JA, Boyle JO, Koch WM, et al. Association between cigarette smoking and mutation of the p53 gene in squamous-cell carcinoma of the head and neck. N Engl J Med 1995;332:712-717.
  30. Koch WM, Lango M, Sewell D, Zahurak M, Sidransky D. Head and neck cancer in nonsmokers: a distinct clinical and molecular entity. Laryngoscope 1999;109:1544-1551.
  31. Bedi GC, Westra WH, Gabrielson E, Koch W, Sidransky D. Multiple head and neck tumors: evidence for a common clonal origin. Cancer Res 1996;56:2484-2487.
  32. Leong PP, Rezai B, Koch WM, et al. Distinguishing second primary tumors from lung metastases in patients with head and neck squamous cell carcinoma. J Natl Cancer Inst 1998;90:972-977.
  33. Califano J, Leong PL, Koch WM, Eisenberger CF, Sidransky D, Westra WH. Second esophageal tumors in patients with head and neck squamous cell carcinoma: an assessment of clonal relationships. Clin Cancer Res 1999;5:1862-1867.
  34. Hong WK, Spitz MR, Lippman SM. Cancer chemoprevention in the 21st century: genetics, risk modeling, and molecular targets. J Clin Oncol 2000;18:Suppl:9S-18S.
  35. Lotan R, Xu X-C, Lippman SM, et al. Suppression of retinoic acid receptor-ß in premalignant oral lesions and its up-regulation by isotretinoin. N Engl J Med 1995;332:1405-1410.
  36. Wan H, Oridate N, Lotan D, Hong WK, Lotan R. Overexpression of retinoic acid receptor beta in head and neck squamous cell carcinoma cells increases their sensitivity to retinoid-induced suppression of squamous differentiation by retinoids. Cancer Res 1999;59:3518-3526.
  37. Mao L, El-Naggar AK, Papadimitrakopoulou V, et al. Phenotype and genotype of advanced premalignant head and neck lesions after chemopreventive therapy. J Natl Cancer Inst 1998;90:1545-1551.
  38. Westra WH, Sidransky D. Phenotypic and genotypic disparity in premalignant lesions: of calm water and crocodiles. J Natl Cancer Inst 1998;90:1500-1501.
  39. Sidransky D. Nucleic acid-based methods for the detection of cancer. Science 1997;278:1054-1059.>
  40. Spafford MF, Koch WM, Reed AL, et al. Detection of head and neck squamous cell carcinoma among exfoliated oral mucosal cells by microsatellite analysis. Clin Cancer Res 2001;7:607-612.
  41. Nawroz H, Koch W, Anker P, Stroun M, Sidransky D. Microsatellite alterations in serum DNA of head and neck cancer patients. Nat Med 1996;2:1035-1037.
  42. Chien Y-C, Chen J-Y, Liu M-Y, et al. Serologic markers of Epstein-Barr virus infection and nasopharyngeal carcinoma in Taiwanese men. N Engl J Med 2001;345:1877-1882.
  43. Lo YM, Chan LY, Lo KW, et al. Quantitative analysis of cell-free Epstein-Barr virus DNA in plasma of patients with nasopharyngeal carcinoma. Cancer Res 1999;59:1188-1191.
  44. Shotelersuk K, Khorprasert C, Sakdikul S, Pornthanakasem W, Voravud N, Mutirangura A. Epstein-Barr virus DNA in serum/plasma as a tumor marker for nasopharyngeal cancer. Clin Cancer Res 2000;6:1046-1051.
  45. Capone RB, Pai SI, Koch WM. Detection and quantification of human papilloma virus (HPV) DNA in the sera of patients with HPV-associated head and neck squamous cell carcinoma. Clin Cancer Res 2000;6:4171-4175.
  46. Lo YM, Chan LY, Chan AT, et al. Quantitative and temporal correlation between circulating cell-free Epstein-Barr virus DNA and tumor recurrence in nasopharyngeal carcinoma. Cancer Res 1999;59:5452-5455.
  47. Sanchez-Cespedes M, Esteller M, Wu L, et al. Gene promoter hypermethylation in tumors and serum of head and neck cancer patients. Cancer Res 2000;60:892-895.
  48. Rosas SLB, Koch W, da Costa Carvalho MG, et al. Promoter hypermethylation patterns of p16, O6-methylation-DNA-methyltransferase, and death-associated protein kinase in tumors and saliva of head and neck cancer patients. Cancer Res 2001;61:939-942.
  49. Ribeiro KCB, Kowalski LP, Latorre MRDO. Impact of comorbidity, symptoms, and patients' characteristics on the prognosis of oral carcinomas. Arch Otolaryngol Head Neck Surg 2000;126:1079-1085.
  50. Johnson JT, Myers EN, Bedetti CD, Barnes EL, Schramm VL Jr, Thearle PB. Cervical lymph node metastases: incidence and implications of extracapsular carcinoma. Arch Otolaryngol 1985;111:534-537.
  51. Stokkel MP, ten Broek FW, Hordijk GJ, Koole R, van Rijk PP. Preoperative evaluation of patients with primary head and neck cancer using dual-head 18fluorodeoxyglucose positron emission tomography. Ann Surg 2000;231:229-234.
  52. Lowe VJ, Boyd JH, Dunphy FR, et al. Surveillance for recurrent head and neck cancer using positron emission tomography. J Clin Oncol 2000;18:651-658.
  53. Alex JC, Sasaki CT, Krag DN, Wenig B, Pyle PB. Sentinel lymph node radiolocalization in head and neck squamous cell carcinoma. Laryngoscope 2000;110:198-203.
  54. Koch WM, Choti MA, Civelek AC, Eisele DW, Saunders JR. Gamma probe-directed biopsy of the sentinel node in oral squamous cell carcinoma. Arch Otolaryngol Head Neck Surg 1998;124:455-459.
  55. Brennan JA, Mao L, Hruban RH, et al. Molecular assessment of histopathological staging in squamous-cell carcinoma of the head and neck. N Engl J Med 1995;332:429-435.
  56. Partridge M, Li SR, Pateromichelakis S, et al. Detection of minimal residual cancer to investigate why oral tumors recur despite seemingly adequate treatment. Clin Cancer Res 2000;6:2718-2725.
  57. Laccourreye H, Laccourreye O, Weinstein G, Menard M, Brasnu D. Supracricoid laryngectomy with cricohyoidopexy: a partial laryngeal procedure for selected supraglottic and transglottic carcinomas. Laryngoscope 1990;100:735-741.
  58. Brazilian Head and Neck Cancer Study Group. End results of a prospective trial on elective lateral neck dissection vs type III modified radical neck dissection in the management of superglottic and transglottic carcinomas. Head Neck 1999;21:694-702.
  59. Urken ML, Buchbinder D, Costantino PD, et al. Oromandibular reconstruction using microvascular composite flaps: report of 210 cases. Arch Otolaryngol Head Neck Surg 1998;124:46-55.
  60. Lyos AT, Evans GR, Perez D, Schusterman MA. Tongue reconstruction: outcomes with the rectus abdominis flap. Plast Reconstr Surg 1999;103:442-447.
  61. Horiot JC, Le Fur R, N'Guyen T, et al. Hyperfractionation versus conventional fractionation in oropharyngeal carcinoma: final analysis of a randomized trial of the EORTC cooperative group of radiotherapy. Radiother Oncol 1992;25:231-241.
  62. Pinto L, Canary P, Araujo CM, Bacelar SC, Souhami L. Prospective randomized trial comparing hyperfractionated versus conventional radiotherapy in stages III and IV oropharyngeal carcinoma. Int J Radiat Oncol Biol Phys 1991;21:557-562.
  63. Datta NR, Choudhry AD, Gupta S, Bose AK. Twice a day versus once a day radiation therapy in head and neck cancer. Int J Radiat Oncol Biol Phys 1989;17:Suppl 1:132-133.
  64. Jackson SM, Weir LM, Hay JH, Tsang VH, Durham JS. A randomised trial of accelerated versus conventional radiotherapy in head and neck cancer. Radiother Oncol 1997;43:39-46.
  65. Maciejewski B, Skladowski K, Pilecki B, et al. Randomized clinical trial on accelerated 7 days per week fractionation in radiotherapy for head and neck cancer: preliminary report on acute toxicity. Radiother Oncol 1996;40:137-145.
  66. Overgaard J, Sand Hansen H, Sapru W, et al. Conventional radiotherapy as the primary treatment of squamous cell carcinoma (SCC) of the head and neck: a randomized multicenter study of 5 versus 6 fractions per week -- preliminary report from DAHANCA 6 and 7 trial. Radiother Oncol 1996;40:Suppl 1:S31-S31.
  67. Dische S, Saunders M, Barrett A, Harvey A, Gibson D, Parmar M. A randomised multicentre trial of CHART versus conventional radiotherapy in head and neck cancer. Radiother Oncol 1997;44:123-136.
  68. Fu KK, Pajak TF, Trotti A, et al. A Radiation Therapy Oncology Group (RTOG) phase III randomized study to compare hyperfractionation and two variants of accelerated fractionation to standard fractionation for head and neck squamous cell carcinomas: first report of RTOG 9003. Int J Radiat Oncol Biol Phys 2000;48:7-16.
  69. Horiot J-C, Bontemps P, van den Bogaert W, et al. Accelerated fractionation (AF) compared to conventional fractionation (CF) improves loco-regional control in the radiotherapy of advanced head and neck cancers: results of the EORTC 22851 randomized trial. Radiother Oncol 1997;44:111-121.
  70. 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;32:567-576.
  71. van den Bogaert W, van der Schueren E, Horiot JC, et al. Early results of the EORTC randomized clinical trial on multiple fractions per day (MFD) and misonidazole in advanced head and neck cancer. Int J Radiat Oncol Biol Phys 1986;12:587-591.
  72. Lee D, Pajak T, Stetz J, Order SE, Weissberg JB, Fischer JJ. 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;16:465-470.
  73. Overgaard J, Sand Hansen H, Overgaard M, et al. A randomized double-blind phase III study of nimorazole as 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;46:135-146.
  74. Peters LJ, Rischin D, Hicks RJ, Hughes PG, Sizeland AM. Extraordinary tumor control in phase I trial of concurrent tirapazamine cisplatin and radiotherapy for far advanced head and neck cancer. Int J Radiat Oncol Biol Phys 1999;45:Suppl:148-149.abstract
  75. Peters LJ, Goepfert H, Ang KK, et al. Evaluation of the dose for postoperative radiation therapy of head and neck cancer: first report of a prospective randomized trial. Int J Radiat Oncol Biol Phys 1993;26:3-11.
  76. Awwad HK, Khafagy Y, Barsoum M, et al. Accelerated versus conventional fractionation in the postoperative irradiation of locally advanced head and neck cancer: influence of tumour proliferation. Radiother Oncol 1992;25:261-266.
  77. Trotti A, Klotch D, Endicott J, Ridley M, Cantor A. Postoperative accelerated radiotherapy in high-risk squamous cell carcinoma of the head and neck: long term results of a prospective trial. Head Neck 1998;20:119-123.
  78. Ang KK, Trotti A, Garden AS, et al. The importance of overall time factor in postoperative radiotherapy of head and neck cancer. In: Proceedings of the Fourth International Conference on Head and Neck Cancer, Toronto, July 28–August 1, 1996:231-5.
  79. Bachaud J, Cohen-Jonathan E, Alzieu C, David JM, Serrano E, Daly-Schweitzer N. Combined postoperative radiotherapy and weekly cisplatin infusion for locally advanced head and neck carcinoma: final report of a randomized trial. Int J Radiat Oncol Biol Phys 1996;36:999-1004.
  80. Al-Sarraf M, Pajak T, Byhardt R, Beitler JJ, Salter MM, Cooper JS. Postoperative radiotherapy with concurrent cisplatin appears to improve locoregional control of advanced, resectable head and neck cancers: RTOG 88-24. Int J Radiat Oncol Biol Phys 1997;37:777-782.
  81. Haffty BG, Son YH, Sasaki CT, et al. Mitomycin C as an adjunct to postoperative radiation therapy in squamous cell carcinoma of the head and neck: results from two randomized clinical trials. Int J Radiat Oncol Biol Phys 1993;27:241-250.
  82. Stevens KR Jr, Britsch A, Moss WT. High-dose reirradiation of head and neck cancer with curative intent. Int J Radiat Oncol Biol Phys 1994;29:687-698.
  83. Weppelmann B, Wheeler RH, Peters G, et al. Treatment of recurrent head and neck cancer with 5-fluorouracil, hydroxyurea, and reirradiation. Int J Radiat Oncol Biol Phys 1992;22:1051-1056.
  84. Ang KK. Radiation injury to the central nervous system: clinical features and prevention. In: Meyer JL, ed. Radiation injury: advances in management and prevention. Basel, Switzerland: Karger, 1999:145-54.
  85. Vokes EE, Haraf DJ, Brockstein BE, Weilchselbaum RR. Paclitaxel, 5-fluorouracil, hydroxyurea, and concomitant radiation therapy for poor-prognosis head and neck cancer. Semin Radiat Oncol 1999;9:Suppl 1:70-76.
  86. Browman GP, Cronin L. Standard chemotherapy in squamous cell head and neck cancer: what we have learned from randomized trials. Semin Oncol 1994;21:311-319.
  87. Rooney M, Kish J, Jacobs J, et al. Improved complete response rate and survival in advanced head and neck cancer after three-course induction therapy with 120-hour 5-FU infusion and cisplatin. Cancer 1985;55:1123-1128.
  88. Forastiere AA, Metch B, Schuller DE, et al. Randomized comparison of cisplatin plus fluorouracil and carboplatin plus fluorouracil versus methotrexate in advanced squamous-cell carcinoma of the head and neck: a Southwest Oncology Group study. J Clin Oncol 1992;10:1245-1251.
  89. Dassonville O, Formento JL, Francoual M, et al. Expression of epidermal growth factor receptor and survival in upper aerodigestive tract cancer. J Clin Oncol 1993;11:1873-1878.
  90. Mendelsohn J, Fan Z. Epidermal growth factor receptor family and chemosensitization. J Natl Cancer Inst 1997;89:341-343.
  91. Grandis JR, Melhem MF, Gooding WE, et al. Levels of TGF-alpha and EGFR protein in head and neck squamous cell carcinoma and patient survival. J Natl Cancer Inst 1998;90:824-832.
  92. Mendelsohn J, Shin DM, Donato N, et al. A phase I study of chimerized anti-epidermal growth factor receptor (EGFr) monoclonal antibody, C225, in combination with cisplatin (CDDP) in patients (PTS) with recurrent head and neck squamous cell carcinoma (SCC). Prog Proc Am Soc Clin Oncol 1999;18:389a. abstract.
  93. Ezekiel MP, Bonner JA, Robert F, et al. Phase I trial of chimerized anti-epidermal growth factor receptor (anti-EGFr) antibody in combination with either once-daily or twice-daily irradiation for locally advanced head and neck malignancies. Prog Proc Am Soc Clin Oncol 1999;18:388a. abstract.
  94. Patel V, Senderowicz AM, Pinto D Jr, et al. Flavopiridol, a novel cyclin-dependent kinase inhibitor, suppresses the growth of head and neck squamous cell carcinomas by inducing apoptosis. J Clin Invest 1998;102:1674-1681.
  95. Hashemolhosseini S, Nagamine Y, Morley SJ, Desrivieres S, Mercep L, Ferrari S. Rapamycin inhibition of the G1 to S transition is mediated by effects on cyclin D1 mRNA and protein stability. J Biol Chem 1998;273:14424-14429.
  96. Heise CC, Williams AM, Xue S, Propst M, Kirn DH. Intravenous administration of ONYX-015, a selectively replicating adenovirus, induces antitumoral efficacy. Cancer Res 1999;59:2623-2628.
  97. Heise C, Sampson-Johannes A, Williams A, McCormick F, Von Hoff DD, Kirn DH. ONYX-015, an E1B gene-attenuated adenovirus, causes tumor-specific cytolysis and antitumoral efficacy that can be augmented by standard chemotherapeutic agents. Nat Med 1997;3:639-645.
  98. The Department of Veterans Affairs Laryngeal Cancer Study Group. Induction chemotherapy plus radiation compared with surgery plus radiation in patients with advanced laryngeal cancer. N Engl J Med 1991;324:1685-1690.
  99. Lefebvre J-L, Chevalier C, Luboinski B, Kirkpatrick A, Collette L, Sahmoud T. Larynx preservation in pyriform sinus cancer: preliminary results of a European Organization for Research and Treatment of Cancer phase III trial. J Natl Cancer Inst 1996;88:890-899.
  100. Forastiere AA, Berkey B, Maor M, et al. Phase III trial to preserve the larynx: induction chemotherapy and radiotherapy versus concomitant chemoradiotherapy versus radiotherapy alone, Intergroup trial R91-11. Prog Proc Am Soc Clin Oncol 2001;20:2a. abstract.
  101. Calais G, Alfonsi M, Bardet E, et al. Randomized trial of radiation therapy versus concomitant chemotherapy and radiation therapy for advanced-stage oropharynx carcinoma. J Natl Cancer Inst 1999;91:2081-2086.
  102. Merlano M, Benasso M, Corvo R. Five-year update of a randomized trial of alternating radiotherapy and chemotherapy compared with radiotherapy alone in treatment of unresectable squamous cell carcinoma of the head and neck. J Natl Cancer Inst 1996;88:583-589.
  103. Adelstein DJ, Adams GL, Li Y, et al. A phase III comparison of standard radiation therapy (RT) versus RT plus concurrent cisplatin (DDP) versus split-course RT plus concurrent DDP and 5-fluorouracil (5FU) in patients with unresectable squamous cell head and neck cancer (SCHNC): an Intergroup study. Prog Proc Am Soc Clin Oncol 2000;19:411a. abstract.
  104. Wendt TG, Grabenbauer GG, Rodel CM, et al. Simultaneous radiochemotherapy versus radiotherapy alone in advanced head and neck cancer: a randomized multicenter study. J Clin Oncol 1998;16:1318-1324.
  105. Brizel DM, Albers ME, Fisher SR, et al. Hyperfractionated irradiation with or without concurrent chemotherapy for locally advanced head and neck cancer. N Engl J Med 1998;338:1798-1804.
  106. Al-Sarraf M, LeBlanc M, Giri PG, et al. Chemoradiotherapy versus radiotherapy in patients with advanced nasopharyngeal cancer: phase III randomized Intergroup study 0099. J Clin Oncol 1998;16:1310-1317.
  107. Chan ATC, Teo PML, Johnson PJ. Controversies in the management of locoregionally advanced nasopharyngeal carcinoma. Curr Opin Oncol 1998;10:219-225.
  108. Cooper JS, Pajak TF, Forastiere A, et al. Precisely defining high-risk operable head and neck tumors based on RTOG #85-03 and #88-24: targets for postoperative radiochemotherapy? Head Neck 1998;20:588-594.
  109. Laramore GE, Scott CB, al-Sarraf M, et al. Adjuvant chemotherapy for resectable squamous cell carcinomas of the head and neck: report on Intergroup Study 0034. Int J Radiat Oncol Biol Phys 1992;23:705-713.
  110. Arbuck SG, Eisenhauer EA, Ivy SP, et al. Common toxicity criteria (CTC), version 2.0. Rev. Bethesda, Md.: Cancer Therapy Evaluation Program, April 1999. (Accessed November 9, 2001, at
  111. Lazarus CL, Logemann JA, Pauloski BR, et al. Swallowing disorders in head and neck cancer patients treated with radiotherapy and adjuvant chemotherapy. Laryngoscope 1996;106:1157-1166.
  112. List MA, Mumby P, Haraf D, et al. Performance and quality of life outcome in patients completing concomitant chemoradiotherapy protocols for head and neck cancer. Qual Life Res 1997;6:274-284.
  113. Deleyiannis FW, Weymuller EA Jr, Coltrera MD. Quality of life of disease-free survivors of advanced (stage III or IV) oropharyngeal cancer. Head Neck 1997;19:466-473.
  114. Trotti A. Toxicity in head and neck cancer: a review of trends and issues. Int J Radiat Oncol Biol Phys 2000;47:1-12.
  115. Brizel DM, Wasserman TH, Henke M, et al. Phase III randomized trial of amifostine as a radioprotector in head and neck cancer. J Clin Oncol 2000;18:3339-3345. [Erratum, J Clin Oncol 2000;18:4110-1.]
  116. Johnson JT, Ferretti GA, Nethery WJ, et al. Oral pilocarpine for post-irradiation xerostomia in patients with head and neck cancer. N Engl J Med 1993;329:390-395.
  117. Trotti A. Toxicity antagonists in head and neck cancer. Semin Radiat Oncol 1998;8:282-291.



The New England Journal of Medicine is owned, published, and copyrighted © 2002 Massachusetts Medical Society. All rights reserved.

Sprechstunde Würzburg

97074 Würzburg
Walther von der Vogelweidestraße 33 A
+49 931 200410
Ambulanz Sprechstunde
Montags 8:00 bis 14:00 Uhr

DRK Manniske Krankenhaus

Abteilung für Viszeralchirurgie
An der Wipper 2
06567 Bad Frankenhausen / Thüringen
Mobil:
+49 170 3607780 (werktags)

консультация

На предварительном собеседовании Вы должны иметь при себе все актуальные лабораторные заключения, рентгеновские снимки, результаты компьютерной и магнитно-резонансной томографии, описание операций.