Management of Skin Cancer

June 3, 2024
0
0
Зміст

Skin Cancer. Melanoma.

 

 

  The skin is the largest organ of the body and consists of three layers: epidermis, dermis, and subcutis (i.e., hypodermis). It is a specialized structure with a wide range of functions that include protection from the environment, synthesis of vitamin D, production of a large number of cytokines, antigen presentation, thermoregulation, and sensation of touch and temperature. Several different cell and tissue types, originating from all three embryonic layers, participate in the formation of the three layers of the skin and its associated appendages. These cell and tissue elements can transform to produce a large number of benign and malignant growths. This chapter summarizes the nonmelanoma skin cancers and precancerous conditions.

EPIDEMIOLOGY

Nonmelanoma skin cancers are the most common cancers in the US white  population. More than 700,000 new cases are diagnosed annually,  according to the data from the American Cancer Society. The mortality rate for the nonmelanoma skin cancers is approximately 2100 per year. Because most skin cancers are diagnosed and treated  in private office settings or outpatient clinics, the available  statistics are thought to grossly underestimate the actual number of  nonmelanoma skin cancer cases. 

Basal cell carcinoma is the most frequently diagnosed skin cancer in whites, accounting for approximately 75% to 80% of all reported cases. Squamous cell carcinoma is the second most common skin cancer and is estimated to represent 20% to 25% of all reported skin cancer cases.

  Persons with fair skin who sunburn easily are more susceptible to developing basal cell carcinoma on the sun-exposed areas of their skin. Basal cell carcinoma occurs less often in darkly pigmented persons, in whom squamous cell carcinoma is the most common skin cancer. Basal cell carcinoma is seen more frequently in men than in women, and it mostly occurs later in life. However, recent  observations indicate an increased incidence of basal cell carcinoma  in younger age groups. 

  Other skin cancers, such as soft tissue sarcomas involving the dermis and subcutis and the adnexal carcinomas, are much less frequent and, although they are encountered in clinical practice, are not as common as basal cell carcinoma or squamous cell carcinoma.

ETIOLOGY AND PATHOGENESIS

The two major factors influencing the development of skin cancers are exposure to ultraviolet radiation and type of skin. Chemical carcinogens have been extensively studied, especially in laboratory animals, as etiologic factors for cutaneous malignancies.  Ionizing radiation and primary chronic irritation play major roles in  skin cancers. Attention has recently been focused on viruses that may cause skin cancers, specifically human papillomavirus causing skin and  mucous membrane carcinomas. Host genetic makeup and host immunity also play roles in the development of skin cancers.

EXPOSURE TO ULTRAVIOLET LIGHT AND SKIN TYPE

Ample evidence supports the combined influence of ultraviolet (UV) light and skin type on the incidence of skin cancer. Exposure to UV radiation in the form of sunlight is overwhelmingly the most important  etiologic factor for development of nonmelanoma skin cancer. The wavelengths of 290 to 320 nm (UVB), and to a lesser extent, 320 to 400 nm (UVA) have been implicated. The amount of exposure, the  timing of exposure, and the skin type being exposed determine the rate  of carcinogenesis. Light-skinned people develop skin cancer much more  readily at a given rate of sun exposure than dark-skinned individuals,  who are protected by the melanin pigment in their skin. As can be  expected, nonmelanoma skin cancers occur most frequently on skin sites  that are most often exposed to the sun. 

  Squamous cell carcinoma usually occurs in the sun-exposed areas of  head and neck. Persons with outdoor occupations, such as sailors and  farmers, have a higher incidence of skin cancers than those with  indoor occupations. Epidemiologic studies worldwide suggest that UV  radiation is the most important etiologic agent for skin cancers. The incidence of nonmelanoma skin cancers directly  correlates with the proximity to the equator. A quantitative  association has been observed between lifetime sun exposure and the  risk of developing nonmelanoma skin cancers. In outdoor workers, the  most common sites for skin cancers are on the head, neck, and dorsum  of the hands, which are the sites of maximal chronic sun exposure.  Chronic UV exposure has changed from occupational to a more  recreational pattern, and younger and younger persons are being  diagnosed with skin cancers. Recent studies have further delineated  risk factors for development of UV light-induced skin cancers. There  is an increased incidence of skin cancer in Australia as compared with  Scandinavian countries, where the population has similar skin but UV  light exposure is different. In addition there is a higher incidence  of skin cancer in albinos than iormally pigmented persons in  Africa. 

  The risk for basal cell carcinoma is increased in light-skinned people, in those who freckled or suffered severe sunburn in childhood.  A large increase in the risk for basal cell carcinoma is seen in individuals with increased sun exposure in childhood and adolescence. This relationship was strongest among individuals who burned rather than tanned upon sun exposure. Another study suggested that  intense sun exposure delivered intermittently to poorly tanning  light-skinned individuals carried more risk than the same dose of sun  exposure delivered more continuously over the same total period of  time.  

  Risk for squamous cell carcinoma is also strongly related to UV  radiation. Again, it occurs more frequently in light-skinned  individuals and is related directly to the amount of solar radiation  they receive. Sailors, farmers, and others with outdoor occupations  have a higher incidence of squamous cell carcinoma than those with  indoor occupations. The temporal relationship to occupational solar  exposure was further delineated by a recent study that found a strong  trend toward increased risk with increasing occupational solar  exposure in the 10 years prior to diagnosis of squamous cell  carcinoma. Other forms of UV radiation, such as the one used  in conjunction with psoralens for the treatment of psoriasis, also  increase the risk of skin cancers, especially that of squamous cell  carcinoma. 

  There is considerable environmental concern about the depletion of the ozone layer by certain chemicals. The ozone layer acts as a strong  barrier in absorbing a major portion of UV radiation and preventing it  from reaching the earth. The chemicals responsible for the depletion of the ozone layer include chlorofluorocarbons, which are found in aerosol sprays and refrigerators. It is thought that the depletion of ozone over the next few years will rapidly increase the  incidence of skin cancers. 

  Persons with light complexions have a higher chance of developing skin cancers than those with darker skin, who are protected from solar  damage by the melanin pigment in the skin. Cutaneous malignancies are  rare in dark-skinned persons, but African albinos have a high  incidence of skin cancers on sun-exposed areas. 

  UV light may influence the development and progression of skin cancers  by affecting the host immune system. The classic work of Kripke and  colleagues showed that UV-induced cancers in mice are highly antigenic  and that most are rejected by the host’s immune system after  transplantation into a normal, genetically identical animal. However, the primary host in whom the tumor was induced by UV  light becomes tolerant to the tumor and allows its rapid growth.  Kripke’s work indicates the development of suppressor factors and  cells that suppress the host’s immune system and prevent rejection of  UV-induced skin cancer. These observations have not yet been confirmed  in humans, but it is likely that UV light affects Langerhans cells in  human skin, which may alter the host immune system, allowing the  development and progression of skin cancer. Recent studies have noted  an intriguing finding that may elaborate on Kripke’s work. A tumor-suppressor protein, designated p53, is present in very small amounts for very short periods of time iormal keratinocytes, such that it is normally undetectable. Long-lived mutants of p53, which may have lost the tumor-suppressor activity, have been detected in sun-damaged epidermis adjacent to basal cell carcinomas, which also contain the mutant p53. This suggests that UV damage to  keratinocytes may result in mutation of p53 to an ineffective form,  allowing carcinogenesis to proceed. Mutant p53 has also been seen in  squamous cell carcinomas, suggesting that the above model  may have wide application.

CHEMICAL CARCINOGENESIS

Most information about cutaneous carcinogenesis has been obtained from  studies using laboratory animals, especially mice. After topical application of a carcinogen at regular intervals, the animals develop multiple squamous papillomas, most of which regress spontaneously.  Some of the lesions develop the cytologic criteria of malignant cells,  and these tumors may become locally invasive. 

  The multistage carcinogenesis model developed on mouse skin tumors has  provided a useful tool for study of carcinogenesis. Three stages of progression have been identified for chemical carcinogenesis:  initiation, promotion, and carcinogenesis. During the initiation  phase, the DNA configuration of the cells undergoes some basic  changes. The process of initiation may remain unchanged for  the life of the tissue; or it may progress to malignancy. The  epidermal cells in psoriasis share many features with initiated cells.  The initiated cells are usually terminally differentiated but may lose their pattern of differentiation and retain their ability to multiply.  For the promoter to be effective and cause malignancies, the cell must  have been initiated previously. Most promoters usually cause  inflammation and hyperplasia, and their effects are reversible.  Promoters can induce tumors only after initiation. Some initiators and promoters are listed in. RasHa and N-ras  have been found to be possible initiation events in chemically induced  epidermal cancers in mice. Promoter compounds have beeoted to  induce increased production of transforming growth factor alpha in  mice. [ref: 16] This has been postulated to provide an environment  that allows clonal expansion of initiated cells. Progression to malignant transformation has beeoted in initiated cells containing rasHa that were given the activated v-fos oncogene. Cells containing  either oncogene alone did not progress to carcinoma, suggesting the  need for multiple activational events before progression occurs. As further exciting details come to light it should be kept in mind that these studies were undertaken in special populations of  mice, and application to humans should be undertaken with caution. 

  Tar, which contains polycyclic aromatic hydrocarbons, is an initiator,  and it has been used for treating psoriasis. Nitrogen  mustard is an accepted treatment for cutaneous T-cell lymphoma.  Phorbol esters, which are found in croton oil, are known promoters.  Anthralin, used in the treatment of psoriasis, and benzoyl peroxide,  used in the treatment of acne, are also known promoters. Long-term use  of these agents in humans has not been associated with an increased  incidence of malignancy. No chemical carcinogen has been  identified that gives rise to basal cell carcinoma or malignant  melanoma in animals, but experiments with cutaneous chemical  carcinogenesis in laboratory animals cannot be directly applied to  humans. 

  The well-documented cases of scrotal carcinoma in chimney sweepers  offer the classic example of chemical carcinogenesis in humans. Arsenic is recognized as a chemical carcinogen. Increased  incidence of cancers is reported in localities where there is a high  level of arsenic in the drinking water. Medical exposure to  arsenic in the form of Fowler’s solution, Donovan’s solution, and  Asiatic pills in the treatment of asthma, psoriasis, and syphilis  predisposes to the development of arsenical keratosis, skin cancer,  and possibly lung cancer.

IONIZING RADIATION

Exposure to ionizing radiation can induce cutaneous malignancies in  humans, usually basal cell carcinoma, squamous cell carcinoma, and  spindle cell carcinoma. Radiation-induced cancers of the  skin have been reported in patients receiving ionizing radiation as  therapy. In the past, acne, facial hair, and tinea capitis were  treated with x-ray therapy. Patients receiving these types of therapy  later developed severe radiodermatitis in the form of skin atrophy,  telangiectasia, hypopigmentation, or hyperpigmentation. Some of these  patients developed large, invasive, deforming skin cancers. These  inappropriate uses of x-ray therapy have been discontinued, but  accidental exposure to x-rays and exposure for medical reasons  continues to cause radiation dermatitis. Occupational exposure and the  resultant radiodermatitis and skin cancers, such as squamous cell  carcinoma on the fingers of dentists, is no longer seen. The use of  fractionated doses of radiation has reduced the long-term side effects  of radiation therapy. Rare cases of squamous cell carcinoma caused by  radioactive gold jewelry have also beeoted.

CHRONIC IRRITATION OR INFLAMMATION

Skin cancers can develop in areas of chronic inflammation or  irritation. Squamous cell carcinoma has been noted to occur in many  skin diseases with a chronic inflammatory or irritant course, such as  long-standing granulomas, venereal granulomas, syphilis, lupus  vulgaris, leprosy, SLE, chronic ulcers, osteomyelitis sinuses, old  burn scars, hidradenitis suppuritiva, poikiloderma congenitale,  dystrophic epidermolysis bullosa, and porokeratosis of Mibelli. Squamous cell carcinoma of the oral cavity can be induced by  chronic irritation secondary to chewing tobacco or betel nuts.

VIRAL ONCOGENESIS

Many malignant neoplasms are caused by viruses in animals. In humans,  such associations have rarely been documented. Human papillomavirus  (HPV) has been identified in lesions of verrucous carcinoma, bowenoid  papulosis, epidermodysplasia verruciforms, and in situ epidermoid  carcinoma. Many papillomavirus subtypes have been  identified, and HPV types 5, 8, 14, 16, 17, and 33 are associated with  various epidermal carcinomas and carcinoma of the cervix.  BPV 6 and 11 have been associated with verrucous carcinoma of the  genitals (Buschke-Lowenstein tumor), while HPV 16 has been associated  with squamous cell carcinoma of the nail bed. [ref: 39] HPV 5 and HPV 8 have been found in most cases of epidermoplasia verruciformis that  have progressed to squamous cell carcinoma. It is possible that other inducing factors are needed to produce the malignant tumor from some of these papillomaviruses. These papillomaviruses appear potentially oncogenic in humans.  There are other viruses, such as human T-cell lymphotrophic virus-I (HTLV-I) in leukemia-lymphoma and Epstein-Barr virus in Burkitt’s lymphoma and nasopharyngeal carcinoma, that may be associated with some of the human malignancies, but conclusive information is not available.

CLASSIFICATION OF SKIN TUMORS

To describe the various tumors of the skin in an orderly fashion, the  cellular origin and the location in the three layers are followed summarize the tumors of the skin based on  their cell of origin, biologic behavior, and location in the three  layers of the skin.

BENIGN TUMORS ARISING FROM EPIDERMAL KERATINOCYTES

Benign tumors are numerous and very common. They rarely give rise to  malignant tumors, but they may be confused with some of the malignant  lesions. It is important for clinicians to be able to recognize and differentiate them from malignant skin tumors.

SEBORRHEIC KERATOSIS

Seborrheic keratosis or basal cell papilloma is a common benign lesion produced by the overgrowth of epidermal keratinocytes. It occurs most often on the trunk, face, and neck of middle-aged and elderly persons.  Lesions may be found on the extremities but not on the palms and soles. Essential diagnostic features include multiple domed or flat-topped, round or ovoid, verrucous papules that are brown to black. These slightly raised, sharply demarcated lesions appear to be  stuck on the surface of the skin. There may be a genetic tendency toward these lesions, but sun exposure has not be found to be an  etiologic factor. A possible role for alpha-transforming growth factor has been evaluated. Histologic features are characteristic and consist of acanthosis, hyperkeratosis, and papillomatosis with  keratin horn and pseudocysts. The lesion consists mostly of squamous  and basaloid epidermal cells. Histologic types include acanthotic,  hyperkeratotic, reticulated (adenoidal), clonal, and irritated  (inflamed) lesions. Other types include melanoacanthoma, dermatosis  papulosis nigra, stucco keratosis, and seborrheic keratoses of the  Leser-Trelat sign. The sign of Leser-Trelat is characterized by a  sudden appearance of innumerable new lesions of seborrheic keratosis,  which may be a sign of an internal malignancy. Rare  transformations of seborrheic keratosis to basal cell carcinoma and  squamous cell carcinoma have been reported.  Differential  diagnosis consists of intraepidermal epitheliorna, verruca vulgaris,  dermal nevus, pigmented basal cell carcinoma, well-differentiated  squamous cell carcinoma, and malignant melanoma. If histologic  confirmation is necessary for differential diagnosis, shave excision  is the method of choice. Because of its relatively superficial  location, seborrheic keratoses may be removed by curettage and light  electrodesiccation or by cryosurgery with liquid nitrogen or carbon  dioxide ice.

EPIDERMAL NEVI

Epidermal nevi are benign, congenital, hyperplastic lesions with a  smooth or hyperkeratotic surface. These are formed by hyperplasia of  epidermal structures, classifying them as hamartomas. They typically  appear at birth or in early childhood. Epidermal nevi are uncommon and  may be solitary or multiple or form a plaque covering a large area of  skin, usually in an asymmetric or linear distribution. The lesions vary in color from pale to deeply pigmented brown to black. Lesions may continue to enlarge through puberty. Clinical variants include  nevus verrucoses, linear epidermal nevus, systematized epidermal  nevus, ichthyosis hystrix, inflammatory linear epidermal nevus, and  epidermal nevus syndrome. Epidermal nevus syndrome or large  verrucous epidermal nevus may present with widespread involvement but  predominantly on one side of the body. It frequently coexists with  central nervous system and skeletal abnormalities.   Differential diagnosis includes incontinentia pigment (verrucous stage), lichen striatus, linear lichen planus, and linear  porokeratosis. Epidermal nevi remain benign and require treatment only  if cosmetically indicated. There have been rare cases of  transformation to carcinomas.

CLEAR CELL ACANTHOMA

Clear cell acanthoma appears as a solitary, pink to red, scaly nodule on the lower leg. The lesion is slow growing, sharply delineated, and 1 to 2 cm in diameter. Lesions appear stuck on, like  seborrheic keratosis, and vascular, like pyogenic granuloma. The growth is usually asymptomatic, but the thin, crusty cover may ooze some moisture. Multiple lesions are uncommon. Histologically, the lesion consists of a proliferating population of slightly enlarged keratinocytes with abundant, clear cytoplasm, rich  in glycogen. The nuclei appear normal. The lesion is usually excised  for histologic diagnosis.

KYRLE’S DISEASE

Kyrle’s disease (i.e., hyperkeratosis follicularis et parafollicularis  in cutem penetrans) appears as multiple follicular or perifollicular  yellowish brown to brownish red papules and plaques with a central  keratinous plug. The lesions are seen mostly on the lower  extremities but appear on upper limbs, trunk, soles, and rarely on  other areas of the body. It is thought that the condition is produced by abnormal keratinization progressing downward from the epidermis and  penetrating from the epidermis into the dermis, forming a keratotic  plug that stimulates inflammatory and foreign-body giant cell  reactions. The histologic picture includes hyperkeratosis, mainly  around hair follicles, lichenoid reactions, and lymphocytic  infiltration. Kyrle’s disease and perforating folliculitis are clinically and  histologically hard to differentiate. Uremic follicular hyperkeratosis  is used to describe lesions that have features of Kyrle’s disease and  perforating folliculitis. Unlike the rarity of Kyrle’s  disease, this new entity is fairly common and occurs mostly in  patients with diabetes and renal failure. Kyrle’s disease is  one of the group of perforating dermatoses. These lesions must be  differentiated from actinic keratosis and squamous cell carcinoma, and  they require biopsy and histologic confirmation. No effective  treatment is available, but cryotherapy may be of some value.

CYSTS

Cysts are relatively common lesions occurring in young and middle-aged  persons, and they are usually of great concern to patients. Two of the  more common forms are epidermal cyst and trichilemmal (i.e., pilar)  cyst. Epidermal cysts are single or multiple lesions, mostly found on the face, neck, chest, and back. They arise spontaneously or may form as a result of trauma. A cyst normally enlarges to about 1 to 5 cm but remains as a firm, asymptomatic mass unless it becomes infected.  Histologically, epidermal cysts are intradermal or subcutaneous cavities lined with normal surface epidermis and filled with fluid and epithelial debris. Development of basal cell carcinoma, squamous cell carcinoma, Meckel cell carcinoma, and Bowen’s disease from epidermal cysts has been reported. Effective treatment requires  complete removal of the cyst and its sac. Trichilemmal cysts are clinically indistinguishable from epidermal cysts. Trichilemmal cysts may be single or multiple lesions, frequently seen on the scalp but rarely on the face and neck.  Histologically, the pilar cyst is lined by epithelium resembling hair follicle epithelium. The absence of the granular layer differentiates it from the epidermal cyst, which consists of all the layers of the epidermis. Trichilemmal cysts may have a more hyperplastic presentation, and they are referred to as proliferating  trichilemmal cysts. Malignant degeneration with metastasis of proliferating trichilemmal cysts has been reported. Treatment is surgical excision of the entire cyst.

BIRT-HOGG-DUBE SYNDROME

Birt-Hogg-Dube syndrome is inherited as an autosomal dominant trait  and consists of the triad of trichodiscoma, fibrofolliculoma, and  acrochordan. The patient has multiple, asymptomatic, flesh-colored papules on the face, mainly around the nose.  Histologically, the lesions may show proliferation of the superficial hair follicle (i.e., trichodiscoma) or proliferation of the dermal part of the hair follicle (i.e., fibrofolliculomas) or features of  fibrous tags (i.e., acrochordon). Birt-Hogg-Dube syndrome is associated with medullary carcinoma of the thyroid and renal cell carcinoma. Lesions are removed only if cosmetically indicated.

BECKER’S NEVUS

Becker’s pigmented hairy epidermal nevus (i.e., Becker’s melanosis) is a relatively common condition presenting with a patch of hyperpigmented, coalescing macules on the shoulders and upper extremities, mostly in young men. Hypertrichosis is  frequently associated with the tan or brown patches. This condition is usually preceded by a sunburn of the involved area. Histologically,  the skin appears normal except for an increased number of melanocytes,  especially in the dermis, and the increased melanin in the basal layer  of the epidermis. The hair follicles appear normal, but the  diameter of the hair may be thicker thaormal. Smooth muscle  hartomas are occasionally associated with this lesion as  well as hypoplasia of underlying structures such as breast tissue  or arm bones and coexistence with a connective  tissue nevus. A malignant melanoma has cooccurred in the same anatomic site as a Becker’s nevus. There is no need for treatment except if cosmetically indicated.

WARTY DYSKERATOMA

Warty dyskeratoma presents in middle-aged patients as a solitary  lesion on the scalp, face, or neck, but it may occur in unexposed skin  and the oral mucosa.  Multiple lesions have been reported.  It occurs as a slightly elevated, circumscribed papule or nodule with a raised border and keratotic umbilicated or pore-like  center. The lesion typically grows to about 1 to 10 mm in diameter.  Common complaints are itching, burning sensation, recurrent drainage,  and bleeding due to trauma. Histologically, the lesion appears as a  large, cup-shaped invagination with a keratotic and parakeratotic  plug. The upper portion of the invagination shows numerous acantholytic and dyskeratotic cells. One or more pilosebaceous  structures are commonly associated with warty dyskeratoma, but they  are not etiologic, because lesions may also occur on the oral mucosa. In older patients, these lesions are frequently associated  with premalignant and malignant lesions, such as solar keratosis,  squamous cell carcinoma, basal cell carcinoma, and adnexal carcinomas.  Differential diagnosis includes Darier’s disease, sebaceous cyst, solar keratosis, basal cell carcinoma, verrucae, nevocellular nevus, and folliculitis. Surgical excision is the most commonly recommended treatment approach.

PREMALIGNANT TUMORS OF THE EPIDERMIS

Because of the absence of good epidemiologic data, it is difficult to  determine with certainty whether a lesion is a true precancerous  lesion and gives rise to a malignant condition or if the association  is a coincidental finding. An example of this is keratoacanthoma,  which is usually a benign, self-healing lesion, but it may predispose  the patient to squamous cell carcinoma. The following are conditions  that are thought to precede or are associated with cancerous lesions.

  ACTINIC OR SOLAR KERATOSIS

Actinic or solar keratoses are common asymptotic lesions seen mostly on sun-exposed areas of light-skinned persons. Those who burn easily, tan poorly, and freckle (type I and type II skin prototypes) are at the greatest risk for development of actinic keratosis. The  lesions are commonly multiple and appear on sun-damaged skin as  skin-colored to yellow-brown, firm, raised papules with scaly, rough,  keratotic surfaces and erythematous bases. Unlike seborrheic  keratosis, actinic keratosis appears to arise from within the  epidermis, rather than being “stuck on” the skin. Common sites for actinic keratosis are the face, dorsum of the hands, upper chest,  upper back, and lower lip. The lesions are typically a few millimeters to 1 cm in diameter. In Australia, 40% of persons over age 40 have one or more actinic keratoses. Histologic features include epidermal dysplasia sparing the skin appendages, hyperkeratosis and inflammation with lymphocytic infiltration, and evidence of actinic elastosis and telangiectasia in the dermis. Abnormal keratinocytes  appear less basophilic thaormal keratinocytes and vary in size and  shape.

    The percentage of patients with actinic keratoses that progress to have squamous cell carcinoma has been reported to be from 1% to 20% in various studies. The tendency of squamous cell carcinoma arising from an actinic keratosis  to metastasize is low, 0.5% to 3%, except in  squamous cell carcinoma arising from actinic cheilitis on the lower  lip, which has been found to metastasize in 11% of cases. If induration, erythema, or erosion is observed, possible progression  to squamous cell carcinoma should be suspected. If malignant  transformation is suspected, shave excision of the lesion and  histologic examination is recommended. Lesions may transform into  squamous cell carcinoma, more rapidly in patients who are  immunosuppressed or have genetic defects of DNA repair enzymes (e.g.,  xeroderma pigmentosum). The report from Australia indicates a much  lower number, approximately 1%, developing into basal cell carcinoma.

  Differential diagnosis includes seborrheic keratoses, lichenoid  keratoses, warts, pigmented basal cell carcinomas, lentigo maligna,  malignant melanomas, and early squamous cell carcinomas. Existing  lesions are treated by curettage and electrodesiccation or liquid  nitrogen cryotherapy. Multiple and widespread lesions may be treated  with application of a cream or solution containing 5-fluorouracil  (5-FU) for approximately 3 weeks, but this treatment may produce  considerable discomfort. Dermabrasion may be an  alternative. Some reports indicate spontaneous regression of lesions.  Protection against excessive sun exposure by the use of  protective clothing and sunscreens helps to prevent actinic keratosis. 

CHEMICAL AND OTHER KERATOSES

Other keratoses that may be considered as premalignant cutaneous  dysplasia are arsenical keratosis induced by exposure to  arsenic, tar keratoses induced by exposure to tars and other  polycyclic aromatic hydrocarbons, thermal keratoses induced after  prolonged (less than 20 years) exposure to infrared radiation that  causes chronic cutaneous thermal damage, chronic radiation keratoses  induced by exposure to x-rays, and chronic cicatrix keratoses or scar  keratoses that may degenerate to scar carcinoma with metastatic  potential. Excision of these lesions is usually the treatment of  choice.

CUTANEOUS HORN

Cutaneous horn appears as a protuberant, raised, hard, hyperkeratotic  nodule with an erythematous base commonly occurring on the sun-exposed  areas of pale-skinned persons. It usually develops due to the underlying dysplasia or frank neoplasm, and it is therefore considered a marker of skin cancer. The histopathology of the underlying lesion includes actinic keratosis, seborrheic keratosis,  filiform verruca, trichilemmoma, basal cell carcinoma, squamous cell  carcinoma, molluscum contagiosum, or keratoacanthoma. A  histopathologic study indicates that 39% of underlying lesions were  premalignant and malignant epidermal lesions, and 61% were benign  lesions. This study also found that lesions were most  likely to have a malignant base if located on the nose, pinna of the  ear, or dorsum of the hand. The lesion with a portion of the base should be removed for histologic diagnosis. Final treatment  is usually determined by the pathology of the underlying tissue.

RADIATION DERMATITIS

Radiation dermatitis is the term used to describe skin damage due to  x-irradiation, whether exposure occurs as a result of occupational  hazard or therapeutic treatments. Involved skin is dry,  scaly, erythematous, thin, and discolored. Areas of telangiectasia,  hyperpigmentation and hypopigmentation, and hyperkeratosis and  ulceration are common. Histologically, the damaged area shows atrophy  and loss of polarity of epidermal keratinocytes, changes in elastic  and collagen bundles of the dermis, and destruction of hair follicles,  sebaceous glands, and sweat glands. There is a potential for  developing actinic keratosis, basal cell carcinoma, and squamous cell  carcinoma on this kind of skin. Management includes  removal of any overlying skin lesion for histologic diagnosis and  definitive treatment as well as continuous skin care. Soft tissue  sarcomas may develop in the irradiated tissue.

BOWEN’S DISEASE

Bowen’s disease or intraepidermal carcinoma in situ is an intraepidermal squamous cell carcinoma that may involve any area of the skin but tends to favor sun-exposed areas of the face, neck, and extremities. Primarily, older persons are affected. In one study, 80% of the patients were older than 60. There is a predominance of Bowen’s disease on the lower limbs in women  and on the scalp and ears in men. In one third of the  patients, the lesions may be multiple. Bowen’s disease is  papulosquamous and appears as a slowly enlarging, sharply defined,  round to irregular plaque with a rough, scaly, hyperkeratotic,  erythematous surface. Other surface characteristics include  pigmentation, fissures, erosion, and ulceration. Similar to in situ  epidermoid carcinoma, Bowen’s lesions may progress to invasive  squamous cell carcinoma. This transformation to squamous cell  carcinoma is reported in 3% to 5% of the cases, with most cases  remaining as carcinoma in situ.  Histologic features of Bowen’s disease include gross dysplasia of the  upper layers of epidermal cells with the basal cell layer relatively  normal, individual cell keratinization, and giant keratinocytes with  atypical mitoses. The cause of Bowen’s disease is unknown, but UV radiation, HPV-induced epidermodysplasia verruciformis, and history of ingestion of inorganic arsenic (e.g., water or medication) are  considered etiologic factors. Bowen’s disease has  been seen oonexposed areas of the skin in younger persons without a  history of arsenic ingestion and without HPV infections.   It has been suggested that patients with Bowen’s disease are more  likely to develop other cancers, including basal cell carcinoma,  adnexal carcinoma, melanoma, or cancers of the lung, gastrointestinal  tract, genitourinary tract, and reticuloendothelial system, but the  role of Bowen’s disease as a marker of internal malignancies is  unclear. A report from the Armed Forces Institute of Pathology  indicates that as many as 80% of cases of Bowen’s disease have other  cutaneous and noncutaneous malignancies. Another report  suggests that as many as 29% of the patients have internal  malignancies. Many of the published reports contain major deficiencies, such as lack  of sufficient control groups. In view of the existing  controversies, it is recommended that patients with Bowen’s disease  have a complete clinical examination combined with the necessary  diagnostic work-up. Differential diagnosis includes basal cell carcinoma, psoriasis, extramammary Paget’s disease, and actinic keratosis. Excisional biopsy of the most indurated area is recommended for histologic confirmation of Bowen’s disease and detection of squamous cell carcinoma. Cryotherapy or curettage and desiccation have been recommended, but there is a higher chance of  recurrence. In recent years carbon dioxide laser vaporization has been  used for lesions located on the hand and finger.  Photodynamic therapy has been used for larger lesions or areas that are anatomically difficult to excise. Surgical excision of  the entire lesion provides the highest cure rate.

ERYTHROPLASIA OF QUEYRAT

Erythroplasia of Queyrat is a clinicopathologic variant of Bowen’s  disease, occurring almost exclusively on the glans penis of  uncircumcised, middle-aged and older men. It may occur  on the penile shaft, scrotum, and vulva, but it is questionable  whether these presentations are Bowen’s disease of the anogenital  region. The lesion appears as an asymptomatic, sharply demarcated,  bright red, shiny plaque with a smooth, velvety surface. The histologic features are identical to those of Bowen’s  disease, especially of the anogenital region. However, the rate of  transformation into invasive squamous cell carcinoma is much higher  for Bowen’s disease, and the resultant squamous cell carcinoma tends  to be more aggressive. Erythroplasia of Queyrat has  progressed into squamous cell carcinoma in up to 30% of patients, with  metastases in 20% of patients with invasive erythroplasia. It thus has a greater tendency toward invasion and metastases  than does Bowen’s disease. Radiotherapy, 5-fluorouracil (5-FU), CO(2) laser, cryotherapy and curettage, and electrodesiccation  have been used. One recent study reported temporary  regression with low-dose isotretinoin. However, due to its  aggressive nature, it is advisable to use surgical excision of the  tumor with microscopic control of the margin (Mohs micrographic  surgery) and to circumcise the penis.

BOWENOID PAPULOSIS

Bowenoid papulosis is a multicentric anogenital dysplasia that  presents as verrucous or lichenoid pink to reddish brown papules,  erythematous macules, or leukoplakia-like lesions in young sexually  active men and women. Lesions of the oral mucosa have been reported. Although pruritus and inflammation with  tenderness and pain are occasionally reported, most patients with  bowenoid papulosis have no symptoms. The lesions typically present as inconspicuous, noncondylomatous papules with mostly smooth and only slightly papillomatous surfaces. A homosexual patient with a  long-standing history of pruritus and black perianal hyperpigmentation  extending out from the anal margin proved to have bowenoid papulosis  on skin biopsy. In women, the lesions tend to be bilateral,  hyperpigmented, and confluent, involving the labia majora, labia  minora, clitoris, perianal region, and inguinal folds; in men, the  lesions usually present as small discrete papules on the glans, shaft,  and preputium. The lesions are 0.2 to 3.3 cm in diameter with small papules coalescing into large pigmented plaques. Bowenoid papulosis is a cutaneous manifestation of the sexually transmitted HPV. It is mostly linked with HPV type 16, but types 18, 34, and 39 have  also been linked to its pathogenesis. This conditioormally follows  a benign but persistent course over many years. Occasional spontaneous  regression, particularly in pregnant women, and progression to  anogenital carcinoma have been reported. These  lesions are frequently confused with genital warts. The histology is  similar to that of Bowen’s disease. Bowenoid papulosis does not show the full-thickness epidermal involvement and disorderly keratinocyte  maturation characteristic of Bowen’s disease and in situ squamous cell  carcinoma. It has dysplastic keratinocytes scattered throughout the  epidermis in a background of orderly keratinocyte maturation. These  atypical keratinocytes have crowded nuclei that appear large,  hyperchromatic, and pleomorphic and are often in metaphase. The  epidermis exhibits papillated hyperplasia, with focally prominent,  hypergranular areas. There are atypical mitotic figures, individual  cell necrosis, dyskeratosis, and multinucleated keratinocytes  scattered in the epidermis. If podophyllin has been used topically for  the treatment of these lesions, histologic sections may exhibit a  pattern of pseudoepitheliomatous hyperplasia, with bizarre  keratinocytic forms, because podophyllin causes metaphase arrest. 

  The clinical features usually differentiate bowenoid papulosis from  Bowen’s disease. Bowenoid papulosis usually presents in young,  sexually active men and women between 20 and 40 years of age, and the  lesions appear as multiple small papules.  Bowen’s disease occurs in patients older than 50 years of age and is  usually a single plaque. Bowenoid populations normally  follow a benign but persistent course over many years. The lesions  respond well to local destructive therapy, such as electrodesiccation,  laser surgery, cryosurgery, and ablation. [ref: 160] Topical treatment  with 5-FU and intramuscular or intralesional interferons have also  been used in the treatment of bowenoid papulosis. Recurrences after treatment are common, due to the life-long nature of  HPV infections. Patients with this condition, especially women, and  their sexual partners are at high risk for anogenital carcinomas and  should have frequent evaluations, including Pap smears, because of the  high risk for anogenital carcinoma, which is usually a squamous cell  carcinoma.

EPIDERMODYSPLASIA VERRUCIFORMIS

Epidermodysplasia verruciformis is a rare, chronic, and often  hereditary disease (autosomal recessive, familial, or sporadic) characterized by widespread eruption of flat, wart-like lesions  and reddish brown plaques with slightly scaly surfaces and irregular  borders. The wart-like lesions are mainly distributed  on the hands, feet, and face, sometimes in a linear arrangement, and  the pigmented plaques preferentially involve the trunk, neck, and  proximal parts of the extremities. Epidermodysplasia verruciformis is  an extremely protracted disease that usually begins in infancy or  early childhood (5 to 11 years) with various types of warts and  plaques. Later, it may progress to form verrucous plaques and nodules  or transform to in situ squamous cell carcinoma. The rate of  appearance of new lesions varies considerably, with some lesions  disappearing in some areas as more appear in other areas. Malignant  tumors typically develop in the third or fourth decade of life in  approximately one third of the patients. These patients are not  inconvenienced by these transformations, which are neither painful nor  itchy. Malignant lesions are numerous and continue to progress as  noninvasive in situ carcinoma without metastasis. They  are locally destructive if not treated. These premalignant lesions  have many characteristics of actinic keratoses. Malignant  transformation to squamous cell carcinoma in predominantly sun-exposed  areas occurs in 30% of patients. In addition, more than 50% of  patients with epidermodysplasia verruciformis develop actinic  keratoses; they are increased iumber, occur at a younger age, and  more frequently become malignant when compared with the general  population. In epidermodysplasia verruciformis, the cancers  that develop usually occur on the forehead or in areas of trauma. They  are slow growing, microinvasive or invasive, and may be locally  destructive. They do not metastasize.

  The etiologic factors associated with the development of  epidermodysplasia verruciformis are impaired cellular immunity, HPV  infection, and the genetic makeup of the host. Most patients with epidermodysplasia verruciformis show defects in cell-mediated immunity characterized by anergy to dinitrochlorobenzene and common skin  antigens, depressed lymphocyte blastogenic reactivity to mitogens,  abnormal T-lymphocyte populations, and decreased number of T  lymphocytes. However, the humoral immune system is  left intact.  HPV 5, 8, 9, 12, 14, 15, 17, and 20 have been isolated from  epidermodysplasia verruciformis lesions and are thus felt to  contribute to the development of epidermodysplasia verruciformis.  HPV types 5 and 8 have consistently been found in benign  and malignant lesions of patients with epidermodysplasia  verruciformis. HPV appears to take advantage of the  immunologic state to induce epidermodysplasia verruciformis. The occurrence of HPV infections and  epidermodysplasia verruciformis in immunosuppressed renal allograft recipients, in an immunosuppressed patient with systemic lupus  erythematous, and in a patient with Hodgkin’s disease provides support  for this hypothesis. Another line of evidence comes  from the family members of patients with epidermodysplasia  verruciformis, in whom the wart-like lesions started to appear but  eventually regressed. However, there are patients with  epidermodysplasia verruciformis without immune dysfunction. Some investigators believe that the impaired cellular  immunity could be a result of the HPV infection. It has been  speculated that patients with epidermodysplasia verruciformis are  susceptible to widespread infection with HPV and skin cancers because  of a limited, specific immunologic defect, such as a defect in an  unidentified suppressor gene that prevents infection with the HPVs  associated with epidermodysplasia verruciformis. 

  On histologic examination, the epidermal changes resemble that of  verruca plana but are more dramatic. Affected cells may be laden with  keratohyalin granules and are often swollen and irregularly shaped.  Dyskeratotic cells may be present in the lower portion of the epidermis. 

  There is no effective therapy for epidermodysplasia verruciformis, but  aromatic retinoids, such as etretinate, may have some beneficial  effects. Interferons produce only partial responses during  therapy, with lesions returning in the same locations after therapy is  discontinued. Surgical removal, electrosurgical approaches,  and cryotherapy are used, as is skin grafting of areas with many  precancerous or cancerous lesions, such as the forehead, with  uninvolved, non-sun-exposed skin.  Radiation therapy is  contraindicated. Prevention by the use of sunscreens in  those with the autosomal recessive gene is essential. It is important for HPV-infected, immunosuppressed patients to have regular dermatologic examinations.

LEUKOPLAKIA

Leukoplakia is a clinical description that denotes white patches or  plaques that cannot be rubbed off located on the mucous membranes of  the oral, anal, and genital mucosa. About 20% of the  patients have histologic findings that consist of epithelial dysplasia  and hyperkeratosis of the mucosa. Approximately 15% of the dysplastic  cases develop carcinoma in situ, and 3% to 6% develop invasive  squamous cell carcinoma within the involved area.  Differential diagnosis includes lichen planus, candidiasis, hairy leukoplakia, white sponge nevus, pachyonychia congenita, and  dyskeratosis congenita. Treatment consists of excision, 5-FU, laser  surgery, and cryosurgery. Some have advocated using 13-cis-retinoic  acid, but the potential benefit has not conclusively been shown to  justify the risk in its use to control oral leukoplakia. Some have proposed the use of antioxidants and beta-carotene to control leukoplakia. Nearly all agree that abstinence from  tobacco and alcohol are important in reducing precancerous leukoplakia  of the oral mucosa.

ORGANOID OR SEBACEOUS NEVI

Organoid or sebaceous nevi are skin lesions composed of abnormal  numbers of pilosebaceous structures and apocrine glands. These lesions  appear as raised, yellowish papules that grow gradually to develop a  papillomatous surface. They usually occur on the scalp  and are present at birth or appear soon afterwards. Basal cell  carcinoma has been reported in as many as 10% of patients, and  squamous cell carcinoma occurs rarely. Syringocystadenoma  papilliferum, derived from the apocrine gland and basal cell  carcinomas may be associated with organoid nevus.  Histologically, the lesion consists of large numbers of skin  appendages, including sebaceous, eccrine, and apocrine glands, hair  follicles, and smooth muscle. Because of these structures, the term  organoid nevus appears more appropriate than sebaceous nevus. In  syringocystadenoma papilliferuim, cystic lesions of the epidermis are  lined by a double layer of columnar epithelium showing decapitation  secretion, suggesting apocrine gland origin. After histologic confirmation of the diagnosis, the lesion is usually  removed by surgical excision. Ideally, the lesion should be excised before puberty, because the risk of malignancy increases following  puberty.

 

MALIGNANT EPIDERMAL TUMORS

 

  BASAL CELL CARCINOMA

Basal cell carcinoma (BCC) is the most common cancer among whites. It  accounts for 77% of the 700,000 new cases of nonmelanoma skin cancer  in the United States each year. 90% of BCC occurs on sun-exposed areas such as the face, neck, ears, scalp, and arms. BCC rarely metastasizes and is usually curable through various modalities of treatment. This tumor is mostly seen in elderly persons,  especially those with fair skin and long-standing sun exposure. 

  Basal cell carcinomas can be seen in association with several  conditions including basal cell nevus syndrome (multiple BCCs, palmar  pits, characteristic facies, odontogenic cysts), Bazex’s syndrome  (basal cell carcinoma resembling nevi, follicular atrophoderma,  anhidrosis, hypotrichosis), xeroderma pigmentosum (premature skin  cancers, photosensitivity, freckling, neuronal degeneration, faulty  repair of UV radiation damage to DNA), occulocutaneous albinism (skin  cancer, nystagmus, photophobia, decreased or absent melanin), linear  basal cell nevi, and Rombo syndrome (multiple basal cell carcinomas,  trichoepitheliomas that may give rise to basal cell carcinomas, grainy  skin, vermicular atrophoderma, hypotrichosis, and acrocyanosis). 

  The incidence of basal cell carcinoma increases with age, but because  of easy access to the sunbelt areas, outdoor recreational activities,  and the popularity of suntanning, basal cell carcinoma is increasingly  seen in younger people. It is no longer surprising to see basal cell  carcinoma in patients in their third or fourth decade of life.   Most basal cell carcinomas occur on sun-exposed areas such as the face  (Figs. 40.2-9 and 40.2-10), especially the nose, the nasolabial fold,  and the inner canthus areas. Solitary basal cell carcinomas are seen  in the geographic areas with temperate climate. Multiple tumors and  tumors on areas other than the face are seen usually in tropical and  equatorial regions. Basal cell carcinoma occurs more commonly in men  than in women, with the incidence increasing with age and with  latitude.

Clinical Presentation

Typical basal cell carcinoma appears as a slowly growing, shiny,  skin-colored to pink, translucent papule with telangiectasias. As the  lesion enlarges, it may ulcerate and develop a rolled border and  crusted center. The lesion may then regress to a smaller and less  visible size. Normally, basal cell carcinomas are reported on only  hair-bearing skin, but in cases of basal cell nevus syndrome and  linear basal cell nevi, they may be seen on the soles and palms.  Several clinicopathologic variants of basal cell carcinoma have been described:

1. Nodular and nodular ulcerative are the most common   variants of basal cell carcinoma, and they show central   depression and a rolled border; they may bleed with trauma,   ulcerate, and form crust, and they are usually located on   the head and neck.     2. Pigmented basal cell carcinoma appears nodular with brown   to black pigment and must be differentiated from melanoma;   it often occurs in darker-complexioned patients. 

   3. Superficial basal cell carcinoma is flat, erythematous,   and scaly and has a thread-like border. It is often found on   the trunk, extremities, and neck, and one must rule out   psoriasis and eczema. 

   4. Sclerosing or morphea-type basal cell carcinoma appears   as an infiltrated yellowish sclerotic scar-like patch with   indistinct margins typically on the face and neck; it lacks   telangiectasias, translucency, and a distinct border.   Morphea-type basal cell carcinoma may undermine skin and go   undetected for an extended period of time. Physical   evaluation of the lesion and curretage are poor at defining   the extent of this tumor, and Mohs surgery is frequently an   appropriate treatment.     5. Cystic basal cell carcinoma often appears as a   translucent blue-grey nodule on the face.     6. Fibroepithelioma of Pinkus usually presents as a sessile   growth on the lower trunk and thighs. 

   7. Hyperkeratotic basal cell carcinomas may occur on the   head, trunk, and extremities, and these must be   differentiated from actinic keratoses, seborrheic keratoses,   squamous cell carcinoma, Bowen’s disease, and psoriasis. 

   8. Wildfire or multicentric basal cell carcinoma occurs on   the face as a plaque made of nests of lesions with   ulceration and scars. 

   9. Other variants include giant exophytic basal cell   carcinoma and giant pore type basal cell carcinoma.  Red dot or halo basal cell carcinoma appear as 1- to   2-mm red papules on the face with or without a halo. Another form of basal cell carcinoma is the so called   basosquamous cell carcinoma. This lesion has biologic   behavior and pathologic features intermediate between basal   cell and squamous cell carcinoma. 

  Many interesting and unusual cases of basal cell carcinoma have been  reported. Pedal basal cell carcinomas have been described, and these  are felt to arise from pluripotential epithelial cells, since there  are no pilosebaceous or apocrine glands in the feet; it remains  speculative whether these pedal basal cell carcinomas are  posttraumatic. Basal cell carcinomas have also been  reported in association with vaccination and burn scars,  nevus sebaceous, nevus flameus, hemangiomas, and a host of other  conditions. Linear basal cell carcinoma is a rare form in  which lateral spread typically occurs beyond clinically apparent  borders. Some question if koebnerization by deep scratching with  strornal implantation may occur.

  Some basal cell carcinomas may have a hyperkeratotic surface and scaly  appearance and may be confused with actinic keratosis, sebaceous  keratosis, squamous cell carcinoma, and Bowen’s disease. The  morphea-type basal cell carcinoma has a scar-like sclerotic appearance  and lacks telangiectasia and translucency, and the usual distinct  border is not seen. The lesion is larger and more indurated in  palpation than on inspection. Another form of basal cell carcinoma is  the so-called basosquamous cell carcinoma. This lesion has  biologic behavior and pathologic features intermediate between basal  cell and squamous cell carcinomas. 

 

Most basal cell carcinomas are diagnosed when tumors are a few  millimeters to 1 to 2 cm in diameter.  However, much larger, ulcerated tumors may be seen. Basal cell carcinoma does not usually metastasize, but if left untreated the tumor may invade underlying tissues and organs slowly and cause destruction of skin, cartilage, soft tissue, and bone.  Rarely, if basal cell carcinoma reaches the bone or blood circulation,  it may metastasize. The world literature indicates fewer  than 300 cases of metastatic basal cell carcinoma. After it reaches  metastatic levels, it behaves as aggressively as other types of  cancers and can spread to vital organs such as lymph nodes and lung.  Pathogenesis

  Most reports indicate the importance of fair complexion, blue eyes,  and fair hair in the development of basal cell carcinoma. This is especially true if the people have fair complexions and live in a  sunny region, such as Scandinavians who live in Australia. The role of genetic susceptibility to the development of  basal cell carcinoma has been suggested and is well documented in  cases of xeroderma pigmentosum and basal cell nevus syndrome. The role of HLA antigens in the  development of basal cell carcinomas remains unclear, although DR1 has  been increased with multiple basal cell carcinomas; interesting  differences in frequencies of HLA DR1, DR4, DR7, and DR53 have been  noted from patients with basal cell carcinomas as compared with  patients with both basal cell carcinomas and squamous cell carcinomas. 

Basal Cell Carcinoma,  T1N0M0 (growth pattern)

 

The association of chronic UV light exposure and development of basal  cell carcinoma is well accepted. It is the  cumulative effect of UV light over many years that causes the  development of basal cell carcinoma. After the necessary level of sun  exposure (threshold) is reached, the exposed person continues to  develop basal cell carcinomas; the process appears to be irreversible.  UVB is the major carcinogen, with UVA playing a lesser role. Ionizing radiation, whether received from the environment or as part  of therapeutic modalities, can promote the development of basal cell  carcinoma. X-irradiated skin shows evidence of chronic dermatitis  several years later and may give rise to basal cell carcinomas. In the  1950s and 1960s, x-irradiation was used for the treatment of acne and  hypertrichosis. Many patients who received therapeutic x-rays  developed radiation dermatitis and multiple skin cancers of the face  20 to 30 years later. 

  The chance of developing basal cell carcinomas increases directly in  relation to the number of previous basal cell carcinomas. Other conditions that may predispose to the development of basal cell carcinoma include other primary malignancies, especially lymphoreticular types, immunodeficiency states, and trauma such as  traumatic injuries or smallpox vaccination. While the risk for basal cell carcinomas is somewhat increased  following exposure to inorganic arsenic or industrial tars and  following immunosuppression, the relative risk for developing squamous  cell carcinomas following these exposures is much higher. Although basal cell carcinomas are most commonly seen on the  sun-exposed areas of the skin, many basal cell carcinomas are seen in  less exposed areas, such as the nasolabial fold and the inner canthus,  suggesting that other factors (e.g., embryonic closure lines) may  contribute to the development of the tumor. While the role of p53 and  tumor-suppressor gene, in the pathogenesis of basal cell carcinomas  has not yet been elucidated, decreased wild-type p53 protein has been  found in basal cell carcinomas, squamous cell carcinomas, and  keratoacanthomas with a concomitant loss of growth suppression.  Histopathology  In all forms of basal cell carcinomas, the histologic features include  masses of compactly arranged basaloid cells resembling cells in the  basal layer of epidermis extending down from the epidermis into the  dermis. Basal cell carcinomas arise from immature  pluripotential cells of the epidermis or from the outer root sheath of  hair follicles, growth is dependent upon the stroma, and basal cell  carcinoma may differentiate towards sebaceous, apocrine, or eccrine  structures or remain undifferentiated. There is usually a variable dermal stromal reaction with connective tissue  proliferation, increased acid mucopolysaccharides, and stromal  retraction or peritumoral lacunae. In the sclerosing type of basal  cell carcinoma, the cells are more fusiform and are admixed with a  more pronounced stromal reaction. In most cases, especially in the  nodular, cystic, and adenoid types, there is a clear peripheral  palisading of the dark epithelial cells that is characteristic of  basal cell carcinoma. The differential diagnosis includes  trichoepithelioma, which may also show some palisading of the cells. Pigmented basal cell carcinoma is due to aggregation of  melanin within the tumor. Basosquamous cell carcinoma consists of a  fibrous stroma admixed with a downward proliferation of epithelial  cells that lack palisading and have some features of squamous cell  carcinoma and more frequent mitosis.

  Metastasizing Basal Cell Carcinoma.

Basal cell carcinoma usually spreads by direct extensions from the  primary tumor site and has little tendency to metastasize. Recurrences of basal cell carcinoma usually occur within 4 to 12 months from either aggressive tumor or inadequate treatment.  Aggressive behavior in basal cell carcinomas may be forewarned by  recurrences, histologically aggressive tumors, location in the H-zone  of the central face, size larger than 2 cm, perineural invasion, and  history of prior ionizing radiation. [ref: 208] Metastases occur in  0.0028 to 0.1% of basal cell cancers, with the higher incidence being  in surgery clinics and usually seen in very large and recurrent  tumors. Stroma appears necessary for metastases, and large tumor  emboli may be responsible. Lymph nodes are involved in two thirds of  the cases, but vital organs may also become involved. Basal cell  carcinoma behaves aggressively after metastases occur with a median  survival of 10 to 14 months.

 

Management of Basal Cell Carcinoma

  Several methods are available for the treatment of basal cell  carcinoma  including surgical  excision, Mohs  surgery,  electrodesiccation and curretage, cryosurgery, radiation therapy,  interferon, laser treatment, and use of sun block and oral  retinoinds in high-risk groups.  The choice is usually based on the location, type of lesion, and experience of the physician. Smaller lesions can be treated with surgical excision or curretage and desiccation. Radiation therapy, although used less frequently, is the most appropriate treatment for basal cell carcinoma of the eyelid, nose, and lips. Cryosurgery and topical use of 5-FU have also been  suggested, but the cure rate is not as high as with other techniques.  Tumors with poorly defined margins and recurrent basal cell carcinomas  are best treated by microscopically controlled excisional surgery that  is known as Mohs micrographic surgery. This technique  allows the removed tissue to be mapped in relation to the underlying  site. Additional tissue removal is carried out at sites where tumor  cells are present microscopically. The treatment is considered  complete when there is no tumor found in removed tissue. Cure rates of  90% to 95% are achieved with most methods of treating primary basal  cell carcinoma. Mohs surgery has a cure rate of 96% to 99%.   Recurrent lesions are larger than 1 to 2 cm, are located over  embryonic cleavage planes, or are the sclerosing type. Some lesions are more aggressive and have multiple recurrences, and Mohs surgery is  the treatment of choice. The superficial spreading type of basal cell  carcinoma usually recurs, no matter what method of treatment is  chosen, because there are small nests of basaloid cells away from the  visible margin of the tumor.   Intralesional injection of interferon has been used for treating basal  cell carcinoma. The available data are not convincing, and  this approach is still experimental. Systemic retinoids have also been  used in the treatment of multiple skin cancers and basal cell  carcinomas, but the results are not promising. Avoidance of excessive and long periods of sun exposure is effective  in the prevention of this cancer.

 

SQUAMOUS CELL CARCINOMA

Squamous cell carcinoma is a malignant skin cancer arising from  epidermal keratinocytes with the potential for metastasis. Squamous cell carcinomas are usually seen in fair-skinned persons who have had excessive sun exposure and developed actinic keratosis. It is more common in men than women. The incidence of squamous cell carcinoma is reported to be 289  per 100,000 in women and 890 per 100,000 in men. Approximately 80,000 to 100,000 people develop squamous cell carcinoma  each year in the United States. The incidence is estimated to be 20%  to 25% of that of basal cell carcinoma. The true incidence of squamous  cell carcinomas is not available because many practitioners remove and  treat these forms of skin cancer without reporting them to the  appropriate cancer registry. Therefore any figure is likely to be an underestimation. In Australia, where data are perhaps more accurate,  the incidence of squamous cell carcinoma is 166 per 100,000 people,  652 per 100,000 for basal cell carcinoma, and 19 per 100,000 for  melanoma. In blacks and whites who are treated with  psoralen-UV light (PUVA), the incidence of basal cell and squamous  cell carcinoma is approximately the same.

Pathogenesis

Many factors may contribute to the genesis of squamous cell carcinoma.  As in basal cell carcinoma, it is the cumulative amount of lifetime UV  exposure that influences the development of squamous cell carcinoma.  Fair skinned, red-haired individuals and albinos are at  especially high risk for developing squamous cell carcinoma early in  life if they are exposed to excessive amounts of UV light, indicating  the protective value of pigmentation. Occupational and  nonoccupational exposure to sunlight are significant risk factors. It  is reported that there is an increased incidence of squamous cell  carcinoma in persons who have been treated with PUVA. PUVA  treatment has been available in the United States for the past 20  years and consists of the systemic intake of psoralen, which is a  photosensitizing drug, and exposure to long-wave UV radiation (UVA of  320 to 360 nm). In one study, males receiving more  than 200 treatments with PUVA had a 30-fold increase in incidence of  squamous cell carcinoma over the general population. Squamous cell carcinoma also developed in rodents exposed to UVB wavelength radiation. In Europe, however, the use of this  photochemotherapy has not been associated with increased squamous cell  carcinoma.

  The incidence of squamous cell carcinoma is increased among patients  who are on immunosuppressive therapy for organ transplantation as well  as in patients infected with HIV. In these  patients, the ratio of basal cell carcinoma to squamous cell carcinoma  is decreased or reversed, suggesting that the immune system may  influence the control of early squamous cell carcinoma.  Chronic ulceration, chronic sinus disease, chronic inflammation, and  scar tissues increase the chance of developing squamous cell  carcinoma. Exposure to ionizing radiation can result in the  development of squamous cell carcinoma several years later.   Certain topical agents used for the treatment of chronic skin  conditions are carcinogenic and perhaps cause squamous cell carcinoma.  Tar causes skin cancer in laboratory animals, but there is no evidence  of tar’s causing skin cancer in humans. Squamous cell  carcinoma was considered an occupational hazard among cotton spinners  due to contact with the cotton oil. Industrial exposure to chemicals  (e.g., cutting oils) is now recognized as a risk factor for squamous  cell carcinoma. The historic reports of scrotal malignancies in  chimney sweeps was assumed to be due to contact with carcinogenic  soot. Arsenic was also recognized to be a carcinogen  producing skin cancer and cancers of other organs. In the presteroid  era, arsenic was used in the treatment of many diseases. Direct exposure of the skin by industrial workers to  hydrocarbons such as paraffin, tar, shell oil, and creosote oil may  cause squamous cell carcinoma.

  Many HPV types have been found in some squamous cell carcinoma. HPV types 16 and 18 were found in cancers of the  cervix. HPV infections are normally found in patients who are not immunosuppressed. Squamous cell carcinoma of mucocutaneous  sites usually occurs in patients with a history of heavy alcohol and  tobacco use. Lip and mouth lesions are also associated with chewing  tobacco and betel nuts. The lower lip is a common location of squamous  cell carcinoma associated with solar exposure.

Clinical Presentation

Squamous cell carcinoma usually appears on the areas of the skin that  are damaged by sun exposure. The most common sites of squamous cell  carcinoma are the face, neck, back, forearm, and dorsum  of the hand in elderly white persons with sun-damaged skin. The primary lesion  manifests as a red, indurated papule that appears de novo or on an  actinic keratosis and expands rapidly, producing a large nodule that  eventually ulcerates and metastasizes to a local draining lymph node.  Bowen’s disease, cutaneous horn, chondrodermatitis  nodularis helicis, chronic ulcers, scar tissues, and radiodermatitis  may be precursor sites of squamous cell carcinomas and basal cell carcinomas. The patient may have several primary lesions developing at the same  time or in a rapid succession. Tumors arising from sun-damaged skin  have relatively lower rates of metastasis, but tumors arising on the  mucocutaneous surface or those arising from skin with previous tissue  changes (e.g., scars of burns, sites of trauma) tend to be more  aggressive, invade locally, and metastasize rapidly. 

Squamous Cell Carcinoma,  T4N0M0

 

Squamous cell carcinoma of mucocutaneous sites usually occurs in  patients with a history of heavy smoking and heavy alcohol intake.  These lesions may arise from an area of leukoplakia or an  indurated or ulcerated plaque and metastasize rapidly.  Chewing tobacco and betel nuts can influence the development of oral  squamous cell carcinoma. Verrucous carcinoma may occur in the oral  cavity or sole of the foot and may present as a persistent, firm, or  vegetating plaque on these areas that enlarges slowly, invades  locally, and rarely metastasizes. 

Squamous Cell Carcinoma of lower eyelid with invasion in bulbar conjunctiva,

T4N0M0

 

Giant condylomata of Buschke-Lowenstein usually presents as warty  lesions on male genitalia. This condition is associated with HPV.  Squamous cell carcinoma may develop in a preexisting  condition of Buschke-Lowenstein condylomata. Epithelioma cuniculaturn  usually presents as a small ulcer with peripheral hyperkeratosis on  the soles of the feet. This lesion is difficult to  eradicate.

  Diagnosis, Classification, and Prognosis

Diagnosis of squamous cell carcinoma ultimately requires histologic  conformation. The characteristic features of squamous cell carcinoma  include proliferation of atypical squamous cells invading the  underlying dermis producing an irregular, disorganized architecture.  Individually keratinized cells and horn cysts are present. In verrucous carcinoma, giant condyloma of Buschke-Lowenstein, and  epithelioma cuniculatum, there is hyperplasia of the mucous membrane  epidermis but no evidence of invasion into the dermis. Mitotic figures are infrequent. 

  Attempts have been made to classify squamous cell carcinoma based on  the percentage of the undifferentiated cells and to relate this to the  prognosis. There are four grades of squamous cell carcinoma. Grade I has fewer than 25% undifferentiated cells, and grade IV  has more than 75%. The higher the number of undifferentiated cells,  the worse the prognosis. Differential diagnosis of squamous cell  carcinoma includes keratoacanthoma, seborrheic keratosis, pyogenic  granuloma spindle cell melanoma, and soft tissue sarcoma with spindle  cells. Antikeratin antibodies can be used to identify and confirm the  keratinocyte origin of a given tumor. This is even possible  in formalin-fixed, paraffin-embedded specimens. Antibodies against  S-100 proteins or NKIC3 indicate a possible melanoma.

Management

Most practitioners use similar treatment modalities for basal cell  carcinoma and squamous cell carcinoma.  Squamous cell carcinoma requires a more aggressive approach, such as  wider excision. Tumors on the mucous membranes must be excised with  good margins. Mohs micrographic surgery may be used. Close follow-up for recurrences and metastatic spread is highly  recommended. In most cases, surgery is preferred to radiation therapy. 

The selection of a treatment modality for squamous cell carcinoma  depends on several factors: size, shape, and location of lesion, depth  of infiltrate, desired cosmetic result, and physician experience.  Excision with adequate margins, radiation, and electrodesiccation are  the most frequently used. Radiation treatment in verrucous carcinoma  is, however, probably contraindicated. Radiation or  chemotherapy may be used in local or distant metastasis. Cryosurgery  has been used with reasonable cure rates by experienced physicians.

  Methods of treatment currently under development or in early clinical  use include laser radiation, photodynamic therapy, and local  immunotherapy. Trials involving interferon-alpha and humaatural  leukocyte interferon have shown good response rates in both basal cell  carcinomas and squamous cell carcinomas.

 

Squamous Cell Carcinoma of Cheek, T4N0M0

 

Results after half course of gamma-therapy, 45 Gy

 

Surgery approach:

    

Skin Squamous Cell Cancer of Neck, T4N0M0. Postoperative wound is temporary covered with pig skin xenografts

 

  

Granulation wound after removing of xenografts.  Skin autografting.

 
Complete recovery. 12 months later.

Screening and Prevention

Approximately 700,000 nonmelanoma skin cancers are diagnosed and  treated in the United States each year, accounting for a significant  portion of the health care budget. The incidence of  squamous cell carcinoma and basal cell carcinoma will likely continue  to climb. This is in large part due to our society’s continued heavy  exposure to UV radiation through sunlight, tanning booths, and the  chronic cumulative effect of the exposure. This problem is compounded  by the continuous depletion of the ozone layer. Although many chemicals known to destroy the ozone layer such as halogenated  chloroflourocarbons and carbon tetrachloride are being eliminated, the  depletion is expected to continue for many years. One study estimated  that a 10% decrease in the ozone layer will result in a 16% to 18%  increase in the incidence of squamous cell carcinomas in men and  women.

  An additional factor contributing to the increasing incidence of skin  cancer is the ballooning number of elderly people who are at increased  risk for skin cancers due to a lifetime of UV radiation exposure.  Theoretically, if everyone were fully protected from UV radiation  beginning today, basal cell carcinomas and squamous cell carcinomas  secondary to solar exposure would continue to appear for several  generations due to damage already perpetrated.   Currently, the most logical and effective approach is to identify  those at risk for skin cancer, provide early diagnostic guidelines,  and educate the public about the need for regular skin cancer  screening by an experienced physician. Excessive sun damage can be  prevented with commercially available sunscreens. A sun protective  factor (SPF) of at least 15 is recommended in the general population.  Although sunscreen use has not yet been proven to decrease the  incidence of skin cancer, it has been shown to decrease the incidence  of precancerous actinic keratoses. Increased pigmentation  in light-skinned persons could be an alternative approach. Several  laboratories are experimenting with a melanocyte-stimulating hormone  analogue that would produce greater melanization of the skin after  topical or systemic use.

  Another possible approach is the reversal of actinic damage before the  development of skin cancers. It is thought that UV-induced photoaging  is the precursor of actinic keratoses that are precursors of  nonmelanoma skin cancers. By reversing photoaging, it may be possible  to decrease, prevent, or delay the development of these nonmelanoma  skin cancers. Studies have demonstrated that with use of sunscreens,  the number of actinic keratoses in patients are decreased.

 

  CANCER-ASSOCIATED GENODERMATOSES

Several inherited dermatoses are associated with the development of  cutaneous malignancies. These include xeroderma pigmentosum, nevoid  basal cell carcinoma, familial dysplastic nevus syndrome, and multiple  self-healing epitheliorna of Ferguson-Smith.

XERODERMA PIGMENTOSUM

Xeroderma pigmentosum is a rare autosomal recessive disease that  occurs in approximately 1 in 250,000 persons in the United States and  Europe and up to 1 in 40,000 persons in Japan. It is characterized by  cutaneous photosensitivity with a decreased ability to repair UV  radiation damage to DNA. The disease presents in early childhood with  photophobia, erythema, and cutaneous pigmentary changes. This stage is  followed by early development of cutaneous malignancy (basal cell  carcinoma, squamous cell carcinoma, and melanoma). Ocular  changes are common and occur in 60% to 90% of patients. Some forms may  have neurologic changes such as low IQ, spasticity, ataxia,  sensorineural deafness, and microcephaly. 

  The underlying abnormality is a defect in the first steps of  nucleotide excision repair. UV radiation induces damage to  DNA, known as photoproducts, which iormal persons are excised and  replaced. In xeroderma pigmentosum, the rate of enzymatic removal of  these photoproducts is substantially decreased, so that the occurrence  of spontaneous mutations is increased. [ref: 283] The faulty enzyme  may be an endonuclease in some cases or an accessory proteieeded to  initiate the repair process in others.

  Xeroderma pigmentosum is a heterogenous disease. Seven complementation  groups, A through G, have been identified. The sensitivity to UV  radiation and the degree of neurologic disease vary between each  group. Approximately 20% of xeroderma pigmentosum patients do not fall  into one of the complementation groups. These people, known as  xeroderma pigmentosum variants, are clinically indistinguishable from  classic xeroderma pigmentosum. However, the nucleotide excision repair  system is not defective in these patients. It is  hypothesized that these patients may have a postreplication repair  defect.  Although the clinical picture is very characteristic in older  children, a young child may present with only sun sensitivity. In this  case, it is necessary to rule out the possibility of porphyria with  the proper porphyrin studies. The clinical diagnosis of xeroderma  pigmentosum can be confirmed by DNA repair study of fibroblast  culture. Management centers on avoidance and protection from UV light  exposure and adequate treatment of precancerous and cancerous skin  lesions. The use of topical 5-FU or cryotherapy for multiple lesions  is beneficial. Radiation therapy is not recommended. Oral retinoids  have been successful in the prevention of new skin cancers, but  withdrawal of therapy results in reversal of this effect.  The severity of the disease correlates closely with the residual  repair capacity. Avoidance of the sun and use of total sunblocks as  early in life as possible should be initiated. With the use of  sunscreens and other protection mechanisms, these patients live much  longer, and they usually die of neurologic involvement and infection  rather than UV-induced cancer. Neurologic involvement and progressive  mental dysfunction are seen in more than 50% of those who live longer.

 

  NEVOID BASAL CELL CARCINOMA SYNDROME

Basal cell nevus syndrome is inherited as an autosomal dominant gene  with complete penetrance and variable expression. It is a relatively common genodermatosis, but 30% to 50% of the patients have no family history, suggesting a high rate of spontaneous mutation. The gene defect is mapped to chromosome 9q, but the abnormal gene product  has not yet been delineated. Clinical features are seen in  skin, bone, optic tissue, and the central nervous system. Skin lesions  consist of basal cell carcinoma that have atypical features resembling  skin tags or pink or flesh-colored small papules and palmar and  plantar pits. Bone cysts are seen in the mandible, and abnormalities  of the ribs and vertebrae occur. The ophthalmic evidence includes  coloboma or cataracts. Other manifestations include intracranial  calcification, ocular hypertelorism, enlarged occipitofrontal head  circumference, macrocephaly, and increased risk of developing certain  tumors, especially medulloblastoma, meningioma, and ovarian fibromas. 

  It is thought that the expression of the autosomal dominant gene  requires solar radiation. Skin lesions usually develop in large  numbers between puberty and age 35. They may become nodular or  ulcerative and locally aggressive and cause significant destruction of  normal tissue. After basal cell carcinoma appears, various modes of  therapy should be used to remove the tumors completely. Radiation  therapy is not recommended, since there is evidence that it may lead  to the development of additional basal cell carcinomas as well as  other malignancies.

 

Cutaneous Melanoma

Cutaneous melanoma is becoming a more common disease. In 1995, an  estimated 32,100 individuals developed melanoma and 7200 died of the  disease in the United States. In 1996, it is estimated that 38,300 new  cases will be diagnosed, a 12% increase in the incidence of the  disease from 1995. No other tumor is increasing faster iumber of  new cases diagnosed. The death rate for melanoma has doubled in the  last 35 years with increases approximately 5% per year in the older  caucasian population. Twelve women and seven men die each day of  melanoma and $1.25 billion is spent each year on the care of the  melanoma patient. The reasons for this epidemic are still unclear, but  may result from combinations of increased recreational exposure to  sunlight, an increased amount of ultraviolet B (UVB) irradiation from  sunlight that reaches the earth’s surface, and earlier detection of  melanoma. Worldwide, 40% of the cases occur ionwhite populations,  but in the United States melanoma is largely a disease confined to the  caucasian population, where the age-adjusted incidence rate nationally  is about 12 per 100,000. It may be threefold higher (30 per 100,000)  in some geographic areas.  In 1935, only 1 in 1500 individuals  developed melanoma. By 1980 this ratio had dramatically increased to 1  in 250, to 1 in 135 by 1987 and assuming present trends, 1 in 75 by  the year 2000. 

  Fortunately, most new patients are diagnosed early in the disease’s clinical course, when it can be cured with simple surgical treatment.  While some subtlety can be involved in the diagnosis of “thin” melanomas, over 90% of intermediate and thick melanomas can be recognized as malignant by experienced observers; therefore, it  behooves each physician to know the clinical characteristics of  melanoma so that biopsies can be performed on suspicious moles or skin  lesions as early as possible. Histologic verification and microstaging with an accurate tumor thickness are essential before embarking on treatment. The options for therapy range from very conservative surgical treatment for early lesions to more radical approaches for  biologically aggressive melanomas. Judgment, experience, and knowledge of the prognostic factors are essential for choosing the most  appropriate treatment for individual patients.

CLINICAL CHARACTERISTICS

HIGH-RISK POPULATIONS

Investigators have identified risk factors for the development of  melanoma and mathematic models have been used so that “high-risk”  populations can be identified for potential screening programs.  Counting the number of raised nevi on the arms has been  suggested as a simple self-screening test to identify persons  requiring physician examination. Prospective randomized controlled  trials should assess the feasibility of this procedure so that models  can be produced for the population screened. These models can then be  applied to statewide and national programs. For example, MacKie and  colleagues followed 116 patients with 3 or more clinically  atypical nevi for at least 5 years. Patients were examined and  photographed every 3 to 6 months and nevi undergoing change were  excised for histologic diagnosis. Among 85 patients with no personal  or family history of melanoma, 5 invasive melanomas developed during  583 person-years of follow-up. The expected number in this population  was 0.05 and the calculated relative risk was 92 (P < .001). There was  a similarly increased risk among 24 patients with atypical nevi and a  family or personal history of melanoma, with a relative risk reported  to be 91. By comparison, no second melanoma developed among 25  patients with previous melanoma and a normal nevus pattern during 213  person-years of similarly intensive follow-up. The risk of melanoma  was highest among seven patients with atypical nevi and a family  history of melanoma (relative risk, 444). The median thickness of  surveillance-detected melanomas was 0.75 mm. This study illustrates  the ability to identify a high-risk population that can be followed to  detect early, “thin,” curable melanomas.

  A patient with a past history of melanoma has a higher risk of  developing a second primary melanoma than an individual in the general  population has of developing a melanoma. [ref: 9] The risk of  developing a second and third melanoma varies from 3% to 7% in  different series, a 900 times higher risk than that of  the general population. A patient who has multiple  dysplastic nevi or has a familial form of melanoma has an even greater  risk of developing multiple primary melanomas than do other patients.  Familial melanomas are uncommon but have been well  documented, and individuals in such families constitute an  identifiable high-risk group. Between 4% and 10% of  patients describe a history of melanoma among their first-degree  relatives. Clark and colleagues described an  autosomal dominant hereditary occurrence of melanoma, originally  termed the B-K mole syndrome and now referred to as the dysplastic  nevus syndrome, familial type (FAM-M). Patients with this  syndrome typically have between 10 and 100 pigmented lesions,  predominantly on the trunk, buttocks, or lower extremities. Genetic  factors may also play an important role in the predisposition to  melanoma, perhaps by genes controlling some aspect of immune response  to melanoma antigens. Melanoma-prone patients appear to  have increased frequencies of certain blood, complement, and human  leukocyte antigen (HLA) phenotypes.

SIGNS AND SYMPTOMS 

Melanomas can be located anywhere on the body, but most commonly occur  on the lower extremities in women and the back (between the scapulae)  in men. Several articles have described and illustrated some of the  clinical characteristics of melanoma. Typical features of  intermediate and thick cutaneous melanoma include (1) variegation in  color; (2) an irregular raised surface; (3) an irregular perimeter  with indentations; and (4) ulceration of the surface epithelium.

Although melanomas may have a variety of clinical appearances, the common denominator is their changing nature.  Therefore, any pigmented lesion that undergoes a change in size, configuration, or color should be considered a melanoma, and an  excisional biopsy should be performed. Recently, emphasis has been  placed on the early diagnosis of “thin” melanoma: lesions that are  malignant but curable with simple surgical excision. This increased  emphasis is due to the ability to identify high-risk populations and  the proliferation of screening programs for the disease, both on a  national and regional basis. The diagnosis of “thin” melanomas may be subtle, but physicians and primary care providers need to familiarize  themselves with early signs of malignant melanoma. These include the  ABCD’s of early diagnosis with A denoting asymmetry of the lesion, B  border irregularity, C color variation, and D a diameter greater than  6 mm. These lesions may not have an antecedent history of change.  On figure is a panel of examples of “thin” melanomas published by the Skin Cancer Foundation for physician education:

 

  GROWTH PATTERNS

A convenient way to categorize melanomas is by their growth patterns. These growth patterns represent distinct pathologic entities and have unique clinical features that should be identifiable  by the experienced clinician. Histologic confirmation is obviously  essential before making any definitive treatment plans. The four major  growth patterns are superficial spreading melanomas, nodular  melanomas, lentigo maligna melanomas, and acral lentiginous melanomas.

Superficial Spreading Melanoma

Superficial spreading melanoma (SSM) constitutes the majority of  melanomas (about 70% in most series). The lesions  generally arise in a preexisting nevus. A history of slowly evolving  change of the precursor lesion over 1 to 5 years is not uncommon, with  more rapid growth developing months before diagnosis. SSMs can occur  at any age after puberty. A typical SSM first appears as a deeply  pigmented area in a brown junctional nevus. The lesion may take on a lacy appearance.

 

Often there are patches of regression recognizable by an amelanotic area. Early in its evolution, an SSM is generally a flat lesion. It may develop an irregular surface, usually asymmetrically,  depending on the vertical growth phase that occurs as it enlarges. As the lesion grows, the surface may become glossy. There is characteristic notching or indentation of the perimeter, especially as the SSM enlarges.

  Nodular Melanoma

Nodular melanoma (NM) is the second most common growth pattern in most  series (15% to 30% of patients). NMs are more aggressive  tumors and usually develop more rapidly than SSMs. They can occur at  any age (but usually in middle age and are most common on the trunk or  head and neck). Men tend to have more NMs than women, whereas the  opposite is true for SSMs. NMs are usually 1 cm to 2 cm in diameter,  but can be much larger. They begin more commonly in uninvolved skin  rather than arising from preexisting nevi. 

  Nodular melanomas are generally darker than SSMs, more uniform in  coloration, and more raised or dome shaped. The typical NM is a  blue-black lesion that often resembles a blood blister or hemangioma.  It may have other shadings of red, gray, or purple. About 5% of NMs  lack pigment altogether (i.e., are amelanotic) and have a fleshy  appearance. NMs are often symmetric, but sometimes they appear as  irregularly shaped plaques. They lack the radial (horizontal) growth  phase so typical of the other growth patterns and therefore have  discrete, sharply demarcated borders, often with irregular perimeters.  NMs that are polypoid, with a stalk or cauliflower appearance, are  particularly aggressive.

  Lentigo Maligna Melanoma

Lentigo maligna melanoma (LMM) appears to be a separate entity from  melanomas with the other growth patterns, because LMMs do not have the  same propensity to metastasize. LMMs constitute a small  percentage of melanomas (usually 4% to 10%) and are typically located  on the face in older caucasian women. Usually LMMs have been  present for long periods of time (5 to 15 years). They are generally  large (more than 3 cm), flat lesions that occur in an older age group,  being uncommon before the age of 50 years.

Almost all are located on the face or neck, although a few may occur on the back of the hands or the lower legs. They are typically tan lesions with differing shades of brown. Irregular mottling or flecking may appear as the lesions  enlarge, with areas of dark brown or black in some parts and areas of  regression in others. LMMs can have extremely convoluted borders with  prominent notching and indentation, which generally represent areas of  regression. The diagnosis of LMM requires the presence of sun-related  changes in both the epidermis and dermis. 

  Acral Lentiginous Melanoma

Acral lentiginous melanoma (ALM) characteristically occurs on the  palms or soles or beneath the nail beds. However, not all  plantar or volar melanomas are ALMs: a minority are SSMs or NMs. ALMs occur in only 2% to 8% of caucasian melanoma patients but in a substantially higher proportion (35% to 60%) of  dark-skinned patients, such as blacks, Asians, and Hispanics. 

  The majority of ALMs are located on the sole of the foot. They are generally large, with an average diameter of about 3 cm. ALMs  generally occur in older people, with the average patient’s age in the 60s. The lesions’ evolution is relatively short, ranging from a few  months to several years, with an average of 2.5 years. Initially,  these lesions appear as tan or brown flat stains on the palm or sole  and often resemble LMMs. The haphazard array of color is  characteristic. A minority of such lesions take on a flesh-colored  appearance that can be misdiagnosed as granuloma. Ulceration is not  uncommon, and fungating masses can result from neglected lesions. As  with LMMs, these lesions often have very irregular, convoluted  borders. However, ALMs are much more aggressive than LMMs and are more  likely to metastasize. 

  Acral lentigines melanoma also include subungual melanoma, an  infrequently seen presentation of cutaneous melanoma. It  develops in only 2% to 3% of caucasian patients but in a higher  proportion of dark-skinned patients. It occurs equally in men and  women and is most often diagnosed in older patients (median age: 55 to  65 years). Over three-quarters of subungual melanomas involve either  the great toe or the thumb. The most common sign of an early subungual  melanoma is a brown to black discoloration under the nail bed.

 

  SCREENING FOR MELANOMA

Malignant melanoma is theoretically conducive to screening because a  detectable preclinical phase corresponds to the noninvasive radial  growth phase that may last months or even years. This  clinically apparent radial phase consists of horizontal growth of the  melanoma without the potential for metastases. Since the radial growth phase may last for years, the changes may be found  during interval screening examinations. Another line of evidence to  suggest the efficacy of screening and early removal of “atypical”  pigmented lesions is the finding that if patients are kept free of  “atypical” moles, they are also kept free of melanoma. Cohen  has recently reviewed his 14-year experience with prophylactic skin  nevus removal. Seventy-five of 250 patients with a past history of  melanoma underwent prophylactic removal of multiple skievi and 28%  of the lesions showed some atypia. Twelve melanomas were discovered  and it was estimated from previous reports of prevention of melanoma  with dysplastic nevus removal that four to six additional  melanomas were prevented by excising precursor lesions. During the  same time period, another 112 patients without a history of melanoma  underwent prophylactic lesion removal. Three cases of melanoma were  found and it was estimated that another three to five cases were  prevented with the removal of the precursor lesion. An average of 2.7  lesions were removed among all screened patients. No melanoma-related  deaths were reported during this 14 year period. 

  Another fact that supports melanoma screening is that there is an  “intermediate” end point for efficacy: the tumor thickness at  diagnosis. Multiple regression analyses have repeatedly confirmed  tumor thickness to be the most powerful prognostic factor for stage 1  and 2 melanoma. Tumor thickness helps the clinician  estimate the chance of occult nodal and systemic metastases, the need  for nodal staging, and the prognosis for the individual patient. There  is a high correlation between tumor thickness and survival. Estimated  5-year survival rate for persons diagnosed with melanoma less than  0.76 mm is 96%, compared with 30% for persons diagnosed with lesions  greater than 4 mm. [ref: 65] Although the true power of a screening  examination can only be eventually judged by survival data,  investigators do not have to wait 5 or 10 years to measure the  effectiveness of a program. If there is a steady drop in mean tumor  thickness at diagnosis and a drop in absolute numbers of thick  lesions, this should lead to increasing survival of the screened  population. 

  This phenomenon is illustrated by data from the Sydney, Australia,  Melanoma Project. Australia has the highest incidence of melanoma of  any location in the world, due to an equatorial climate and an  immigrant Celtic population with fair skin. The mean tumor thickness  at diagnosis in 1960 for this area was greater than 2.5 mm. In 1960,  an intensive education campaign was instituted for the lay public and  community physicians on the early signs of malignant melanoma. Almost  immediately, more patients were referred to skin cancer specialists,  more melanomas were diagnosed, and the mean tumor thickness began to  fall. By 1986, the mean tumor thickness at  diagnosis had decreased to 0.8 mm. [ref: 67] This was associated with  an increased diagnosis of superficial spreading melanoma in the radial  growth phase, a decrease in the diagnosis of ulcerated lesions that  have a worse prognosis, and an increase in survival. The 5-year survival before 1960 for stage I and II melanomas was 82%, improving  to 94% in the last decade. Thus, the earlier decrease in tumor thickness at diagnosis seen in data from Australia eventually influenced the survival data, although lead time bias may have  contributed to some of this increase in survival. 

  Studies from other parts of the world confirm the increase in survival noted with the diagnosis of thinner melanomas. In Scotland, early work showed that a high percentage of patients were presenting  with thick melanomas, defined as tumor thickness greater than 1.5 mm.  Eighty-four percent of the patients delayed seeking medical advice for more than 3 months after noticing a new or changing pigmented lesion.  Patient delay was attributed to a lack of knowledge of the  consequences of a new or changing pigmented lesion. Armed with this  data, a public and professional education campaign began in Glasgow. The response and the resulting publicity led to a 380%  increase in the number of patients referred to pigmented lesions  clinics. This was associated with a doubling in the number of  melanomas diagnosed per month compared to the period before the  intervention. There was an overrepresentation of younger women and an  underrepresentation of older men in the population attending the  clinics. An analysis of thickness of melanomas at diagnosis in the whole of Scotland from 1985 has shown a sustained and significant  shift in favor of the diagnosis of thinner lesions. The percentage of  tumors detected less than 1.5 mm in thickness rose from 38% in 1979 to  1984 to 54% in 1985 to 1989. [ref: 70] Melanoma mortality trends show  a downward slope for women in the west of Scotland, but the trend for  men is still upward.  MacKie and colleagues have shown  that the mortality of melanoma is decreasing in the female  subpopulation in Scotland, implying that the publicity with education  and early detection programs in Scotland is having a survival effect.  This is an important study, since this is the first population that  has shown a leveling off and actual decrease in mortality of melanoma  due directly to an interventional program. In contrast, until  recently, the mortality rate for melanoma in Australia has continued  to rise, with perhaps a leveling off of the rate in the last 2 to 3  years. 

  Screening for melanoma in the United States has been largely performed  under the auspices of the American Cancer Society and the American  Academy of Dermatology (ACS/AAD) Skin Cancer Education and Screening  Demonstration Project. In this effort, local and national media  disseminate public information about melanoma/skin cancer. In  addition, volunteer dermatologists provide educational materials and  free screenings (visual examinations) in special sessions open to the  public. To achieve maximal effects, education and screening for early  detection are offered together because of the unique external and  visible nature of the disease and to make use of the “captive”  audience who registers for the screening. 

  From 1985 to 1993, this volunteer effort has grown rapidly, providing  more than 650,000 free screenings. Screenings have averaged nearly  100,000 persons per year since 1990. 

  Mass media have been involved in the educational component of this  demonstration project. For example, in May 1992, 238 television  stations in more than 150 US cities (with populations greater than  50,000) carried skin cancer broadcasts during prime-time news  coverage. These messages penetrated all 30 of the top TV markets in  the United States, reaching an estimated 50 million Americans. Further  data are needed to determine how these messages affect knowledge,  attitude, and behavior of people toward sunlight exposure.

Screening for Melanoma (Continued)

The free screening programs are generally conducted in community  hospitals, physician offices, area workplaces, and local shopping  malls. Volunteer screening dermatologists provide educational  materials and visually examine the skin for cancer (melanoma, basal  cell carcinoma, squamous cell carcinoma) or precancerous lesions  (actinic keratosis, atypical mole/dysplastic nevus). No biopsies or  diagnostic studies are performed. Screening participants found to have  lesions suspected of being cancer or precancerous lesions are asked to  consult a dermatologist or their own family physician after the  screening to obtain definitive diagnosis and appropriate treatment.  AAD staff encourage persons to seek follow-up medical care and provide  names of physicians in the area. All persons across the country with  suspected melanoma receive telephone calls and letters to ensure rapid  follow-up. 

The results of the national screening program in the United States are  as follows. The program has registered an increasing number of people  each year, suggesting the demand for this public service. The ACS/AAD  program was not designed as a formal screening study and the  evaluation has been limited to preliminary analyses. Nevertheless,  important questions can be answered with information from areas in  which local follow-up was possible. Koh and colleagues achieved 85% follow-up of 2560 patients screened in Massachusetts

  Thirty percent of the screened population had an abnormal examination  and 0.3% had melanoma. An appropriate self-selection took place: 86%  of the registrants had one of the risk factors for melanoma and 78%  had two. The screened population had a predominance of white, female  college-educated people, and these investigators have questioned how  to attract older white men of low socioeconomic status. Rigel reported on a follow-up study from the 1986 Manhattan program,  concentrating exclusively on the yield of biopsy proven melanomas. Of  the 2239 persons screened by dermatologists, 14 cases (0.63%) of  melanoma were verified by biopsy. Other issues such as sensitivity,  specificity, and compliance with follow-up were not addressed. Further evaluations must address questions such as the compliance with the  referral recommendation, the rate of pathologically confirmed results,  the sensitivity and specificity of the skin examination performed by  various health care providers, and the cost effectiveness of the  program. 

  As the first step to achieve comprehensive ascertainment of those  screened nationwide, the AAD used several follow-up systems for people  with a screening diagnosis of melanoma in the 1989 to 1993 programs. A  centralized follow-up program has now successfully contacted 97% of  people in the 1992 to 1993 screened population with a suspected  diagnosis of melanoma at the time of screening. Preliminary  results find that AAD skin cancer screenings appear to detect early  melanoma (almost 99% of all screen-detected melanomas are stage I and  II), with fewer advanced melanomas identified compared to the 1990  SEER registry. Specifically, of those persons with a screening  diagnosis of melanoma attending the 1992 to 1993 AAD national  programs, 257 melanomas have been confirmed, with all but four cases  being diagnosed at a localized stage of disease (American Joint  Committee on Cancer [AJCC] stage I and II). Only 18 of the 257 melanomas (7.0%) discovered during this time have had thickness  greater than 1.5 mm. Lesions that were either in situ or had tumor  thickness less than 0.76 mm accounted for 77% of the screened detected  melanomas. All but four persons had localized disease. In comparison  with 1990 SEER population-based data, screen-detected persons (1992 to  1993) had fewer advanced melanomas (P = .003). The  investigators caution that the results are preliminary. Lacking a  formal control group and without longer follow-up, they cannot make  any projections about improved mortality. There is also the strong  likelihood that self-selection bias and other screening biases exist. 

  ACS/AAD screening programs for melanoma/skin cancer in the United  States may serve as the sole opportunity for skin cancer examinations  for some participants. In an analysis of insurance status and prior  dermatologic care among participants (1992 and 1993), 80% of  registrants reported not having a regular dermatologist, 52% would not  have seen their primary care physician without the free screening, and  9% had no health insurance. Eighty percent of persons were attending  their first program, suggesting that screenings also offer an  opportunity for many persons to receive their first complete skin  examination by an expert. Of significance is the fact that 36% of  persons with screen-detected melanomas stated that they would not have  sought a doctor without the free screening. 

  During the next few years, further studies should examine issues  including: 

   The benefit of educational campaigns (e.g., can one measure   an increase in high-risk persons seeking skin cancer   examinations from nondermatologist physicians?)     Melanoma mortality in intensely screened states versus   states without intensive screening.  Proportions of high risk persons (i.e., men 50 years of age   or over) attending screening programs. Rigorous analysis of these and other issues should help determine the ultimate role of education and screening in reducing melanoma  mortality. The ideal study for skin cancer screening would be a  randomized prospective trial. No such randomized trial currently exists in the United States or worldwide and it may never be done because of educational contamination in many areas of the United States and the time and expense involved in a prospective high-volume trial. Viable public health strategies for reducing the morbidity and  mortality rate for melanoma involve educating primary care providers  to incorporate the skin examination as part of their routine physical  examination and recognize the early signs of melanoma diagnosis, since  less than 20% of the population have a regular dermatologist. Fletcher  [ref: 74] has found that 85% of the population in the United States  see a physician every 2 years for a routine physical examination,  which is among the 10 most common reasons for seeing a physician.  [ref: 75] Primary care physicians already manage skin problems and  dermatologic complaints constitute 7% of all ambulatory patient  visits. [ref: 8] It makes public health sense that the primary care  visit be an opportunity for the screening examination to take place.   Screening for melanoma may not work. Certainly, the finding of  amelanotic melanoma in 1% or 2% of the cases may decrease the  sensitivity of the skin examination. A fraction of the melanomas,  perhaps as many as 60% of the lesions, arise de novo with no  antecedent lesion. The radial growth phase may be too short to detect  in a screening interval, although the weight of evidence is to the  contrary. Finally, the early subtle changes of “thin” melanomas may at  times be too subtle to be clinically apparent.  Nevertheless, the increasing incidence of the disease, the growing  understanding of the natural history of melanoma, the public health  problem in years of life lost, the well-established effectiveness of  treating early melanoma, and the availability of an acceptable, safe  screening test simply does not exist for other cancers and favors the  investigation of screening as an early detection for melanoma. 

PRIMARY PREVENTION OF MALIGNANT MELANOMA

Primary prevention of malignant melanoma is a longer-term exercise.  From what is known about the growth kinetics of malignant melanoma, it  is likely that trends of decreasing melanoma mortality and decreasing  tumor thickness might be seen within 3 to 5 years of mounting a public  education campaign aimed at early detection. However, the latent  interval between receipt of an insult on the skin and development of  melanoma may be longer than 20 years. Programs in many countries  currently educate the public on safe sun exposure, on the  assumption that excessive exposure to natural ultraviolet radiation is  the most important causative agent in developing malignant melanoma.   Epidemiologic studies strongly support this hypothesis, with  increasing evidence that sunlight exposure in early childhood is a  significant risk factor for subsequent development of malignant  melanoma. The exact wavelength and action spectrum for the development  of malignant melanoma are, however, not yet established. 

  Because of the recently recognized importance of avoiding excessive  sun exposure in early life, emphasis also includes the education of  young mothers and school-aged children. A wealth of educational  material is now available for primary school children in a variety of  many languages. Educational materials stress safe sun approaches  including avoidance of noonday sun, the use of shade (such as trees or  sun umbrellas), protective clothing (e.g., hats and t-shirts) and  frequent application of a sunscreen with a high sun protection factor  (SPF). 

  The field of assessment of primary prevention of melanoma is a new  one, pioneered by Drs. Robin Marks and David Hill in Australia. These investigators stress that knowledge and attitude changes  precede behavioral change and emphasize the importance of  age-appropriate material that does not arouse fear or alarm. These  programs emphasize enjoyment of outdoor activities with appropriate  precautions to prevent excessive sun exposure. These activities are  inherently more difficult to monitor and audit than early detection  activities, but preliminary surveys in Australia indicate a greater  awareness of the hazards of sun exposure. 

  However, there is still considerable room for improvement in the  public’s knowledge of dangerous sun exposure. For example, a survey of  22,000 individuals in the United Kingdom in 1993 found sunscreens with  an SPF of 4 to be the most popular. Recent surveys conducted by Marks’  group in Australia have shown that the effect of their 35-year public  education campaigns has resulted through the years in an increased  awareness to the disease in Australia, with more people performing sun  avoidance activities. But the message has saturated the public at this  point, with recent surveys showing a leveling off of the educational  effect and an inability to reach any more of the population. 

  Effecting structural change is the next strategy that will be employed  in Australia. This is defined as changing the social, political, or  economic structure of the community that would then lead to behavioral  change. Examples of this strategy would include the passage of laws so  that employers are require to provide sun protection to their outdoor  employees, the distribution of free sunscreens, and the building of  public shade structures for the community or tree-planting campaigns  to provide shade for schools. 

  Assessment of the efficacy of primary prevention campaigns is a  long-term activity, involving rigorous monitoring of incidence and  mortality trends. With increasing availability of leisure time, and  the possible effects of a fall in ozone levels in the Northern  Hemisphere, it is possible that incidence rates will continue their  upward trend of the last 20 years. The importance of this observation  has recently been recognized with the awarding of the 1995 Nobel Prize  in Chemistry to three researchers for their work in atmospheric  chemistry, particularly concerning the formation and decomposition of  ozone and the ozone layer.  Primary prevention activities also refer to the avoidance of excessive  exposure to artificial ultraviolet radiation. In the northern United  States and Europe sunbeds and sun lamps have been popular during the  winter months and, more recently, at all times of year, to promote the  “year-round” tan. Three case control studies, from Canada, the United  Kingdom, and Sweden, all show that excessive use of UV sunbeds is an  independent risk factor for malignant melanoma. In the United States,  the tanning salon industry has grown tremendously in recent years with  estimates as high as 1 million people visiting daily and 2 million  regular patrons. There are over 20,000 salons with another 40,000  units in beauty salons and health clubs. [ref: 78] Female users  outnumber men by 3 to 1 and the peak incidence of use is during  adolescence and early adulthood. People with a skin type I phenotype  (those who cannot tan) are just as likely to visit a tanning salon as  those with a type III skin phenotype (people with some natural  protection). Motivation for visiting tanning salons seems to be  self-image improvement, protection from subsequent sun exposure, and  mood modification. The bulbs used in the tanning salons emit UVA  radiation, at a peak wavelength of 350 nm. The amount of UVB  radiation, the wavelength that is implicated in skin cancer and  melanoma development, ranges from 0.025% to 4% in the bulbs, thus  tanning salons have advertised a “safe” tan. 

  Although UVA radiation is more efficient at producing a tan, the tan  that develops is inefficient for subsequent protection against  burning. Short-term problems such as eye injuries (UVA is  predominantly absorbed by the lens of the eye) and erythema are  common, and long-term problems such as cataract development, skin  photoaging, and skin cancer are expected to surface with time. It  appears that the training of operators and warning of patrons is  inadequate. 

  Education and legislation are addressing these concerns. The Federal  Trade Commission has recently ruled that patrons need to use eyewear  and health risk notices need to be placed on the tanning units. In  addition, tanning salons cannot advertise the ability to provide a  “safe” tan or any other health benefit associated with the activity.  In Texas, children under the age of 14 must be accompanied by an  adult, adolescents under the age of 18 need parental permission, and  clients have to sign an informed consent form stating that they have  been warned about the potential risks of using the salon. Primary  prevention activities also should advise against excessive exposure to  artificial UV radiation. 

  Targeting primary prevention messages to the high-risk population has  been recommended. In Europe this appears an appropriate strategy since  the incidence of melanoma, although rising rapidly, is still  relatively low. In contrast, policy decisions in high-incidence  countries (e.g., Australia) target all caucasians. MacKie and  colleagues [ref: 6] have identified four important independent risk  factors; total number of banal nevi, presence of freckling, presence  of three or more clinically atypical or dysplastic nevi, and a history  of three or more episodes of severe sunburn. A melanoma risk factor  chart, in regular use in a number of clinics, categorizes the  population into four main groups. The group with the most  significantly increased risk of developing melanoma receives  additional advice against excessive sun exposure, and possibly  surveillance. The chart has recently been extended and confirmed in a  German population.  Sun avoidance and sensible sun exposure is the mainstay of advice on  primary prevention. Currently monitors to measure UV penetration of  the skin are being evaluated. Since no safe level of sun  exposure has been determined, such monitors must be treated  cautiously. Tumor Thickness

  The total vertical height of a melanoma is the single most important  prognostic factor in stage I and II melanoma. [ref: 80] It is a  quantitative parameter that can define subsets of patients with  different survival rates (P < .00001). Numerous groups of thickness  subsets have been analyzed for their prognostic value (e.g., smaller  than 1 mm, 1 to 4 mm, larger than 4 mm), without much improvement on  the subdivisions of tumor thickness originally proposed by Brelow.  [ref: 80] However, there are apparently no “natural breakpoints,”  [ref: 86] but rather statistically defined subgroups that will vary  from one data set to another depending on the number of patients, the  duration of follow-up, and the distribution of other factors (e.g.,  ulceration, anatomic site of primary lesion, and gender). The large  number of patients in the UAB-SMU series has permitted the derivation  of a simple nonlinear mathematic model that describes the relationship  between tumor thickness and 10-year mortality as a continuous event.

 Level of Invasion

There is an inverse correlation between increasing level of melanoma  invasion and survival. The level of invasion is a significant  prognostic factor by single-factor analysis (P < .00001), and it will  differentiate patients at various risks for metastases. A direct  comparison of the level of invasion and thickness microstaging methods  was made by subdividing each level of invasion into thickness  categories. Within levels III, IV, and V, gradations of thickness  influenced survival. Converse relationships were not observed when  analyzing sets of melanoma thickness subdivided by levels of invasion. For example, the 5-year survival rates for patients  with level III, IV, or V lesions that measured 1.5 to 4.0 mm were not  significantly different. These observations demonstrate that the tumor  thickness measurement is a more accurate prognostic factor than level  of invasion.

Ulceration

Ulcerated melanomas appear to be more aggressive lesions biologically,  since they invade through the epidermis rather than pushing it  upwards. Thus, the presence of ulceration in microscopic sections of  melanoma was a significant adverse determinant of survival (P <  .00001) in the UAB-SMU study. [ref: 85] Stage I and II melanoma  patients with ulceration had a 10-year survival rate of 50%, whereas  those without ulceration had a 79% 10-year survival rate (P < .0001).  Even after correction for tumor thickness in prognostic models, the  variable ulceration continues to add prognostic information.

Other Prognostic Factors Based on the Primary Tumor

Clinical variables such as age, gender, race, and primary site; and  pathologic factors such as histologic type, mitotic rate, tumor  ploidy, S-phase fraction, DNA content, lymphatic or vascular invasion,  host response, and regression have all been analyzed in multiple  regression analyses. Occasionally these factors will be important in  the mathematic model that predicts prognosis in individual  populations. [ref: 89,90] However, once tumor thickness and ulceration  are controlled for, many if not most of the other prognostic factors  lose much of their significance noted in a multivariate analysis.

Multifactorial Analysis

Clinical and pathologic parameters can be simultaneously compared for  their prognostic strength using multifactorial analysis. The influence  of these factors on survival was examined in the UAB and SMU  population using a cohort of 4568 AJC stage I and II patients for whom  information was available for all prognostic factors being analyzed.  There were three dominant factors predicting survival: (1) thickness  of the melanoma, (2) melanoma ulceration (presence or absence), and  (3) anatomic location (upper extremity, lower extremity, trunk, or  head and neck). Four other variables also correlated to a lesser  extent with survival in certain subgroups of patients: (1) the type of  initial surgical management (primary excision alone versus excision  plus elective lymph node dissection), (2) pathologic stage (I and II  versus III), (3) level of invasion, and (4) gender.   Most other major studies utilizing multifactorial analysis have  established tumor thickness to be the most important prognostic factor  in their series of stage I and II melanoma patients. [ref: 88]  Ulceration was likewise found to be another strong predictor of  survival in other patient series. [ref: 89-91] Some of these series  confirmed that anatomic location of the primary lesion was another  major predictor of survival. Primary site location may be  more predictive of survival in specific sites of the body, such as  melanomas that occur beneath the nail bed.

  Other factors analyzed by multifactorial analysis that emerged as  important variables overall or within selected subgroups in other  series included gender, race, [ref: 83] and age of the  patient, as well as tumor features such as lymphocytic  infiltration, [ref: 91] tumor diameter, [ref: 91] cell type, microscopic satellites, [ref: 96] mitotic activity, [ref: 88] and  level of invasion. The timing of the biopsy prior to first  definitive treatment was also found to have a significant influence on  survival in one series. Flow cytometry analyses of DNA  content (aneuploidy versus diploidy) correlated with survival in  several recent studies.

  The impression that melanoma arising during a pregnancy is associated  with virulent disease and poor survival is derived from case reports  and clinical studies of small numbers of patients. Wong and associates demonstrated that the clinical course of 66 patients with  stage I and II melanoma diagnosed during a pregnancy was no different  than that of the 619 nonpregnant controls. In a larger series from  Duke University, 100 women were identified who were  pregnant at the time of diagnosis of their melanoma and compared to a  nonpregnant control population. Mean follow-up was 6.8 years and  overall mortality between the two groups was the same (25% in the  pregnant group and 23% in the control arm). Among the pregnant group,  there was an increased incidence of lymph node metastases and this  increased the overall incidence of recurrent or metastatic disease in  this population. Multivariate analysis demonstrated that pregnancy at  diagnosis was significantly associated with the development of  metastatic disease (P = .008), when controlling for tumor site,  thickness, and Clark level. The failure to document a survival  difference between the two groups may be explained by the need for a  greater statistical power, or by the fact that nodal disease can be  surgically excised and may not have the lethal implications that a  systemic recurrence would in this population. 

  Other studies have addressed the impact of a subsequent pregnancy  after diagnosis on the clinical course of patients with melanoma.  Reintgen and associates have found that the disease-free  interval did not differ between 43 patients who had a pregnancy within  5 years of their diagnosis of melanoma and those women who never  became pregnant during their clinical course.

METASTATIC MELANOMA IN REGIONAL LYMPH NODES

(AJCC STAGE III)

Once patients develop metastatic melanoma to their regional nodes,  prognostic factors based on the primary melanoma contribute very  little to the prognostic model.  Number of Metastatic Nodes  There was a direct correlation betweeumber of metastatic nodes and  survival in the UAB and SMU database. Patients with one metastatic  node had a better survival rate than patients with two metastatic  nodes or more. Data from patients with different numbers of  nodal metastases were analyzed for survival differences. The greatest  differences in this series were between patients with one metastatic  node, patients with two to four nodes, and patients with five or more  nodes (Fig. 41.2-8). Thirty-seven percent of patients had one  metastatic node, 38% had two to four, and 25% had five or more. Their  3-year survival rates were 40%, 26%, and 15%, respectively (P < .001).  Ten-year survival rates demonstrated that only patients with one  positive node had a reasonable prospect of cure (40% were alive at 10  years), whereas only about 15% of patients with two or more metastatic  nodes were alive at 10 years. 

  The number of metastatic nodes was first shown to be of prognostic  significance in a multifactorial analysis by Cohen and associates. Later, both Day [ref: 104] and Callery and associates demonstrated that tumor thickness and the number of metastatic  nodes were dominant independent variables in stage III patients.  Cascinelli and colleagues identified the extent of nodal  metastases (i.e., confined to or invading through the lymph node  capsule) and the number of metastatic nodes as the most significant  factors in a multifactorial analysis of 530 stage III patients.

Molecular Biology Techniques for the Staging of Melanoma

For most solid tumors, including melanoma, the most powerful predictor  of survival is the presence or absence of lymph node metastases. The  presence of lymph node metastases decreases the 5-year survival of  patients approximately 40% compared to those that have no evidence of  nodal metastases. Much time, effort, and expense are placed on  identifying prognostic factors based on the primary tumor, while not  enough emphasis is given to identifying which patients really have  signs of micrometastatic disease in their nodal basins. For instance,  there are currently 26 prognostic factors (Table 41.2-3) for melanoma  based on variables from the primary tumor. Yet in multiple regression  analysis performed on many collected populations in the literature,  the lymph node status of the patient with melanoma is the most  powerful factor for predicting recurrence and survival. Primary tumor  variables such as Breslow thickness, ulceration, primary site, and  gender may add to the prognostic model, but only after nodal status is  considered. It is also clear that routine histologic examination of  the regional lymph nodes, an examination that typically involves  making one or two sections of the central area of the node and  staining with a standard hematoxylin and eosin stain, examines less  than 1% of the submitted material and will miss micrometastatic  disease. The sensitivity of this examination is finding one abnormal  melanoma cell in a background of 10**4 normal lymphocytes. If serial  sectioning and immunohistochemical staining are added, the yield of  positive dissections may double and the sensitivity becomes  identifying one abnormal melanoma cell in a background of 10**5 normal  lymphocytes. Serial sectioning and immunohistochemical staining  techniques have been available for years, yet have not been  incorporated into the everyday practice of the pathologist for  examining nodes, because of the time and expense involved. 

  New technology that enables the surgeon to map the cutaneous lymphatic  flow from the primary tumor and identify the sentinel lymph node (SLN,  see below) in the regional basin could contribute to better nodal  staging of the melanoma patient. This procedure, as initially proposed  by Morton and colleagues [ref: 107,108] has shown that the SLN is the  first site of metastatic disease. If the SLN is negative, the  remainder of the lymph nodes in the basin should also be negative.  Selective lymphadenectomy also allows for the detailed  examination of the SLN, since it is an examination of one or two  nodes. This advance allows the pathologist to section the node  serially and use immunohistochemistry to look for micrometastatic  disease. Nevertheless, 25% of the histologic node-negative stage I and  II melanoma patients will experience recurrence and die of their  disease within 5 years of diagnosis, suggesting that some of these  patients have missed nodal micrometastases or these patients have  hematogenous metastases. A more sensitive method was needed to  identify accurately the presence or absence of metastatic disease in  the node. 

  A study was initiated by investigators at Moffitt Cancer  Center to develop a highly sensitive method to detect micrometastases  by examining lymph nodes for the presence of tyrosinase messenger RNA.  The hypothesis was that if messenger RNA for tyrosinase was found in  the lymph node preparation, that finding is good evidence that  metastatic melanoma cells are present. 

  The assay was refined using the combination of reverse transcription  and double round polymerase chain reaction (RT-PCR) on preparations  from lymph nodes. The amplified samples were examined on a 2% agarose  gel and tyrosinase cDNA was seen as a 207 base-pair (bp) fragment.  SLNs from 124 patients with primary melanomas greater than 4.0 mm  thick were analyzed by standard pathologic staining and RT-PCR.  Fifty-eight percent of the histologic-negative SLN population were  upstaged with the RT-PCR assay. This subgroup had a worse prognosis  than the patients whose SLN were negative for micrometastases with  both routine histology and the RT-PCR assay to suggest clinical  correlation. In a spiking experiment, one SK-Mel-28 melanoma cell in 1  million normal lymphocytes could be detected, thus indicating the  sensitivity of this method to be two orders of magnitude greater in  sensitivity than routine histology. In addition, analysis by  restriction enzyme mapping showed that the amplified 207 bp PCR  product was a part of the tyrosinase gene sequence. From this study it  was concluded that the RT-PCR method was an extremely sensitive,  reproducible, and efficient technique for the identification of  micrometastases in patients with melanoma and could be widely  applicable. 

  Other investigators have also used molecular biology techniques to  improve the staging of the melanoma patient. Dale and colleagues used an RT-PCR technique to detect occult circulating tumor cells  in the blood of melanoma patients. Four different markers were used to  improve the sensitivity and specificity of the occult metastases  assay. Preliminary studies of this multiple marker RT-PCR assay, which  utilized the four melanoma markers tyrosinase, MAGE-3, Muc-18, and p97  showed the presence of all four markers in 10 melanoma cell lines and  none detected in the blood of 14 normal volunteers. The assay was used  to detect circulating tumor cells in 74 melanoma patients of various  clinical stage. The pattern of marker detection was as follows:  tyrosinase (59%), MAGE-3 (9%), Muc-18 (66%), and p97 (65%). For all  AJCC stages, the ability to detect circulating tumor cells was  significantly higher (P = .025) in the 53 patients alive with disease  than the 21 disease-free patients. 

  For all patients alive with disease, as the clinical stage increased  from I to IV there was a corresponding increase in the ability of the  assays to detect circulating melanoma cells. It was concluded that the  detection of circulating occult tumor cells could provide a tumor  marker for the early detection of metastatic or recurrent disease or  evaluate the response to specific therapeutic modalities. 

  The staging of the melanoma patient is becoming more important with  the recent publication of a multicenter, prospective randomized trial  that purports to show a benefit for the adjuvant treatment of T4 or  stage III melanoma patients with interferon alpha (IFN-alpha).  Adjuvant therapy should be applied early when tumor burden is minimal  and in a selective fashion so that only those patients with a proven  benefit are exposed to the toxicities and expense of the adjuvant  therapies. The accurate staging of the melanoma patient will identify  the subgroup of patients who have the most to benefit from IFN-alpha.

 

METASTATIC MELANOMA AT DISTANT SITES (AJCC STAGE IV)

The data for 200 patients with distant metastases treated at UAB were  analyzed for predictive factors affecting survival rates.

  Site of Distant Metastases

The locations of distant metastases were an important prognostic  factor when examined by single-factor analysis (P = .0001). The skin,  subcutaneous tissues, and distant lymph nodes were the most common  first sites of relapse, which occurred in 59% of patients. In 23% of  patients, nonvisceral metastases at these sites were the sole  manifestation of disease (14% for skin, 5% for subcutaneous sites, and  4% for distant lymph nodes). The median survival duration for the  entire patient group was 7 months, with 25% alive at 1 year. There was  no difference in survival among patients with metastases at any  combination of these three sites. The next most common site of first  relapse was the lungs (36% of patients). Patients with isolated lung  metastases had the longest median survival duration (11 months) of  patients with metastases at distant sites. The brain, liver, and bone  were the next most common sites of first relapse. The median duration  of survival for these patients was very poor, ranging from 2 to 6  months, with a 1-year survival rate of only 8% to 10%. 

  The presence of visceral metastases had an overriding influence on  survival, since those patients with combined visceral and nonvisceral  metastases had the same poor prognoses as those with visceral  metastases alone.

SURGICAL MARGINS OF EXCISION

Local control of a primary melanoma requires wide, local excision  (WLE) of the tumor or biopsy site with a margin of normal-appearing  skin. The reason for a WLE is that approximately 5% of the primary  melanomas will have a satellite focus of melanoma separated from the  main lesion. Until recently, the routine surgical approach was to  excise all primary melanomas with a 3- to 5-cm margin and apply a  split-thickness skin graft to the defect. However, it has become  increasingly clear that the risk of local recurrence correlates more  with the tumor thickness than with the margins of surgical excision.  It therefore seems more rational to excise melanomas using  surgical margins that vary according to tumor thickness and  ulceration, since these factors correlate best with the risk for local  recurrence.   The earliest lesion is a melanoma in situ.  This is a  noninvasive tumor that does not metastasize but is capable of  recurring locally. Although the natural history of these  noninvasive lesions is not completely understood, there is a risk of  local recurrence (either as an in situ melanoma or an invasive  melanoma) if they are not reexcised after biopsy. It is  therefore recommended that the biopsy site of an in situ melanoma be  excised, usually with a 0.5 to 1 cm margin of skin.   For thin melanomas (smaller than 1 mm thick), there has been only  minimum risk of local recurrence in all reported patient series,  despite wide variations in margins of excision. In other  words, survival is not influenced by the size of the resection  margins. In 1991, the World Health Organization (WHO) published their  series on 612 patients with melanomas less than 2.0 mm who were  randomized to receive a 1.0 cm compared with a 3.0 cm WLE. With a mean  follow-up of 8 years, the disease-free and overall survival were  similar in the two groups. [ref: 133,134] There were no local  recurrences in the group with melanomas less than 1 mm thick. However,  there was a 6% local recurrence rate in patients with melanoma 1 mm to  2 mm thick who received narrow surgical margins, compared with 1% in  those that received wider margins (3 cm). [ref: 135] These results  demonstrate conclusively that a narrow excision for thin melanomas  (less than 1 mm) is safe, but no conclusion could be reached about the  appropriate margin of excising melanomas more than 1 mm thick. This  study set the standard of care of a 1 cm WLE for melanomas less than 1  mm thick. The 1 cm WLE can be performed as a generous elliptical  excision and a primary skin closure. 

 

      

     

Wide local excision of skin melanoma with wound plastic.

  A randomized prospective study conducted by the Intergroup Melanoma  Committee has evaluated 2-cm versus 4-cm radial margins of excision  for intermediate-thickness melanomas (1 to 4 mm). Local recurrences  increased in frequency as the tumor thickness increased, but there was  no significant difference between the different WLE groups. Patients  with melanoma between 1 and 2 mm had a 1.3% incidence of local  recurrence, while patients whose melanomas were between 2 and 3 mm and  3 and 4 mm had local recurrence rates of 3.7% and 6.5%, respectively.  Lesions without ulceration had a local recurrence rate of 1.10% and  those with ulceration had a rate of 6.67% (P = .001). The 15 patients  who suffered a local recurrence had a dismal survival rate at 5 years  of 28%, illustrating the clinical impact of a single local recurrence.  Some physicians believe that a local recurrence may be a marker of  systemic disease. This study set the standard for WLE for patients  with “intermediate” thickness melanomas (1 mm < t < 4 mm) as a 2-cm  WLE.  The risk of local recurrence may exceed 10% to 20% for those melanomas  over 4 mm thick, and no study has addressed the proper  WLE margin for thick melanomas.

 

ELECTIVE LYMPH NODE DISSECTION

RATIONALE. The issue of ELND is probably one of the most important controversies  in the management of patients with melanoma. The debate is finally  coming to a close since the results of the Intergroup Melanoma  Surgical Trial have shown significant improvement in subgroups of  patients based on age and thickness (see later) and because of the  increasing applicability of intraoperative lymphatic mapping. Two  randomized prospective studies involving only extremity melanomas did  not demonstrate any survival advantage for ELND, while  three nonrandomized studies involving melanomas from all anatomic  sites showed a statistically significant improvement in survival for  the subgroup of intermediate thickness melanomas. While  there is unanimous opinion that all melanoma patients do not need  ELND, there is still a continuing debate that centers around two  issues: (1) Is it possible to identify accurately a subgroup of  melanoma patients at high risk for microscopic regional nodal  metastases and a low enough risk of occult systemic metastases to  justify a regional node procedure? (2) What is the optimal timing of  dissection (immediate versus delayed) if such a high-risk group can be  identified? Prospective randomized studies have recently been  completed to resolve this issue.

  SELECTION OF PATIENTS

Prognostic Factors.  Tumor thickness provides a quantitative estimate of the risk  for occult metastatic melanoma at regional and distant sites. In fact, melanoma thickness is the most important but  not the sole guide for selecting patients who might benefit from ELND. The major advantage of using tumor thickness for these  surgical decisions is that it can provide a quantitative estimate of  the risk for occult metastatic melanoma in both regional and distant  sites. Thin melanomas (less than 1 mm) are associated  with localized disease and a 98% or greater cure rate. ELND would  provide no therapeutic benefit in such patients. Patients with  intermediate-thickness melanomas (1 mm to 4 mm) have an increased risk  (up to 60%) of harboring occult regional metastases, but have a  relatively low risk (less than 20%) of distant metastases. Patients with these lesions might therefore benefit  from ELND. Patients with thick melanomas (more than  4 mm) not only are at high risk for regional nodal micrometastases  (more than 60%), they also have a high risk (greater than 70%) of  occult distant disease at the time of initial presentation. These patients do poorly as a group, since the distant  metastases in most instances negate the benefit of surgically excising  the regional lymph nodes. The treatment goal of removing these nodes  is palliative, and the operation might be deferred until nodal  metastases become clinically evident. Some surgeons prefer to perform  ELND as expectant palliation in patients with thick melanomas to avoid  the probability (about 40%) of a second operation for lymph node  metastases. [ref: 175] ELND might also be justified as a staging  procedure to document the pathologic status of the lymph nodes in  patients with thick melanomas prior to entry into clinical trials  involving systemic adjuvant chemotherapy or immunotherapy. Ulcerative  melanomas have a higher risk for micrometastases than their  nonulcerated counterparts, even when matched for other prognostic  parameters such as tumor thickness. Tumor thickness should not be the sole criterion for making surgical  treatment decisions. Other factors, such as the presence or absence of  tumor ulceration, the patient’s gender and age, the anatomic location  of the melanoma, and the operative risk should all be considered when  making the decision to perform ELND on any individual patient. For  instance, for melanomas less than 1.0 mm thick, a nodal procedure may  be considered if the primary lesion is Clark level IV or greater, is  ulcerated, or shows regression, suggesting that at one time the lesion  was thicker.

 

Identifying the Regional Lymph Node.  Basins at Risk for Metastases.

Since melanomas located on the trunk and on the head and neck area  have unpredictable lymphatic drainage, it is difficult to decide which  nodal basin is at risk for metastatic disease. In many patients, this  problem has been overcome by performing a radionuclide cutaneous scan  that can accurately define the location of nodes that are the primary  drainage site for a melanoma located anywhere on the body. Lymphoscintigraphy, a nuclear medicine test that involves a  radiocolloid injection around the primary site and a scan of the  regional basins, has been used to redefine cutaneous lymphatic flow  from classic anatomic descriptions. [ref: 153]   The understanding of cutaneous lymphatic flow remains grounded in the  work performed by Sappey in 1874. By the injection of  mercury into the skin of cadavers, he was able to show lines of  demarcation running along the midline, and from just above the  umbilicus curving slowly backward to the second lumbar vertebra.  Lesions within 2.5 cm of these lines could show bidirectional  drainage. 

  Lymphoscintigraphy, a dynamic study performed in live patients, has  shown that the watershed areas of the body are much expanded compared  to Sappey’s original description and one does not see straightforward  lymphatic flow until a melanoma is located 10 cm off the midline or 10  cm off Sappey’s line. Lymphoscintigraphy, using  technetium antimony trisulfide, has been performed in the Sydney  Melanoma Unit (SMU) [ref: 168] to show the great variability in lymph  node drainage patterns with the identification of several new  lymphatic drainage pathways. Twenty-six percent of melanomas on the  back drain into an in-transit node located in the triangular  intramuscular space, 20% of the periumbilical melanomas show an  initial drainage into an internal mammary node, direct drainage has  beeoted from the back into paraaortic and mediastinal lymph nodes,  and at least one forearm melanoma showed direct drainage to a  supraclavicular node. A series from Moffitt Cancer Center (MCC) has reported on over 400 studies of patients with clinical stage  I and II disease. With a mean follow-up of 5 years, there has beeo  metastases in regional nodal basins not visualized by  lymphoscintigraphy. 

  Lymphoscintigraphy is an extremely accurate method of identifying all  nodal basins at risk for metastatic disease, and unless it is  performed preoperatively, the ELND or sentinel node biopsy may be  misdirected in up to 50% of the cases. The advantage of  lymphoscintigraphy over classic anatomic guidelines is the scan’s  reflection of functional and not just structural anatomy. The results  from the MCC and the SMU show that lymphoscintigraphy can be used as a  road map so that the surgeon does not perform too little or too  aggressive surgery. The results of these series also cast into doubt  the conclusions of studies of ELND in which preoperative  lymphoscintigraphy was not used as part of the protocol. Of the  recently completed prospective randomized trials evaluating the  efficacy of ELND, only the Intergroup Melanoma Study required  lymphoscintigraphy prior to the ELND. 

  Either all of the regional nodes should be removed or a policy of  monitoring multiple nodal sites at risk should be followed. An ELND of  two nodal basins (e.g., bilateral axillary dissection) for trunk  melanomas or ipsilateral anterior and posterior neck dissections for  head and neck melanoma may be warranted in select cases, but removing  more than two nodal basins or performing a bilateral cervical  dissection as ELNDs is never indicated. A bilateral inguinal  dissection is usually not performed electively because of its  attendant side effects, especially lymphedema of the extremities and  genitalia.

 

Intraoperative Lymphatic Mapping and Sentinel Node Biopsy

A new procedure has been developed to assess the status of the  regional lymph nodes more accurately and decrease the morbidity and  expense of a complete ELND. The technique termed intraoperative  lymphatic mapping and selective lymphadenectomy relies on the concept  that regions of the skin have specific patterns of lymphatic drainage  not only to the regional lymphatic basin, but also to a specific lymph  node (SLN) in the basin. Morton initially proposed the technique [ref:  107,108] using a vital blue dye method and showed that in animals and  initial human trials, the SLN is the first node in the lymphatic basin  into which the primary melanoma drains. He showed that the SLN  histology reflected the histologic status of the remainder of the  nodal basin, so that complete nodal staging could be obtained with an  SLN biopsy. 

  These data have been confirmed by four other institutions, including  the MCC, MD Anderson Cancer Center, the Sidney  Melanoma Unit, and University of Connecticut. These studies have demonstrated an orderly progression of melanoma  nodal metastases. Most solid tumors are thought to demonstrate a  random nodal metastatic pattern. The incidence of “skip” nodal  metastases, such as reported in breast cancer in up to 15% of the  cases, [ref: 190] precluded the use of sampling procedures of first  statioodal basins to achieve accurate pathologic staging. The data  from these surgical trials demonstrated that the pattern of nodal  metastases from cutaneous melanoma is not random. The SLNs in the  lymphatic basins can be individually identified and they reflect the  presence or absence of melanoma metastases in the remainder of the  nodal basin. This information is being used to change the standards  for melanoma surgical care so that only those patients with evidence  of nodal metastatic disease are subjected to the morbidity and expense  of a complete node dissection. [ref: 182] These findings demonstrate  accurate pathologic staging, no decrease in standards of care, and a  reduction of morbidity (no lymphedema, early return to work or normal  activity) with a less aggressive, rational surgical approach and lower  costs for the health care system. [ref: 191] The status of the SLN can  then be used as a prognostic factor to identify which patients need  the more radical complete node dissection. 

  With the addition of intraoperative radiolymphoscintigraphy to vital  blue dye lymphatic mapping, [ref: 192] the SLN localization becomes  easier and more widely applicable. In the initial study [ref: 192]  from MCC comparing the vital blue dye and radiocolloid mapping  technique, 450 microCuries of technetium-labeled sulfur colloid was  mixed with the standard isosulfan blue and injected at the site of the  primary cutaneous melanoma. The nuclear probe (Neoprobe, Columbus, OH)  was used to trace lymphatic channels from the primary site to lymph  nodes in the regional lymphatic basin. The SLN in the basin was  identified by intense radioactivity, and when the node was excised the  high levels of residual activity in the node and the low background  activity in the rest of the basin confirmed that the SLN was removed. 

  This study consisted of 106 consecutive patients with cutaneous  melanoma greater than 0.76 mm at all primary site locations. A total  of 200 SLNs and 142 neighboring nonsentinel nodes (non-SLN) were  harvested from 129 basins in 106 patients. When correlated with the  vital blue dye mapping, 70% of the SLNs demonstrated blue dye staining  while 84% of SLNs were defined as being hot by radioisotope  localization. With the use of both intraoperative mapping techniques,  identification of the SLN was possible in 96% of the nodal basins  sampled. Micrometastases were identified in SLNs in 15% of the  patients by routine histology while 2 patients had micrometastatic  disease in a hot but not blue-stained nodes. This suggests that the  radiocolloid localization identifies more SLNs, some of which are  clinically important since they contain micrometastatic disease. The addition of the radiocolloid mapping allows the  identification of the location of the SLN through the skin, without  making a skin incision. This allows many of the procedures to be  performed under local anesthesia without the need for extensive skin  flaps and thus more conservative operations.   In a subsequent trial from MCC, only those patients who had a positive  SLN were subjected to a complete node dissection. Further  micrometastatic disease was found in 22% of the patients who had a  positive SLN and eventually had a complete node dissection,  reinforcing the need for a complete node dissection in patients with a  positive SLN. Data from MD Anderson and MCC have recently been  compiled on 423 patients who had lymphatic mapping and a negative SLN  biopsy. These patients were observed and with a mean follow-up of 18  months, 12 (2.8%) have recurred in the regional basin as their first  site of recurrence. A detailed examination of the SLN in these 12  patients with serial sectioning, immunohistochemical staining, and  RT-PCR analysis [ref: 111] found the metastatic cells in 83% of the  cases, suggesting that these cases do not represent a failed mapping,  but the inability of routine histologic examination of the SLN to find  the metastatic disease. [ref: 193] 

  An NCI-sponsored prospective randomized trial is currently  being performed that randomizes patients to receive either WLE of the  primary melanoma site or WLE and SLN biopsy of the regional lymphatic  basins at risk for metastases. The endpoint of the study is whether  this surgical strategy can extend the survival of the melanoma  patient. The study is different from the previous randomized trials  addressing the efficacy of ELND, since only a percentage of the  patients with melanomas greater than 1.0 mm will receive a complete  node dissection. Even if this trial is a negative trial, with the  emerging data on the efficacy of adjuvant IFN-alpha in patients with  nodal metastases, [ref: 113] it may be necessary to perform a nodal  staging procedure on all patients with melanomas greater than 1.0 mm.  Lymphatic mapping and selective lymphadenectomy make up the less  morbid, less costly approach than ELND in obtaining nodal staging. 

  It is crucial for the surgeon who performs intraoperative mapping to  have adequate nuclear medicine and pathology support. Lymphatic  mapping requires the close collaboration of nuclear medicine, surgery,  and pathology in order to perform the procedure accurately. The  preoperative lymphoscintigraphy is a road map for the  surgeon and is used for four distinct reasons in planning the surgical  procedure: 

   1. To identify all nodal basins at risk for metastatic   disease.

   2. To identify any intransit areas of nodal collections   thatcan be tattooed by the nuclear medicine colleague for   later harvesting. Intransit nodal areas occur in 5% of the   melanoma population and, by definition, are the SLN.

  3. To identify the location of the SLN in relation to the   rest of the nodes in the basin. 

   4. To estimate the number of SLN in the regional basin that   will need to be harvested.

  Intraoperative vital blue dye and radiocolloid mapping with the  Neoprobe can identify the blue-staining afferent lymphatic draining  into the blue-staining and hot SLN. Both intraoperative  mapping techniques are needed and complementary since primary sites  near regional basins are difficult to map with the radiocolloid alone  because of enormous radioactivity at the injection site (primary  site). Likewise, radiocolloid mapping adds to the blue dye technique  by the ability to locate the so-called “hot” spot in the regional  basin through the skin, lessening the need for extensive flaps and  dissections, and providing a measure that all the SLN have been  removed from the basin (activity in basin at background levels after  the removal of the SLNs). The SLN can then be harvested, allowing the  pathologist time to make a detailed examination of the SLN with  multiple sections, immunohistochemical staining with melanoma-specific  monoclonal antibodies, HMB-45 and S-100, and perhaps RT-PCR  determination. This strategy will invariably increase the yield of  positive dissections and more accurately stage the patient with  melanoma. 

  The question then becomes what constitutes the standard of surgical  care for the patient with melanomas greater than 1.0 mm thick. With  five reports in the literature that illustrate how the histology of  the SLN is reflective of the histology of the rest of the nodes in the  basin, if surgeons have adequate support from nuclear medicine and  pathology services, there is no need to perform an ELND. In  communities and centers that do not have the collaboration in place,  or in circumstances in which intraoperative mapping cannot be  performed (such as when a previous WLE of the primary melanoma has  already been performed) or when the results of the mapping are  equivocal, the guidelines for ELND from the Intergroup Melanoma  Surgical Trial should be used. 

ADJUVANT THERAPY FOR THE TREATMENT OF HIGH RISK FOR RECURRENT STAGE II AND III DISEASE

  ROLE OF ADJUVANT RADIATION THERAPY

  The role of radiotherapy as a surgical adjuvant after therapeutic node dissection or as an alternative to ELND in the regional treatment of  patients with intermediate to thick melanomas has not been clearly  defined. The rationale for its consideration in this context is as  follows. ELND, while effectively reducing regional recurrence rates,  carries varying degrees of morbidity and does not offer any survival  advantage in patients with thick primary tumors. [ref: 176] On the  other hand, therapeutic dissection of pathologically involved nodes is  associated with a local recurrence rate of up to 50% in patients with  head and neck melanomas. [ref: 155,195] In view of the management  problems associated with uncontrolled local/regional disease and the  extent of elective dissections required for scalp and facial primary  sites, a study was initiated at M.D. Anderson Cancer Center in 1983 to  evaluate the role of radiotherapy in the treatment of clinically  uninvolved lymph drainage areas in patients at high risk of nodal  metastases, or as an adjunct to surgery in patients undergoing  therapeutic nodal dissections. 

  The preliminary results of this trial, indicating an apparent  advantage in terms of local regional recurrence for adjuvant  radiotherapy, were published by Ang and colleagues in 1990. [ref: 196]  The following is an update of the study with a median follow-up of 38  months. Through July, 1994, 224 patients (171 men and 52 women) were  enrolled. Their ages ranged from 16 to 89 years (median: 55 years).  Patients were organized into three groups. Group I consisted of 118  previously untreated patients who presented with primary lesions  either greater than 1.5 mm thick or Clark level IV or V who had no  palpable lymphadenopathy. Twenty-seven percent had lesions greater  than 4.0 mm thick. After WLE, these patients received radiotherapy to  the tumor bed and at least two echelons of the draining lymphatics of  30 Gy (D(max)) delivered in five fractions with electron beams of  appropriate energy. This group of patients had an overall 5-year  actuarial rate of local regional control of 86% and a 5-year survival  of 63%. It was interesting, that 71% of patients with primary lesions  1.6 to 4.0 mm thick were 5-year survivors after elective radiotherapy. 

  Group II consisted of 39 patients with previously untreated disease  who presented with clinically positive lymphadenopathy. These patients  mostly received postoperative radiotherapy (30 Gy D(max) delivered in  five fractions over 2.5 weeks); the remainder received preoperative  treatment (2 Gy D(max) delivered in four fractions over 2 weeks).  These patients achieved a 5-year local regional control rate of 92%  and 5-year survival of 41%, with survival being inversely proportional  to the number of pathologically involved lymph nodes. 

  Group III consisted of 67 patients who presented with recurrent  regional and/or local disease, but without evidence of distant  metastases. These patients were treated in the same manner as group II  patients. Their 5-year local regional control rate was 88% and  survival 45%. With the addition of adjuvant radiotherapy, the  local-regional control rate of group II and III patients was not  affected by the number of positive nodes or extracapsular extension. 

  These results demonstrate that adjuvant radiotherapy, either alone in  clinically node-negative patients or as a surgical adjuvant in  pathologically node-positive patients, can achieve local regional  control in excess of 85%. This is substantially better than rates  previously reported with surgery alone in comparable patients. The  radiotherapy schedule used was not associated with any significant  morbidity. In total, 3 of the 224 patients treated to date have  sustained mild to moderate sequelae. There has beeo severe  complications. Although these data are impressive, proof of a  therapeutic benefit from adjuvant radiotherapy can be obtained only  from a prospective randomized trial. Such a trial, based on this pilot  study, has been activated by the Radiation Therapy Oncology Group  (RTOG).

 

ROLE OF ADJUVANT SYSTEMIC THERAPY FOR PATIENTS

WITH MELANOMA AT HIGH RISK FOR RECURRENCE

 

  Interferon

Advances in melanoma treatment over the past decade have evolved  primarily from more detailed knowledge about prognostic factors of  primary and metastatic lesions. Within the larger group of melanoma  patients who undergo potentially curative treatment by surgical  resection, subgroups can be identified who are at high risk for  recurrence and for development of systemic metastases. Patients with  melanomas 1.51 to 3.99 mm deep have a variable and intermediate risk  of relapse, while patients with thick primary melanomas (more than 4  mm thick), in-transit lesions, and regional lymph node involvement are  at particularly high risk: the 5-year relapse risk of these stage  groupings generally exceeds 50%. Once distant metastases develop,  median survival is only 6 to 9 months. The investigation of adjuvant  systemic treatment that can prevent melanoma recurrence has become a  critical area of investigation, focused upon either the intermediate  or high-risk stage groups. The rationale and general principles for  adjuvant treatment of cancer are based on the premise that treatment,  whether chemotherapy or immunotherapy, is more effective when the  tumor cell population is small and host immune and other resistance  mechanisms are still intact. To date, randomized trials using  dacarbazine, nitrosoureas, a variety of combination chemotherapy  regimens, BCG, Corynebacterium parvum, transfer factor, and  combinations of immunotherapy and chemotherapy have not demonstrated  any advantage for treatment.   Adjuvant treatment for melanoma should be considered whenever possible  within clinical research protocols that seek to improve on the results  of current standard therapy in a systematic manner. Patients with a  high risk of recurrence and death due to melanoma in the first several  years following surgery include those patients with AJCC stage III  melanoma (positive regional lymph node pathology) or those with deep  primary invasion (greater than 4 mm Breslow depth). Patients in these  two categories have at least a 50% chance of recurrence and death  within 5 years of their diagnosis with just observation and are  candidates for adjuvant trials. Investigations over the past decade  have focused upon the interferons in this category of high-risk  melanoma, and four trials conducted in this interval have recently  been published or presented iational and international forums. The  Eastern Cooperative Oncology Group has completed a trial of adjuvant  therapy with high-dose IFN-alpha2b compared versus observation in  stage IIb or III (AJCC) patients, the results of which have served as  a pivotal basis for Food and Drug Administration (FDA) approval of  IFN-alpha2b for the adjuvant treatment of high-risk melanomas. The  treatment protocol adopted for this early trial is notable for its use  of an initial month of daily intravenous therapy at 20 MU/m**2/d for 5  days a week for 4 weeks. This 1-month period of induction therapy was  designed to deliver peak levels of circulating interferon-alpha  unattainable by other routes, and was followed by 11 months of  subcutaneous therapy at 10 MU/m**2 three times a week, designed to  sustain maximal tolerable levels of interferon in an outpatient or  home therapy setting. An analysis of 280 of 287 patients for whom full  data were available has demonstrated a significant prolongation of  median time to relapse (1.7 versus 0.98 years, P(2) = .005) and  overall survival (3.8 versus 2.8 years, P(2) = .047). Treatment was  associated with an increment in the 5-year survival from 36% to 47%,  and with an increment in the 5-year continuous relapse-free survival  from 26% to 37% (Fig. 41.2-16A and B). These data have suggested the  possibility of a curative impact of therapy with IFN-alpha2b as given  in this trial. [ref: 113] Cox multivariate analysis of this trial  showed IFN therapy to be a highly significant independent prognostic  factor and, after stage of disease, the most significant favorable  prognostic factor. 

  Toxicity of this therapy is significant, with nearly ubiquitous  flulike symptoms of moderate to severe degree, and the need for dose  delay or attenuation in more than one-third of patients during  induction, and again during the subcutaneous maintenance phase of  treatment. The completion of 1 year of therapy was feasible in the  majority (74%) of patients who did not experience progression. Proper  attention to hematologic and hepatic function has been sufficient to  avoid lethal hepatic toxicities observed early during the application  of this intensive protocol. The ECOG has led an intergroup  confirmatory sequel study (E1690/SWOG9111/CALGB9190) that completed  accrual in mid-1995 and will require up to several years of maturity  for evaluation. This protocol was designed not only to corroborate  E1684 but also to test the influence of a lower dose of IFN-alpha2b, 3  MU/d given subcutaneously three times weekly for 2 years. It closed in  1995 with accrual of 642 patients. This Intergroup Trial differed from  the initial trial E1684 in that lymph node dissection was no longer  required for patients with T4 (greater than 4.0 mm) primary melanomas. 

  In an accompanying editorial on the E1684 ECOG trial, [ref: 198] Balch  and Buzaid questioned the role of adjuvant IFN-alpha in a high risk  for recurrence patient whose tumor is greater than 4.0 mm thick and  lymph node examination is clinically negative. Although excluded from  the ECOG eligibility criteria for the E1684 trial, these patients have  a greater than 60% chance of harboring microscopic nodal metastases  and therefore may benefit from adjuvant high-dose interferon therapy.  Knowing the pathologic status of the regional nodes in patients with  thick melanomas provides important prognostic information. Patients  with thick melanomas who have negative nodes have a significantly  better prognosis than those with positive nodes (58% to 71% versus 32%  to 42%). [ref: 199] The available data from the ECOG E1684 trial were  derived from patients whose lymph node pathology was known. The  benefit of this therapy was seen for patients who entered the trial  with lymph node involvement by comparison with the small subgroup of  patients with thick, lymph node negative tumors. These authors  therefore recommended that patients with melanomas greater than 4.0 mm  have a nodal staging procedure, preferably intraoperative lymphatic  mapping and sentinel lymph node biopsy, since this is the least morbid  approach. Extrapolation of the results of E1684 may be considered for  patients with local recurrences, satellite lesions, or in-transit  metastases. Important questions that remain to be answered are whether  the high-dose intravenous induction is necessary (or may in fact be  sufficient to obtain the benefit of the E1684 treatment) and whether  more prolonged interferon administration would enhance the benefit. In  addition, the question remains whether early IFN-alpha therapy (after  microscopic nodal metastases are removed) is better than late adjuvant  therapy (after grossly positive nodal disease is resected with a  therapeutic node dissection).

    Only a small subset of patients who receive high-dose IFN-alpha2b  therapy actually derived benefit from it. Serum markers of tumors may  provide a key to the more selective application of adjuvant therapy.  Tyrosinase (a key enzyme in melanin biosynthesis whose gene is actively expressed only in melanocytes, melanoma cells, and Schwann  cells) may serve as a marker of early dissemination of disease to  allow more selective use of adjuvant therapy in the future. Because  melanocytes and melanoma cells do not usually circulate in the blood,  the detection of tyrosinase mRNA by reverse transcriptase-polymerase  chain reaction (RT-PCR) in blood is considered a potential indication  of the presence of circulating melanoma cells. Patients who  have a positive RT-PCR before starting therapy that subsequently  becomes negative may be those who benefit from the therapy. [ref: 198]   The North Central Cancer Treatment Group (NCCTG 83-7052) evaluated the  adjuvant role of IFN-alpha2a given at 20 MU/IM three times weekly for  3 months, versus observation in patients with 1.69-mm Breslow depth  T3+ or T4, and N1 patients (subsets of AJCC stages IIA or IIB and  III). An analysis of 260 evaluable patients who entered this trial  demonstrates no significant prolongation of survival, or of  relapse-free interval overall. Subset evaluation of patients in stage  II as opposed to those in stage III participating in this trial  reveals a potential impact in the latter, by Cox analysis.

  The WHO Melanoma Program Trial 16 has evaluated the efficacy of a  lower, less-toxic dosage of IFN-alpha2a, given 3 MU/d subcutaneously  three times weekly for 3 years versus observation. Of 444 patients who  entered this trial, a majority exhibited extracapsular extranodal  involvement, a pathologic variable that made patients ineligible for  the trials of the US Cooperative groups previously noted. The analysis  of WHO Trial 16 has been reported preliminarily at 22 months of  follow-up, and in a subset analysis an interaction between age and  gender and treatment was found. This interim analysis of subsets,  defined by the presence or absence of extranodal extension, has  suggested an influence of this therapy upon intranodal disease, and  lack of any trend to benefit among patients with extracapsular  extension. Subsequent reports of interim analyses of this trial at  intervals up to 39 months median follow-up have suggested the absence  of a significant impact upon either relapse-free or overall survival.

  In summary, the analyses of E1684, NCCTG 83-7052, and WHO 16, taken  together, argue that IFN-alpha2b at higher dosages, delivered  intravenously, may be necessary for the benefit observed in E1684.  Equivalent dosages of IFN-alpha2a administered by the IM route for  shorter periods have been less effective, and lower dosages of  IFN-alpha2a administered by the subcutaneous route for longer periods  have been ineffective to date. The approval of IFN-alpha2b for  adjuvant therapy within 2 months of surgery of high-risk node-positive  and deep primary melanoma is the first adjuvant therapy approved for  melanoma, and the first new agent approved for therapy of this disease  in any stage or setting, since dacarbazine. 

  The evaluation of newer and potentially more effective biologic agents  in the adjuvant setting has been pursued in multiple trials. SWOG  86-42 is a trial of IFN-alpha administered at dosages projected to be  the optimal immunomodulatory dosages (0.2 mg/d sc qod for 1 year).  IFN-alpha is among the most potent immunomodulators yet tested, and  one for which initial hopes for adjuvant and metastatic disease  applications were elevated even beyond IFN-alpha2. As previously  noted, the therapeutic activity of IFN gamma in the advanced disease  setting has beeegligible. The recently published SWOG 86-42 trial  demonstrates the lack of therapeutic benefit for this agent at the  dosage tested, in either intermediate-risk stage II or stage III  resected melanoma. Of concern is that the initial report of this trial  suggested a potentially adverse impact of treatment that has not yet  been confirmed. In any case, the unequivocal failure of IFN-gamma to  improve relapse-free or overall survival in the adjuvant setting  argues that the effects of the dose, route, and schedule of IFN-gamma  and the monocyte activation and NK activation associated with  IFN-alpha administered as in this trial are not sufficient to alter  the outcome of this disease. [ref: 203] The pleiotropic effects of  IFN-alpha2b, which account for its therapeutic benefit in melanoma,  have yet to be proven. In vivo activities that may be important range  from a direct antiproliferative and cytotoxic action to  immunopotentiation of T- or B-cell host responses, dendritic cell  antigen presentation, anti-angiogenesis factor, and tumor-restricted  antigen expression. These are being evaluated prospectively in the  context of the recently completed intergroup trial (ECOG 1690/SWOG  91-11, CALGB 91-90).

Immunotherapy

  Tumor vaccines have a long history, but the search for effective  methods to induce active immunity against tumors has been difficult.  Repeated attempts have been made to inhibit the outgrowth of tumors  and influence the natural history of melanoma by immunization against  tumor cells or extracts of tumor cells. More recently, purified  antigens recognized by the host antibody response, as well as the  epitopes defined by T-cell responses, have been identified and  prepared for use as vaccines. In addition, genetically modified  melanoma cells, recombinant vaccines, and antiidiotype antibodies that  represent the antibody-recognized antigens of melanoma have entered  into clinical trials. Evidence is mounting that vaccination can induce  immune responses to melanoma. Efforts to vaccinate patients against  the antigens of melanoma can be classified categorically into trials  of allogeneic tumor cell immunization, autologous tumor cell  immunization, and immunization against synthetic chemically defined  antigens. 

  The last category can be subclassified into approaches directed at the  induction of antibody responses, such as the gangliosides noted below,  and approaches directed at the induction of a T-cell response to  antigens that may include either proteins or peptides. Formal phase  III trials have been initiated to evaluate the potential efficacy of  relatively few tumor vaccines as adjuvant therapy in high-risk  patients. 

  At this point, it remains unclear which approaches will be most  effective. The multitude of different ongoing trials listed indicate  that there is no consensus on vaccine strategies. Early clinical  trials of each of the categories of vaccines have been conducted in  patients with metastatic disease, for reasons of safety evaluation.  Despite early concerns, no reports of tumor enhancement have been  documented with the whole-cell vaccination trials conducted to date.  Antitumor responses have been recorded in a small fraction of subjects  with metastatic disease, receiving varied programs of tumor cell  immunization against allogeneic tumor cells incorporating two of three  cell lines of hapten-coupled autologous [autochthonous] tumor cells.  [ref: 205,206] Most subsequent clinical studies have been performed in  patients after resection of regional lymph node metastases or  high-risk primary lesions. A more interesting observation is that  patients who develop evidence of immune responses have improved  disease-free survival or overall survival. This intriguing  correlation does not directly imply that vaccines improve the clinical  course (for instance, patients who a priori have a better prognosis  may be more likely to develop an immune response to vaccination), but  is consistent with the hypothesis that an immune response induced by  vaccination may have antitumor benefit. A number of problems exist in  the construction of tumor vaccines, including the weak immunogenicity  of tumor antigens, heterogeneity of tumor-restricted antigen  expression in tumors, and the ability of tumors to escape any immune  responses that may be induced naturally, or therapeutically. [ref:  208] Most melanoma antigens on tumor cells are not tumor-restricted or  specific (i.e., present only on cancer cells) but are shared with  certaiormal cells.

  Active specific vaccination trials in melanoma (as opposed to  nonspecific vaccination with BCG) have progressed from the use of  unmodified, irradiated allogeneic or autologous melanoma cells for  immunization, either alone or in combination with BCG and other  nonspecific immunomodulators. It has been difficult to demonstrate any  benefit or specific immune responses in the course of these trials.  [ref: 210,211] One exception has been immune responses to  gangliosides, the major acidic glycolipid of melanoma cells, in some  patients immunized with selected allogeneic melanoma cells.

  Efforts have been made to improve immunogenicity of allogeneic tumor  cells by treating with the enzyme neuraminidase to remove sialic acid  residues from the cell surface. [ref: 214] Strategies have been  developed to augment the immune response to tumor antigens by  infecting tumor cells with nonpathogenic viruses. Viral proteins  presented on the cell membrane of tumors have been shown to augment  the immunogenicity of tumor antigens. [ref: 215]   Several centers have investigated the immunogenicity of lysates  derived from tumor cells infected with Newcastle disease virus,  vaccinia virus, and vesicular stomatitis virus. A multicenter  randomized controlled prospective clinical trial that enrolled 250  patients with resected moderate- to high-risk melanoma was recently  reported demonstrating the lack of any therapeutic benefit of  immunization with the Newcastle disease virus viral oncolysate  approach. This is the only randomized controlled multicenter trial of  a melanoma tumor cell vaccine reported in the literature. [ref: 216]  Our understanding of molecular biology of immune response to melanoma  has increased greatly since the design of this trial. We know the  importance of tumor cell expression of histocompatibility antigens  (e.g., HLA-A2) that were lacking in this tumor vaccine, or  costimulatory molecules (B7.1) that may have been suboptimal in the  vaccine. Yet this trial demonstrates the approach that will be  necessary to evaluate the most promising vaccine candidates in the  adjuvant setting. As with the SWOG 9035 trial (see later), the  eligibility criteria for this trial included T3 (AJCC IIA) lesions for  which prognosis is more heterogeneous than for stage IIb or a stage  III disease, and the sample size was insufficient to detect small  benefits. 

  The allogeneic tumor cell vaccine reported first by Mitchell and  colleagues, admixed with the immunologic adjuvant Detox (Ribi  Immunochem Res, Hamilton, MT) has been studied in a second large  multicenter randomized trial that is enrolling moderate-risk T3 N0  patients, and will be completed in 1996. This study has been enlarged  from its original goal to 600 patients to increase the power of the  trial, and its ability to detect more subtle movements in relapse-free  survival. In addition, the understanding of the nature of melanoma  antigens recognized has moved forward rapidly, including the discovery  of a number of peptide antigens whose recognition is restricted by the  HLA-A2 allele, which was absent in the tumor cell lines employed in  this vaccine trial.   Another approach to increasing immune response has been to alter tumor  cells by binding haptens to autologous tumor cells. [ref: 217] Pilot  single-institution data with haptenated autologous tumor cells have  been intriguing but, to date, this type of vaccine has not been  evaluated in a randomized controlled or multicenter trial, owing in  part to the difficulty of manipulating autologous tumor cells for the  vaccine. [ref: 218] 

  A variety of efforts are also underway to immunize melanoma patients  with purified or partially purified preparations of potentially  immunogenic molecules from melanoma cells. The rationale for this approach comes from the identification of antigens on melanoma cells  that can be recognized by antibodies in sera or lymphocytes from  patients with melanoma. A large experience using partially purified  cultured allogeneic tumor cell antigens has been the stimulus for the  initiation of controlled trials of one vaccine preparation derived  from shed antigens of cultured melanoma cell lines.  Gangliosides have been shown to induce antibody responses after  immunization. Of particular interest is that the  ganglioside GM2 induces IgM antibody responses in more than 80% of  patients immunized with GM2 and BCG following cyclophosphamide  treatment at low dose as an immunomodulator. A prospective randomized  study evaluated the role of vaccination for 6 months against GM2 with  BCG, in comparison to BCG alone, using low-dose cyclophosphamide  pretreatment in both groups. 

  Serologic studies have shown a favorable prognostic significance for  the presence of anti-GM2 ganglioside antibodies, suggesting that the  induction of antibodies against gangliosides may confer protection  from relapse and mortality due to melanoma. However, native autologous antibody reactivity against GM2 is  infrequently detected in patients with melanoma. A variety of vaccination programs utilizing intact autologous and allogeneic cells, and cell fractions derived from allogeneic melanoma cell lines, have  been employed to increase immune responses to autologous melanoma  cells. The most promising results from randomized controlled trials of  vaccination have emerged from studies of purified ganglioside GM2.  Livingston and colleagues have performed a series of trials employing  autologous tumor cells, and more recently purified ganglioside  preparations, with various immunologic adjuvants. The combination of  BCG given together with ganglioside GM2 has been shown to induce the  formation of increased titers of IgM antibodies in the majority of  patients immunized (33 of 44). 

  A randomized adjuvant trial of GM2/BCG given following surgery for  stage III melanoma (AJCC staging) has been conducted in 122 patients  at Memorial-Sloan-Kettering Cancer Center (MSKCC) between 1987 and  1988. This trial, reported at more than 5 years of follow-up, has  demonstrated the induction of IgM anti-GM2 antibodies in a majority of  patients who were immunized, and the presence of native antibodies  against GM2 in a small (fewer than 5%) subset of unvaccinated patients  in the control group. Antibody response against GM2 was again  demonstrated to be a highly significant and favorable prognostic  factor. In addition, vaccination was associated with a prolongation of  relapse-free survival of borderline statistical significance,  confounded by the occurrence of native anti-GM2 antibody responses  among six patients in the total, five of whom were entered into the  control group. Analysis of the efficacy of immunization among  seronegative entrants into this protocol revealed a significant  improvement of relapse-free survival associated with GM2/BCG  vaccination (P = .02). 

  To further augment the immunogenicity of GM2, a variety of measures  have been explored. Covalent attachment of GM2 to carrier proteins,  such as keyhole limpet hemocyanin (KLH), has improved the titer and  durability of the IgM response seen with BCG and GM2 and induced  antibody of the IgG isotype for the first time. A phase-III  evaluation of the GM2-KLH vaccine with the QS21 adjuvant  has been planned in the ECOG and US Intergroup mechanism, as well as  in England.

 

NOTICE.

The education materials were taken from “CANCER: Principles & Practice of Oncology. 5th Edition. USA”.

 

 

 

 

 

 

 

 

 

 

 

 

 

Head and Neck Cancers

 

Tumors of the Nasal Cavity and Paranasal Sinuses, Nasopharynx,

Oral Cavity, and Oropharynx

 

The analysis of cancers within the upper aerodigestive tract reveals a heterogeneity of neoplastic processes. Each bears its own unique set of epidemiologic, anatomic, pathologic, and treatment considerations. This chapter will review such considerations based on four anatomically defined regions: the nasal cavity and paranasal sinuses; the nasopharynx; oral cavity; and oropharynx, respectively. However, there exist general principles regarding these cancers which may be considered here. Such principles involve anatomy (i.e., primarily the anatomy of regional lymph nodes within the head and neck), pathology, staging and screening, as well as general principles of treatment involving either single modality or multimodality therapy that are relevant to all sites.

 

ANATOMY An understanding of the regional lymph node anatomy is critical to the care of head and neck cancer patients. There are several major lymphatic chains in the neck containing nearly 200 lymph nodes that run parallel to the jugular veins, spinal accessory nerve, and facial artery and into the submandibular triangle. To facilitate communication regarding cervical lymph node anatomy, the regions of the neck have been characterized by levels. Level I includes nodes within the submental triangle and the submandibular triangle. The submental triangle extends from the midline anteriorly to the anterior belly of the digastric muscle posteriorly. Its third border is formed by the hyoid bone inferiorly. The submandibular triangle is bounded by the mandible superiorly. The anterior and the posterior belly of the digastric muscle complete the triangle. Level II includes the jugular nodes extending from the subdigastric area down to the carotid bifurcation and the nodes surrounding the spinal accessory nerve from the jugular foramen to the posterior border of the sternocleidomastoid muscle. It includes the lymph nodes in the upper posterior cervical triangle above the entrance of the spinal accessory nerve into this triangle.

 

 

Level III represents the nodal area principally along the jugular vein between the carotid and its bifurcation, the posterior border of the sternocleidomastoid muscle, and the omohyoid muscle. Level IV constitutes nodal areas below the omohyoid muscle above the level of the clavicle and between the carotid vessels anteriorly and the omohyoid muscle posteriorly. Level V represents nodes in the posterior cervical triangle. Its borders are formed by the posterior edge of the sternocleidomastoid muscle, the level of the entrance of the spinal accessory nerve, the trapezius muscle, and the posterior belly of the omohyoid muscle. Specific sites within the aerodigestive tract have a predetermined drainage pattern. A knowledge of this pattern will aid in diagnosis. It will also impact on therapy. Drainage patterns will be addressed in each of the anatomic subsites detailed in this chapter.

 

PATHOLOGY The predominant lesion within these anatomically defined regions is squamous cell carcinoma. Squamous cell carcinoma can be categorized into three classic differentiations. Well differentiated disease shows greater than 75% keratinization; moderately differentiated disease contributes to the bulk of squamous cell carcinoma and is characterized by 25% to 75% keratinization; poorly differentiated disease demonstrates less than 25% keratinization. Other variants of squamous cell carcinoma include verrucous carcinoma, sarcomatoid squamous cell carcinoma, and lymphoepithelioma. Additional pathologic criteria of squamous cell carcinoma, which are believed clinically relevant, were developed by Jacobsson and others. [ref: 4-7] This includes the number of mitoses, presence of vascular invasion, size of nuclei, degree of inflammatory infiltrate, and pushing or infiltrating borders (Table 29.2-1). Premalignancy A series of pathologic changes from premalignant disease to frank malignancy can occur in the oral cavity. Among the premalignant diseases are leukoplakia, erythoplakia, hyperplasia, and dysplasia. Each of these types has a propensity for malignant transformation. [ref: 8] Histopathologic assessment of leukoplakia reveals hyperparakeratosis, which is variably associated with an underlying epithelial hyperplasia. Leukoplakia without underlying dysplastic changes is rarely associated with progression to malignancy (i.e., less than 5% probability of malignant changes). Erythroplakia is a condition within the oral cavity and pharynx characterized by red superficial patches adjacent to normal mucosa. Distinct from leukoplakic lesions as identified above, erythroplakia is commonly associated with underlying epithelial dysplasia. Likewise, it can be associated with carcinoma-in-situ to frank malignancy in nearly 40% of lesions.

Dysplasia as compared to the above two clinical descriptives is a true histopathologic term, which is characterized by several morphologic changes including the presence of mitoses, pleomorphism, and prominent nucleoli. When dysplasia involves the entire thickness of the mucosa, it is commonly referred to as carcinoma in-situ. Dysplasia has been associated with a subsequent risk of progression to frank malignancy ranging from 15% to 30% of cases. [ref: 12,13] Significant advances have occurred over the last several years in defining phenotypic and genotypic characteristics of oral premalignancy. Premalignant lesions can be characterized by their increased proliferative capacity. For instance, Coltrera and colleagues [ref: 14] have demonstrated that premalignant lesions of the oral cavity demonstrate increased expression of proliferating cell nuclear antigen. Other studies have demonstrated abnormalities in differentiation pathways, primarily in the aberrant expression of cytokeratin content. [ref: 15,16] Premalignant lesions of the upper aerodigestive tract have been shown to express genotypic abnormalities as well. This includes aneuploidy, increased DNA index, and specific mutational abnormalities. [ref: 17-30] Mutations involving the p53 gene have perhaps been the most extensively characterized of the aberrant genetic events involving the premalignant state of the upper aerodigestive tract.

STAGING AND SCREENING Staging The role and current status of staging for head and neck cancer is undergoing continuous analysis. Though standard staging has been defined, there is a growing debate as to what the primary function of staging should be. The American Joint Committee on Cancer Staging (AJCC), however, has described the principal goal of staging as a means of defining the natural history of disease. Additional goals include the ability to judge therapeutic results between various centers as well as a means of defining patient prognosis. Revisions to standard staging have continually been offered, but today it is based on the tumor-nodes-metastasis (TNM) classification. T stage represents extent of primary disease. N represents the extent of regional lymph node metastasis, and M is a measure of distant metastasis. The AJCC staging system, used for classifying TNM status, is periodically revised. The current clinical staging system, although based principally on physical examination, has also incorporated specific radiographic observations of disease status. Thus, invasion through cortical bone by an oral cavity tumor will upstage a T2 or T3 lesion to T4 (i.e., from stage II or III to stage IV). Radiographic assessment of cervical lymph node metastases has not been integrated into clinical staging. The benefits of these diagnostic techniques beyond that provided by standard physical examination are under investigation. Most would consider the combination of radiographic and clinical assessment to be more accurate than either one alone. Furthermore, growing emphasis is being placed on the advantages of one imaging technique versus another (i.e., the relative merit of magnetic resonance imaging versus computed tomography). Summarizes the relative value of these two techniques in head and neck imaging. The criteria for T staging within the upper aerodigestive tract differs dependent on the primary site. N staging and M staging, however, are uniform and will therefore be considered here.

REGIONAL LYMPH NODES (N) NX: Regional lymph nodes cannot be assessed N0: No regional lymph node metastasis N1: Metastasis in a single ipsilateral lymph node, 3 cm or less in greatest dimension N2: Metastasis in a single ipsilateral lymph node, more than 3 cm but not more than 6 cm in greatest dimension; or in multiple ipsilateral lymph nodes, or in bilateral or contralateral lymph nodes, none more than 6 cm in greatest dimension N2a: Metastasis in a single ipsilateral lymph node more than 3 cm but not more than 6 cm in greatest dimension N2b: Metastasis in multiple ipsilateral lymph nodes, none more than 6 cm in greatest dimension N2c: Metastasis in bilateral or contralateral lymph nodes, none more than 6 cm in greatest dimension N3: Metastasis in a lymph node more than 6 cm in greatest dimension

 

DISTANT METASTASIS (M) MX: Presence of distant metastasis cannot be assessed M0: No distant metastasis M1: Distant metastasis Table 29.2-3 represents the most current stage classification as defined by the AJCC. [ref: 31] Screening for Primary Cancers The significance of screening is emphasized by reports that note that most oral cancer lesions could have been detected several months earlier by appropriate dental examination. [ref: 39,40] However, although it is intuitive that early detection of disease would benefit an individual patient, firm evidence to support routine screening of head and neck cancer has not been demonstrated. Indeed, the report of the United States Preventive Task Force does not recommend such a strategy for assymptomatic persons. [ref: 41] Several considerations may account for the pessimism. Head and neck cancer is a chronic disease process. It may exist in a subclinical state for a prolonged period of time and elude detection. Furthermore, head and neck cancer is a disease of patients who are in the fifth and sixth decades of life. These individuals visit the dentist infrequently and far less frequently than they visit a physician. [ref: 42,43] Some studies have suggested that dentists are more effective than physicians in routinely performing a complete mouth examination and detecting early stage oral cancer. [ref: 44] Thus, an effective approach to the screening of head and neck cancer may be in the education of primary care providers in high-risk areas on effective clinical examination. Indeed, education designs have been established and implemented. [ref: 45] Current limitations have been identified and relate to physician time constraints for prevention activities generated by other health care demands within the practice setting. The most extensively studied screening procedure has been oral exfoliative cytology, which has been recommended by the American Dental Association. [ref: 46] A high proportion of false-negative examinations have beeoted with this procedure. [ref: 47] Those individuals who are most at risk (i.e., individuals with high tobacco and alcohol consumption patterns) are least likely to voluntarily participate in such efforts. This has been emphasized by a recent report of the Metropolitan Detroit Oral Cancer Screening Control Program in which 5679 individuals were evaluated in a 27-month period. [ref: 48] Eighteen cancers were identified. High-risk populations were, however, felt not to be benefited. Another screening method frequently utilized has been toluidine blue staining of aerodigestive mucosa. [ref: 49,50] Toluidine blue is a metachromatic nuclear stain that is taken up by dysplastic and cancerous epithelium. Mashberg [ref: 50] has extensively detailed the appropriate use of toluidine blue and has noted false positive results to approximate 9% and false negative result approaching 5%. Its value is that it is quick, inexpensive, and noninvasive. Standardized physical examination is the best means of detecting lesions of the upper aerodigestive tract. The care and thoroughness of the examiner are paramount. [ref: 51,52] The need for careful assessment was emphasized by Jesse and colleagues [ref: 51] in their report on patients who present with regional metastatic disease from a presumed unknown primary. Many individuals were subsequently found to have a primary cancer with careful examination. Host Susceptibility and Screening Perhaps the most effective means of improving screening strategies is through the better identification of individuals at increased risk of developing disease. More intensive follow-up can be undertaken. Likewise, the use of potentially invasive or costly procedures can be reserved for those who would most benefit. The risk of performing such diagnostic tests on those who are unlikely to benefit would also be minimized. In that regard, the relative risk of disease as a function of duration and intensity of tobacco exposure has been well defined. The confounding influence of alcohol on tobacco-induced cancers has been extensively investigated. New developments in genetics and molecular biology, however, are now providing information as to which smoker may be at increased risk of developing head and neck cancer. Summaries of our current understanding of genetic susceptibility to head and neck cancer development have been provided. [ref: 53-55] Susceptibility factors can be placed into broadly-defined categorical groupings including genetic polymorphisms involving carcinogen-metabolizing enzymes, heritable characteristics associated with race and gender, HLA phenotypes, cancer family syndromes, as well as DNA repair deficiencies. When considering polymorphisms involving carcinogen metabolizing enzymes, one must account for innate host systems that serve to activate both carcinogens and enzyme systems that act to protect the host. In the latter category, it is the glutathione S-transferases and the acetyl transferases that have been most extensively investigated regarding the risk of head and neck cancer. Considerable variation in the general population exists in the individual capacity of an individual to express these protective carcinogen-detoxifying enzymes. Those persons who have deficient activity would be at greatest risk. Although the data are somewhat sparse at this time, several epidemiologic studies have been conducted that support this contention. [ref: 56,57] It is relevant that epidemiologic studies are defining an increased incidence of disease among certain populations within the United States. Recent data from the Surveillance, Epidemiology, and End Results Program of the National Cancer Institute have revealed that the incidence of carcinoma of the pharynx among African-American males has been increasing at a highly significant rate of 6% per year since 1973 (Fig. 29.2-3). [ref: 58] Factors that contribute to that increase have been poorly defined. However, it should be recognized that different ethnic groups and racial populations may differ in their capacity to metabolize carcinogens, and this may contribute to disease incidence. [ref: 59] Hecht and colleagues [ref: 60] have reported initial results on the capacity of various racial groups to metabolize tobacco specific carcinogens. Aberrant metabolizing capacity was found within certain individuals and African-Americans were more likely to express this deficiency. [ref: 60] Aberrant metabolism of tobacco nitrosamines may contribute to increased incidence of disease. Evidence is now accumulating that suggests that women may be more susceptible to tobacco-induced cancers. [ref: 61-64] For instance, in a case-control study involving 2829 males and 1348 females, Harris and colleagues [ref: 63] noted that female smokers had a nearly two-fold higher risk for lung cancer, after adjusting for intensity and duration of smoking. The increased incidence of second primary malignancies among women with head and neck cancer suggests an increased susceptibility to tobacco-induced disease. [ref: 61,62] In that regard, Ryberg and colleagues [ref: 64] analyzed levels of aromatic DNA adducts in the normal lung tissue of 63 lung cancer patients and found significantly higher adducts in smokers than ionsmokers. When considered on a pack-per-day exposure basis, women had significantly higher levels of adducts than men. [ref: 64] DNA adducts may set the stage for subsequent mutational events and cancer development. Perhaps the most compelling information on host susceptibility resides in the concept of DNA repair. [ref: 55] Those individuals with deficiencies in their ability to repair DNA damage would be at increased risk of developing tobacco-induced disease. It is beyond the scope of these chapter to detail the current understanding of DNA repair. The concept has been espoused for many years. [ref: 65] There is, however, supportive evidence as to the role DNA repair deficiencies play in head and neck cancer development. Perhaps the most supportive evidence comes from the understanding of chromosome fragility syndromes. These syndromes include xeroderma pigmentosum, Bloom’s syndrome, ataxia-telangiectsia, and Fanconi’s anemia. Individuals with these diseases characteristically die at an early age from multiple causes, including infections and neoplastic diseases. Despite their shortened lifespan, individuals with these diseases have been known to develop head and neck cancer. [ref: 66] The underlying mechanisms that contribute to DNA repair deficiencies is under intensive investigation. Additional evidence for the role of aberrant DNA repair as a determinant of head and neck cancer comes from several case case-control studies that examined mutagen sensitivity expressed by an individual’s peripheral blood lymphocytes. [ref: 55] Lymphocytes obtained from individuals with head and neck cancer express increased chromosome fragility to clastogens in vitro as compared to age- and sex-matched healthy controls. Furthermore, those patients who expressed the greatest sensitivity are the individuals who are most likely to develop second cancers. [ref: 67] Studies to confirm these observations are underway. Though these various studies on host susceptibility suggest important underlying causes of head and neck cancer that are independent of tobacco consumption, no conclusions can yet be stated as to the ultimate value of including these markers into screening programs to identify high-risk individuals. Such information should be forthcoming in subsequent years. Screening for Second Primary Cancers Head and neck cancer patients are characterized by their high risk of developing second primary malignancies. The majority of these cancers occur within tobacco-exposed tissue, including the esophagus, lung, and remaining upper aerodigestive tract. The risk has been well characterized and is known to occur at a rate of 4% per year. [ref: 61] Given the high rate of second malignancies within these individuals, numerous screening strategies have been described. Available screening modalities include laryngoscopy, esophagoscopy, contrast studies of the esophagus, chest x-ray, sputum cytology, and bronchoscopy. Newer modalities are under investigation including the use of molecular assessments of cells within saliva and sputum. [ref: 68] Despite this effort, no agreement exists as to the optimal screening means, including the timing and duration of follow-up. Perhaps the greatest controversy revolves around the role of panendoscopy at the time the patient presents for treatment of their index cancer. Proponents of this procedure, which includes laryngoscopy, esophagoscopy, and bronchoscopy, cite the high rate of identified second cancers, reported in one prospective study to occur in 10% of patients. [ref: 69] Disease found in this setting is presumed to be of an earlier stage and more responsive to treatment. Opponents of routine panendoscopy site the relatively low yield and questionable value in actually altering disease course and survival. [ref: 70-72] A recent prospective study by Benninger and colleagues [ref: 71] provides compelling evidence to support the use of screening procedures based principally on symptomatology (i.e., so-called symptom-directed selective endoscopy). [ref: 71] A careful history and physical examination on all newly-diagnosed head and neck cancer patients represents the most effective screening method. Though routine panendoscopy cannot be advocated, there exist certain individuals in whom its use may be more beneficial. This, in our experience, would include patients whose index cancer resides within the pharynx and who admit to a long history of tobacco and alcohol abuse. Special attention should be given to the assessment of the esophagus in these individuals, as well as the remaining upper aerodigestive tract.

 

TREATMENT Pretreatment Considerations The comprehensive care of the head and neck cancer patient begins with pretreatment considerations including the assessment of general medical conditions, nutritional status, dental health, and the appropriate choice of medical therapies designed to minimize treatment-related complications. It is beyond the scope of this chapter to detail the numerous associated medical illnesses that are typically identified in the head and neck cancer patient. Given the prolonged history of tobacco and alcohol abuse that can be typically identified, diseases involving the pulmonary, cardiovascular, and digestive systems are common. There remain, however, important considerations that should be stressed. These include the significance of pretreatment dental care, nutritional support, the impact of therapy on the elderly patient, and the choice of preoperative medications (i.e., antibiotics for the patient about to undergo major surgical procedures). The standard of care today for the head and neck cancer patient is the reduction of oral diseases prior to initiation of treatment. [ref: 73-75] Periodontal diseases, infections, and caries are common in this patient population. This can lead to loss of integrity of the gingival-crevicular tissues. Left unchecked, significant morbidity can result to structural elements of the oral cavity in the face of aggressive therapy. Following initial evaluation by the oncologic team, dentulous patients should be referred to dental colleagues for appropriate oral hygiene. Pretreatment radiographic dental surveys should identify caries and periapical lesions. Other factors that should be considered include defective restorations, ill-fitting prostheses, and impacted molars. It is generally considered prudent to perform necessary dental rehabilitation, including extractions, approximately 2 weeks prior to the initiation of any radiotherapy. This allows for the appropriate healing of extraction sites and mucosal coverage of exposed bone. To minimize delays in the initiation of radiation, dental care can be performed at the time of surgical resection in a patient who is to undergo multimadality therapy. The assessment of nutritional status and the choice of pretreatment nutritional regimens are more controversial. Several authors report the common finding of malnutrition in the head and neck cancer patient. [ref: 76-78] Severe malnutrition has been identified in over 25% of the patients. [ref: 76] Furthermore, the presence of severe malnutrition was associated with increased operative morbidity. [ref: 78] This has led to efforts to appropriately quantitate nutritional status through the use of documentation of pretreatment weight loss, the measurement of triceps skin fold thickness, and the inclusion of various laboratory measures such as plasma protein levels and the creatinine/height index. To date there has beeo conclusive randomized trial of pretherapy nutritional restoration to minimize treatment morbidity. However, Goodwin and colleagues [ref: 76] have stressed that in the severely malnourished patient, attention should be given to a 2-week pretreatment course of nutritional support. Such attempts can most often be achieved through enteral means with the placement of either a nasogastric tube or a percutaneously-placed gastrostomy tube. The care of the elderly patient represents a commonly occuring dilemma. [ref: 79-81] The tendency to deny a patient optimal treatment because of his or her advanced years should be avoided. In a study by McGuirt and Davis, [ref: 80] operative mortality was 4% in 217 patients greater than 65 years of age and not significantly different than those less than 65 years. In a subset of patients over the age of 75 and with stage III or IV disease, however, the mortality rate increased to 6%. A prospective case-control study by Kowalski and colleagues [ref: 79] on elderly patients undergoing head and neck surgery failed to identify any increased frequency of postoperative complications or mortality as compared to younger patients. The choice of treatment should not be predicated on the age of the patient. Rather, it is the patient’s general medical condition that remains the most critical consideration regardless of age. Increasing attention over the last 5 years has been given to the significance of continued tobacco use in the head and neck population. The data would support the notion that continued tobacco use following diagnosis of the index cancer will lead to an adverse patient outcome. This relates to not only the more obvious problem of progressive cardiopulmonary disease, but considerations related to head and neck cancer progression as well. Day and colleagues [ref: 82] have recently provided more information regarding the risk of second primary malignancies in patients who continue to smoke following treatment of their index cancer. The risk of second primary malignancies was significantly higher in the smoking population as compared to those who achieved smoking cessation. This difference was apparent only after 5 years following initial treatment. In another study by Browman and colleagues [ref: 83] continued tobacco use was associated with a decreased likelihood to respond to primary therapy. This latter study represents an initial report and is limited by its small population size. It does, however, raise an important consideration in the overall care of these patients, namely, the systematic approach toward achieving smoking cessation. In that regard, several studies have recently identified characteristics of patients who are likely to continue smoking habits. [ref: 84-86] Interestingly, Ostroff and colleagues [ref: 86] have recently reported that it is the patients with the best outlook who seem to be the most recalcitrant. Patients with early staged disease continued to use tobacco at higher rates than patients with higher staged disease. Strategies are being explored to effectively support the patient through this critical period. [ref: 84] The hallmark of any approach should include surgeon-delivered advice, as well as the judicious use of nicotine replacement. Finally, head and neck surgical oncologists should critically assess the need for supportive therapies in the patient who is to undergo operative procedures. This has principally related to the use of the choice of perioperative antibiotics. The use of prophylactic antibiotics can only be supported for those individuals undergoing clean-contaminated surgery (i.e., when it is anticipated that the aerodigestive tract is to be entered, or in those circumstances in which there is frank contamination). Postoperative infectious complications may occur in up to 30% of the patients. Several studies have addressed risk factors for infectious complications and found duration of the operative procedure, blood loss, and complexity of the reconstructive procedure to be high-risk variables. [ref: 87-89] Common microbials isolated from infected wounds include both aerobes and anaerobes with Bacteroides fragilis, Escherichia coli, beta-hemolytic Streptococcus, Staphylococcus, and Pseudomonas being frequently identified. Perioperative antibiotics should be started prior to the operative procedure and should continue for 72 hours postoperatively depending on the likelihood of infection. Antibiotic regimens should allow for broad coverage. Regimens include combination sulbactam and ampicillin or metronidazle combined with a cephalosporin. General Principles of Surgery In the execution of effective surgical management, the single most significant principle is the adequate preoperative assessment of disease extent. Precise and methodical physical examination of the patient is paramount. Such examination allows for the assessment of adequate extent of surgical excision, which remains for most cancers the fundamental tenet for achieving cure. An extension of adequate preoperative assessment is optimal intraoperative exposure of disease. The surgeon should consider appropriate means to achieve operative exposure. The choice of incision and the ability to mobilize surrounding anatomical structures to achieve exposure will be considered later in the text for each anatomical subsite. Additionally, exposure is facilitated by careful hemostasis. Besides allowing for better operative exposure, minimizing blood loss prevents potential sequelae associated with blood transfusion. Weber and colleagues [ref: 90] have recently reported expected blood loss for various surgical procedures involving cancers of the upper aerodigestive tract. Electrocautery dissection had been utilized by Weber and colleagues, which may explain the relatively infrequent need for blood transfusion. Electrocautery dissection has been adopted by many experienced surgeons as the preferred extirpative technique. Additional methods of surgical excision have included the use of the Mohs technique and laser ablation. [ref: 91-93] These techniques, however, cannot be considered standard surgical procedure at this time. There exist general principles of surgery involving regional lymph nodes. The standard in the surgical control of cervical metastases by which various procedures are judged is the radical neck dissection. The radical neck dissection involves complete removal of the lymphatic pathways within the neck. To assure complete extirpation, anatomical structures including the sternocleidomastoid muscle, spinal accessory nerve, and jugular vein are routinely sacrificed. Recent developments in the management of cervical lymph node disease involve more conservative surgical procedures. [ref: 3,94-96] These procedures differ from the classic radical neck dissection principally in the sparing of specific anatomical structures (i.e., principally the spinal accessory nerve and the sternocleidomastoid muscle). Table 29.2-4 provides a classification of currently used selective neck dissection and details removed lymph node regions. Controversy exists as to the value of elective neck dissection in the face of clinically N0 disease. Arguments in favor of such procedures are based on the finding that greater than 20% of clinically negative necks harbor histopathologic evidence of metastases. [ref: 97-99] Whether resection of this disease improves survival beyond that afforded by delayed neck dissection when disease becomes clinically evident cannot be stated with certainty. In one of the few prospective trials of elective neck dissection, Vandenbrouck and colleagues [ref: 100] failed to find survival benefit in oral cavity cancer patients randomized to receive elective dissection. The study is limited by the small number of individuals entered. More conclusive studies are in order. In those individuals who are not likely to receive careful postoperative assessment, it is recommended that elective neck dissection be performed. General Principles of Radiation Therapy For early-stage disease, both radiation and surgery are frequently curative and can produce similar rates of cure. Selection of treatment must be individualized to each patient and must consider issues such as cosmetic and functional outcome, quality of life, speed with which treatment can be completed, sequelae of each modality, patient reliability, risk of subsequent cancers, and capacity of salvage therapy should there be a recurrence. For advanced head and neck cancer, surgery and radiation are frequently combined. Tupchong and colleagues [ref: 101] reported the RTOG experience comparing preoperative radiation therapy (5000 cGy) versus postoperative radiation therapy (6000 cGy) for supraglottic and hypopharynx cancers. Locoregional control was significantly better for the postoperative radiation group. The results were most significant for the supraglottic cancers. Marcial and colleagues [ref: 102] found that 5000 cGy given preoperatively did not adversely affect the complication rate compared to patients who had surgery alone for advanced but resectable lesions. Nowadays, if surgery is planned for an otherwise resectable lesion, there is no indication for preoperative radiation therapy. Definitive surgery should be performed, and postoperative radiation is used. In planning treatment for advanced lesions, it is essential to separately consider the primary site and the neck and to integrate a management strategy that considers both locoregional control and quality of life. For example, an early T stage lesion with N2 or greater neck disease is a stage IV lesion. However, it is likely that the primary site can be managed by radiation alone, and the neck should receive combination therapy with surgery and radiation. Such a patient can be treated with definitive radiation therapy to the primary and neck, followed by neck dissection. This strategy maximizes locoregional control, while attempting to optimize functional outcome and quality of life. Similarly, some T3 to T4 patients may be managed by definitive radiation therapy, including brachytherapy, reserving surgery for salvage. This is especially true for oropharynx lesions, where major surgery has significant impact on speech and swallowing. Therefore, it must be emphasized that treatment of advanced, resectable disease must be individualized; whereas most patients may benefit from surgery, followed by postoperative radiation, many may be treated by definitive irradiation, with or without a neck dissection, in an attempt to obtain maximal locoregional control and quality of life. In general, when making decisions about postoperative radiation, it is important to consider the local site as well as the regional volume. In terms of the local site, reasons to add postoperative radiation include inadequate margins, significant local invasion, and a large tumor in the T3 or T4 category. Studies have demonstrated that adequate doses of well-delivered postoperative radiation therapy can sterilize cancer-positive surgical margins. [ref: 103,104] Regional issues that determine the need for postoperative radiation therapy include the presence of involved lymph nodes at one or more levels, extracapsular extension, the risk of contralateral nodal metastases requiring elective irradiation to the contralateral neck, risk of disease in undissected nodal areas such as the retropharyngeal region, and risk of disease in the lower neck. Huang and colleagues [ref: 105] identified 125 patients who underwent radical head and neck operations, and were found to have either extracapsular lymph node extension in the neck, positive resection margins, or both. Surgery alone was used in 71 patients, whereas 54 received postoperative radiation therapy. Treatment was based on physician preference, not tumor characteristics, as one group of surgeons at their institution preferred not to refer patients for postoperative radiation. The two groups were quite comparable except for the delivery of postoperative radiation. The 5-year, locoregional control and adjusted survival were significantly better in patients who received postoperative radiation. This finding was true for those with extracapsular extension as well as those with positive margins. For those with both, locoregional control was 68% in the postoperative radiation group, and 0% (P = .001) in the surgery alone group. Survival was 72% versus 41% (P = .001) for the postoperative radiation patients versus the surgery alone group. Peters and colleagues [ref: 106] reported an important prospective randomized trial from the M.D. Anderson Cancer Center. This trial attempted to determine the optimal dose of postoperative radiation therapy for patients with advanced head and neck cancer. Patients were assigned to either a low-risk or high-risk category based on prognostic factors such as T stage, N stage, resection margin status, presence or absence of perineural invasion, extracapsular extension of neck nodes, and direct invasion of tumor into adjacent structures. Low-risk patients were randomized to receive 52.2 to 54.0 Gy versus 63.0 Gy, and high-risk patients were randomized between 63.0 and 68.4 Gy. Fraction size was 1.8 Gy/fraction. It was noted early in the study that those patients receiving 52.2 to 54.0 Gy had an increased recurrence rate, so the dose for that arm was increased to 57.6 Gy. The study demonstrated that doses of 52.2 to 54.0 Gy were clearly suboptimal, that at least 57.6 Gy was required in the postoperative setting, and that patients with extracapsular extension should receive 63.0 Gy. Doses above 63.0 Gy added to complications but did not improve local control. In practice, standard procedure should be to deliver at least 57.6 Gy to the entire operative bed, and a boost to 63.0 Gy to all areas at high risk at the primary site and in the neck. Locally advanced, unresectable cancer presents a particular challenge. Although radiation alone has seen the standard of care, combined chemotherapy and radiation therapy has been demonstrated to be superior in both single arm studies and prospective randomized trials. Many of these trials will be reviewed in the following section on chemotherapy. However, particularly promising results have been reported using concomitant cisplatin and radiation. Harrison and colleagues [ref: 107] reported a series of patients using concomitant cisplatin/ radiation, using a delayed, accelerated fractionation program of 70 Gy in 6 weeks, with the last 2 weeks containing a twice daily program of 1.8 Gy in the morning and 1.6 Gy in the evening. A preliminary report revealed local control and survival of 56% and 69%, respectively, at 1 year. [ref: 107] The subgroup with unresectable skull base lesions had a particularly promising 92% local control in a separated preliminary report. [ref: 108] These data have been updated recently (not yet published) for 52 patients who have a minimum of 3-year follow-up, and a mean follow-up of 45 months. Local control is 58%. For the cohort with advanced, unresectable skull base lesions of the paranasal sinuses and nasopharynx (all T4) local control was 78%. General Principles of Radiation Therapy (Continued) (1 of 1) Merlano and colleagues [ref: 109] reported a prospective, randomized trial comparing radiation alone with an alternating chemotherapy/radiation approach. This trial is reviewed later in this chapter, but revealed an improved outcome in the chemotherapy/radiation group. [ref: 109] Side effects of radiation therapy are usually separated into acute and late effects. Acute effects generally are related to inflammatory reactions in the tissues within the radiation field (i.e., epidermitis and mucositis). Acute changes from irradiation of tissue that includes the taste buds can cause loss or diminution of taste, irradiation of the salivary glands causes xerostomia, irradiation of the lacrimal glands can cause dryness in the eye, and epilation can result from irradiating hair bearing skin. Whether these effects are temporary or permanent are usually dose and site related. Because radiation therapy to the salivary glands and oral cavity can have significant impact on dentition, all patients receiving this treatment should be seen by a dentist prior to radiation therapy. Any required dental work should be done prior to the initiation of radiation, and patients should be placed on dental prophylaxis with fluoride applications. It has been clearly shown that fluoride application significantly reduces dental sequela after radiation therapy. [ref: 110] It has also been clearly shown that dental extractions in an irradiated mandible can lead to osteonecrosis. [ref: 111] Advances in radiotherapy techniques have had a significant impact on head and neck patients. For external beam treatments, three-dimensional conformal radiation therapy is a particularly exciting new area. This allows the physician to plan radiation therapy based on three-dimensional reconstruction of the target area and three-dimensional planning of the radiation beams. As a result, it is frequently possible to lower the dose to surrounding normal tissue while potentially escalating the dose to the tumor. [ref: 112] Efforts to use this technique iasopharyngeal cancer have been particularly interesting and will be discussed in that section. The development of multileaf collimators and on line portal imaging techniques should make the delivery of three-dimensional radiation therapy more efficient. Efforts to increase the total dose and shorten the overall treatment time have been shown to be important in improving local control. [ref: 113] In this regard, there has been considerable interest in utilizing twice-a-day (b.i.d.) radiation programs. [ref: 114-116] Whether these fractionation programs truly produce better tumor control than conventional radiation are still unproved. However, considerable retrospective data are suggestive of a benefit that may be most significant in the more advanced disease patients. [ref: 114,116] A prospective randomized trial by the RTOG failed to show a benefit to the b.i.d. program, but was faulted by the fact that the total dose in the b.i.d. group was too low and significantly below the dose level in the conventional group. In the area of brachytherapy, there are also new developments. Combined external beam irradiation plus brachytherapy boost is a commonly employed strategy for cancers of the oral cavity and oropharynx, nasal vestibule, buccal mucosa, and other sites. This strategy allows very high doses to the tumor with the implant, which also serves to minimize radiation to surrounding normal tissue such as salivary gland, bone, and soft tissue. This combination of higher tumor dose with lower normal tissue dose serves to increase the local control, as well as to improve the functional and quality of life outcomes. Also, brachytherapy alone can be used for early lesions of the oral tongue, floor of mouth, lip, nasal vestibule, and other sites. This approach also allows for external beam irradiation to be held in reserve, for potential future use, should the patient develop a second primary cancer that requires that modality. External beam radiation is usually delivered once a day, five days per week, continuous course until the desired dose is achieved. For postoperative radiation, the dose requirements have already been discussed. In general, doses of 50 to 54 Gy in 5 to 6 weeks are given to areas at risk for microscopic disease that have not been operated on. Fletcher [ref: 117] has shown that doses of 50 Gy in 5 weeks are very effective in sterilizing microscopic disease in areas at risk that have not had surgery. Table 29.2-5 documents the results of elective neck irradiation. Because the anatomic separation in the head and neck region is thinner than in other parts of the body, a low-energy linear accelerator is preferred. This means that cobalt 60, 4 Mv, or 6 Mv photons will usually be required. Electron beams will also be utilized for the posterior neck or other regions where it is preferable to treat superficial structures and avoid irradiating deeper ones. Higher-energy linear accelerators have too much skin sparing to be useful in treating most head and neck cancers. Patients with involved nodal metastases are usually managed by a combination of surgery and radiation. Although the N1 neck can frequently be managed by either surgery or radiation (see Table 29.2-5), those with more advanced disease are better served by combined treatment. Table 29.2-5 summarizes the regional control rates with either radiation alone or combined with neck dissection. Table 29.2-6 shows neck control as a function of radiation dose. Hirabayashi and colleagues [ref: 118] has pointed out that 31% of the N1 patients in their series had evidence of extracapsular spread. Snyderman and colleagues [ref: 119] reported that 38% of their N1 patients had extracapsular spread. A similar cohort will be found to have multiple positive nodes. Therefore, it is clear that a significant percentage of patients who have neck dissection for N1 disease will require postoperative radiation anyway due to the presence of these prognostic features. Principles of Chemotherapy (Recurrent Disease) The median survival for patients with local or disseminated recurrent squamous cell carcinoma of the head and neck is 6 months; 20% of patients survive at 1 year. These statistics have not been affected by the use of chemotherapy. Consequently, patients with recurrent squamous cell carcinoma of the head and neck are candidates for Phase I and II trials of new drugs and new combinations of agents.

 

SINGLE-AGENT CHEMOTHERAPY TRIALS. The most active agents and their response rates are listed in Table 29.2-7 (120-143) and include methotrexate, bleomycin, cisplatin, 5-fluorouracil (5-FU), and paclitaxel. Methotrexate. Methotrexate is the standard palliative therapy for recurrent squamous cell carcinoma of the head and neck. The standard dose for initiation is 40 mg/m**2/wk to be escalated weekly by 10 mg/m**2 increments to 60 mg/m**2/wk or until dose-limiting toxicity or an objective response is reached. Therapy with this drug is relatively nontoxic, inexpensive, and convenient. Higher doses of methotrexate in single-arm studies were shown to produce higher response rates. [ref: 120-123] Five randomized trials have showo significant difference in survival rates between higher doses of methotrexate with leucovorin (as much as 5000 mg) and standard-dose methotrexate. [ref: 121-126] Bleomycin. Bleomycin has been studied extensively as a single agent and in combination in recurrent-metastatic squamous cell carcinoma of the head and neck. Response rates as a single agent vary from 6% to 45%, with a pooled average of 21%. [ref: 127,128] Because of its distinct toxicity (skin, mucous membranes, and lung but no significant myelosuppression), it has an advantage in combination chemotherapy. Bleomycin can be given in full dosage in combination with other agents with a different spectrum of toxicity. Continuous infusion of bleomycin produces less pulmonary toxicity than bolus injection. Cisplatin. Cisplatin is perhaps the most important chemotherapeutic agent in squamous cell carcinoma of the head and neck. Most of the studies have used a dose of 80 to 100 mg/m**2 every 3 to 4 weeks as standard dose. Response rates have ranged from 14% to 41%, with a pooled average of 28%. [ref: 127,128] Whether there is a dose-response relation is not yet proved. Single-agent cisplatin in doses of up to 200 mg/m**2 produced higher response rates in pilot trials, [ref: 129,130] but a randomized trial comparing 60 mg/m**2 doses with 120 mg/m**2 doses found no difference in response or survival. [ref: 131] 5-Fluorouracil (5-FU). 5-FU was studied initially in recurrent squamous cell carcinoma of the head and neck as second or third line chemotherapy to be used after other drugs failed. Response rates ranged from 0% to 33%, with an average of only 15%. [ref: 127,128] In these studies, 5-FU was usually given as an intravenous bolus daily for 5 days or weekly. The dose-limiting toxicity of this method of administration was myelosuppression. In the 1970s, 5-FU was studied as a prolonged infusion for 96 to 120 hours, usually in a dose of 1000 mg/m**2/day. The dose-limiting toxicity was found to be mucositis; myelosuppression was significantly lower. When 5-FU was administered as an infusion instead of a bolus, it was found to have increased activity in squamous cell carcinoma of the head and neck, synergistic interaction with cisplatin, and enhanced cytotoxicity with modulators such as leucovorin. Taxanes. The taxanes paclitaxel and docetaxel represent a new class of compounds that bind to the beta-subunit of tubulin, induce the formation of stable microtubule bundles, and inhibit microtubule depolymerization. In vitro, docetaxel is the more potent analogue. Docetaxel appears to be schedule independent, whereas paclitaxel appears to be more effective with prolonged exposure. [ref: 144-147] These two agents are undergoing intensive study in patients with head and neck cancer. Trials conducted in the United States and Europe consistently report response rates in the 30% to 40% range in patients with recurrent squamous cell carcinoma. [ref: 132-136] The doses used in published studies to date are: paclitaxel, 250 mg/m**2 as a 24-hour infusion every 3 weeks; docetaxel, 100 mg/m**2 intravenous bolus every 3 weeks. The dose-limiting toxicity for both drugs is myelosuppression, specifically neutropenia. Paclitaxel doses of 200 to 250 mg/m**2 are recommended only for patients with excellent performance status. At these doses a nadir absolute neutrophil count of less than 500 cells/microliter will occur in the majority of patients on day 8 after treatment. The use of colony-stimulating factors is recommended to shorten the duration of neutropenia and lower the risk of infection. [ref: 148] For patients with poor performance status or when treating without growth factor support, doses of 135 to 150 mg/m**2 by 24-hour infusion are recommended. It is not known whether the response rate or duration of response are improved by using higher dosage. It is known that at doses of 135 mg/m**2, adequate plasma concentrations of paclitaxel can be achieved to induce polymerization of microtubule. Studies are in progress evaluating other infusion schedules (1 hour, 3 hour, and 96 hour), but the relative efficacy compared to the 24-hour infusion schedule has not been established. Other toxicities associated with paclitaxel include sensory neuropathy, cardiac conduction disturbances causing bradycardia or arrhythmias, and anaphylaxis that requires premedication with corticosteroids. To date, docetaxel has been studied more in Europe and Asia than in the United States. In addition to myelosuppression, its toxicity profile includes peripheral neuropathy, fluid retention, asthenia, and skin toxicity. Trials using 100 mg/m**2 have been limited to patients with excellent performance status. Lower doses (60 to 75 mg/m**2) are much better tolerated and need to be further evaluated. Cisplatin Analogues. Carboplatin has significantly less renal, otologic, neurologic, and gastrointestinal toxicity than does cisplatin, but response rates are lower, in the range of 14% to 30%, with an average of 22%. [ref: 127,137] Carboplatin should be reserved for treatment of patients with peripheral neuropathy or renal dysfunction that prohibits use of cisplatin. Iproplatin trials have yielded much lower response rates and major toxicity in early phase II trials. [ref: 149,150] Methotrexate Analogues. The methotrexate analogues trimetrexate, [ref: 151] edatrexate, [ref: 152] and piritrexim [ref: 153-155] have all been tested in small numbers of patients and seem to be active but have no particular advantage over methotrexate. In a randomized comparison of methotrexate and edatrexate was more toxic. [ref: 152] Ifosfamide is another agent that deserves mention. At least five studies have been reported that document complete and partial response rates ranging from 6% to 43%, median 26%. [ref: 138-143] Response rates appear to correlate with extent of prior treatment; thus, the highest response rates were observed in chemotherapy-naive patients. Other Single Agents. Three other new drugs with activity that need further study are the pyrimidine antimetabolite gemcitabine, [ref: 156] the topoisomerase inhibitor topotecan, [ref: 157] and vinorelbine. [ref: 158] Drugs with an uncertain level of activity because they were studied in broad phase I to II trials before uniform response criteria were established include doxorubicin, cyclophosphamide, and hydroxyurea. Response rates of less than 10% have been reported for the plant alkyloids: vinblastine and etoposide in phase II trials. [ref: 159,160] Principles of Chemotherapy (Recurrent Disease) (Continued) (1 of 1)

 

COMBINATION CHEMOTHERAPY TRIALS. Combination chemotherapy regimens have been developed for treating recurrent squamous cell carcinoma of the head and neck in an effort to improve response rates and survival. Over the last two decades, numerous phase II trials of methotrexate or cisplatin-based regimens have been published. [ref: 127,128] Most contain small numbers of patients and often the results suggest greater efficacy than would be expected with single agent cisplatin or methotrexate. In the early 1980s, researchers at Wayne State reported a response rate of 70% and a CR rate of 27% using cisplatin (100 mg/m**2/day) and 5-FU (1000 mg/m**2/day) for 96 hours repeated every 3 weeks. [ref: 161] Twelve trials in patients with recurrent or newly diagnosed metastatic squamous cell carcinoma are shown in Table 29.2-8. [ref: 161-172] The average response rate was 50% for the 365 evaluable patients. Attempts to improve on the cisplatin plus 5-FU regimen include the addition of leucovorin, [ref: 173] continuous infusion bleomycin, [ref: 174,175] bleomycin and methotrexate, [ref: 176] interleukin-2, [ref: 177] and interferon. [ref: 178] These 3- and 4-drug combinations result in enhanced toxicity without indications of survival benefit.

 

RANDOMIZED TRIALS OF SINGLE AND COMBINATION CHEMOTHERAPY REGIMENS. Two trials directly compared the single agents methotrexate and cisplatin. [ref: 179,180] No differences in response rate or survival were observed. Randomized trials comparing combination regimens to either single agent methotrexate or cisplatin are shown in Table 29.2-9. [ref: 181-192] Survival benefit was reported in 3 trials. [ref: 184,187,189] Morton and colleagues [ref: 184] reported median survivals of 4.2 and 4 months for patients treated with cisplatin or cisplatin plus bleomycin, respectively, compared to a 2-month median survival for a no treatment control group. Campbell reported a significant improvement in survival of patients treated with cisplatin (8.7 months), compared to those treated with methotrexate (2.7 months). [ref: 187] The Liverpool Head and Neck Oncology Group randomized 200 patients to receive cisplatin alone, methotrexate alone, cisplatin and infusional 5-FU, or cisplatin and methotrexate. [ref: 189] Whereas there were no differences in response rate, survival was significantly better in the three cisplatin-containing arms. Three more recent large multicenter trials reported by Jacobs and colleagues, [ref: 190] Forastiere and colleagues, [ref: 191] and Clavel and colleagues [ref: 192] compared cisplatin and infusional 5-FU to the single agents cisplatin, 5-FU, or methotrexate. The results of the 3 trials were remarkably similar. The response rate to cisplatin plus 5-FU was 32% in 2 of the trials and 31% in the third; all studies demonstrated a significantly higher response rate for cisplatin plus 5-FU compared to the single agents. However, there were no differences in median survival rates for any of the treatment arms. Randomized trials comparing 2 combination regimens are shown in Table 29.2-10. [ref: 163,193-196] Kish and colleagues [ref: 163] compared cisplatin plus infusional 5-FU versus cisplatin plus bolus 5-FU. The response rate was significantly higher for infusional 5-FU, but median survivals were 6.7 and 5.0 months, respectively. Response rates were also significantly higher for methotrexate plus bleomycin plus vincristine plus cisplatin combination chemotherapy compared to methotrexate plus bleomycin plus vincristine reported by Clavel, [ref: 192] but survivals were similar. In an attempt to put into prospective the many trials that have been reported for palliation of patients with recurrent and metastatic squamous cell carcinoma of the head and neck, Browman and Cronin [ref: 197] performed a metaanalysis using randomized trials published between 1980 and 1992. Based on this pooled analysis of response and survival data, they concluded that single agent cisplatin is more efficacious than single agent methotrexate; the combination of cisplatin plus 5-FU is more efficacious than treatment with single-agents; and cisplatin plus 5-FU is more efficacious than other combinations. In summary, cisplatin-based combination chemotherapy is more effective than single agents. Response to cisplatin plus infusional 5-FU occurs in approximately 32%, with complete response in 5% to 15%. However, median survival is approximately 6 months, with 20% of patients alive at 1 year. This underscores the need for new therapies to treat this population. The advantage for choosing combination chemotherapy seems to be limited to patients with excellent performance status, no prior chemotherapy for treatment of recurrent disease, and minimal tumor burden. Durable complete responses and prolonged survival appear possible in this small subset of patients.

 

BIOLOGIC THERAPY FOR RECURRENT SQUAMOUS CELL HEAD AND NECK CANCER. Patients with head and neck cancer characteristically have moderate to severe depression of cellular and hormonal immunity, which may be reduced even further by surgery or radiation therapy. Early clinical trials adding nonspecific immunoadjuvant therapy (BCG or levamisole) were negative. Cytokines (interferon [FN] alpha and gamma interleukin-2 [IL-2], and combination IL-2 and IFN) have been used in squamous cell head and neck cancer with disappointing results. [ref: 198] Combinations of interferon alpha-2a and the single agents cisplatin and 5-FU have also been disappointing, with no improvement in either response rates or response duration and substantial increase in toxicity. [ref: 199] Several human squamous cell head and neck cell lines express high-affinity IL-2 receptors and are growth inhibited by IL-2 in vivo and in vitro. It has been shown in squamous cell head and neck cancer that tumor-infiltrating lymphocytes can be increased 30,000-fold by IL-2. Systemic IL-2 combined with IFN-gamma or IFN-alpha has antitumor activity but with substantial toxicity. [ref: 198,200] Retinoids have been studied in recurrent squamous cell head and neck cancer in two trials for a total of more than 30 heavily treated patients; in one, 13-cis-retinoic acid (13-cRA) produced a 15% response rate. [ref: 201] Research currently focuses on the best way to combine cytokines, retinoids, cytotoxic agents, and radiation therapy. A small series using combinations of the biologic agents IFN-gamma and IL-2 and another using IFN-gamma and retinoids have reported objective responses in advanced squamous cell head and neck cancer. [ref: 200-202] Squamous cell carcinoma of skin appears particularly responsive to the combination of 13-cRA and IFN-alpha. [ref: 203] Principles of Chemotherapy (Previously Untreated Disease)

 

NEOADJUVANT (INDUCTION) CHEMOTHERAPY. Chemotherapy may be given before or after ablative surgery or radiation therapy. The main advantage of induction chemotherapy (i.e., chemotherapy before surgery or radiation therapy) is the preservation of organ function. Induction chemotherapy eliminates the pharmacologic sanctuary problems (i.e., poor vascularity leading to poor concentrations of chemotherapy) after surgery or radiation therapy. Initial chemotherapy results in delivery of chemotherapy to the best possible host, resulting in increased compliance, responsiveness, and tolerance of higher doses. Downstaging of the primary and regional node disease has successfully allowed for organ preservation and a reduction in distant micrometastases. Nonrandomized Trials of Neoadjuvant Chemotherapy. Many neoadjuvant trials have been reported in which the single agents methotrexate, bleomycin, or cisplatin were used. [ref: 204-212] The CR rate with single agents was low (5%). In the late 1970s, a combination of cisplatin and bleomycin used by Randolph and associates [ref: 213] obtained a 71% response rate with a 19% CR rate. Many other investigators since then have used the regimen and have confirmed the high overall response rate, but reported lower CR rates. [ref: 214-218] To achieve higher response rates, investigators added methotrexate [ref: 219-226] or vinca alkaloids [ref: 227-233] to the cisplatin and bleomycin regimen. The CR rate improved from 7% with cisplatin and bleomycin to 16% with cisplatin, bleomycin, and methotrexate and to 20% with cisplatin and bleomycin and vinca alkaloid. The next major advance occurred in the early 1980s when Wayne State investigators reported induction trials with a cisplatin and infusional 5-FU regimen. [ref: 234,235] They achieved a response rate of 88% (CR, 19%) [ref: 234] with two courses and 93% (CR, 54%) with three courses of the same regimen. [ref: 235] Over the last decade and a half, hundreds of patients have received this regimen (cisplatin, 100 to 120 mg/m**2 day 1, and 5-FU, 1000 mg/m**2/day, infused over 96 to 120 hours) in clinical research trials. Response occurs in 85% of patients, on average, with complete response in approximately 40%. Two thirds of clinical complete responses are pathologically confirmed in biopsy or resection specimens. Response varies by site with the larynx and nasopharynx being most responsive and the oral cavity less responsive. Cisplatin and infusional 5-FU remains the most active regimen in previously untreated patients. [ref: 236,237] Trials with a modification of this regimen have been conducted in an effort to increase CR rates [ref: 238-246] (Table 29.2-11). Five trials of cisplatin, 5-FU, and leucovorin (PFL) have been reported. Overall and complete response rates range from 59% to 95% and 24% to 65%, respectively. The initial reports by Vokes and colleagues [ref: 238] using oral leucovorin and Dreyfuss and colleagues [ref: 239] using continuous infusion leucovorin appeared promising; however, long-term follow-up has not demonstrated any striking improvement in survival over cisplatin + 5-FU. Severe myelosuppression, diarrhea, and mucositis occur frequently, necessitating dose reductions in most patients. A few non-cisplatin-containing chemotherapy combinations have been tried as induction therapy, but CR rates with these remain low. Hill and colleagues [ref: 247] used a combination of vincristine, bleomycin, methotrexate, 5-FU, and hydrocortisone in 200 untreated patients with advanced disease. They described a 66% response rate after two cycles (CR not reported). The CR rate after local therapy was significantly greater in chemotherapy responders (78%) than in chemotherapy nonresponders (49%). Overall survival duration was 32 months for all patients, 37 months for chemotherapy responders, and 69 months for all patients achieving CR after all therapy. Survival was longer in patients with nasopharyngeal and laryngeal tumors. Toxicity was minimal with this regimen. Randomized Trials of Neoadjuvant Chemotherapy. Many randomized trials have been conducted to determine whether the addition of chemotherapy improves survival, and these are detailed in several reviews. [ref: 236,248-250] A listing of 17 randomized trials that utilized platinum-based chemotherapy regimens is shown in Table 29.2-12. [ref: 218,251-268] From the 1970s through the mid-1980s neoadjuvant trials consisted of a randomization between definitive local treatment (surgery with pre- or postoperative RT for operable patients or radiation for inoperable patients) and 1 to 4 cycles of neoadjuvant chemotherapy followed by definitive local treatment. Improved survival through a decrease in local-regional failure and distant metastatic rates was the primary goal. Studies designed in the latter half of the 1980s up to the present have focused on organ preservation specifically for primaries in the larynx and hypopharynx. [ref: 260,268] None of the trials shown in Table 29.2-12 has been able to demonstrate an overall survival advantage for patients receiving neoadjuvant chemotherapy. Two trials demonstrated a significant improvement in survival for chemotherapy treated patients on subset analysis only. [ref: 251,261] One trial showed statistically equivalent survival results, [ref: 268] and one preliminary report in abstract form only showed a significantly worse survival for the neoadjuvant chemotherapy group. [ref: 261] With regard to pattern of failure, five trials showed a decrease in the rate of distant metastases. [ref: 218,239-261,268] The one trial reported by Paccagnella and colleagues [ref: 261] that showed a significant increase in survival for the inoperable subset of patients reported a significant decrease in both local-regional failure and distant metastatic rates. This supports the theory that improvement in local-regional control is necessary for the survival of patients with head and neck cancer to be improved. Aside from this one study, neoadjuvant chemotherapy has had no impact on local-regional control of disease, despite the dramatic tumor reduction often observed prior to definitive local therapy. Its only indication is as an alternative to surgery for laryngeal preservation in patients with cancers of the larynx or hypopharynx. Nevertheless, important issues were resolved from these neoadjuvant trials including the following: 1. Induction chemotherapy results in significant tumor regression in 60% to 90% and complete regression in 20% to 50% of patients with locally advanced squamous cell carcinoma of the head and neck. 2. Patients who achieve a complete response have much better survival than do partial and nonresponders. 3. Response to chemotherapy continues through at least three courses of treatment. 4. Pathologic CR is documented by biopsy or surgery in 30% to 70% of clinical complete responders. 5. Chemotherapy responders demonstrate further response to radiation therapy and chemotherapy nonresponders do not. 6. Induction chemotherapy does not significantly increase surgical or radiation therapy complications. 7. TN stage and type of chemotherapy regimen are important prognostic factors for response to chemotherapy, whereas performance status, tumor site, TN stage, and response to chemotherapy are predictors for survival. 8. Although all sites of head and neck are lumped together in almost all the studies, there is evidence that biologic behavior differs with primary site. 9. A significant reduction in distant metastases results when chemotherapy is part of combined modality treatment of head and neck cancer. 10. No significant difference in overall survival has been demonstrated with the use of induction chemotherapy compared with surgery or radiation alone. 11. Organ preservation and improved quality of life can result from induction chemotherapy.

 

POSTOPERATIVE ADJUVANT TREATMENT Adjuvant Chemotherapy. Chemotherapy administered after a patient has been rendered disease free with surgery and/or radiation has been evaluated in three large multicenter randomized trials. [ref: 269-271] The rationale for posttreatment adjuvant chemotherapy rather thaeoadjuvant chemotherapy is three-fold. First, for resectable disease definitive surgery is not delayed. Second, wheeoadjuvant chemotherapy is successful, tumor margins are blurred and the extent of required surgery may be uncertain. Third, in pilot trials of neoadjuvant chemotherapy, up to 20% of patients refused surgery once response was achieved and their symptoms abated. The sequence of surgery, followed by adjuvant chemotherapy and then radiation, was a strategy developed to avoid the disadvantages of neoadjuvant chemotherapy. The Radiation Therapy Oncology Group tested this strategy using three cycles of cisplatin and infusional 5-FU adjuvant chemotherapy. [ref: 269] From 1985 to 1990, 499 patients with stage III and IV resected squamous cell carcinoma of the oral cavity, oropharynx, hypopharynx, and larynx were randomized through six cooperative groups. Patients were excluded if margins of resection were positive. Randomization was to immediate radiation or adjuvant chemotherapy, followed by radiation. Patients were stratified into high- and low-risk groups with additional RT (10 Gy) given to high-risk patients (close surgical margins less than 5 mm, carcinoma-in-situ at margins, or extracapsular nodal extension). The overall comparison of the two treatments showed no significant differences in survival, disease-free survival, or time to local-regional failure. However, there was a significant decrease in time to development of distant metastases for patients receiving adjuvant chemotherapy. The most important outcome of this trial was a difference that approached statistical significance showing improved survival and decreased local-regional failure for the high-risk subset of patients who received adjuvant chemotherapy compared to radiation alone. No benefit for adjuvant chemotherapy could be detected for the low-risk patients. Thus, this large intergroup trial identified a high-risk population, which is now being studied in postoperative adjuvant chemoradiotherapy trials. Two other large randomized trials performed by head and neck cancer study groups in Japan and France have reported preliminary results in abstract form only. [ref: 270,271] Japanese investigators used a combination of tegefur and uracil known as UFT as adjuvant chemotherapy. Four hundred twenty-four patients with locally advanced head and neck cancer were randomized to surgery alone or surgery followed by 1 year of adjuvant UFT. Compliance was greater than 75% for the 1 year of UFT, but no survival benefit could be detected. [ref: 271] French investigators limited eligibility for their adjuvant trial to patients with extracapsular nodal extension. Two hundred eighty-seven patients were randomized to receive postoperative radiation alone or to receive adjuvant cisplatin, bleomycin, and methotrexate after completion of RT. Chemotherapy-treated patients had significantly worse overall survival and an increased rate of distant metastases, but better local-regional control. [ref: 271] A number of randomized trials that utilize both neoadjuvant and adjuvant chemotherapy have been conducted in an attempt to further intensify therapy. [ref: 218,272-278] The Head and Neck Contracts Program, a three-arm trial, compared surgery and postoperative radiation to neoadjuvant cisplatin and bleomycin and to treatment with neoadjuvant chemotherapy plus adjuvant cisplatin for 6 months after surgery and radiation. [ref: 218] No differences in overall survival were observed, but a significant decrease in distant metastases was reported for the adjuvant group. Ervin and colleagues [ref: 278] treated 114 patients with neoadjuvant cisplatin, bleomycin, methotrexate, and leucovorin. After definitive local treatment, patients were randomized to observation or three cycles of adjuvant chemotherapy. The 3-year disease free survival rates were 55% and 88%, respectively; partial responders to neoadjuvant chemotherapy showed the greatest benefit. In summary, currently there is no role for adjuvant chemotherapy in low-risk patients (negative margins of resection, neck disease staged N0 or N1 without extracapsular extension). Patients with positive or close margins of resection, two or more involved regional nodes, or extracapsular extension of disease are at increased risk for both local-regional recurrence and distant metastases. Based on the results of the Head and Neck Intergroup trial, [ref: 269] adjuvant cisplatin and infusional 5-FU can significantly reduce the rate of developing distant metastases and may improve local-regional control and overall survival as well in this high-risk group. How to optimally sequence postoperative chemotherapy and radiation to achieve the highest compliance rate and greatest impact on survival outcome is as yet unclear. This may be achieved best with concomitant rather than sequential adjuvant chemotherapy and radiation. Adjuvant Chemoradiotherapy. Improvement in local-regional control through the use of concomitant chemotherapy and radiotherapy may be most feasible in the patient with locally advanced but “resectable” disease. Local-regional recurrence is the most frequent site of failure after surgery and postoperative radiation. Extracapsular extension of tumor in cervical node metastases is well established as an adverse prognostic factor for recurrence. [ref: 279-281] The rationale and potential role of chemotherapy to enhance radiation effects are well known and the clinical experience in head and neck cancer is extensive. [ref: 282-284] Until recently, this experience has been almost exclusively limited to patients with bulky unresectable disease. The RTOG conducted a feasibility pilot study treating 51 patients with resected stage IV disease and/or positive margins with postoperative RT and concurrent cisplatin 100 mg/m**2 every 3 weeks for three cycles. [ref: 285] Local-regional control was significantly better compared to matched historic controls. This pilot trial and the positive results of a randomized trial by Bachaud and colleagues [ref: 286] have led to a Head and Neck Intergroup Phase III trial now in progress. Resected patients with positive margins, metastases to multiple regional nodes, or the presence of extracapsular spread of disease are randomized to receive postoperative RT with or without concurrent cisplatin using the previously piloted dose and schedule. [ref: 285] Four randomized trials of postoperative chemoradiation have been published. [ref: 286-289] Bachaud and colleagues [ref: 286] randomized 88 patients with extracapsular spread of disease in regional neck node metastases to receive postoperative RT alone or postoperative RT plus cisplatin 50 mg intraveneously weekly. After a minimum follow-up of 5 years, the median survival (22 months versus 40 months) and 5-year survival rate (13% versus 36%) were significantly better for the chemoradiotherapy group. Radiation treated patients had a higher local-regional failure rate compared to the chemoradiotherapy group, whereas no differences were observed in the incidence of distant metastases. Randomized trials reported by Weissberg and colleagues [ref: 287] and Haffty and colleagues [ref: 288] evaluated mitomycin C and postoperative RT versus RT alone. Both investigators reported improvement in local-regional control but not survival. A third very small trial conducted by Weissler and colleagues found no benefit for cisplatin and 5-FU plus RT compared to RT alone. [ref: 289] These last three trials were not targeted to high-risk patients but included all resected patients with stage III and IV disease. All postoperative chemoradiotherapy studies reported increased acute toxicity, primarily mucositis, and weight loss with combined therapy. Careful monitoring of patients receiving combined chemoradiotherapy along with psychosocial and nutritional support is essential for these therapies to be successful and to have a high level of patient compliance. Postoperative adjuvant chemoradiotherapy for high-risk patients is promising. Completion of the current Head and Neck Intergroup Trial is crucial to confirm the improved survival reported by Bachaud and colleagues [ref: 286] and to change standard of care.

 

CONCURRENT CHEMORADIOTHERAPY Single Agents and Radiotherapy. The purpose of simultaneous chemoradiotherapy is to increase local-regional control and to prevent distant metastases. The possible synergistic effect has been explained by supposing that the drugs (e.g., bleomycin, cisplatin, and others) interfere with cell repair after sublethal or potentially lethal damage or with tumor cell synchronization. [ref: 282,283] Many clinical trials testing simultaneous chemoradiotherapy have been conducted since the 1960s (Table 29.2-13). [ref: 287,290-308] All the agents active in head and neck cancer have been combined with radiation therapy to enhance its effect and possibly prevent distant metastases. Methotrexate. Methotrexate has been used with concomitant radiation therapy based on its high single-agent activity in head and neck cancer. Two randomized trials have been published with opposing results. Condit [ref: 293] reported no improvement in the combined chemotherapy and radiation therapy group, whereas Gupta and colleagues [ref: 294] reported a significantly better primary control and survival, especially in oropharyngeal cancers. In the study by Gupta and colleagues, there was more cutaneous and mucosal toxicity in the chemotherapy and radiation therapy group, but no patients had their treatment interrupted or required nasogastric tube or intravenous feedings. [ref: 294] There was no increase in late toxicity. Bleomycin. Bleomycin has been combined with radiation therapy frequently in head and neck cancer. Many studies of bleomycin and radiation therapy report CR rates in the range of 38% to 79%. [ref: 282] There are at least nine randomized trials [ref: 295-303] comparing bleomycin and radiation therapy with radiation therapy alone. Three of the trials showed a benefit in response rate or survival or both. [ref: 295,296,302] However, the other five trials, including the large European Organization for Research on Treatment of Cancer (EORTC) randomized trial, did not reveal any significant benefit in the bleomycin and radiation therapy arm. [ref: 303] Most of the studies did report increased acute toxicity, especially mucositis and skin reaction. 5-Fluorouracil. Many studies have used 5-FU with concurrent radiation therapy. Three randomized trials have been reported. [ref: 304-306] In a study from Japan, intraarterial 5-FU was used with radiation therapy in maxillary sinus carcinoma. [ref: 304] Disease-free survival was better, but overall survival was similar in both groups. In the study by Lo and associates, [ref: 305] 5-year survival was significantly better only in the oral cavity group. 5-FU did increase the acute and late tissue toxicity of radiation therapy. Browman and colleagues [ref: 306] evaluated infusional 5-FU and radiotherapy versus RT alone. The complete response rate was higher with combined treatment, but there was no difference in overall survival. Other Agents. Hydroxyurea has been used concurrently as a radiosensitizer without benefit. [ref: 290-292] One randomized trial from Yale used mitomycin-C with radiation therapy and showed improved local control [ref: 287] but not a significant difference in survival. A high incidence of pulmonary complications was reported in the experimental arm. The new agents paclitaxel, topotecan, and gemitabine, which have antitumor activity against squamous cell carcinoma of the head and neck, all have radiation enhancing properties. [ref: 308-312] Phase I trials are in progress to assess dose-limiting toxicities using various schedules of these drugs combined with radiation. Cisplatin and Its Combinations. Recently, cisplatin has been combined with RT because mucositis is not a primary toxicity, and experimental data support its effectiveness as a radiosensitizer. [ref: 284] As a single agent, cisplatin has been administered using two schedules in head and neck cancer: weekly low doses (20 mg/m**2) or intermittent high doses (100 mg/m**2 every 3 weeks) concomitant with RT. Preliminary results of intergroup randomized trials comparing low-dose weekly cisplatin (20 mg/m**2) during radiation with conventional radiation were reported by Haselow. [ref: 312a] The overall response rate was significantly higher for the cisplatin-treated patients (73% versus 59%; P = .007), but there was no difference in complete response rate (34% versus 30%) or survival. The lack of benefit may be attributable to the low total dose of cisplatin received (120 to 140 mg/m**2 over the 6 to 8 weeks of RT). The Radiation Therapy Oncology Group (RTOG) evaluated the intermittent high-dose cisplatin regimen (100 mg/m**2 every 3 weeks for 3 doses) during RT in 124 patients. [ref: 313] The complete response rate was 71% and estimated 4-year survival 34%. Compared to a historic control database, local-regional control and survival rates appeared superior to treatment with RT alone. This treatment regimen is currently being formally compared to RT alone in a Head and Neck Intergroup trial for patients with locally advanced, unresectable disease. [ref: 284] Al-Sarraf [ref: 307] reported the preliminary findings of an intergroup trial for patients with stages III and IV cancer of the nasopharynx. Patients were randomized to receive standard radiotherapy or cisplatin (100 mg/m**2 on days 1, 22, and 43) during radiation, followed by three cycles of adjuvant cisplatin and 5-FU. The trial was terminated early after an interim analysis showed a significant improvement in 2-year survival (80% versus 55%) with combined treatment. Local and distant failure rates were also significantly reduced with combined treatment. These results cannot be generalized to other sites in the head and neck and the contribution of each component (concurrent chemoradiotherapy therapy and adjuvant chemotherapy) to the improvement in survival cannot be determined. Concurrent Chemoradiotherapy (Continued) (1 of 1) Combination Chemotherapy and Radiotherapy. Administering multiple cytotoxic drugs during radiation substantially increases toxicity and ofteecessitates frequent interruptions in radiotherapy. Thus, investigators have developed regimens that use split course RT providing planned breaks in therapy, or regimens that alternate chemotherapy and radiation to minimize normal tissue toxicity. The mode of combining the two therapies is an important issue. For head and neck cancer, it has been demonstrated that protracted radiation therapy as single modality treatment results in decreased local control rates. [ref: 314,315] This is thought to be due to accelerated repopulation of tumor cells surviving the initial treatment. The failure of induction chemotherapy to show any survival benefit in randomized trials may have a similar cause. If radiation therapy is considered a noncrossresistant tumor-killing agent, then alternating it with chemotherapy might circumvent the problem of heterogenous tumor cell repopulation and primary drug resistance. This hybrid of chemotherapy alternating with radiation therapy would reduce normal tissue toxicity compared with the simultaneous approach. Many concomitant chemotherapy and radiation pilot trials have been reported. [ref: 236,316] Some with the longest follow-up that utilize cisplatin-based combination chemotherapy have reported promising survival and response data but also severe mucosal toxicity. [ref: 317-321] The advantage of multiagent chemotherapy is that in addition to improved local-regional control, distant metastases may also be decreased. Taylor and colleagues [ref: 317] used a regimen of concurrent radiation and cisplatin/5-FU chemotherapy administered every other week. Severe mucositis was reported in one half of the 53 unresectable patients. Complete response was achieved in 55% and median survival was 37 months. Adelstein and colleagues [ref: 318] conducted two pilot trials of cisplatin and 5-day induction 5-FU every 3 weeks for 4 courses given simultaneous with split course radiation. Between the second and third courses of chemotherapy there was a break in treatment during which optional surgery was performed in responding patients. A 77% complete response rate was reported and an actuarial 4-year survival of 49%. [ref: 318] Severe mucositis occurred in one half the patients. This regimen is currently being compared to RT alone in a Head and Neck Intergroup trial for locally advanced, unresectable disease. [ref: 284] Wendt and colleagues [ref: 319] reported on an aggressive regimen consisting of split course hyperfractionation and simultaneous cisplatin, 5-FU, and leucovorin. Eighty-one percent of the 59 patients studied achieved a CR; actuarial survival rate was 52%. This regimen was subsequently tested in a randomized comparison to RT alone, but results have not been published as yet. Haraf and colleagues [ref: 320] have investigated a regimen of 5-FU and hydroxyurea with radiation administered every other week. In an advanced group of untreated patients, the complete response rate was 71% and median survival 14 months. To increase complete response rates and survival, this group has used sequential cisplatin, 5-FU, leucovorin, and IFN induction chemotherapy, followed by organ preservation surgery and then 5-FU, hydroxyurea, and simultaneous radiation. [ref: 321] Survival appears to be better than achieved with prior regimens, but severe toxicity occurred in 70%. These intensive and toxic regimens are not recommended for use outside of an investigational treatment program in which intensive support systems can be provided. Six randomized trials of combination chemotherapy and alternating or concurrent radiation have been published (Table 29.2-14). [ref: 322-328] Two trials compare the combined treatment to standard fractionation radiotherapy [ref: 322,323]; one reported positive results. Merlano and colleagues [ref: 323] compared alternating courses of cisplatin/5-FU and radiotherapy to radiotherapy alone in stage III and IV patients with unresectable disease. After a minimum follow-up of 4 years, 5-year survival rates were 10% for RT and 24% for combined treatment (P < .02). Progression-free survival and time to local-regional relapse were also superior with the alternating regimen. Mucosal toxicity was similar for both treatment groups. Four randomized trials [ref: 324-328] have addressed the question of treatment strategy: sequential (neoadjuvant) chemotherapy and radiation versus concurrent or alternating chemotherapy and radiotherapy. The trial by Merlano and colleagues [ref: 327] showed a significantly increased survival for patients randomized to vinblastine, bleomycin, and methotrexate alternating with radiotherapy compared to sequential treatment. The SECOG [ref: 328] evaluated vincristine, bleomycin, methotrexate with or without 5-FU, and concomitant or sequential RT in a 2 x 2 trial design. Overall survival was not different, but on subset analysis, disease-free survival was improved for patients with larynx primaries receiving the concomitant treatment. Taylor and colleagues [ref: 324,325] and Adelstein and colleagues [ref: 326] each compared their cisplatin/5-FU and concomitant radiotherapy regimen to sequential treatment. Adelstein and colleagues reported a significant improvement in disease-free survival but not overall survival with concomitant treatment. [ref: 326] Taylor and colleagues observed a significant improvement in local-regional control on subset analysis for patients with T(3-4) N0 and T1-2, N2 disease receiving concomitant treatment but no differences in overall survival or disease-free survival. [ref: 324] In summary, patients with locally advanced, unresectable disease continue to challenge physicians. Randomized trials show positive results for the strategy of alternating chemotherapy and radiotherapy compared to either radiotherapy alone or sequential (neoadjuvant) chemotherapy followed by radiotherapy. [ref: 323,327] In addition, the mucosal toxicity associated with this strategy appears acceptable and similar to that occurring with daily standard fractionation radiation. More intensive regimens and protracted treatment schedules have not as yet demonstrated survival benefit although randomized trials are in progress. [ref: 284]

 

ORAL CAVITY EPIDEMIOLOGY Oral cavity cancer represents a multiplicity of diseases. Epidemiology as it relates to each of these disease processes likewise differs. However, given that squamous cell carcinoma represents the preponderance of cancers that occur in this region, greater attention will be focused on its etiologic determinants. It is estimated that 30,000 new cases of oral cavity cancer will have occurred in 1991. [ref: 508,509] The relationship between tobacco exposure and disease development has been clearly demonstrated. [ref: 510,511] A clear dose-response relationship has been identified with a greater risk being directly proportional to intensity and duration of exposure. Alcohol has been identified as a coagent, most probably through a topical effect. [ref: 512] The mucosal areas, which are exposed to prolonged contact with alcohol, are at greatest risk of cancer development. Readers are referred to reviews on mechanisms of tobacco-induced carcinogenesis for in-depth understanding. [ref: 513] Likewise, reviews regarding the role of alcohol in cancer development are available. [ref: 512,514] Cigarette smoking cannot be considered the sole etiologic agent for oral cavity cancer. This fact is made evident by the observation that over 50 million individuals in the United States consume cigarettes. The percentage of the total population that is using tobacco in its various forms is even greater. As mentioned, however, only 30,000 individuals develop oral cavity cancer annually. Other factors must therefore be considered. Arguably, it is genetic susceptibility that may be the most significant. Genetic factors associated with increased risk include mutagen sensitivity, which is potentially reflective of an underlying DNA repair deficiency. [ref: 515] Syndromes that are characterized by mutagen sensitivity, including xeroderma pigmentosum, Fanconi’s anemia, and ataxia telangiectasia, have all been associated with oral cavity cancers. [ref: 516] Other relevant genetic markers may include inducibility of cytochrome P450 enzyme system. [ref: 517] Additional risk factors for oral cavity cancer include diet. [ref: 518,519] Patients with vitamin A deficiency have been considered at high risk of malignant transformation of oral mucosa. High dietary consumption of fruits and vegetables have been found to provide a protective effect. Chronic irritants have been considered an etiologic factor, including mouth wash, poor dental hygiene, and syphilis. [ref: 520,521] Recent reports have incriminated marijuana smoking as a contributing factor to oral cavity cancer. [ref: 522] Recent studies have focused on the viral etiology of cancers within the upper aerodigestive tract. Herpes simplex virus type 1 (HSV-1) has long been considered an etiologic agent. The inability to identify HSV-1-related proteins within oral cavity cancers has, however, raised questions as to the significance of this virus. [ref: 523] Recent investigations have identified human papillomavirus within head and neck cancers, specifically types 2, 11, and 16. [ref: 524-527] Papilloma transcriptional factors when inserted within human DNA can alter normal gene replicative control mechanisms.

 

ANATOMY The anatomy of the oral cavity will be covered with each specific site.

STAGING T staging for oral cavity cancer applies to all subsites within the oral cavity unless otherwise stated. For the oral cavity, the definition of T staging is as follows:

PRIMARY TUMOR (T) TX: Primary tumor cannot be assessed T0: No existence of primary tumor Tis: Carcinoma in situ T1: Tumor 2 cm or less in greatest dimension T2: Tumor more than 2 cm but not more than 4 cm in greatest dimension T3: Tumor more than 4 cm in greatest dimension T4(lip): Tumor invades adjacent structures (e.g., through cortical bone, tongue, skin of neck) T4(oral cavity): Tumor invades adjacent structures (e.g., through cortical bone, into deep (extrinsic) muscle of tongue, maxillary sinus, skin) The regional lymph node (N) and distant metastases (M) staging are identical to other sites within the upper aerodigestive tract and are as stated in the beginning of this chapter.

 

PATHOLOGY The most common cancer within the oral cavity is squamous cell carcinoma. Additionally, cancers can arise from minor salivary glands; these latter cancers include adenoid cystic carcinoma, mucoepidermoid carcinoma, and adenocarcinoma. Rare soft tissue neoplasms include mucosal melanoma, plasmacytoma, and soft tissue sarcomas. Also found within the oral cavity are cancers arising from bone, including osteosarcomas. There also exist neoplastic lesions that are not truly malignant disorders of bone growth, such as ameloblastoma. These lesions, however, have a propensity for local expansion and destruction. The principles of sound oncologic surgery and radiation thus apply to these latter processes.

 

NATURAL HISTORY Earliest changes associated with squamous cell carcinoma are associated with erythema and slight mucosal surface irregularities. Many times a punctate lesion will also be identified. As disease progresses, several growth patterns emerge that can be typically characterized as exophytic or infiltrative. The latter is more characteristically associated with destruction of surrounding anatomical structures. The exophytic lesions have a less aggressive growth pattern. It should be realized that both patterns are capable of producing metastasis and disease progression. Therapy should be planned accordingly. Characteristics of the disease will be reflected in certain histopathologic criteria. When considering tumor differentiation, it has been reported that poorly-differentiated disease will have a greater propensity for metastasis than well-differentiated disease. This has, however, not been universally accepted. Jacobsson’s criteria as outlined in Table 29.2-2 will likewise reflect the natural history of the disease. More specific information regarding natural history will be covered in each anatomical subsite. LIP Epidemiology Carcinoma of the lip is second only to skin cancer as a site of neoplasia within the head and neck region. It is noted to occur in approximately 3600 cases per year (i.e., 1.8 persons per 100,000 population annually). [ref: 508] The majority of these lesions occur on the lower lip and 95% occur in males. A principal etiologic factor, similar to other upper aerodigestive cancer, has been the use of tobacco, including both pipes and cigars. [ref: 528] Sun exposure has also been incriminated and may represent the most significant factor. The latter fact is of potential relevance given the increase incidence of other skin cancers as well as lip cancer. Patients genetically susceptible to skin cancers following sun exposure, (i.e., patients with xeroderma pigmentosum) are likewise susceptible to lip cancer. [ref: 516] Such an observation emphasizes ultraviolet radiation as an etiologic agent. Disease has likewise beeoted in renal and homograft recipients, implicating immune suppression as a determinant. [ref: 529] Anatomy The lip is composed of the orbicularis oris muscle and is delineated by the junction of the vermillion border with the skin. Blood supply and sensory nerve supply are by means of the labial artery (a branch of the facial artery) and by cranial nerve V, respectively. The primary lymph node drainage is to levels I and II. Pathology The principal cancer involving the lip is squamous cell carcinoma. Other lesions include basal cell carcinoma. Rarely, minor salivary gland cancers can occur. Natural History Patients most frequently present with either an exophytic or ulcerative lesion of the lower lip. Occasionally these lesions are associated with bleeding and pain. The latter symptom, however, is a late feature of the disease. These lesions are typically slow growing lesions. With progression there may be associated numbness of the skin of the chin secondary to involvement of the mental nerve, a branch of the third division of cranial nerve V. Furthermore, progression of disease along the mental nerve may extend into the mental foramen of the mandible. Such involvement leads to enlargement of the foramen with bone destruction and widening of the inferior alveolar canal. A Panorex examination of the mandible is recommended as part of each diagnostic evaluation. Lymphatic spread occurs relatively infrequently in lip cancer; approximately 5% to 10% of patients will develop evidence of nodal involvement. [ref: 530-532] Lymph node spread is typically to submandibular nodes or submental nodes. Lesions in the midline may spread bilaterally. The incidence of metastases has been related to histologic grading, with high grade lesions being at greatest risk. The upper lip tends to metastasize earlier than the lower lip. Upper lesions will metastasize to periparotid nodes (i.e., preauricular nodes), in addition to submandibular nodes. The prognosis from lip cancer principally depends on the size of the primary tumor. [ref: 533-535] T1 lip cancers have a 5-year survival of 90%. T2 survival is 84%. With evidence of lymph node metastases, survival will decrease to 50%. Perineural invasion represents a bad prognostic sign. [ref: 536] Likewise, prognosis appears worse in younger adults. [ref: 537] Tumors have a greater tendency to metastatic spread in these latter patients. Treatment

EARLY DISEASE. Surgery and radiotherapy are the mainstay of therapy. Dysplasias and carcinoma in situ can be handled by lip shave (i.e., vermilionectomy with advancement of a mucosal flap). Those lesions that involve less than 30% of the lip can be resected with a V excision and primary closure of resulting defects. For larger lesions, transposition flaps are required for reconstruction. Undoubtedly, the challenge in the surgical management of lip cancer resides in the best means of reconstruction.

 

Oral competence remains the primary goal. Those lesions that require resection of 30% to 50% of the lip can be best handled with a transposition flap drawn from the uninvolved opposing lip. This reconstruction technique is termed Abbe-Estlander. [ref: 538-540] A detailed description of the Abbe-Estlander flap has been provided elsewhere. [ref: 538] When the near total lip is involved (i.e., 50% to 75%), the Karapandzic advancement flap can be utilized. [ref: 541] This has the benefit of providing a competent oral sphincter with an associated neurovascular integrity. The problem with the Karapandzic reconstruction is that the reconstructed lip is tight and significantly foreshortened. Other methods have been devised. [ref: 542-544] These vary from simple nasolabial flaps based inferiorly or superiorly to more formal fan-type flaps such as the Gillies flap and Webster cheek advancement flap. [ref: 544] Most T1-3 squamous cell carcinomas of the lip can be managed by either radiation therapy or surgery. The choice of radiation or surgery may depend on the size and location of disease. If the lesion is quite small and can be easily excised without functional sequelae, surgery would be the chosen treatment. Lesions involving commissures can be irradiated, without the functional sequelae of surgery. However, involvement of the commissure under such circumstances is rare. Brachytherapy alone can be used for early T1 and small T2 lesions. Temporary implantation with iridium-192 is the treatment of choice. Doses in the 6000 cGy range are usually adequate, with the dose rate being in the 40 to 60 cGy per hour range. Figures 29.2-10a, 29.2-10b, and 29.2-10c show a clinical example of the brachytherapy procedure. Similarly, external beam can be used either alone or with an implant for T3 lesions. Given the infrequency with which early cancers spread to regional lymph nodes, elective treatment of the neck is not necessarily required.

 

ADVANCED DISEASE. Stages III and IV lip disease are optimally managed with combined surgery and postoperative radiation therapy. Reconstructive options are as described above. Doses in the 6000 to 6300 cGy range, delivered at 180 to 200 cGy per fractions over 6 to 7 weeks, is preferred. If the patient has lymph node metastases in the neck, a neck dissection would be done along with the resection of the primary site. The postoperative radiation therapy would then be delivered to both the neck and the primary site. Even if the patient were N0, one should still utilize elective radiation therapy or elective node dissection as part of the management, given the increased risk of microscopic lymph node metastases in these patients. For patients with T(1-3) disease who have had an operation, sometimes the radiation oncologist is faced with a positive margin of resection. This can either be managed with brachytherapy alone, or localized superficial external beam irradiation, with similar doses and techniques as when radiation therapy alone is used. Results of Treatment The radiation therapy results are similar to the reported results of surgical management. [ref: 545] Heller and Shah [ref: 533] reported approximately 90% local control for T1, T2, and T3 lesions treated with surgery alone. A significant problem in surgical management is local recurrence, which may approach 40% for T3 and T4 lesions. [ref: 531] Fitzpatrick [ref: 546] has reported the Princess Margaret Hospital experience, which also documents that the results of radiation therapy and surgery are basically equivalent. Jorgensen and colleagues [ref: 547] have reported a series of 869 lip cancers. In 766 patients, treatment was performed was entirely with brachytherapy. The remaining cases had external beam radiation, mainly with orthovoltage beams. The majority of patients had either T1 or T2 lesions, with only 75 having T3 lesions. Local control was in the 90% range. External beam radiation can also be used. Petrovich and colleagues [ref: 548] reported on 250 patients, most receiving 5100 cGy in 3 weeks with daily fraction sizes of 300 cGy. There were a total of 896 patients in the study, most of whom were treated with ortho-voltage beams. Local control was obtained in 94% of T1 and T2 lesions, and in 90% of T3. For T4 disease, 47% of the patients obtained local control with radiation alone, pointing to the need for combined modality treatment for this subset.

 

FLOOR OF MOUTH Epidemiology The annual incidence of cancers of the floor of mouth is 0.6 cases per 100,000 in the United States. [ref: 508] It occurs in males approximately three times as frequently as females. Recent reports, however, have demonstrated an increasing incidence of the disease among women. [ref: 560] The median age of individuals developing squamous cell carcinoma is approximately 60 years. Anatomy The floor of mouth is delineated by the free margin of the mucosa because it extends from the junction of the mobile tongue to the alveolar process. This margin extends from one anterior tonsillar pillar to the other. Within the floor of mouth anteriorly are the openings of bilaterally located submandibular salivary glands, called Wharton’s ducts. These ductal openings are significant because anterior lesions of the floor of mouth can frequently obstruct associated salivary flow, leading to tenderness and enlargement of the respective submandibular gland. Also within the floor of mouth are minor salivary glands and the sublingual glands. Distinct from mucosa of the tongue, mucosa of the floor of mouth is nonkeratinizing stratified squamous epithelium under nonpathologic situations. Musculature constituting the floor of mouth includes the genioglossus, geniohyoid, and mylohyoid muscles. Blood and nerve supply are principally from the paired lingual arteries and lingual nerves, respectively. Pathology Cancers of the floor of mouth account for approximately 10% to 15% of all oral cavity cancers. Squamous cell carcinoma constitutes the majority of lesions within the floor of mouth, with the majority of these lesions being moderate to well differentiated. There are several variants of squamous cell carcinoma of the floor of mouth. These include verrucous and sarcomatoid squamous cell carcinoma. Cancers derived from salivary gland tissue also are encountered including mucoepidermoid carcinomas, adenocarcinomas, and adenoid cystic carcinomas. Natural History Squamous cell carcinomas of the floor of mouth typically present as infiltrative lesions that are characteristically painful. These lesions may extend anteriorly to invade bone, deeply to infiltrate muscles of the floor of mouth, or posteriorly to invade tongue. On occasions it is an enlarged lymph node in the neck, which is the presenting symptom. Floor of mouth cancers can grow to massive size without metastasizing to cervical lymph nodes. However, approximately 12% of T1 lesions are associated with occult metastatic disease depending on thickness of lesion. [ref: 97] Metastatic rates to cervical lymph nodes occur in 30%, 47%, and 53% of T2, T3, and T4 cancer, respectively. [ref: 97] Lymph nodes in the submandibular and submental triangles represent the first echelon of metastatic sites. Distant metastasis is infrequently observed in patients who present with previously untreated disease. Prognosis is influenced principally by disease stage and presence or absence of histopathologically confirmed regional lymph node metastases. The overall 5-year survival for stage I disease approximates 85% to 90%. [ref: 561,562] For stage II, III, and IV disease, 80%, 66%, and 32% of patients, respectively, are alive at 5 years. Other factors considered to reflect a worse prognosis include evidence of perineural invasion, depth of primary tumor invasion, and poor tumor differentiation. [ref: 563-565] The latter factor, however, has not been universally accepted as having prognostic significance. Treatment

EARLY DISEASE. Treatment of floor of mouth cancers has been principally surgical resection but may either be surgery or radiation alone. As is true for cancers of the alveolar ridge, superficial involvement of the mandible can be handled by marginal mandibulectomy (Fig. 29.2-12). When radiation is used for early disease, it has been shown that results are improved when at least a portion of the treatment is delivered by an interstitial implant. [ref: 566,567] Interstitial implant alone can also be used. [ref: 568] Lesions that abut or are tethered to the periosteum of the mandible are not good candidates for primary radiotherapy. Implants against the mandible can lead to osteonecrosis. The treatment of the neck for early cancer of the floor of mouth is controversial. Most would advocate elective neck treatment for clinically N0 disease. The value of this approach as compared to observation with neck dissection and radiation therapy being performed for clinically developing disease remains unproven. Some have advocated performing neck dissection dependent on the thickness of the primary lesion (i.e., more than 4 mm thickness).

 

ADVANCED DISEASE. For advanced lesions, combined therapy of surgery and radiation is the treatment of choice. Surgical resection generally entails partial glossectomy and segmental mandibulectomy. Identification of the inferior alveolar nerve and frozen section histopathologic assessment should be performed during the operation.

This is to ensure that disease has not extended beyond surgical margins by perineural spread. Resection for most advanced lesions will require removal of the entire thickness of the floor of mouth. New reconstructive techniques have greatly facilitated rehabilitation following surgical excision of advanced tumors. Techniques include myocutaneous flaps as well as osteomyocutaneous free flaps with microvessel anastomoses. [ref: 569] Elective and/or therapeutic neck dissections are considered necessary in each case. Bilateral neck dissections are indicated for those lesions that approach or cross the midline. Postoperative radiation entails doses in the range of 6000 to 6300 cGy at the primary site. In instances of positive surgical margins, our policy is to treat the area of positive margins to 6300 cGy. Patients are treated with opposed lateral fields for the primary site and upper neck, which junctions to a low neck field at approximately the thyroid notch. A midline block is used for the low neck field to protect the larynx and spinal cord. The dose to the low neck is usually 5000 cGy given in 5 weeks. The upper neck and primary site are generally treated to 4500 cGy in 5 weeks, after which a spinal cord block is placed. The primary site and upper neck are generally taken to 5400 cGy, after which a boost to the primary site and the involved region of the neck is taken to approximately 6300 cGy. Electron fields are used posterior to the spinal cord block, as needed, to bring the posterior neck to the appropriate dose. Results of Treatment Local recurrences after surgical resection of T1 and T2 floor of mouth cancers are noted io less than 10% of the patient population. [ref: 561,562,570] As tumors increase in size, the pattern for failure becomes predominately a regional problem. Nearly 40% of failures will be solely within regional cervical lymph nodes. [ref: 561] Mazeron and colleagues [ref: 568] have reported a large recent radiotherapy series. The majority of patients were treated to 65 Gy with Iridium-192 brachytherapy alone. Local control was 94% for T1, N0, and 74% for T2, N0 lesions and was dependent on size of lesion as well as the presence or absence of gingival extension. Wang and colleagues [ref: 571] have reported excellent results with the use of intraoral cone electron boost and no brachytherapy. The daily dose of radiation via the cone is frequently greater than the conventional 180 to 200 cGy range. Local control was obtained in all 13 patients with T1 lesions, and in 19 of 20 (95%) with T2 lesions. Fu and colleagues [ref: 572] have published an extensive radiation therapy experience with floor of mouth cancer. When implant was either the only treatment or a part of the treatment, local failure occurred in only 2% (1 of 39 patients) with T1 lesions, 7% (4 of 54 patients) with T2 lesions, and 14% (5 of 35 patients) with T3 lesions. The use of primary radiation may be associated with improved functional outcome as compared to surgery, but this requires more investigation. [ref: 573] When examining results of treatment for advanced disease, a retrospective review of the results at the Memorial Sloan-Kettering Cancer Center [ref: 104] highlights the fact that local control is improved with combined surgery and radiotherapy as compared to surgery alone, for patients with stage III and IV head and neck lesions. In a prospective randomized trial, Kramer and colleagues [ref: 574] compared preoperative radiation (5000 cGy) plus surgery, surgery plus postoperative radiation (6000 cGy), and radiation therapy alone (6500 to 7000 cGy) with surgical salvage for patients with certain stage II, and all stage III and IV squamous cancers of the oral cavity and oropharynx. There was no difference in the 4-year actuarial survival, or locoregional control, regardless of treatment type. However, this study did not stratify the results by anatomic subsite within the oral cavity and oropharynx. A recent report from Memorial Sloan-Kettering Cancer Center [ref: 104] highlights the importance of analyzing treatment results by anatomic subsite in patients with advanced disease. With a median follow up of 6 years, the 5-year actuarial local control was 74%. However, the local failure rate was 38% versus 11% for oral tongue (29 patients) versus floor of mouth (22 patients); the difference was statistically significant. TONGUE Epidemiology Tongue cancer is estimated to occur in 6,200 individuals per year in the United States. [ref: 508] Excluding lip, it exceeds all other sites in the oral cavity. The median age for individuals with tongue cancer is approximately 60 years. The male:female ratio is similar to other disease sites, approximately 3:1. It is of interest that an increase in tongue cancer has been reported among young adult males. [ref: 575,576] Some have suggested marijuana use as a contributing factor in this latter population. [ref: 527] Anatomy The oral tongue represents the mobile portion of the tongue musculature that extends from the line demarcated by the circumvallate papilla posteriorly to the junction of the floor of mouth anteriorly. It is composed of the genioglossus, hyoglossus, styloglossus, and palatoglossus muscles. All of these muscles are innervated by the hypoglossal nerves. Taste and sensation within the tongue are provided by the lingual nerve, a branch of the third division of the cranial nerve five. Blood supplies principally from the external carotid artery through the paired lingual arteries. Lymphatic drainage is principally to levels II, III, and I in decreasing order. Pathology The primary cancer of the tongue is squamous cell carcinoma. Other cancers are frequent and include minor salivary gland cancers such as adenoid cystic carcinoma and adenocarcinoma. Myeloblastoma represents a rare tumor of the tongue. Natural History These cancers can grow in both an exophytic and an infiltrative fashion. The primary presenting symptom is pain, although many of these lesions can be painless. Difficulty in speech and deglutition is occasionally elicited. It tends to be more rapid in its onset than other cancers within the oral cavity. There may be a history of long-standing leukoplakia prior to the development of symptoms, especially in younger females. As compared to other cancers within the oral cavity, tongue cancers have a greater propensity for lymph node metastases. This occurs in ranges from 15% to 75% depending on the extent of primary disease. [ref: 539,577,578] Lymph nodes most frequently involved lie within level II (i.e., jugulodigastric nodes). Nodes within levels I, III, and IV are involved in decreasing order. It is of note, however, that all nodes can be involved. The incidence of bilateral nodal metastases occur up to 25% of cases. Contralateral nodal metastases are present in 3% of cancers. Prognosis is principally reflected in extent of nodal metastases ranging from 75% in early stage node negative disease to 30% in those with advanced lesions. Other factors portending more aggressive disease include perineural and vascular invasion and infiltrative versus pushing borders. [ref: 579,580] Depth of invasion has likewise been considered significant. Whether tongue cancer in young adults represents a worse disease has not been demonstrated. [ref: 581] Treatment

EARLY DISEASE. It is generally considered that disease control rates for early disease when using either surgery or radiation therapy are equivalent depending on treatment bias. Early stage I and II lesions can usually be removed intraorally. Excision usually entails an hemiglossectomy. Special attention to surgical margins should be exercised, since disease may spread along muscle bundles beyond that expected by clinical assessment. Most T1 lesions can be managed with brachytherapy alone. This generally consists of an iridium-192 implant. Although needles are still used by some groups, this is really an old technique that should be avoided. It introduces unnecessary exposure to the physicians, nurses, and other staff, and it does not allow the physician to optimize the dose distribution of the brachytherapy procedure. Iridium-192 is inserted via after loading catheters. The catheters themselves are placed in the operating room under general anesthesia. The iridium-192 is loaded 1 to 2 days postoperatively. Localization films are taken, and computerized dosimetry is performed. The usual dose rate is in the 40 to 60 cGy per hour range, and the usual total dose is 6000 to 7000 cGy. The patient wears a tongue prosthesis during the dwell time of the implant to protect the hard and soft palates as much as possible. Because the lesion increases in size when using radiation as primary therapy, it is preferred to combine external beam irradiation with implant. First, the external beam can be used as elective neck irradiation simultaneous with irradiation to the tongue. The implant then serves as the boost to the tongue. Second, the external beam allows a wider margin of tongue to be treated than does the implant. In these situations, it is typical to treat the primary site and the neck to doses in the 5000 cGy range, followed by a 2000- to 3000-cGy implant boost to the tongue. For N0 patients, this treatment program manages both the primary site and the neck. For those patients with palpable neck nodes, a neck dissection can be performed at the same anesthesia as the implant, thereby completing the treatment to the primary site and the neck. This can usually be done about 3 weeks after the completion of the external beam irradiation. Radiation therapy is certainly suitable for most T1 lesions. For T2 and T3 lesions, it is most suitable for those tumors that are exophytic or have minimal infiltration. Tumors that are deeply infiltrative are preferably managed with a primary surgical approach, usually with postoperative radiation therapy.

 

ADVANCED DISEASE. The surgical management of more extensive lesions requires either a mandibulotomy or a lingual releasing procedure to gain access to disease. The latter procedure entails removal of neck contents prior to primary cancer resection. The tongue is delivered into the neck by releasing musculature attachments posteriorly and mucosal attachments within the oral cavity. Large lesions with mandibular involvement will require composite resection. The term composite resection refers to the removal of tissue involving multiple anatomically defined structures, one of which includes mandible (Fig. 29.2-13). Typically, it refers to resection of a portion of tongue, floor of mouth, and segment of mandible. For patients who require postoperative radiation therapy to the primary site alone, this can frequently be done with brachytherapy. This is especially true for the smaller lesions. The decision of how to deliver this irradiation is integrated with the management of the neck. We prefer to use neck dissection as part of the management of all deeply infiltrative or advanced tongue lesions. For the N0 patients, this generally means a staging procedure or a functional neck dissection. For patients who have involved lymph nodes, this means either a radical neck dissection or one of its modifications. Results of Therapy Decroix and Ghossein [ref: 582] from the Curie Institute in Paris have reported on over 600 patients treated with primary radiation therapy for T1, T2, or T3 squamous cell carcinoma of the oral tongue. Although the majority of patients had implants alone, a large cohort had combined external beam irradiation plus implant. Almost all patients at this center received radiation therapy as their primary treatment. Primary control was obtained at 86% for T1, 80% for T2, and 68% for T3. These data compare quite favorably with the results obtained with partial glossectomy. Pernot and colleagues [ref: 583] have recently reported a series of 448 patients with brachytherapy alone with or without neck dissection (181 patients) or combined external beam plus brachytherapy (267 patients) for oral tongue cancer. The 5-year local control was 93% for T1, 65% for T2, and 49% for T3 lesions. For T2 lesions managed by brachytherapy alone, local control was 90% versus 50% for those managed by external beam plus implant. These data emphasize the importance of using brachytherapy as a major part of the radiation program, but patient selection factors clearly play a role as well. The 5-year overall survival for T1, T2, and T3 lesions (all N stages) was 69%, 41%, and 25%, respectively. Severe complications were uncommon. Whereas 15% experienced grade 1 soft tissue injury and 3% had grade 1 bone necrosis, only 1% and 2% had grade 3 soft tissue and bone complications, respectively. Spiro and colleagues [ref: 584-586] have reviewed the Memorial Sloan-Kettering Cancer Center experience using partial glossectomy as primary management. As was true for the Curie Institute series, the Memorial Hospital series is also relatively unselected, with primary surgery being offered to the great majority of patients at that institution. Local control was obtained in 85% for T1, 77% for T2, and 50% for T3. These data highlight the similarity in local control rates for surgery or irradiation for most early tongue lesions. There are relatively few recent studies that report the results of therapy for surgery alone for advanced disease. [ref: 584,586,587] Radiation therapy is generally utilized in the postoperative setting. Failure is most often within regional lymph nodes and leads to a 5-year survival of 35% for stage III and stage IV disease. [ref: 585,588] Ange and colleagues [ref: 589] have reviewed their experience using implants after excisional biopsy for patients with positive or close margins. Local control was obtained iearly 100% of the cases. Similar results have been reported by others. When the primary site and the neck require postoperative radiation therapy, external beam techniques must be used. Just how radiation is required remains an unanswered question. Bamberg and colleagues [ref: 590] make the point that patients treated to the equivalent of 7000 cGY in 7 weeks do better than those treated to lower doses. Million [ref: 591] recommends 6500 to 7000 cGY, based on the retrospective experience at the University of Florida.

 

REHABILITATION OF THE ORAL CANCER PATIENT Over the last decade there has developed an increased effort to address the rehabilitation of the oral cancer patient, the major considerations reflected in speech and swallowing disorders. [ref: 601-606] There is no question as to the impact of surgical resection of anatomical components of the oral cavity including the mandible, tongue, and other soft tissue components. The ability to preserve function through appropriate reconstructive measures is becoming increasingly apparent. Likewise, rehabilitation efforts have been enhanced in recent years by improved quantitative assessments of functional outcomes, as well as through improving rehabilitation techniques. No medical center or treating physician can truly be considered as providing state-of-the-art therapy unless the are prepared to systematically address these issues.

 

Tumors of the Larynx and Hypopharynx

LARYNX Considering that cancer occurs in the larynx 14 times less frequently than in the lung, 15 times less frequently than in the breast, 16 times less frequently than in the prostate gland, and 9 times less frequently than in the colon, the number of publications on laryngeal cancer that appear in the North American literature seems excessive. This considerable body of writing probably reflects the perceived importance of this disease relative to its potential impact on people’s communication skills. Among oncologists, there seems to exist a new attitude that is characterized by a keener concern for quality of life and death; curing the cancer at any cost is no longer accepted casually, and more than ever before, a premium is now placed on return to a productive and useful lifestyle after cancer treatment. Nowhere in oncology is this changed attitude more vividly demonstrated than in the treatment of laryngeal cancer. It would seem that the mere threat to a patient’s vocal organ is associated with profound psychological overtones. Investigations continue, therefore, into the methods of conservation laryngeal surgery, different radiation therapy strategies, and combined chemotherapy/radiation therapy protocols designed for larynx preservation. [ref: 1-3] Although the cure rates of the various laryngeal malignancies have not changed dramatically during recent years, [ref: 4] the treatment options and the sequencing of those options have, and a higher percentage of laryngeal cancer patients are retaining their larynx in the process.

EPIDEMIOLOGY AND ETIOLOGY Even though considerable differences between countries exist in the incidence of larynx cancer, its distribution within each country is consistent. For example, the disease most commonly affects middle-aged or older men who have smoked tobacco [ref: 5,6] and have drunk alcohol. [ref: 7] Laryngeal cancer rarely occurs in people who have done neither. In the United States during 1994, more than 12,500 new larynx cancers were diagnosed, and over 10,000 of those were in men. This disease has always been more common in men. In 1956, this ratio was 15:1, whereas current studies show a 4.5:1 ratio of men to women. This shifting trend is probably due to the long-term effects of the changing smoking patterns of the sexes. [ref: 7] Compared with whites, blacks in America have a significantly higher incidence of larynx cancer. [ref: 8] The peak incidence of larynx cancer is in the sixth decade. The disease occurs in young people only rarely. [ref: 9] The etiologic factors that have been implicated in laryngeal cancer are voice abuse and chronic laryngitis, [ref: 10,11] certain dietary factors, [ref: 12-14] chronic gastric reflux, [ref: 15] and exposure to wood dust, nitrogen mustard, asbestos, and ionizing radiation. [ref: 16-19] Most consistently seen, however, is the association between larynx cancer and smoking, whether by pipe, cigarette, or cigar. [ref: 16] There seems to be an association with heavy alcohol intake and larynx cancer and an enhancement of the already present risk factors associated with smoking. [ref: 20] On the other hand, some studies have failed to demonstrate an interdependent causal effect for alcohol intake and larynx cancer. [ref: 21] The issue of alcohol, smoke, and carcinogenesis is complicated by the nutritional deficiencies that usually occur in alcoholics. [ref: 14] In the larynx, this complex issue is more specifically defined by the fact that whatever the role of alcohol, it is apparently more significant in supraglottic than in glottic cancers. [ref: 22-25] With the maturation of the current generation of those youngsters using smokeless tobacco, there may be some alteration of the relative incidence in the United States of supraglottic and glottic cancers. Those worldwide data that show large variations of laryngeal cancer statistics consistently reflect the smoking and drinking habits of the individual countries. [ref: 26] Also, the sites within the larynx affected by cancers vary considerably between countries. This distribution is shown in Table 29.3-1, [ref: 10,25,27-29] which represents a compendium of worldwide data that addresses the relative distributions of cancer within the larynx.

 

SURGICAL AND DEVELOPMENTAL ANATOMY The larynx is a uniquely complicated organ that is strategically located so that significant alteration of its anatomy by either surgery or cancer can have a noticeable impact on vocal, digestive, and respiratory physiology. The organ consists of three subsites, which are the glottis (paired true vocal cords), the supraglottis, and the subglottis. Because of different embryologic development and different lymphatic patterns that are subsite specific, discussing larynx cancers without specific reference to the exact location(s) within that structure invites inaccuracies in staging and miscalculations in treatment planning. The larynx consists of four cartilages: the cricoid, the epiglottis, the paired arytenoids, and the shield-like thyroid cartilage. Suspended within the endolarynx are the mobile true vocal cords, which are collectively known as the glottis.

      

That portion above the glottis, the supraglottis, consists of the false vocal cords, the epiglottis, and the aryepiglottic folds. These folds form the junction with the hypopharynx. The medial wall of the aryepiglottic fold is within the endolarynx, and its lateral wall makes up the medial wall of the adjacent pyriform sinus (Fig. 29.3-1). Those lesions that arise on the rime of the aryepiglottic folds, therefore, have been appropriately referred to as marginal cancers, because they bridge the junction between the larynx and the hypopharynx. Those marginal lesions that extend predominantly into the endolarynx behave more like supraglottic cancers, whereas those lesions that spill into the pyriform sinus tend to follow the natural history of the hypopharynx. The subglottis is that portion of the larynx between the underedge of the true vocal cords and the cephalic border of the cricoid cartilage. [ref: 30] The true vocal cords are a marvel of engineering and are attached anteriorly to the inner lamina of the thyroid cartilage and posteriorly to the arytenoids. The muscles of the vocal cords are complex in their activity, and the relation between them and the overlying mucosa is critical to voice production. Any loss of mucosal mobility relative to the underlying muscle, such as that produced by surgery or, to a lesser extent, by radiation therapy, alters the voice. An appreciation of this fundamental fact is an important component in the selection of treatment of vocal cord cancer. The lining of the endolarynx consists of respiratory epithelium except on the vibratory edges of the true vocal cords, which typically are lined with pseudostratified squamous epithelium. The paired arytenoid cartilages each sit on the cephalic rim of the cricoid cartilage and rotate in a relatively horizontal axis around a central pivot point. Each arytenoid is attached anteriorly to a true vocal cord, and the clockwise and counter-clockwise rotation of these cartilages pulls the respective vocal cord attachment with it, causing abduction and adduction of those structures. Any or all of the muscles that are responsible for arytenoid rotation and also the branches of the recurrent laryngeal nerve fibers that innervate them can be damaged by invading cancer. The posterolateral aspect of the larynx is particularly vulnerable to the invasion of cancer because of the adjacency of the medial wall of the pyriform sinus. When cancers of this part of the hypopharynx extend through the mucosa, they gain direct access to the important laryngeal compartment known as the paraglottic space, which leads to all parts of the endolarynx, including the vocal muscles and the preepiglottic space (Fig. 29.3-2). Treatment options for such a tumor are altered significantly because of the paraglottic space involvement. Tumors that invade the endolaryngeal muscles or the nerve fibers that innervate them usually create a noticeable effect on vocal cord motion. Of all the findings on laryngeal examination during cancer evaluation, the state of endolaryngeal mobility is one of the most important. This fact that has been substantiated by the separate designation that the American Joint Committee on Cancer (AJCC) has ascribed to the immobile vocal cord in the staging categorization of this disease. [ref: 31] Another type of motion alteration pertains to the anatomic relation between the vocal cord musculature and the overlying mucosa. This relatively recent knowledge has enhanced our understanding of the pathogenesis and treatment of early glottic cancer. [ref: 32] The vibratory mechanism that produces the voice is due to the mobility of the mucous membrane overlying the musculature of the vocal cord. The free edge of the true vocal cord consists of a pseudostratified squamous epithelium, under which is a lamina propria of fibroelastic and gelatinous consistency. This arrangement allows a sliding motion of the mucous membrane that creates a mucosal wave, the fluidity of which is a direct reflection of the freedom of that layer from the underlying muscle. Any surface cancer that invades through the basement membrane, such as any cancer deeper than carcinoma in situ, affects the mucosal wave by creating a tethering effect. These subtle differences are hardly appreciated by routine laryngeal examination but are obvious with laryngostroboscopic evaluation. [ref: 33] An appreciation of these subtleties translates into the practical matter of determining whether to radiate or microscopically excise certain minimal vocal cord cancers. Because of the different embryologic origins of the supraglottic from the glottic and subglottic larynx, and also because of the independent lymphatic drainage patterns from each of these subsites, the larynx can be thought of as a compartmentalized structure. These features are important influences in determining the spread of various cancers within that organ. [ref: 34] The lymphatics of the supraglottic larynx are profuse, and the frequency of metastasis associated with cancers of this subsite reflects that fact. [ref: 35] Lymphatic spread from the epiglottis is to the false cords, and these channels are directed bilaterally. The drainage from the false cords and the remainder of the supraglottic larynx is lateral and superior, and these channels exit the larynx bilaterally through the thyrohyoid membrane. They then proceed to the adjacent deep cervical nodes. The lymphatics of the infraglottic larynx drain laterally and inferiorly, out of the cricothyroid membrane into the lower deep cervical lymph nodes. The true vocal cords, on the other hand, are unique because they possess little or no lymphatic drainage. [ref: 34] From a lymphatic drainage standpoint, the left half of the larynx is essentially independent from the right and the supraglottic larynx is independent from the structures below. These facts are clinically demonstrated; in their early stages, supraglottic cancers have little affinity for extension into the lower structures, and those beginning below do not tend to extend cephalad. [ref: 36,37] Knowledge of this unique pathogenesis has substantial impact on our ability to predict metastasis into various parts of the neck and on our planning of the various partial laryngectomies, which are those conservation operations that allow removal of laryngeal parts while preserving vocal and swallowing functions. Additionally, radiation therapy planning, especially for occult cervical metastasis, is predicated on a thorough knowledge of these and other drainage tendencies of laryngeal cancers.

 

PATHOLOGY, PATHOGENESIS, AND NATURAL HISTORY General Considerations A variety of malignancies, most of which are primary to the larynx, and others that are metastatic from other sites have been reported. A comprehensive classification is shown in Table 29.3-2. More than 95% of all primary laryngeal malignancies are squamous cell carcinomas, with the remainder being sarcomas, adenocarcinomas, neuroendocrine tumors, and other types. [ref: 38] It should be noted that knowledge and recognition of the neuroendocrine family of malignancies has changed considerably during recent years, and the exact percentage of these within the overall population of larynx cancers is unknown. In the past, certain tumors were vaguely classified as poorly differentiated malignancies, when they actually were neuroendocrine in origin. Modern techniques of immunohistochemical and morphologic analysis will almost certainly lead to the recognition and accrual of more of these tumors in the future. [ref: 39,40] Separate consideration must be given to the spectrum of premalignant squamous lesions, carcinoma in situ, and superficially invasive carcinomas. To discuss the epithelial changes that precede and probably lead to carcinoma of the larynx is of considerable importance, because it is with this group of lesions that cancer prevention and conservative management are most effective. As our knowledge of this subject has increased, so too has our sophistication in applying the minimal techniques necessary to achieve excellent cure rates in these disorders. The value of this philosophy of treatment minimalism is that it leads to the least physiologic change. Investigators have studied the occurrence of aberrant squamous epithelium in various areas of the larynx, and there seems to be a correlation between that metaplasia and the predilection for carcinogenesis in those respective sites; [ref: 41] however, because only lesions that began on the true vocal cords produce early symptoms and signs, the opportunity to treat minimal disease is largely limited to that structure, a fact that leads to spectacular cure rates for lesions of that site.

 

The mucosal changes that lead to cancer take years to develop, and that evolution probably follows a consistent pattern. Most laryngeal squamous cell carcinomas result from prolonged exposure to recognized carcinogens that stimulate mucosal hyperplasia and metaplasia. Some of these changes are associated with keratosis, and others are not. In some situations, epithelial atypia or dysplasia may exist, the degree of which probably determines whether a lesion is destined to become malignant. [ref: 42-44] In one large study by Slamniker, 3% of those patients who demonstrated vocal cord keratosis without atypia and 7% with mild atypia developed invasive carcinoma [ref: 45]; however, in those patients with moderate and severe atypia, 18% and 24%, respectively, developed carcinoma. Another study by Hjslet and colleagues showed a similar probability of cancer evolution in the group with less dysplasia and a strikingly higher probability in those patients with severe atypia. [ref: 46] In addition to the morphologic appearance of mucosal alteration, DNA changes seem to show a correlation between cancer potential and cellular aneuploidy. In a study by Munck-Wirland and coworkers, for instance, all of those patients with dysplastic laryngeal lesions that later went on to become carcinoma demonstrated an aneuploid DNA pattern. [ref: 47] These surface lesions, whether premalignant or not, have an inconsistent gross appearance. Some of these lesions are white, and others are hyperemic. Many investigators believe the risk for cancer development is substantially higher in lesions that are soft and red in appearance. [ref: 44] Without histologic study, even the most experienced diagnostician cannot consistently predict the presence of cancer or the likelihood of its evolution in any of these surface lesions. Any given spot within a lesion does not necessarily represent the balance of that lesion. The facts that carcinoma in situ is often surrounded by dysplastic epithelium and that many areas of invasive carcinoma are surrounded by zones of carcinoma in situ and dysplastic epithelium [ref: 48] lend credence to the concept that each of these morphologic categories of epithelial disturbance is but part of a dynamic spectrum of disorders, each probably related and representing different stages of the same process. This rationale means that dysplasia leads to carcinoma in situ, which leads to invasive carcinoma. It is unknown whether those lesions that have achieved the status of carcinoma continue to grow at the same rate as they did during their premalignant state or whether their growth is accelerated. The growth of a cancer through the basement membrane into the lamina propria constitutes the transition from carcinoma in situ to microinvasive carcinoma, and accompanying this is a tethering of vocal cord mucosal motion. [ref: 33] Failure to appreciate these subtle changes can result in the employment of suboptimal treatment. For example, high failure rates that have been reported with mucosal stripping in carcinoma in situ patients almost certainly represent underestimation of these lesions. [ref: 49] Some of those lesions that had been classified in the prestroboscopic era as carcinoma in situ probably contained areas of invasive carcinoma, and the stripping left behind foci of cancer that resulted in recurrence. Actually, pure vocal cord carcinoma in situ is probably an unusual occurrence. The gross appearance of a given laryngeal lesion is suggestive of its general type. Squamous cell carcinomas originate within the mucous membrane and are exophytic or ulcerative, are of surface origin (Fig. 29.3-3), and are frequently adjacent to or surrounded by mucosal keratoses. Neuroendocrine cancers and tumors metastatic to the larynx are usually submucosal and, as such, do not resemble lesions of surface origin. Metastatic lesions of various types and neuroendocrine tumors are seen throughout the various subsites within the larynx, although the latter group shows a predilection for the supraglottic area. The distribution of squamous cell carcinoma within the various laryngeal subsites varies between different countries, a fact that reflects the different social habits within those cultures. For example, in the United States, the ratio of supraglottic to glottic squamous cell carcinomas is 1:2, whereas the reverse is true in certain European countries (see Table 29.3-1). The major differences in the natural histories of the various squamous cell carcinomas of the larynx are related largely to the area anatomy and to the lymphatic drainage patterns of the respective subsite or subsites. Cellular characteristics vary by site; in the supraglottis, lesions are more likely to be nonkeratinizing and poorly differentiated and, in general, they have more aggressive local behavior. [ref: 50] Those lesions of the true vocal cords, on the other hand, are more often well differentiated and tend to be less aggressive locally. Overall, although the degree of cellular differentiation is not thought to be the most significant fact in tumor grading, it does seem to correlate with the probability of cervical metastasis, [ref: 25,37,53,54] which in turn strongly impacts on survival. [ref: 55-57] Other local characteristics, such as tumor-host interface, [ref: 37,53,58] peritumor inflammatory response, [ref: 59] and vascular and perineural invasion [ref: 58] also seem important in determining performance. Finally, the actual tumor thickness and depth of invasion almost certainly have an influence on metastasis and, ultimately, on survival. As in other sites in the aerodigestive tract, a multifactorial analysis of a variety of parameters, as well as patient performance status, may produce a more predictable prognostic indicator than the standard TNM staging outlined by the AJCC. [ref: 60] Specific Subsites

SUPRAGLOTTIS. Lesions of the supraglottic larynx tend to spread locally. If they begin on the epiglottis, they can extend onto the false vocal cords and into the ventricle. Inferior extension beyond the ventricle is initially thwarted, but as growth continues these cancers can penetrate into the paraglottic space from which they gain full access to the length of the endolarynx. These cancers often exit the paraglottic space cephalad and caudad to enter directly into the neck. Most supraglottic lesions arise on the epiglottis, with fewer being seen on the false vocal cords and aryepiglottic folds. Those lesions that occur on the suprahyoid or upper part of the epiglottis are more often exophytic, whereas those that occur on the lower portion of that structure are likely to be endophytic or ulcerative. [ref: 50,51] The characteristic of endophytic growth is especially significant in this particular area of the epiglottis, because there are foramina here that lead directly through the cartilage into the preepiglottic space, which is a compartment that leads to the base of the tongue. What would appear to be a localized tumor in the endolarynx, therefore, can actually involve considerable unrecognized extralaryngeal extension. [ref: 52] Tumors are confined initially to the preepiglottic space by the ligamentous boundaries of that compartment, but once those barriers are overcome, the loosely arranged skeletal muscle fibers of the tongue provide no restriction to further tumor extension. [ref: 61] Modern imaging, especially magnetic resonance (MRI), has greatly improved the ability to recognize tumor extension into the preepiglottic space and base of tongue. Those lesions that occur on the laryngeal surface of the epiglottis are capable of invading and destroying that structure. Supraglottic cancers, on the other hand, almost never destroy the thyroid cartilage. [ref: 62,63] This feature has an influence on the design of treatment plans; for example, an ossified and invaded thyroid cartilage poses a substantial problem for surgeons attempting to perform partial laryngectomy and also for radiation oncologists attempting to deliver curative therapy. Aryepiglottic fold cancers are somewhat different in their behavior, following more the tendencies of the pyriform sinus lesions, that is, spreading in a more diffuse fashion and metastasizing more frequently than their endolaryngeal counterparts. The particularly ominous natural history of these lesions probably relates as much to the more abundant and multidirectional lymphatic drainage of the area as it does to individual cellular peculiarities. [ref: 52] Because of the profuse lymphatic network of the area, supraglottic carcinomas metastasize frequently to the cervical lymph nodes, and failure of treatment is usually a result of metastasis rather than local disease. [ref: 35,64-66] The incidence of patients with clinically positive lymph nodes at the time of diagnosis is 23% to 50% for all supraglottic sites and stages combined. [ref: 51,67-71] A substantial number of those patients with clinically negative necks turn out to have histologic disease if a neck dissection is done or, if left untreated, to convert to clinically positive necks. [ref: 64,65] In supraglottic cancers, the probability of cervical metastasis and the probability of delayed contralateral metastasis increase in direct proportion to the size of the primary (i.e., the T stage). [ref: 50,72,73] Lindberg reported impressive overall metastatic rates with various supraglottic carcinomas: T1 had 63%; T2 had 70%; T3 had 79%; and T4 had 73%. [ref: 35] In that group of patients with supraglottic lesions that present with a clinically positive cervical node 2 cm in diameter or more, the possibility for contralateral neck metastasis is 40% or higher. [ref: 74] The epiglottis is particularly prone to bilateral metastasis, and even in smaller lesions of that site, the incidence of contralateral metastasis is more than 20%. [ref: 72] Much of the data on clinically positive necks and on occult metastasis were compiled before the routine employment of computed tomography (CT) and MRI of the neck. With the employment of these more sophisticated staging methods added to the already 75% to 85% accuracy of physical examination, [ref: 75-78] the overall incidence of metastasis noted at the time of diagnosis will probably be higher than that reported previously. GLOTTIS. Glottic or true vocal cord carcinoma is the most common of all laryngeal cancers encountered in the United States. Although these lesions are usually well differentiated, they often demonstrate an infiltrative growth pattern, even when they appear exophytic and well organized. Most true vocal cord cancers occur on the anterior two thirds of that structure, a small percentage of them develop on the anterior commissure, and they rarely occur on the posterior commissure.

 

To a large extent, growth characteristics and the natural history of glottic carcinomas are determined by the unique anatomy of the true vocal cords. First, the sparcity of the lymphatic drainage of the true vocal cords in all areas other than the posterior commissure makes metastasis of early lesions extremely unlikely. Second, the elastic layers (conus elasticus) within the larynx often divert cancers that begin on the free edge of the vocal cord and continue into the underlying vocalis muscle and paraglottic space, which is an inferolateral pathway that leads out of the larynx through the cricothyroid space.

With penetration into the underlying tissues, all degrees of motion impairment, from subtle mucous membrane stiffness to frank fixation of the vocal cord, can follow. That increasing impairment of motion has a telling effect on local control and survival data, a fact that is reflected in AJCC staging designations. Much discussion continues about mobility change and its therapeutic implications, and it is in the group of glottic cancers that demonstrate this that the clinical judgment of the physician is most tested. The final anatomic factor unique to the glottis that influences the growth pattern of certain cancers is the anterior commissure ligament that forms the bridge between the anterior ends of the true vocal cords. This structure lies immediately against the inner lamina of the thyroid cartilage, and its presence initially retards penetration of cancers of that area, often causing their diversion upward onto the epiglottis or downward onto the cricothyroid membrane. From there, these lesions can escape the larynx into the anterior neck. If the cancer overcomes the ligamentous barrier at the anterior commissure, the cartilage is penetrated. [ref: 62,80] This event is particularly likely in thyroid cartilages that are ossified, and when this does occur, there are substantial therapeutic implications that compromise the radiotherapist and dictate certain surgical approaches. [ref: 81,82]

SUBGLOTTIS. Carcinomas of the subglottic larynx are unusual, making up only about 1% to 8% of all laryngeal cancers. [ref: 83] These lesions tend to be poorly differentiated and often demonstrate an infiltrative growth pattern unrestricted by tissue barriers. These tumors are, therefore, frequently circumferential and can extend down the trachea. The incidence of cervical metastasis in this group of cancers is reported to be 20% to 30%, but that figure is somewhat obscured by the fact that the primary drainage pattern of these lesions is to the less detectable pretracheal and paratracheal nodes. The actual incidence of metastasis may, therefore, be significantly higher. [ref: 84,85] Unusual and Rare Neoplasms The pathology and pathogenesis of verrucous carcinoma is unique and deserves special consideration. This unusual tumor is poorly understood, and its origin, classification, and response to treatment are controversial. [ref: 86,87] Verrucous carcinoma is described as a distinct neoplastic entity of squamous origin that occurs in the oral cavity, larynx, esophagus, and nose and on the genitalia. [ref: 88-90] Some authorities have suggested the human papilloma virus as its cause. [ref: 92] Even though there are views to the contrary, most investigators consider verrucous carcinoma to be an entity unto itself. [ref: 91] Just because some tumors originally thought to be verrucous carcinoma are discovered to have features of squamous cell carcinoma and can metastasize does not, in their opinion, justify combining the two diagnoses; actually, they think that such tumors were always low-grade squamous cell carcinomas rather than verrucous carcinoma. [ref: 90] Other investigators, although conceding verrucous carcinoma to be unique, believe it to be only a variant of well-differentiated squamous cell carcinoma. Different authors believe that because verrucous carcinomas neither fulfill the histologic and cytologic criteria of malignancy nor possess the capability to metastasize, they should be renamed verrucous acanthoma rather than carcinoma. [ref: 93] When this lesion does occur in the larynx, it usually is on the true vocal cord, where it grows slowly and can cause significant local destruction by expanding gradually. Even though these lesions often destroy cartilage, they do not tend to metastasize; aggressiveness is directed locally. Verrucous carcinoma is consistently difficult to diagnose, even when the clinical index of suspicion is high. This observation relates to the fact that these tumors microscopically demonstrate an exuberant and keratinizing hyperplasia that is benign by pure histologic and cytologic criteria. [ref: 94] The diagnosis is largely a clinical one and is most effectively achieved by concern between pathologist and surgeon, but usually only after multiple biopsies have been taken. This tumor is typically a slow-growing but relentless mass, exophytic and warty in appearance, and broad based at its interface with the mucosa (Fig. 29.3-4). Its surface is ofteecrotic and infected, and the associated inflammation of adjacent tissues can be remarkable. This tendency to cause inflammation can erroneously influence treatment planning. For example, the patient with verrucous carcinoma can demonstrate enlarged adjacent cervical lymph nodes that are worrisome when in fact the adenopathy is only secondary to the inflammatory process. Although this finding has been described in other aerodigestive tumors, [ref: 95] the mere presence of lymphadenopathy in the primary drainage area of an impressive primary tumor is worrisome, no matter how benign looking its histology. In such a circumstance, clinical judgment is enhanced greatly by modern imaging and cytopathologic techniques. This discussion of the nature of verrucous carcinoma has substantial therapeutic implications, especially when the lesion occurs in the larynx. Essentially, squamous cell carcinoma is a radiosensitive cancer, a fact that provides treatment options to the physician. On the other hand, verrucous carcinoma seems to be somewhat radioresistant, whether found in the mouth or the larynx. [ref: 86] Additionally, there is anecdotal information suggesting radiation induced dedifferentiation into anaplastic cancer in these lesions. This transformation seems to occur in fewer than 10% of verrucous carcinoma [ref: 67,96-99] and may involve alteration of the DNA that facilitates the integration of the human papillomavirus into host cells. [ref: 92] Both the concept of radiation resistance and the transformation into anaplastic cancers are vigorously disputed. [ref: 90,100-102] The neuroendocrine family of tumors represents an evolving data base. This is true largely because recent diagnostic techniques have allowed pathologists specifically to label as neuroendocrine a variety of previously undefined cancers. Almost certainly, an immunohistochemical reexamination of laryngeal cancers previously diagnosed as atypical or undifferentiated malignancies would result in the reclassification of many as neuroendocrine tumors. The small cell tumors look and act much like their counterpart oat cell lung lesions and as such generally are managed by chemotherapy and radiation therapy. [ref: 3,39,103] Surgical procedures do not seem to enhance the likelihood of survival in patients with these tumors. [ref: 104-108] The other neuroendocrine tumors that occur in the larynx — carcinoids and paragangliomas — are rare and are best managed surgically. [ref: 109-111] Cartilaginous malignancies, [ref: 112,113] adenocarcinomas, [ref: 114] sarcomas, [ref: 115] malignant fibrous histiocytomas, [ref: 116,117] plasmacytomas, [ref: 118,119] granular cell tumors, [ref: 120] and primary lymphomas [ref: 121] have all been reported but are rare. Primary melanomas of the larynx are equally rare; of all of the larynx cancers reported from Memorial Sloan-Kettering Cancer Center between 1949 and 1983, only three were melanomas. [ref: 122]

 

DIAGNOSIS AND EVALUATION Cancers of the supraglottic larynx usually do not produce early symptoms or signs, and it is common for the first hint of such a cancer to be cervical adenopathy. When symptoms do occur, they are often subtle; pain perceived in the primary site or in the ear (otalgia), a scratchy sensation when swallowing, or merely an alteration of one’s tolerance for hot or cold foods may be all that is noticeable. Airway alteration, hoarseness, or a tendency to aspirate liquids are all produced by more advanced lesions. Cancers of the glottis, on the other hand, are often detected early in the course of the disease because even a slight alteration of the vibratory surface of the true vocal cord(s) produces voice change. Smokers are often hoarse, however, and such alteration of the voice may not alarm them. Anyone with a voice change that does not resolve within several weeks should have a laryngeal examination. It is unusual for glottic cancer patients to seek medical attention because of cervical adenopathy. In such lesions, metastasis generally occurs late in the course of the disease, long after the early warning signals. Fortunately, subglottic cancers are uncommon, but when they occur, they fail to produce early symptoms; therefore, the disease is often advanced by the time of diagnosis. Most larynx cancers are squamous carcinomas and, as such, are surface lesions. Most are visible with routine laryngeal inspection, but a small percentage are obscure. The modern generation of flexible endoscopes (Fig. 29.3-5) has provided to a broad range of physicians the capability to examine the larynx; and with the improved optical resolution of these instruments, the overall process of screening and follow-up after treatment has been enhanced by this technology. Importantly, these methods allow the occasional laryngeal examiner to see areas that previously have been visually inaccessible. It is essential that the larynx be examined in the awake patient who is sitting upright. Direct laryngoscopy should be reserved for biopsy and a more detailed tumor mapping (Fig. 29.3-6). Even when done under local anesthesia, the introduction of a direct laryngoscope distorts the natural position and the relaxed motion of the larynx and, by doing so, tends to disguise subtle motion changes that are important in staging of these tumors. Certain subtleties of contour, such as bulging and tethering, are visually not appreciable during direct laryngoscopy. The earliest stage of invasive glottic carcinoma through mucosa into the underlying lamina propria is visible as a tethering of the mucous membrane that is designed to slide over the underlying structures (i.e., a loss of the membrane wave). [ref: 32] The gross abductive capabilities of the vocal cord may be intact, but the early invasive character of a lesion can be appreciated when the stroboscope is employed and demonstrates this restrictive feature. As the process of invasion continues into the vocalis muscle, the actual lateral excursion of the vocal cord is limited and eventually lost. The ability of the clinician to view and interpret this scenario is critical to the sophisticated management of larynx cancer in general and vocal cord cancer in particular. Essentially, lesions of the true vocal cord that do not transgress the basement membrane (i.e., surface lesions) do not cause tethering of the membrane, and those that enter the underlying lamina propria do. [ref: 33] Benign lesions and even carcinoma in situ, therefore, may look extensive topographically, but their lack of depth is revealed by appropriate diagnostic technology. Even though contemporary methods of staging tend to emphasize the bulk and topographic size of tumors rather than depth, recent thinking has begun to focus more on this third dimension. As data are accumulated, more emphasis will be placed on this important matter. Imaging should not be relied upon to detect early larynx cancer, because the routine methods of physical examination are far more suitable. The primary care physician should not, therefore, initially resort to CT or MRI when a laryngeal cancer symptom persists. Instead, that patient should be examined by someone skilled in the appropriate techniques. Once a lesion is discovered, the evaluation of its depth, bulk, and cartilage invasion and the status of the regional lymph nodes are enhanced by CT or MRI. It is not clear which of the two imaging methods is better for larynx cancer evaluation. Both methods have certain advantages over the other, and both are of limited usefulness in evaluating the radiated larynx. CT of the larynx is most effectively achieved in the axial plane, and because of this, the images show well the lateral tumor extension and the relation of that extension to cervical nodal disease. The axial projection is effective in demonstrating the important paraglottic space. CT effectively demonstrates the vertical extension of tumor, especially in the subglottic and anterior commissure areas. MRI offers the advantages of multiplane visualization of the larynx, and therefore, is especially valuable in evaluating the preepiglottic space and the adjacent base of the tongue. Invasion of laryngeal cartilage is important in treatment planning. Determining whether this feature exists has always been difficult because of the inconsistency of the ossification that occurs in the laryngeal framework. Generally, cartilage is vulnerable to tumor invasion in those areas where it is ossified and somewhat resistant where it is not. In fact, healthy, nonossified cartilage provides a considerable natural barrier to cancer invasion. Writings by Castelijns and associates [ref: 123,124] and by Towler and Young [ref: 125] have suggested that MRI is the method of choice for delineating the important finding of cartilage invasion. Other investigators would dispute this. One study correlated MRI findings of cartilage invasion with the effectiveness of radiation treatment, and in so doing, the authors found a surprising number of small glottic lesions with foci of cartilage invasion. [ref: 126] Significantly, it was from this group that most of the radiation failures of the series occurred. Other imaging methods such as tomography and laryngography have been surpassed by more elaborate technology and are only of historic interest.

 

STAGING The AJCC last updated larynx staging in 1983, and that version is presented in Table 29.3-3. [ref: 31] Staging provides a commonality of language that is essential for effective outcomes analysis. The larynx is a complex structure because it involves many anatomic and physiologic factors that impact on performance and, therefore, on staging. Although it is essential that pathologic findings always be compared with preoperative analysis, it should be remembered that the staging referred to and that reported by the AJCC is a clinical one, which is based on performance. The accuracy of clinical staging is periodically updated on the basis of better recognition of performance. For example, Pillsbury and Kirchner studied this question by comparing whole-organ sections of nonradiated larynges and compared the actual pathologic findings with the preoperative staging. [ref: 127] They found that 40% had been categorized incorrectly, most inaccuracies being attributable to understaging. Most commonly, the depth of invasion had been underestimated, and the frequency of cartilage invasion was much higher than had been realized previously. Certainly, as imaging technology improves, so will the ability to stage more accurately. As clinicians employ treatment protocols that use neoadjuvant chemotherapy and in which the assessment of complete versus partial response is required, modern imaging hopefully will enhance the accuracy of that assessment. Survival in larynx cancer decreases in a linear fashion with increasing stage. The most remarkable change is between stages II and III, the zone that generally represents the occurrence of cervical metastasis.

 

TREATMENT AND SURVIVAL Supraglottis Because the supraglottic larynx is composed of multiple sites, referring to it as one unit is not always accurate when discussing treatment results. Because all of the subsites are intimately related and because the supraglottis is continuous with its neighboring hypopharynx, glottic larynx, and oropharynx, it can be difficult to determine the exact site of origin for many larger cancers. For example, when one encounters a lesion that involves the pharyngoepiglottic fold, aryepiglottic fold, and pyriform sinus, it can be difficult to know whether this is a primary hypopharynx cancer extending superiorly or a primary supraglottic cancer extending inferiorly. Unlike glottic cancer, in which cervical metastasis is relatively uncommon for early-stage disease, the probability of nodal spread in all supraglottic lesions is substantial. [ref: 35,50] There is a significant probability of contralateral metastasis in these lesions, and that increases with increasing primary size.

This is especially true for epiglottic lesions, which make up most of the supraglottic carcinomas. It is essential to recognize that the site of treatment failure in supraglottic cancer is usually the neck; therefore, treatment strategies require neck management for virtually all lesions. For the N0 neck, this implies selective neck dissection or elective radiation, and for patients with clinically positive neck or necks, this implies neck dissection, therapeutic radiation, or a combination of both. Early-stage primary disease is highly curable by partial laryngeal surgery or by radiation therapy. More advanced lesions, on the other hand, usually are treated with combined modalities and often require total laryngectomy. Wang and associates have reported an extensive experience with radiating early lesions dating back to 1973. [ref: 128] They have reported 5-year actuarial disease-free survival of 73% and 50% for T1 and T2 lesions, respectively. When surgical salvage was added, this survival increased to 80% and 58%, respectively. A total of 92% of T1 patients and 86% of T2 patients survived with their larynx. Mendenhall and colleagues reported local control of 100% of T1 and 81% of T2 lesions. [ref: 129] When surgical salvage was added, local control for T2 cancers increased to 88%. Importantly, this study revealed no significant differences in local control by anatomic subsite. These data were recently fortified when Mendenhall and his group reported on 209 patients with a minimum 2-year follow-up after radiation therapy, with or without neck dissection. Local control was 100% for T1 lesions and 83% for T2 lesions. In the T2 group, local control was 80% for once daily radiation versus 90% for twice daily. [ref: 130] Olofsson and coworkers reported a 47% 3-year survival for T1 and T2 lesions. [ref: 131] Lederman reported a 53% control rate for T1 and 64% for T2 tumors at 5 years. [ref: 132] Fletcher and associates reported local control in 94% of T1 and 89% of T2 lesions, with no difference noted by anatomic subsite within the supraglottis. [ref: 133] Vermund compared the 5-year survival for primary surgery with primary radiation for T1, N0 to T4, N0 supraglottic carcinoma. [ref: 134] The results for T1, N0 to T3, N0 disease were identical for both groups, but there was an advantage (56% versus 14%) for T4, N0 treated by primary surgery. Those smaller lesions of the supraglottis (i.e., T1 and T2) are equally well treated with surgical procedures that remove only the upper portion of the larynx. The so-called supraglottic laryngectomy is physiologic and allows retention of vocal and swallowing functions. Because of the unique lymphatic drainage patterns of the organ and the presence of certaiatural anatomic barriers to tumor spread, this operation is oncologically sound, yielding the same local control rates as achieved by total laryngectomy in comparable lesions. [ref: 27,51,135] Some authors described a 68% 5-year control rate with supraglottic laryngectomy [ref: 74]; Ogura and Biller reported an 85% 3-year control for epiglottis cancers treated with supraglottic laryngectomy, but this decreased to 71% with extension onto the false cords. [ref: 135] Because of the complications associated with persistent swelling, swallowing difficulty, and wound healing, supraglottic laryngectomy usually is not recommended in patients who have had full-course radiation therapy. The decision to radiate primarily a supraglottic laryngeal lesion should be made with the realization that the backup operation for failure of therapy is almost always a total laryngectomy. Management of the neck is critical to successful therapy of supraglottic cancer. Levendag and coworkers studied elective surgical management of the neck in a group of patients with stages I and II supraglottic carcinomas treated with surgery alone at the Memorial Sloan-Kettering Cancer Center. [ref: 136] In those patients who underwent elective neck dissection, that is, the group with clinically negative necks, 32% were found to have histologically positive cervical lymph nodes. Half the patients with involved nodes eventually failed in the dissected neck. Additionally, 19% of patients with negative elective neck dissections failed in the contralateral neck. Finally, in a group of 48 patients who did not have elective neck dissection, 29% failed in the neck. Therefore, a total of 35% of the T1N0 and T2N0 patients ultimately developed cervical lymph node metastases. Importantly, nearly two thirds of those who relapsed in the neck eventually died from their cancer. On the other hand, none of the patients without neck relapse died from supraglottic cancer. These investigators compared their experience with a similar patient group from the other study groups that showed similar neck failure rates with surgery alone and when radiation therapy was administered to the necks. Harwood and coworkers reported a 3% failure rate in the electively radiated neck. [ref: 137] These reports indicate that even the smallest supraglottic tumors require elective neck management. A relative advantage of radiation therapy in the treatment of early-stage disease is that bilateral elective neck treatment can be done with minimal morbidity. If an adequate dose of elective neck radiation is given, neck relapse should be less than 5%. If surgery is chosen as the treatment for a T1 or T2 supraglottic lesion, the supraglottic laryngectomy should be combined with bilateral selective neck dissections. Postoperative radiation therapy is added in the patients who have metastatic disease in the neck or necks. The obvious disadvantage to this approach is that it becomes necessary to employ two different treatment modalities compared with the strategy in which radiation therapy is employed initially to the primary and necks. The disadvantage of the latter plan is that when failure occurs, total laryngectomy is needed. It should be noted, however, that in the very rare patient that fails locally after supraglottic laryngectomy, total laryngectomy becomes necessary for salvage. Although surgery and radiation strategies appear to yield comparable results for small supraglottic lesions with negative necks, the effectiveness of radiation diminishes markedly in controlling advanced lesions.

Various studies report dismal survival statistics with radiation therapy management alone; Siirala and Pavolainen reported a 20% 5-year survival [ref: 138]; Vermund reported a 32% 5-year survival [ref: 134]; and Wang and colleagues reported a 5-year actuarial survival of 37% for T3 and 23% for T4 lesions. [ref: 139] Mendenhall and colleagues reported local control in 61% and 33% of radiated T3 and T4 lesions, respectively. [ref: 129] These numbers increase to 83% and 66% with surgical salvage. There was no difference in local control in patients suitable for a supraglottic laryngectomy compared with those who were not suitable for this procedure. Supraglottis (Continued) (1 of 1) There are a variety of factors that can help select a patient for primary radiation therapy, even if the patient has an advanced-stage lesion. For example, all T3 lesions are not alike. Freeman and coworkers [ref: 140] evaluated the local control after definitive radiation therapy as a function of the extent of preepiglottic space invasion and a CT-derived measurement of the primary tumor volume. For lesions involving less than 25% of the preepiglottic space, local control was 75% versus 60% when more extensive involvement exists. In addition, for T3 lesions whose volume is less than 6 cm**2, local control was 82% versus 55% for lesions at least 6cm**2. This trend is sustained in a more recent report. [ref: 141] This information suggests that additional factors other than stage should be considered before excluding definitive radiation therapy as a treatment option in T3 lesions. It also suggests that tumor bulk, rather than T-stage designation, is a better predictor of local control. It should also be noted that T3 designation by virtue of preepiglottic space invasion is different from the lesions that achieve T3 status because of vocal cord invasion and immobilization. This latter circumstance is, in fact, often associated with unrecognized cartilage invasion, and as such, these are really T4 cancers, and the performance is probably different from that of the preepiglottic space group. [ref: 62] An important advance has been the development of chemotherapy-radiation therapy programs for larynx preservation for patients who would otherwise require total laryngectomy. The concepts are discussed in some detail later in this chapter, and apply to supraglottic cancers as well. If total laryngectomy is required, postoperative radiation is usually given to the primary site and necks. For those with subglottic extension, the stoma is included in the field. It is not the mission of this text to describe elaborately the various surgical techniques that are used to manage supraglottic laryngeal cancer; however, the student of this disease and its management should have at least a summary knowledge of the precise methods known collectively as conservation laryngeal surgery]. The compartmentalization of the larynx and the directional drainage patterns of the lymph channels within it provide the surgeon with a unique setting for removing that portion of the larynx above the true vocal cords and, with proper reconstruction, to allow retention of the swallowing and vocal functions of the patient. Because of the consistent drainage patterns of the area cancers, the so-called supraglottic laryngectomy is associated with the same cure rates as would be produced by a total laryngectomy in the same lesion. Essentially, this procedure is a horizontially directed hemilaryngectomy in which the surgeon removes the upper half of the thyroid cartilage and the contents within it, namely the false vocal cords, the epiglottis, and the aryepiglottic folds. The edge of the thyroid cartilage is brought up to and approximated to the transected base of the tongue. Because the motor nerve supply of the vocal cords comes from below (recurrent laryngeal nerves), the important glottic function of abduction and adduction are retained; because of this, voice and the important functions of glottic closure are retained. The supraglottic laryngectomy is, however, physiologically challenging, and patients with chronic pulmonary disease do not tolerate the aspiration that can follow. Essentially, this elegant technique is oncologically sound in appropriate tumors, but certain patients are not good candidates for its implementation. The correct use of the supraglottic laryngectomy is accomplished only by surgeons properly trained in this complex methodology and who have the experience to apply the right methods in the right situation. A succinct discussion of the method of selection for conservation procedures and which patients are suitable for them was developed by Sessions and Parish. [ref: 142] Although there have been certain chemoradiotherapy options popularized since that discussion was published, the fundamental principles are the same and can be applied to the current philosophy. As those alternative schema such as the larynx preservation protocols are implemented more frequently, fewer supraglottic laryngectomies will be done, and the number of surgeons accomplished in these methods will diminish. While the idea of saving more larynges without compromising cure is worthwhile, it is worrisome that those subtle skills necessary for achieving excellent functional results with the family of partial laryngectomies is to some extent being lost by the current generation of head and neck surgeons. There are a variety of conservation surgery procedures that are applied to variations of laryngeal cancer, and while a description of each is beyond the scope of this text, a classification of these operations should be helpful to the reader in placing this important surgical methodology into the proper perspective. Such a classification is the following: 1. Hemilaryngectomy a. Horizontal hemilaryngectomy (supraglottic) b. Vertical hemilaryngectomy i. Lateral hemilaryngectomy ii. Frontal hemilaryngectomy 2. Cordectomy 3. Partial laryngopharyngectomy Elaborate details of radiation techniques for the treatment of supraglottic carcinoma are not intended in this writing, but certain principles and concepts are essential and will be described. Patients are generally treated with opposed lateral fields for the primary site and upper neck, and a separate anterior field for the lower neck (including the stoma in appropriate patients). The spinal cord is restricted to 45 Gy. Electrons are used to boost the posterior neck, when appropriate, after the spinal cord has been protected. For T1 and T2 lesions, a total dose of 65 to 68 Gy is used, while more advanced lesions receive at least 70 Gy. Patients receive 1.8 to 2.0 Gy per fraction, continuous course treatment. Generally, either Co-60 or a low energy linear accelerator is used. Care should be taken to ensure dose homogeneity throughout the larynx, usually via a larynx compensator. A variety of fractionation programs have been used. The most traditional is a once daily, continuous course program. Several twice daily (BID) programs have been used, but there is no firm data to suggest their superiority to the traditional, once daily programs. Wang [ref: 128] reported a regimen of 1.6 Gy BID to approximately 38.4 Gy, a 2-week break, and continuation to approximately 64 to 67 Gy. Local control was 66% for T3-4 lesions managed with BID radiation versus 33% for patients treated once daily. These data are retrospective and must be confirmed in a randomized trial before this approach is considered superior. Mendenhall [ref: 129] attempted to compare BID to once daily radiation. Their program involved 1.2 Gy BID, continuous course, to doses generally in the 74.4 to 76.8-Gy range. There is clearly no benefit to BID treatment for early-stage disease. There are too few patients with advanced disease to make definitive judgements. Glottis Carcinoma in situ (CIS) of the true vocal cord is highly curable. It can be cured with equal efficiency by microexcision, laser vaporization, [ref: 143] or radiation therapy. [ref: 144,145] Although pure carcinoma in situ lesions are unusual, there is a frequent association between carcinoma in situ and invasive carcinoma. Those series that have been reported in which there were numerous recurrences after stripping of vocal cord carcinoma in situ lesions almost certainly consisted of a heterogenous group that included lesions containing areas of invasive cancer. True carcinoma in situ, by definition, remains superficial to the basement membrane, and if mucosal excision techniques are confined to that group of patients, the cure rate should be the same as the best that can be achieved by radiation therapy. The advantages of radiation therapy are that the voice is probably better than with surgery, it is a more definitive treatment for invasive cancer that may exist within the lesion, there is no requirement for general anesthesia, and when treatment fails, a conservation surgical procedure can salvage most patients. The advantage of surgery, whether it be microexcision or laser vaporization, is that it is simpler, and radiation therapy is held in reserve for future use. As long as the integrity of the submucosa is not violated, the voice results with microexcision or laser vaporization are probably comparable with that achieved with radiation. All things considered, if the diagnosis of pure carcinoma in situ of the vocal cord or vocal cords is fairly certain, microexcision is probably the treatment of choice. So called “vocal cord stripping” often has a crippling effect on voice and is to be discouraged. The more precise microexcision, on the other hand, is followed by minimal impact on vocal function. Additionally, microincision has the advantage over both laser vaporization and radiation therapy of providing the pathologist with a specimen for complete histologic examination, perhaps revealing small areas of microinvasion that would have otherwise been unappreciated. Radiation therapy is used either as the definitive treatment of CIS when microexcision is not possible or is refused, or as a salvage treatment for patients with recurrent CIS following prior surgical procedures. [ref: 146,147] This is not an issue, however, on which there is complete agreement. There are those who feel that endoscopic cordectomy is the superior method of addressing this problem, both from a curative and from an economic standpoint. [ref: 148] On one hand, function after radiation for CIS of the vocal cord is better than in cordectomy, but considering the value of saving the radiation option for potential need later, there is a case to be made for the surgical option. Importantly, if a surgical option is chosen, microexcision of these lesions must be associated with a thorough pathologic evaluation of the specimen to minimize the possibility of microinvasive areas going undetected. Cordectomy, on the other hand, although associated with a substantial compromise of vocal function, tends to avoid this pitfall. We tend to rely heavily on the preoperative evaluation by videostroboscopy [ref: 33] to estimate the depth of the surface cancer. If we suspect the lesion is truly confined superficial to the basement membrane, we do microexcision and examine the specimen carefully for microinvasion. If there is evidence of microinvasion, the patient usually receives radiation. On the other hand, if the stroboscopic analysis suggests restricted mucus membrane motion and possible invasion, we generally begin with radiation. We do not favor cordectomy because of the poor vocalization results associated with this procedure. Many of these so-called carcinoma in situ lesions have probably been understaged and are riddled with areas of superficial invasive carcinoma. Also, certain carcinoma in situ lesions, such as those on the anterior commissure, in the subglottis, or some that extend into the laryngeal ventricle, do not lend themselves to these surgical methods; in these, radiation is probably a better means of treatment. For early glottic cancer (T1 or T2), excellent local control is achieved by radiation or partial laryngectomy. The relative advantage of radiation over surgery relates to function. With radiation, the voice is unquestionably better. After this treatment, the voice is normal or near normal most of the time. Harrison and coworkers have analyzed the impact on vocal quality by radiation therapy for T1 and T2 glottic lesions, and their conclusion corroborated this tenet. [ref: 149] In contrast, all patients are hoarse to a varying degree after hemilaryngectomy or cordectomy. Hemilaryngectomy can be used successfully for salvage in many patients who fail radiation therapy. [ref: 150-152] In a carefully selected subset of those patients in whom the initial treatment is radiation therapy, there is a second line of defense against glottic cancer that does not involve sacrifice of the larynx. The actual survival results for primary surgery for T1 glottic cancer are 84% to 98% with laryngofissure and cordectomy. [ref: 152-155] Various series have reported similar results with hemilaryngectomy, which is considered to be a better operation, oncologically and functionally, than a cordectomy. Essentially, the 5-year survival rates for primary surgery and primary radiation for T1 lesions are comparable. [ref: 156-160] Local control obtained in this same stage glottic group using conservation surgical procedures is reported to be 78% by Kirchner and Owen [ref: 67] and 87% by Ogura and associates. [ref: 161] Results obtained for comparable (T1) lesions treated by radiation therapy show local control of 91% by Harwood [ref: 144] and 93% by Pellitteri and colleagues. [ref: 160] As is true with any treatment, the local control rate decreases with increasing tumor bulk. This finding is especially true for radiation therapy of early glottic cancer. Dickens and coworkers have reported the results for early glottic tumors of various sizes and extent. [ref: 162] The lesions were categorized by the type of surgical procedure that would have beeecessary had surgery been used. This type of analysis provides an excellent basis for comparing the results of surgery and radiation. The local control with radiation alone in patients suitable for cordectomy was 97%, and in patients who needed hemilaryngectomy it was 94%. In both categories, local control increased to 100% when surgical salvage was added. On the contrary, for patients managed by radiation therapy who would have required total laryngectomy, local control was only 65%, but it increased to 91% when surgical salvage was added. Extension of these glottic carcinomas onto the anterior and posterior aspects of the larynx lessens the local control rates achieved with radiation therapy. [ref: 163] The surgical procedure required for salvage usually was a total laryngectomy rather than a hemilaryngectomy. Specifically, with anterior commissure involvement, Sessions and associates reported a 5-year survival of 74% for T1 and T2 lesions [ref: 164] and usually found that survival and recurrence rates of anterior commissure lesions correlated with the size and stage of the tumor. [ref: 165] Results with radiation therapy for early-stage glottic carcinomas that involve the anterior commissure are reported by Olofsson and colleagues [ref: 166] to be 80% survival at 5 years (including those recurrences salvaged by surgery) and Kirchner and Fischer, [ref: 80] who reported a local control rate of 85%. Those anterior commissure lesions that are thin and of low volume and that do not have substantial subglottic extension probably are treated with equal efficiency by partial laryngectomy or radiation therapy. As lesions become more advanced, the natural barrier of the anterior commissure ligament is overcome and the thyroid cartilage is invaded [ref: 167]; therefore, radiation therapy becomes less appealing than surgery as the front-line treatment. Most tumors involving the anterior commissure occur as a result of spread from the true vocal cord. Lesions actually arising in the anterior commissure are unusual, making up 1% to 2% of glottic cancers. [ref: 165,167] Glottis (Continued) (1 of 4) In most centers, most T1 glottic cancers are treated with radiation therapy, and partial or total laryngectomy is used as a salvage operation in those patients who fail radiation. The management of T2 lesions is more complicated, because this is a somewhat heterogenous group. Surgical management usually consists of vertical hemilaryngectomy and is associated with 3-year survival rates of 83% [ref: 164] and 82% [ref: 161] in two major series. Primary radiation therapy with surgical salvage yields a net 5-year survival rate of 92% in the series of Pellitteri and associates, [ref: 160] 72% in Wang’s series, [ref: 157] 90% in Fletcher and colleagues’ series, [ref: 159] and 72% in Jorgensen’s series. [ref: 158] Because of the heterogeneity of the T2 group, Wang has suggested subdividing these lesions into those with normal mobility (T2A) and those with impaired mobility (T2B). [ref: 157] He showed that local control was obtained in 86% of the former and 63% of the latter when primary radiation was used. Similar observations were made by Harwood, [ref: 144] whose series yielded 77% and 51% local control rates for T2A and T2B lesions, respectively. Essentially, T2B lesions seem to behave more like T3 than T2 lesions. Radiation therapy usually is recommended as the primary treatment modality for T2 lesions with no vocal cord mobility impairment (T2A); however, in those lesions that demonstrate impairment of motion (T2B), hemilaryngectomy usually is preferred. These criteria are variable, however, depending on tumor bulk, and in those T2B lesions with less bulky lesions, radiation can be employed. It should be noted also that when vocal cord motion is restricted by actual surface tumor bulk, rather than by invasion of the underlying muscle, radiation is often effective. The overall 5-year survival rate for T1 glottic carcinomas is 82% to 96%, and for T2 lesions it is 51% to 85%. [ref: 168,170] Those conservation surgical procedures that can be applied to the management of glottic cancer are time honored and tested, and when used in the properly selected case of laryngeal cancer, they consistently yield excellent results functionally and oncologically. The most commonly employed of these procedures is the vertical hemilaryngectomy (see the classification of conservation laryngeal procedures outlined earlier) in which the surgeon bisects the larynx and, to a varying degree, removes a portion or all of the true and false vocal cord along with the respective half of the thyroid cartilage. Because most of the lesions for which this operation is employed are located on the anterior two thirds of the vocal cord, the most posterior resection line usually is in front of the arytenoid cartilage. In those circumstances in which the cancer extends onto the posterior larynx, this cartilage can be resected (Fig. 29.3-7). By using the perichondrium from the external surface of the half of the thyroid cartilage that has been removed, the operated side of the larynx heals in the midline, forming a firm buttress (pseudocord) against which the opposite and normal true vocal cord vibrates. Although a variety of radiation therapy techniques for early-stage lesions have been reported, the most commonly used technique is opposed lateral fields. The patient is simulated in the supine position with maximal head extension. A head cast is made to ensure immobilization, and small portals are designed that treat the larynx alone. Field size is an important prognostic factor. The local recurrence rate is substantially higher for those patients whose field size is 25 cm**2 or larger. Actually, it would seem that field size is best at 5.5 x 5.5 cm to 6 x 6 cm. Another important factor is the daily fractionation schedule. Fraction sizes of 200 cGy or greater should be used. Fraction sizes less than 200 cGy per day should be avoided, because the local recurrence rate is higher when this is done. Rudoltz [ref: 171] reported 91 patients with T1N0M0 glottic cancer with median follow-up of 9 years. Patients treated at less than 2 Gy per fraction had a local control rate of 62%, versus 87% for those treated with at least 2 Gy per fraction. The most common strategy is to deliver 66 Gy in 33 fractions of 2 Gy over 6.5 weeks. A larynx contour should be taken during simulation, and a dose distribution should be evaluated. A larynx wedge is added to keep dose in homogeneity to within 5% to 10% of the reported satisfactory results with 6-mV photons as well, as long as adequate planning is done. Great caution should be used, however, in using 6-mV photons for lesions involving the anterior commissure. Glottic cancers that cause vocal cord or hemilaryngeal immobilization or fixation (i.e., T3) are substantially more ominous than lesions that do not. At a minimum, such motion impairment reflects vocalis muscle invasion; however, there is often further tumor extension into the paraglottic space, opening the entire larynx and adjacent exit areas to cancer spread. Many of these lesions are associated with thyroid or cricoid cartilage destruction, and there is a tendency to underestimate glottic lesions with vocal cord fixation. In Olofsson’s study, [ref: 62] a significant number of tumors originally had been understaged as T3 cancers but were actually T4 lesions with extralaryngeal extension. With the unpredictable extent of the disease and the overall high probability of cartilage destruction in mind, surgery, which usually consists of a total laryngectomy, traditionally has been favored over radiation therapy for the primary management of T3 lesions. The overall 5-year survival rate for T3 glottic cancers treated with total laryngectomy is reported to be 55% to 72% in four major series. [ref: 67,134] However, it has been shown by Kirchner and Som, [ref: 173] Marchetta, [ref: 174] and Martensson [ref: 175] that in carefully selected vocal cord lesions that are clearly limited to the vocalis muscle, and in which there is no associated cartilage destruction, hemilaryngectomy can be performed with oncologic safety. In their series of 22 patients, they demonstrated a 60% cure rate. In another similar study, Som reported a 58% curability in 26 patients with selected T3 glottic tumors treated by partial laryngectomy. [ref: 176] It should be noted that both of these significant studies were done before the use of CT or MRI. Such technology would have made the evaluation of these lesions and the delineation of their dimensions easier and more accurate. Even with this limitation lessened by contemporary technology, the reader should not be led to underestimate the difficulty of judging correctly the small subset of T3 lesions that are suitable for hemilaryngectomy. This is a judgment appropriately left only to those surgeons with considerable experience in partial laryngeal surgery. Although there seems to be a deserved preference for primary surgery in the management of T3 glottic cancers, there also appears to be a subset of patients who can be cured without operation. Van den Bogert and coworkers [ref: 177] reported a local control rate of 53% in a group of patients with operable T3 lesions who had refused surgery. Harwood and colleagues reported a 45% 5-year local control for T3 glottic patients, [ref: 178] and Wang reported a 36% 5-year actuarial local control, which increased to 57% when surgical salvage was added. [ref: 179] There is a suggestion of improvement of these results with hyperfractionated radiation therapy. In his group of 53 T3 glottic cancer patients, Mendenhall and coworkers reported a trend toward improved local control in patients treated with twice daily radiation. [ref: 180] In this group, local control was obtained in 71% compared with 53% with once daily treatment. Terhaard and associates [ref: 181] noticed the same trend as Mendenhall and coworkers, reporting an actuarial 3-year local control of 53% overall, but 67% in the subset of patients who were treated with twice daily radiation therapy. These particular studies suggest that twice daily radiation may control between 60% and 70% of patients with T3 glottic cancer. These data need further maturation and investigation. Glottis (Continued) (2 of 4) There has been some interest in using radiation therapy itself to select those patients who might have radioresponsive tumors. In this strategy, radiation therapy is delivered up to a total dose of 4000 to 5000 cGy. At that point, the patient is critically reassessed. If there has been significant regression of disease with return of vocal cord mobility, then definitive radiation is continued. If the tumor has not responded satisfactorily, however, radiation is stopped, and a laryngectomy is performed. Terhaard and associates reported success using this method. [ref: 181] Mendenhall and coworkers, [ref: 180] however, found no correlation between the return of vocal cord mobility and eventual local control. T4 tumor designation denotes cartilage destruction or extension of the tumor outside of the confines of the larynx. This grouping together of all T4 lesions is misleading, however, because it fails to account for the more favorable tumor of lesser thickness and volume that has achieved a T4 status only because it originated in a marginal zone and extended to an adjacent area, such as pyriform sinus or base of the tongue. Just as with T3 supraglottic lesions, there are favorable and unfavorable T4 lesions, and that determination is made appropriately on the basis of tumor volume, depth, and thickness. With this exception in mind, the performance of the group of T4 glottic lesions is expectedly poorer than that of lesser stages. Traditionally, most of these lesions are treated with total laryngectomy and some sort of a neck dissection, and depending on the status of the neck nodes, they may or may not receive postoperative radiation. The concept of organ preservation by induction chemotherapy identifying those lesions likely to respond to radiation therapy is a recent addition to the treatment armamentarium. Those favorable results of the nonrandomized trials and the results of the Veterans Administration Cooperative Study Group [ref: 1-3,182,183] include as part of the studies a number of patients with T4 lesions. Induction of chemotherapy-integrated larynx preservation protocols are now practical rather than merely investigational, and while we must exercise caution in replacing standard and time-honored therapy with new strategies, the concept of organ preservation is today widely accepted as appropriate therapy in selected larynx and hypopharynx cancers. The unfavorable subgroup of advanced lesions that have destroyed cartilage has been analyzed by Jesse, [ref: 184] who demonstrated an ominous pattern; of the 48 patients, 6% had distant metastasis at presentation, 39% had cervical metastasis, and 30% required an emergency tracheostomy at the time of diagnosis. In Jesse’s series, using total laryngectomy and radiation in some patients, the 4-year cure rate was 54%; however, in that subset of patients who required tracheostomy or who had pharyngeal wall extension, the survival rate decreased to 38%. Other studies have demonstrated 5-year survival rates of 25% to 30% in the T4 subset of larynx cancer patients. [ref: 169,170] Harwood and colleagues [ref: 178] reported local control in 56% of T4, N0 glottic lesions treated with radical radiation therapy. The five-year survival rate for radiation with surgical salvage was 49%. Within this group, the local control rate for patients classified as T4 from cartilage invasion was 67% at 5 years. This was significantly better than for those T4 patients who had pyriform sinus involvement. In this latter group, the actuarial local control rate was only 19%. It does not seem that the T4 patients with hypopharyngeal involvement are good candidates for radical radiation therapy. This circumstance parallels the dismal performance of a similar group treated surgically and reported by Jesse. [ref: 184] It also is similar to a comparably poor performance for those larynx cancer patients with hypopharyngeal extensiooted by Pfister and colleagues [ref: 2] in their larynx preservation (induction chemotherapy) study. The same trend of poor performance for those larynx cancer patients with hypopharyngeal extension was noted by Karp and coworkers. [ref: 3] Whatever the treatment variation, these patients consistently seem to do poorly. In those patients with T3 and T4 cancers of the glottis and other laryngeal sites who are destined for laryngectomy, induction chemotherapy has been used recently in what are collectively referred to as the larynx preservation protocols. Generally speaking, organ preservation trials are designed to improve quality of life by preserving a critical function such as natural speech or swallowing while achieving survival rates that are at least comparable with standard surgical or radiotherapy approaches. The conceptual schema for organ preservation is shown in Figure 29.3-8. A number of feasibility pilot trials have been published using cisplatin-based neoadjuvant chemotherapy, and some are listed in Table 29.3-4. [ref: 2,3,182,185-188] Jacobs [ref: 182] was the first to demonstrate that organ preservation was feasible with neoadjuvant chemotherapy followed by radiation in those patients who achieved a complete response to chemotherapy at the primary site. Nodal metastases were treated surgically when indicated. A 40% organ preservation rate was reported as well as a 2-year survival rate of 53%, which compared favorably with historic controls. Demard and coworkers [ref: 185] treated 71 patients with hypopharyngeal or laryngeal primaries (20 stage II, 61 stage III and IV) with induction chemotherapy. Clinically complete responders received radiation alone for organ preservation. This occurred in 52% and was much higher for T2 patients. Two-year survival rates for patients with primary tumors of the larynx and hypopharynx were 93% and 69%, respectively. Survival rates for nonresponders undergoing primary resection were 66% and 40%. Karp [ref: 3] was able to achieve organ preservation in 57% of patients with glottic and supraglottic primaries and a 50% 2-year survival rate. These early results have been duplicated in more recent trials. [ref: 186-189] Clayman [ref: 186] reported the experience from M.D. Anderson between 1986 and 1991 using induction chemotherapy to preserve the larynx. Fifty-five patients with stage III and IV cancer of the glottis (9 patients), supraglottis (17 patients), or pyriform sinus (29 patients) received two to three cycles of cisplatin and 5-fluorouracil with or without bleomycin. Complete and partial response rates after chemotherapy were comparable for larynx and hypopharynx primaries (75% and 78%, respectively), as was the complete response rate after radiotherapy (88% and 83%, respectively). The laryngeal preservation rates were 65% (17 of 26 patients) for laryngeal primaries and 69% (20 of 29 patients) for hypopharyngeal primaries. Comparisons of patients who did and did not achieve laryngeal preservation for each site with matched surgical controls showed nearly identical survival results. For both laryngeal and hypopharyngeal primaries, local control was significantly better for the surgery controls; differences in the distant metastatic rate did not reach statistical significance. Only five patients in this series had T4 primaries, and none survived. The authors concluded that laryngeal preservation was feasible and did not compromise survival for intermediate stage T2 and T3 squamous cell carcinomas of the larynx and hypopharynx. Glottis (Continued) (3 of 4) At the University of Michigan, Urba and colleagues [ref: 187] tested an intensified induction chemotherapy regimen in patients with locally advanced disease from any site in the head and neck. Patients who were downstaged to T1N1 were treated with definitive radiotherapy. Initial surgery was avoided in 69%, and 40% remained disease-free at 38.5 months of follow-up. Laryngeal preservation was achieved in 8 of 8 patients with laryngeal primaries and 7 of 10 with hypopharyngeal primaries. Survival rates were the same as those achieved by historic controls treated with surgery and radiation. Investigators at Memorial Sloan-Kettering Cancer Center reported their experience using induction chemotherapy to preserve speech or swallowing function in patients with cancers of the larynx, [ref: 2] hypopharynx, [ref: 188] and tongue. [ref: 189] For larynx cancer, Pfister and coworkers [ref: 2] observed a 68% laryngeal preservation rate and a 58% 2-year survival rate using high-dose cisplatin combined with vinblastine or bleomycin. Kraus and colleagues [ref: 188] extended the same concept to the treatment of 25 patients with advanced, operable hypopharyngeal carcinoma. Laryngeal preservation was achieved in 32%, and the 2-year survival rate was 44%. Neck dissection was performed after induction chemotherapy prior to definitive radiotherapy for those patients with substantial nodal involvement. These nonrandomized trials confirm the feasibility of induction chemotherapy followed by definitive radiation to preserve speech and swallowing function for patients with T2, T3, and T4 laryngeal cancer and T2 and T3 hypopharyngeal cancer. These data suggest that survival is uncompromised. Three large multicenter randomized trials evaluating laryngeal preservation for advanced, operable cancers of the larynx or hypopharynx (Table 29.3-5) have been completed. In 1985, the Veterans Administration Cooperative Studies Program launched a landmark trial in which 332 patients with stage III or IV (M0) cancers of the glottis or supraglottis were randomly assigned treatment with surgery (total laryngectomy) and radiotherapy or induction chemotherapy and radiotherapy. [ref: 190] In the experimental arm, responders after 2 cycles of cisplatin and 5-FU received a third cycle of chemotherapy followed by definitive radiotherapy reserving laryngectomy for salvage. Patients demonstrating less than a partial response to two cycles of chemotherapy underwent immediate laryngectomy. The results of this trial indicate that sequential chemotherapy using cisplatin and 5-FU followed by definitive radiotherapy can be an effective strategy for preserving natural voice in a high percentage of patients without compromising survival. No differences in survival have been observed with over 5 years of follow-up. The larynx preservation rate was 64%, and the 2-year survival rate in both treatment groups was 68% after a median follow-up of 33 months. The 3-year survival rates published subsequently were 53% and 56% for the chemotherapy and surgery groups, respectively, after a mean follow-up of 60 months. At that time, the laryngeal preservation rate was 66% in surviving patients. [ref: 183] The disease-free interval was shorter for chemotherapy-treated patients, but with prompt surgical salvage no differences were observed in survival rates. Other important outcomes from this trial were the following: (1) Smaller tumor size and growth pattern (pushing borders or well-formed infiltrating cords) were significant predictors of complete response to chemotherapy. [ref: 191,192] (2) Disease-free survival was significantly better among patients achieving complete response. [ref: 191] (3) Local failure rates were significantly higher in the chemotherapy group and the distant metastatic rate significantly lower; however, causes of death (locoregional failure or distant disease) were nearly identical among patients in both groups. [ref: 190] (4) The survival of chemotherapy nonresponders did not differ from patients initially randomized to surgery. [ref: 191] (5) The only factor associated with a need for salvage surgery was N2 or N3 neck disease that was not in complete response after chemotherapy. [ref: 193] Other investigators [ref: 194] are currently evaluating the efficacy of these same chemotherapy and radiation strategies as they apply to neck disease, and it may ultimately be helpful to our ability to improve management of this aspect of head and neck aerodigestive squamous cancer. These results raised a number of questions to be addressed in future trials. The precise contribution of chemotherapy cannot be assessed without a direct comparison to radiotherapy alone as an alternative organ preservation strategy, particularly for patients with T3N0 cancers. Because local control was inferior in chemotherapy-treated patients and survival was not improved, the strategy of concomitant chemotherapy and radiatioeeds to be explored as a treatment with potential for improving these outcomes. Finally, surgical management of the neck for patients with N2 or N3 disease may reduce the need for salvage surgery and improve survival. [ref: 193] These issues are being addressed in the current head and neck laryngeal preservation intergroup trial (R91-11). [ref: 195] This randomized trial is for patients with locally advanced squamous cell carcinoma of the glottic and supraglottic larynx. T1 lesions and large T4 lesions (tumor eroding cartilage or extending greater than 1 cm into the tongue base) are excluded. Radiation alone (70 Gy), and concomitant radiation (70 Gy) plus cisplatin (100 mg/m**2 on days 1, 22, and 43) are the two experimental arms that are being compared with a control arm of sequential chemotherapy and radiotherapy that is identical to that used in the VA Cooperative Studies Program. Patients in all three treatment arms will undergo elective neck dissection for nodal disease initially staged as N2 or N3. In summary, trials to date demonstrate that induction chemotherapy and radiation are alternative treatments for patients with some locally advanced cancers of the glottis and supraglottis. This treatment must be performed with close monitoring by a head and neck surgeon so that surgery can be performed promptly ionresponders to chemotherapy and for salvage of recurrent disease. Regarding the feasibility of treatment by concomitant chemotherapy and radiation strategies, there are several such endeavors worth noting. Reports of pilot trials of concomitant chemotherapy and radiation, sometimes preceded by several cycles of neoadjuvant chemotherapy, indicate feasibility and high rates of locoregional control [ref: 196-199] (Table 29.3-6). Mucocutaneous toxicity is often severe, requiring aggressive nutritional and analgesic support. Most of these trials were initially conducted in patients with unresectable disease of sites other than the larynx or hypopharynx (see Chap. 29.2) and later expanded to preserve function in patients with resectable disease of the oral cavity, oropharynx, larynx, or hypopharynx. Thus, rates of organ preservation, quality of function (speech and swallowing), and acute and late toxicity data are available on only small numbers of patients with laryngeal or hypopharyngeal primaries treated with any one particular regimen. One trial reported by Koch and colleagues [ref: 196] was specifically designed for patients with resectable squamous cell cancers of the oral cavity, oropharynx, or hypopharynx for whom surgery would significantly compromise speech or swallowing function. Twenty-two patients were treated with split-course standard fractionation radiotherapy, 70 Gy, weekly carboplatin employing pharmacokinetic dosing, and cisplatin, 100 mg/m**2 every 3 weeks. The rationale was to double exposure to the radiosensitizing effects of platinum using these two agents with nonoverlapping toxicities. At the completion of treatment, 91% (20 of 22) of patients had a pathologic complete response at the primary, and organ preservation was achieved in 68% at 2 years. Myelosuppression, mucositis, and nutritional complications were the major acute toxicities encountered. Glottis (Continued) (4 of 4) Leyvraz and colleagues [ref: 197] evaluated a more intensive regimen of alternating courses of hyperfractionated radiotherapy, total dose 48 to 60 Gy, and cisplatin and infusional 5-FU in a phase I/II trial. Sixty-nine of 91 patients were treated for organ preservation (initially resectable), and 33% (30 of 91) of patients had hypopharyngeal or laryngeal primaries. At a median follow-up of 45 months, the organ preservation rate was 64%. Locoregional failures all occurred within 15 months of treatment. Leukopenia and mucositis were dose-limiting toxicities; late toxicities were not considered different from that expected with radiation alone. Glicksman and coworkers [ref: 198] reported a series of 101 resectable patients of whom 36 were treated nonsurgically to preserve organ function. Eighteen of these had primaries of the larynx or hypopharynx and were treated with standard fractionation radiotherapy, 70 Gy, and continuous infusion cisplatin, 20 mg/m**2 for 96 hours every 3 weeks for 3 courses. Patients managed surgically received 45 Gy and 2 courses of infusional cisplatin preoperatively with or without additional cisplatin, radiotherapy, or both after resection. In the resected group, 82% of primary specimens and 86% of nodal specimens contained no residual tumor, supporting the notion that these patients might be curable without surgery. At a median follow-up of 41 months, the actuarial 5-year survival rate was 49% and did not differ between surgically treated and nonsurgically treated patients. Vokes and colleagues [ref: 199] administered three cycles of induction chemotherapy (cisplatin, 5-FU, leucovorin, and interferon) followed by optional organ-sparing surgery and seven or eight cycles of 5-FU, hydroxyurea, and concurrent radiotherapy in a phase I and II trial. The goal was to improve locoregional control with combined chemoradiotherapy and decrease distant metastases with the induction chemotherapy. Seventy-one patients were treated (26 with larynx or hypopharynx primaries). The survival rate at 3 years was 60% for a cohort of 57 patients treated with phase II doses of the protocol. Both local and distant failure rates were lower than observed in prior studies of these investigators. The treatment, however, was associated with an excessive amount of life-threatening toxicity. These investigators are continuing to pursue this approach with modifications to reduce treatment duration and toxicity. Taken together, the results of these pilot chemoradiotherapy trials suggest that this strategy for organ preservation produces survival rates that are at least as good as and perhaps better than surgery. In theory, improved locoregional control, which is suggested by these trials, should favorably impact on survival. Site-specific randomized trials, such as the intergroup laryngeal preservation trial in progress [ref: 195] will be necessary to establish benefit over less toxic organ-preserving therapy with sequential chemotherapy and radiotherapy or radiotherapy alone. In summary, the selection of a treatment strategy for T4 glottic cancers is dependent on a variety of factors, including the following: 1. If there is bulky neck disease, convention dictates laryngectomy, neck dissection, and postoperative radiation therapy, with both necks being included in the treatment plan. 2. If there is bulky neck disease and if the primary tumor does not have extensive hypopharyngeal extension, induction chemotherapy followed by radiation therapy to primary and neck can be employed in chemosensitive tumors. This would be followed by a neck dissection. In such lesions that are not chemoresponsive, laryngectomy and neck dissection plus postoperative radiation therapy would be done. 3. If there is substantial hypopharyngeal extension, a conventional approach with laryngectomy or laryngopharyngectomy plus neck dissection and postoperative radiation therapy is recommended. 4. If there is no clinical neck disease present (i.e., T4N0), induction chemotherapy followed by radiation therapy to primary and necks in chemoresponsive tumors should be considered. If such a lesion fails to respond adequately to induction chemotherapy, indicating the probability of relative radioresistance, then the logical course should be a laryngectomy. The laryngectomy should probably be accompanied by appropriate staging, selective neck dissection, [ref: 200] and, depending on the result of the histologic analysis of the neck nodes harvested, postoperative radiation therapy. 5. In these large lesions, especially lesions that demonstrate subglottic extension, radiation must be directed to the tracheal stoma, because recurrence here can be seen in a substantial percentage of these patients. Other indications for postoperative radiation are cartilage and extensive soft tissue invasion. Most of these strategies for treating the patient with advanced larynx or hypopharynx cancer and clinically positive necks must be analyzed in light of the dynamic state of our knowledge and experience with the organ preservation concepts. Hitherto, we have been comfortable with the notion that substantial neck disease usually required surgery of some sort at some stage in the treatment plan. Armstrong and coworkers, [ref: 194] however, have recently published early and very limited data that are an extension of the concept of larynx preservation and that suggest that patients with clinically palpable cervical nodal metastases who have a complete response to chemotherapy and who receive high-dose radiation therapy have excellent neck control and may not need neck dissection. To address this issue, Armstrong reviewed the neck management on the first 80 patients treated on the larynx preservation trials at Memorial Sloan-Kettering Cancer Center. Of these, 54 patients presented with clinically positive nodes. Of these, 44% had a complete response and 20% had a partial response in the neck to induction chemotherapy. In 63% of the major responders, surgery to the neck was omitted, and radiation alone was used. Neck control was obtained in 91%, with only 1 in 17 complete responders and 1 of 5 partial responders exhibiting neck failure. These data suggest that major responders to induction chemotherapy may possibly avoid neck dissection. These data require supplementation and maturation, and data for concomitant chemotherapy and radiotherapy must also be generated in order to decide if this observation is sustained. Even if these preliminary suggestions are substantiated, however, the definitive question to be asked is whether the morbidity endured with high-dose radiation is justified when moderate dose neck radiation plus a tailored neck operation is so well tolerated. These creative newer strategies for the management of the neck provide a fertile area for clinical investigation. Subglottis Although the preponderance of subglottic tumors are squamous cell carcinomas, adenocarcinomas and adenoid cystic carcinomas are seen occasionally. [ref: 84] The cure rate for these tumors is poor, despite combination therapy. Total laryngectomy and appropriate neck surgery, including thyroidectomy, are probably the surgical treatment most recommended, and they should be followed by radiation therapy. [ref: 201,202] Mediastinal dissection of the paratracheal nodal groups does not seem to add substantially to survival rates. [ref: 84] Considerations for Neck Surgery There are a variety of neck operations currently used in the various treatment plans for laryngeal cancer: the classic radical neck dissection, the modified radical neck dissection, and a group of regional dissections collectively known as selective neck dissections. This group of operations has been standardized and endorsed by the American Academy of Otolaryngology-Head and Neck Surgery, the American Society for Head and Neck Surgery, and the Society for Head and Neck Surgeons; a monograph outlining the recommended terminology should be studied, and oncologists are strongly urged to incorporate this classification into their lexicon in order to facilitate interinstitutional data recording and appropriate comparisons. [ref: 200] There is considerable diversity of opinion in the surgical community as to which of these operations should be applied to which situation. Exact guidelines are not consistently substantiated by the data available. Generalizations are possible, however. The radical neck dissection is almost never used in the clinically negative neck, even though the probability for metastasis is extraordinary. This procedure is usually reserved for necks in which there is gross metastasis. The selective neck dissections often are applied when there is gross disease, but more often they are used as staging procedures in which the goal is to harvest and sample the nodal groups at highest risk for metastasis. This philosophy has evolved as a result of our increasing reliance on radiation therapy as the second half of neck treatment whenever disease is discovered in the neck. The choice of the appropriate selective neck dissection in larynx cancer is based on a knowledge of the consistent patterns of metastasis that are exhibited by the various subsites within the larynx; for example, lesions of the supraglottis are prone to metastasis bilaterally and to levels II and III. However, they rarely spread to level I of the neck. [ref: 33] Based on this knowledge, the logical staging operation for a supraglottic laryngeal cancer with a clinically negative neck would be tailored specifically to clean out those various compartments. This is but one example of this tailored approach, and the reader is referred to the classic work by Lindberg [ref: 35] in which he mapped the patterns of metastases of the various aerodigestive squamous cell carcinomas. More recently, Shah and colleagues [ref: 203] have applied the modern classification to this process of selectivity and have established recommendations for the use of these neck operations.

 

LARYNGEAL REHABILITATION The philosophy that emphasizes a maximal rehabilitative effort to return the patient to a functional role in society is exemplified vividly in the efforts currently being expended on vocal restoration after total laryngectomy. When laryngectomy is done, the normal method of vocal communication is lost. Since the first total laryngectomy by Billroth, [ref: 204] an ideal method of artificially providing voice has been sought. [ref: 205-214] Until recently, most laryngectomy patients have had to rely on mechanical vibratory devices, esophageal speech, or written communication. Only about 20% to 40% of patients master esophageal speech. [ref: 215] It is those postlaryngectomy patients with ineffective or no esophageal speech who are least likely to reenter their social and family environments as full participants.

 

Even laryngectomy patients who have mastered esophageal speech are hampered by certain mechanical problems and stigmas that create difficulty with their ability to communicate well. Even when mastered, esophageal speech produces only short sentences or monotonal words of limited volume, and its production requires swallowing techniques that are often accompanied by facial grimacing, distortions, and embarrassment. The electrovibratory devices are helpful for intermittent use during the immediate postoperative period and as a back-up system, but they are of limited value because one hand is occupied during their use (Fig. 29.3-9). With these instruments, the voice is mechanical and often distracting, and it is difficult for the user to be heard in a noisy environment. The state of the art of vocal rehabilitation involves the employment of air shunting from the trachea to the neogullet, thereby setting up a vibratory column that produces noise that comes out of the mouth where it is articulated as speech. This is accomplished by a tracheoesophageal puncture (TEP) and placement of a silicone valve-like device that is structured to allow air into the neogullet but not to allow food or liquids out (Fig. 29.3-10). What is produced is a form of esophageal speech and voice, but one that is vastly superior to the traditional method of swallowing and burping. The sounds produced are less monotonal, the volume of air is limited only by the inspiratory lung volume capacity (just as normal speech is) rather than by how much air is swallowed and burped, and the product has a more fundamental resemblance to normal voice than other methods. After the establishment of an appropriate fistula and the placement of the TEP prosthesis, a tracheostoma valve and external housing device is used (see Fig. 29.3-10). This device allows normal breathing but shunts the air into the underlying TEP prosthesis. With the increased airflow of voice production, air is routed into the esophagus and up the neogullet that vibrates and causes sound that comes out of the mouth as articulated speech. When an external valve is placed over the stoma with the TEP in place, the final product is a laryngectomized person who usually has exceptional speech with normal articulation and who can dress with clothing that allows complete coverage of the device. [ref: 216] Patients can return to work and a relatively normal social life with a functional method of communication. This method effectively achieves voice in about 80% to 90% of patients. [ref: 217] There are other surgical methods of vocal rehabilitation, [ref: 218,219] but none with the extensive record of clinical application and success achieved by the tracheoesophageal phonation as developed and popularized by Singer and Blom. [ref: 216] The TEP procedure can be done with equal results at the time of laryngectomy (primary TEP) or as a secondary procedure after radiation. The matter of alaryngeal rehabilitation should be undertaken in concert between the head and neck surgeon and the speech pathologist. The patient who has been rushed to laryngectomy without adequate preoperative preparation (i.e., education, counseling) is usually more difficult to rehabilitate. On the other hand, the patient who is well prepared usually achieves successful speech rehabilitation. It is our approach to take the patient methodically through a series of experiences after the determination is made that a laryngectomy is necessary. The laryngectomy patient should have multiple communication options; therefore, the authors endeavor to teach them how to use the electrovibratory device (see Fig. 29.3-9), esophageal speech, and TEP phonation (in a selected subset). The speech pathologist is involved at the outset, giving an in-depth description of the anatomy, the operation, and the proposed plan for rehabilitation. The team relies heavily on video demonstrations of the whole subject and on patient volunteers who have had similar procedures. At this point, an attempt is made to incorporate family members in the instructional sessions, because a constructive support system contributes to a successful outcome. Additionally, the patient is taught how to use the electrovibrating device before surgery so that he or she will be able to communicate during convalescence in the hospital. Contacts are also made with the American Cancer Society and various laryngectomy clubs. During this preoperative educational period, the initial training for esophageal speech is begun. After laryngectomy, the speech pathologist visits the patient in the hospital and encourages the use of the mechanical vibrating devices. The patient’s perception of a continuum of rehabilitative effort is well served by this comprehensive approach in which the various members of the team communicate and function toward preestablished goals. There is an ongoing controversy regarding primary versus secondary TEP. The authors favor the latter; however, others believe the procedure should be done at the time of the laryngectomy. The primary approach shortens the time of rehabilitation and lessens overall expense, and according to the advocates of this approach, the patients have more immediate positive reinforcement in the critical perioperative period. On the other hand, the advocates of the secondary placement of the TEP argue that by delaying the procedure, more time is allowed for the patient to adapt to and learn to care for the new laryngectomy stoma, the wound is allowed to mature, postoperative radiation therapy is completed, and the patient is allowed to begin learning skills of esophageal speech. The authors believe that the motivation to learn esophageal speech is lessened when a TEP is in place. If the patient is properly prepared psychologically and educationally, this orderly step-by-step approach to rehabilitation is more efficient. Finally, healing is a dynamic phenomenon, and the precise placement and angle of the prosthesis are better ensured by a secondary or delayed placement. There are circumstances that make patient selection critical to the success of rehabilitation with TEP. To function at a practical level of phonation, the patient must have the cognitive skills and the dexterity necessary to care for the stoma and the TEP site. Patients who have had laryngopharyngectomies with reconstruction of the gullet have a somewhat unpredictable tracheoesophageal voice rehabilitation. Those patients who undergo free jejunal interposition procedures have a somewhat better and more predictable voice than those who have had a gastric transposition for reconstruction. The experience in voice rehabilitation in both of these groups is limited, and the vocal results achieved to date must be viewed as preliminary. Whether employed primarily or secondarily, the enormous contribution made to the laryngectomy patient by the current methods of TEP phonation should not be underrated. There is a considerable experience currently with this method, and as the plastics technology continues to improve, the process of this type of vocal rehabilitation should be enhanced. It is hoped that the reader appreciates that the current concern among oncologists for quality of life is reflected by the considerable effort being devoted to saving the larynx with conservation operations, innovative radiation strategies, larynx-sparing chemoradiotherapy protocols, and, when these methods fail and laryngectomy is required, a vigorous and thoughtful effort to communication rehabilitation.

 

Prepared by Prof. Igor Galaychuk, MD,

2011

 

Leave a Reply

Your email address will not be published. Required fields are marked *

Приєднуйся до нас!
Підписатись на новини:
Наші соц мережі