National Cancer Institute National Cancer Institute
U.S. National Institutes of Health National Cancer Institute
Send to Printer
Unusual Cancers of Childhood Treatment (PDQ®)     
Last Modified: 12/02/2008
Health Professional Version
Table of Contents

Purpose of This PDQ Summary
General Information
Head and Neck Cancers
Nasopharyngeal Carcinoma
        Treatment options under clinical evaluation
Esthesioneuroblastoma
Thyroid Tumors
Oral Cancers
Salivary Gland Tumors
Laryngeal Cancer and Papillomatosis
Respiratory Tract Carcinoma Involving the NUT Gene on Chromosome 15
Thoracic Cancers
Breast Cancer
Bronchial Tumors
Pleuropulmonary Blastoma
Esophageal Tumors
Thymoma and Thymic Carcinoma
Tumors of the Heart
Mesothelioma
Abdominal Cancers
Carcinoma of the Adrenal Cortex
        Treatment options under clinical evaluation
Carcinoma of the Stomach
Cancer of the Pancreas
Colorectal Carcinoma
Carcinoid Tumors
Gastrointestinal Stromal Cell Tumor
Genital/Urinary Tumors
Carcinoma of the Bladder
Ovarian Cancer
Carcinoma of the Cervix and Vagina
Other Rare Childhood Cancers
Multiple Endocrine Neoplasia Syndrome
        Treatment options under clinical evaluation
Skin Cancer (Melanoma, Basal Cell, and Squamous Cell Carcinoma)
        Treatment options under clinical evaluation
Chordoma
Cancer of Unknown Primary Site
Get More Information From NCI
Changes to This Summary (12/02/2008)
More Information

Purpose of This PDQ Summary

This PDQ cancer information summary for health professionals provides comprehensive, peer-reviewed, evidence-based information about the treatment of unusual cancers of childhood. This summary is reviewed regularly and updated as necessary by the PDQ Pediatric Treatment Editorial Board 1.

Information about the following is included in this summary:

  • Incidence of unusual childhood cancers.
  • Treatment options for unusual childhood cancers.

This summary is intended as a resource to inform and assist clinicians and other health professionals who care for pediatric cancer patients. It does not provide formal guidelines or recommendations for making health care decisions.

Some of the reference citations in this summary are accompanied by a level-of-evidence designation. These designations are intended to help readers assess the strength of the evidence supporting the use of specific interventions or approaches. The PDQ Pediatric and Adult Treatment Editorial Boards use a formal evidence ranking system 2 in developing their level-of-evidence designations. Based on the strength of the available evidence, treatment options are described as either “standard” or “under clinical evaluation.” These classifications should not be used as a basis for reimbursement determinations.

This summary is also available in a patient version 3, which is written in less-technical language, and in Spanish 4.

General Information

The National Cancer Institute (NCI) provides the PDQ pediatric cancer treatment information summaries as a public service to increase the availability of evidence-based cancer information to health professionals, patients, and the public.

Cancer in children and adolescents is rare. Children and adolescents with cancer should be referred to medical centers that have a multidisciplinary team of cancer specialists with experience treating the cancers that occur during childhood and adolescence. This multidisciplinary team approach incorporates the skills of the primary care physician, pediatric surgical subspecialists, radiation oncologists, pediatric medical oncologists/hematologists, rehabilitation specialists, pediatric nurse specialists, social workers, and others to ensure that children receive treatment, supportive care, and rehabilitation that will achieve optimal survival and quality of life. (Refer to the PDQ Supportive Care 5 summaries for specific information about supportive care for children and adolescents with cancer.)

Guidelines for pediatric cancer centers and their role in the treatment of pediatric patients with cancer have been outlined by the American Academy of Pediatrics.[1] At these pediatric cancer centers, clinical trials are available for most types of cancer that occur in children and adolescents, and the opportunity to participate in these trials is offered to most patients/families. Clinical trials for children and adolescents diagnosed with cancer are generally designed to compare potentially better therapy with therapy that is currently accepted as standard. Most of the progress made in identifying curative therapies for childhood cancers has been achieved through clinical trials. Information about ongoing clinical trials is available from the NCI Web site 6.

In recent decades, dramatic improvements in survival have been achieved for children and adolescents with cancer. Childhood and adolescent cancer survivors require close follow-up because cancer therapy side effects may persist or develop months or years after treatment. (Refer to the PDQ Late Effects of Treatment for Childhood Cancer 7 summary for specific information about the incidence, type, and monitoring of late effects in childhood and adolescent cancer survivors.)

The tumors discussed in this summary are diverse; the discussion is arranged in descending anatomic order, from infrequent tumors of the head and neck to rare tumors of the urogenital tract and skin. All of these cancers are rare enough that most pediatric hospitals might see fewer than two in a year. Most of these tumors are more frequent in adults with cancer; thus, much of the information about these tumors may also be sought through sources relevant to adults with cancer.

References

  1. Guidelines for the pediatric cancer center and role of such centers in diagnosis and treatment. American Academy of Pediatrics Section Statement Section on Hematology/Oncology. Pediatrics 99 (1): 139-41, 1997.  [PUBMED Abstract]

Head and Neck Cancers

Note: Some citations in the text of this section are followed by a level of evidence. The PDQ editorial boards use a formal ranking system to help the reader judge the strength of evidence linked to the reported results of a therapeutic strategy. (Refer to the PDQ summary on Levels of Evidence 2 for more information.)

Head and neck cancers include nasopharyngeal carcinoma, esthesioneuroblastoma, thyroid tumors, mouth cancer, salivary gland cancer, laryngeal carcinoma, papillomatosis, and respiratory tract carcinoma involving the NUT gene on chromosome 15. The prognosis, diagnosis, classification, and treatment of these head and neck cancers are discussed below.

Nasopharyngeal Carcinoma

Nasopharyngeal cancer arises in the lining of the nasal cavity and pharynx.[1,2] This tumor accounts for about one-third of all cancers of the upper airways. The incidence of this tumor is approximately 1 in 100,000 persons younger than 20 years in the United States.[3] There is a higher frequency of this tumor in North Africa and Southeast Asia.

Nasopharyngeal carcinoma occurs in association with Epstein-Barr virus (EBV), the virus associated with infectious mononucleosis.[4] The virus can be detected in biopsy specimens of these cancers, and tumor cells can have EBV antigens on their cell surface. Three histologic subtypes are recognized by the World Health Organization. Type 1 is squamous cell carcinoma, type 2 is nonkeratinizing carcinoma, and type 3 is undifferentiated carcinoma.

This cancer most frequently spreads to lymph nodes in the neck, which may alert the patient, parent, or physician to the presence of this tumor. The tumor may also spread to the nose, mouth, and pharynx, causing snoring, epistaxis, obstruction of the eustachian tubes, or hearing loss; it may also invade the base of the skull, causing cranial nerve palsy or difficulty with movements of the jaw (trismus). Distant metastatic sites may include the bones, lungs, and liver. The location of the primary tumor can be made by direct inspection of the nasopharynx. A diagnosis can be made from a biopsy of the primary tumor or of enlarged lymph nodes of the neck. Nasopharyngeal carcinomas must be distinguished from all other cancers that can present with enlarged lymph nodes and from other types of cancer in the head and neck area. Thus, diseases such as thyroid cancer, rhabdomyosarcoma, non-Hodgkin lymphoma, Hodgkin lymphoma, and Burkitt lymphoma must be considered, as should benign conditions such as nasal angiofibroma, which presents with epistaxis, and infections draining into the lymph nodes of the neck.

Diagnostic tests should determine the extent of the primary tumor and whether there are metastases. Visualization of the nasopharynx by an ear-nose-throat specialist using a mirror, examination by a neurologist, and magnetic resonance imaging of the head and neck can be used to determine the extent of the primary tumor. Evaluation of the chest and abdomen by computed tomography and bone scan should also be performed to determine whether there is metastatic disease. The levels of EBV and antibody to EBV should also be measured.[1,5]

Tumor staging is performed utilizing the tumor-node-metastasis classification system of the American Joint Committee on Cancer (AJCC).[6] The majority (>90%) of children and adolescents with nasopharyngeal carcinoma present with advanced disease (stage III/IV or T3/T4).[7] Metastatic disease at diagnosis is uncommon (stage IVC). Outcome is directly related to the stage of the disease, with overall survival ranging from 80% for stage I and stage II to 40% for stage III.[8] Other factors associated with an inferior outcome include node size larger than 6 cm, radiation dose less than 60 Gy, and poor response to chemotherapy.[8]

Surgery has a limited role in the management of nasopharyngeal carcinoma since the disease is usually considered unresectable because of extensive local spread. High-dose radiation therapy alone may have a role in the management of low-stage nasopharyngeal carcinoma; however, studies in both children and adults have shown that combined modality therapy with chemotherapy and radiation is the most effective way to treat nasopharyngeal carcinoma.[8-12] In a meta-analysis of studies adding chemotherapy to radiation therapy in adults with nasopharyngeal carcinoma, concomitant chemotherapy plus radiation therapy offered a significant benefit for survival, locoregional disease control, and reduction in distant metastases.[11] Neoadjuvant chemotherapy resulted in a significant reduction in locoregional recurrence only, while postradiation chemotherapy did not offer any benefit. In children, two studies utilizing preradiation chemotherapy with methotrexate, cisplatin, 5-fluorouracil (5-FU), and leucovorin with or without recombinant interferon-beta reported response rates of more than 90%.[13,14] Radiation therapy doses utilized in both studies were approximately 60 Gy. Additional drug combinations that have been used in children with nasopharyngeal carcinoma include bleomycin, epirubicin, and cisplatin, cisplatin and fluorouracil, and cisplatin, methotrexate, and bleomycin.[2] Incorporation of high-dose-rate brachytherapy into the chemoradiation therapy approach has been reported, but its role in the management of nasopharyngeal carcinoma in children is unknown.[15,16]

A preliminary report of the use of EBV-specific cytotoxic T-lymphocytes revealed minimal toxicity and evidence of significant antitumor activity in patients with relapsed or refractory nasopharyngeal carcinoma.[17] (Refer to the PDQ summary on Nasopharyngeal Cancer Treatment 8 for more information.)

Treatment options under clinical evaluation
  • ARAR0331: This Children's Oncology Group trial (COG-ARAR0331 9) is evaluating the efficacy of induction chemotherapy with cisplatin plus 5-FU followed by concomitant chemotherapy (cisplatin) plus radiation therapy with amifostine as a radioprotectant in patients with AJCC stages IIB to IV nasopharyngeal carcinoma. Patients with stages I to IIA disease will receive only radiation therapy with amifostine.
Esthesioneuroblastoma

Esthesioneuroblastoma (olfactory neuroblastoma) is a very rare, small round-cell tumor arising from the nasal neuroepithelium that is distinct from primitive neuroectodermal tumors.[18-20] Most children present with a nasopharyngeal mass, which may have local extension into the orbits, sinuses, or frontal lobe, with associated symptoms. There appears to be a male predominance, and the average age of presentation is in adolescence. The youngest child reported with this diagnosis was aged 2 years. Metastatic disease is uncommon. The mainstay of treatment has been surgery and radiation. Newer techniques such as endoscopic sinus surgery, radiosurgery, and proton-beam therapy may play a role in the management of this tumor.[21] A retrospective analysis of data from the Surveillance, Epidemiology, and End Results program identified 311 patients with esthesioneuroblastoma.[22] Patients were staged by the extent of the tumor. Disease limited to the nasal cavity was considered the lowest stage and involvement of regional lymph nodes or metastasis was considered the highest stage. This staging system correlated well with outcome. A meta-analysis of 26 studies with a total of 390 patients, largely adults, with esthesioneuroblastoma indicates that higher histopathologic grade and metastases to the cervical lymph nodes may correlate with adverse prognostic factors.[23] Recent reports indicate increasing use of neoadjuvant chemotherapy.[18,19,24,25] Chemotherapy regimens that have been used with efficacy include etoposide (VP-16), ifosfamide, and cisplatin (Platinol),[26] vincristine, actinomycin D and cyclophosphamide without doxorubicin (Adriamycin), ifosfamide/etoposide, and cisplatin plus etoposide or doxorubicin.[24] The long-term survival rate appears to be approximately 60% to 80%. Local recurrences may occur later in life.

Thyroid Tumors

Tumors of the thyroid are classified as adenomas or carcinomas.[27-31] Adenomas are benign growths that may cause enlargement of all or part of the gland, which extends to both sides of the neck and can be quite large. Some of these tumors may secrete hormones. Transformation to a malignant carcinoma may occur in some cells, which then may grow and spread to lymph nodes in the neck or to the lungs.

Although rare, thyroid cancers represent about 1.5% of all tumors seen in the pediatric age group. Most thyroid carcinomas occur in girls.[32] Patients with thyroid cancer usually present with a thyroid mass with or without cervical adenopathy.[33-35] There is an excessive frequency of thyroid adenoma and carcinoma in patients who previously received radiation to the neck.[36,37] When occurring in patients with the multiple endocrine neoplasia syndromes, thyroid cancer may be associated with the development of other types of malignant tumors. (Refer to the Multiple Endocrine Neoplasia Syndrome 10 section of this summary for more information.) The American Thyroid Association Taskforce [38] has developed guidelines for management of thyroid nodules in older adolescents and adults, but it is not yet known how to apply these guidelines to thyroid nodules in children.[27]

Initial evaluation of a child or adolescent with a thyroid nodule should include an ultrasound of the thyroid. Tests of thyroid function are usually normal, but thyroglobulin can be elevated. Fine needle aspiration (FNA) is the initial diagnostic approach, though experience in FNA in pediatric hospitals may be limited, in which case open biopsy or lobe resection should be considered.[39,40] Open biopsy or resection may be preferable for young children as well.

Various histologies account for the general diagnostic category of carcinoma of the thyroid,[41] but the vast majority of tumors are differentiated. These tumors comprise papillary carcinoma (60%–75%),[37] follicular carcinoma (10%–20%), medullary carcinoma (5%–10%), and anaplastic carcinoma (<1%). Follicular carcinoma may be sporadic or familial and medullary carcinoma is usually familial.[42] Papillary carcinoma often has multicentric origins and a very high rate of lymph node metastasis (70%–90%).[41] Follicular carcinoma is usually encapsulated and has a higher incidence of bone and lung metastasis. Follicular carcinoma and papillary carcinoma generally have a benign course, with a 10-year survival rate of more than 95%.[43] Fifty percent of medullary thyroid carcinomas in adults and children have hematogenous metastases at diagnosis.[44] Patients with medullary carcinoma of the thyroid have a guarded prognosis, unless they have very small tumors (microcarcinoma, defined as <1.0 cm in diameter), which carry a good prognosis.[45]

Surgery by an experienced thyroid surgeon is the treatment required for all thyroid neoplasms.[43] Total or near-total thyroidectomy plus cervical lymph node dissection, when indicated, is the most common surgical approach.[33] For patients with obvious metastatic disease or heavy nodal invasion, total thyroidectomy and treatment with radioactive idodine is indicated. For patients with an isolated nodule in the thyroid, treatment may involve only a lobectomy.[33,46] During the 4- to 6-week period following surgery, patients who received a total thyroidectomy may develop hypothyroidism. A radioactive iodine (I-131) scan is then performed to search for residual, functionally active neoplasms. If there is no disease outside of the thyroid bed, an ablative dose of I-131 (approximately 29 mCi) is administered for total thyroid destruction. If there is evidence of nodal or disseminated disease, higher doses (100–200 mCi) of I-131 are required. In children, the I-131 dose may be adjusted for weight and other age-dependent safety factors.[47,48] After surgery and radioactive iodine therapy, hormone replacement therapy must be given to compensate for the lost thyroid hormone and to suppress thyrotropin (TSH) production.[49]

Initial treatment (defined as surgery plus one radioactive iodine ablation plus thyroid replacement) is effective in inducing remission for 70% of patients. Extensive disease at diagnosis and larger tumor size predict failure to remit. With additional treatment, 89% of patients achieve remission.[50] Periodic evaluations are required to determine whether there is metastatic disease involving the lungs. Lifelong follow-up is necessary.[51] Thyroglobulin, T4, and TSH levels should be evaluated periodically to determine whether replacement hormone is appropriately dosed.

Patients with differentiated thyroid cancer generally have an excellent survival with relatively few side effects.[51-53] Recurrence is common (35%–45%), however, and is seen more often in children younger than 10 years and in those with palpable cervical lymph nodes at diagnosis.[29,54,55] Of note, the sodium-iodide symporter (a membrane-bound glycoprotein cotransporter) essential for uptake of iodide and thyroid hormone synthesis, is expressed in 35% to 45% of thyroid cancers in children and adolescents. Patients with expression of the sodium-iodide symporter have a lower risk of recurrence.[56] Recurrent papillary thyroid cancer is usually responsive to treatment with radioactive iodine ablation.[57] Even patients with a tumor that has spread to the lungs may expect to have no decrease in life span after appropriate treatment. (Refer to the PDQ summary on adult Thyroid Cancer Treatment 11 for more information.)

Oral Cancers

Cancer of the oral cavity is extremely rare in children and adolescents.[3,58] The vast majority (>90%) of tumors and tumor-like lesions in the oral cavity are benign.[59-62] Benign odontogenic neoplasms include odontoma and ameloblastoma. The most common nonodontogenic neoplasms are fibromas, hemangiomas, and papillomas. Tumor-like lesions include lymphangiomas, granulomas, and eosinophilic granuloma (Langerhans cell histiocytoma [LCH]). Malignant tumors are found in 0.1% to 2% of a series of oral biopsies performed in children [59,60] and 3% to 13% of oral tumor biopsies.[61,62] Malignant tumor types include lymphomas (especially Burkitt) and sarcomas (including rhabdomyosarcoma and fibrosarcoma). The most common type of primary oral cancer in adults, squamous cell carcinoma (SCC), is extremely rare in children. Only occasional case reports are found in the literature.[63,64] Adolescents with an oral SCC should be screened for Fanconi anemia.[65,66]

Treatment of benign oral tumors is surgical. Management of malignant tumors is dependent on histology and may include surgery, chemotherapy, and radiation.[67] LCH may require other treatment besides surgery. (Refer to the PDQ summaries on adult Oropharyngeal Cancer Treatment 12 and Lip and Oral Cavity Cancer Treatment 13 for more information.)

Salivary Gland Tumors

Most salivary gland neoplasms arise in the parotid gland.[68-72] About 15% of these tumors may arise in the submandibular glands or in the minor salivary glands under the tongue and jaw. These tumors are most frequently benign but on very rare occasions may be malignant.[73] Sialoblastomas are usually benign tumors presenting in the neonatal period but can rarely metastasize.[74] A chemotherapy regimen of carboplatin, epirubicin, vincristine, etoposide, ifosfamide, and dactinomycin has been used in the treatment of metastatic sialoblastoma and has produced a response in one child.[75][Level of evidence: 3iiiDiv] The malignant lesions include mucoepidermoid carcinoma,[76] acinic cell carcinoma, rhabdomyosarcoma, adenocarcinoma, and undifferentiated carcinoma. These tumors may occur after radiation therapy and chemotherapy are given for treatment of primary leukemia or solid tumors.[77,78] Radical surgical removal is the treatment of choice, whenever possible, with additional use of radiation therapy and chemotherapy for high-grade tumors or tumors that have spread from their site of origin.[76,79,80] Prognosis for patients with these tumors is generally good.[71,81-83] (Refer to the PDQ summary on adult Salivary Gland Cancer Treatment 14 for more information.)

Laryngeal Cancer and Papillomatosis

Benign tumors of the larynx are rare. Malignant tumors, which are especially rare, may be associated with benign tumors such as polyps and papillomas.[84,85] These tumors may cause hoarseness, difficulty swallowing, and enlargement of the lymph nodes of the neck. Rhabdomyosarcoma is the most common malignant tumor of the larynx in the pediatric age group. SCC of the larynx should be managed in the same manner as in adults with carcinoma at this site, with surgery and radiation.[86] Laser surgery may be the first type of treatment utilized for these lesions.

Papillomatosis of the larynx is a benign overgrowth of tissues lining the larynx and is associated with the human papillomavirus (HPV), most commonly HPV-6 and HPV-11.[87] The presence of HPV-11 appears to correlate with a more aggressive clinical course than HPV-6.[88] This condition is not cancerous, and primary treatment is surgical ablation with laser vaporization.[89] Frequent recurrences are common. If a patient requires more than four surgical procedures per year, treatment with interferon should be considered.[90] A pilot study of immunotherapy with HspE7, a recombinant fusion protein that has shown activity in other HPV-related diseases, has suggested a marked increase in the amount of time between surgeries.[91] These results, however, must be confirmed in a larger randomized trial. These tumors can cause hoarseness because of their association with wart-like nodules on the vocal cords and may rarely extend into the lung, producing significant morbidity. Malignant degeneration may occur with development of cancer in the larynx and squamous cell lung cancer. (Refer to the PDQ summary on adult Laryngeal Cancer Treatment 15 for more information.)

Respiratory Tract Carcinoma Involving the NUT Gene on Chromosome 15

Researchers have described a group of young patients with midline carcinomas with a very poor prognosis. The tumors arise in midline epithelial structures including the thymus, mediastinum, airway structures, and bladder. They exhibit squamous differentiation. Tumors from 8 of 11 patients exhibited a balanced chromosomal translocation t(15;19) involving the BRD4 and the NUT genes. These patients had no response to chemotherapy and died very quickly. Tumors from the remaining three patients had a chromosomal break in the NUT gene on chromosome 15 but had normal chromosome 19. These patients were older and had a slightly longer survival than the eight patients exhibiting t(15;19).[92]

References

  1. Vasef MA, Ferlito A, Weiss LM: Nasopharyngeal carcinoma, with emphasis on its relationship to Epstein-Barr virus. Ann Otol Rhinol Laryngol 106 (4): 348-56, 1997.  [PUBMED Abstract]

  2. Ayan I, Kaytan E, Ayan N: Childhood nasopharyngeal carcinoma: from biology to treatment. Lancet Oncol 4 (1): 13-21, 2003.  [PUBMED Abstract]

  3. Young JL Jr, Miller RW: Incidence of malignant tumors in U. S. children. J Pediatr 86 (2): 254-8, 1975.  [PUBMED Abstract]

  4. Bar-Sela G, Ben Arush MW, Sabo E, et al.: Pediatric nasopharyngeal carcinoma: better prognosis and increased c-Kit expression as compared to adults. Pediatr Blood Cancer 45 (3): 291-7, 2005.  [PUBMED Abstract]

  5. Naegele RF, Champion J, Murphy S, et al.: Nasopharyngeal carcinoma in American Children: Epstein-Barr virus-specific antibody titers and prognosis. Int J Cancer 29 (2): 209-12, 1982.  [PUBMED Abstract]

  6. American Joint Committee on Cancer.: AJCC Cancer Staging Manual. 6th ed. New York, NY: Springer, 2002. 

  7. Casanova M, Ferrari A, Gandola L, et al.: Undifferentiated nasopharyngeal carcinoma in children and adolescents: comparison between staging systems. Ann Oncol 12 (8): 1157-62, 2001.  [PUBMED Abstract]

  8. Laskar S, Sanghavi V, Muckaden MA, et al.: Nasopharyngeal carcinoma in children: ten years' experience at the Tata Memorial Hospital, Mumbai. Int J Radiat Oncol Biol Phys 58 (1): 189-95, 2004.  [PUBMED Abstract]

  9. Al-Sarraf M, LeBlanc M, Giri PG, et al.: Chemoradiotherapy versus radiotherapy in patients with advanced nasopharyngeal cancer: phase III randomized Intergroup study 0099. J Clin Oncol 16 (4): 1310-7, 1998.  [PUBMED Abstract]

  10. Wolden SL, Steinherz PG, Kraus DH, et al.: Improved long-term survival with combined modality therapy for pediatric nasopharynx cancer. Int J Radiat Oncol Biol Phys 46 (4): 859-64, 2000.  [PUBMED Abstract]

  11. Langendijk JA, Leemans ChR, Buter J, et al.: The additional value of chemotherapy to radiotherapy in locally advanced nasopharyngeal carcinoma: a meta-analysis of the published literature. J Clin Oncol 22 (22): 4604-12, 2004.  [PUBMED Abstract]

  12. Venkitaraman R, Ramanan SG, Sagar TG: Nasopharyngeal cancer of childhood and adolescence: a single institution experience. Pediatr Hematol Oncol 24 (7): 493-502, 2007 Oct-Nov.  [PUBMED Abstract]

  13. Mertens R, Granzen B, Lassay L, et al.: Treatment of nasopharyngeal carcinoma in children and adolescents: definitive results of a multicenter study (NPC-91-GPOH). Cancer 104 (5): 1083-9, 2005.  [PUBMED Abstract]

  14. Rodriguez-Galindo C, Wofford M, Castleberry RP, et al.: Preradiation chemotherapy with methotrexate, cisplatin, 5-fluorouracil, and leucovorin for pediatric nasopharyngeal carcinoma. Cancer 103 (4): 850-7, 2005.  [PUBMED Abstract]

  15. Nakamura RA, Novaes PE, Antoneli CB, et al.: High-dose-rate brachytherapy as part of a multidisciplinary treatment of nasopharyngeal lymphoepithelioma in childhood. Cancer 104 (3): 525-31, 2005.  [PUBMED Abstract]

  16. Louis CU, Paulino AC, Gottschalk S, et al.: A single institution experience with pediatric nasopharyngeal carcinoma: high incidence of toxicity associated with platinum-based chemotherapy plus IMRT. J Pediatr Hematol Oncol 29 (7): 500-5, 2007.  [PUBMED Abstract]

  17. Straathof KC, Bollard CM, Popat U, et al.: Treatment of nasopharyngeal carcinoma with Epstein-Barr virus--specific T lymphocytes. Blood 105 (5): 1898-904, 2005.  [PUBMED Abstract]

  18. Kumar M, Fallon RJ, Hill JS, et al.: Esthesioneuroblastoma in children. J Pediatr Hematol Oncol 24 (6): 482-7, 2002 Aug-Sep.  [PUBMED Abstract]

  19. Theilgaard SA, Buchwald C, Ingeholm P, et al.: Esthesioneuroblastoma: a Danish demographic study of 40 patients registered between 1978 and 2000. Acta Otolaryngol 123 (3): 433-9, 2003.  [PUBMED Abstract]

  20. Dias FL, Sa GM, Lima RA, et al.: Patterns of failure and outcome in esthesioneuroblastoma. Arch Otolaryngol Head Neck Surg 129 (11): 1186-92, 2003.  [PUBMED Abstract]

  21. Unger F, Haselsberger K, Walch C, et al.: Combined endoscopic surgery and radiosurgery as treatment modality for olfactory neuroblastoma (esthesioneuroblastoma). Acta Neurochir (Wien) 147 (6): 595-601; discussion 601-2, 2005.  [PUBMED Abstract]

  22. Jethanamest D, Morris LG, Sikora AG, et al.: Esthesioneuroblastoma: a population-based analysis of survival and prognostic factors. Arch Otolaryngol Head Neck Surg 133 (3): 276-80, 2007.  [PUBMED Abstract]

  23. Dulguerov P, Allal AS, Calcaterra TC: Esthesioneuroblastoma: a meta-analysis and review. Lancet Oncol 2 (11): 683-90, 2001.  [PUBMED Abstract]

  24. Eich HT, Müller RP, Micke O, et al.: Esthesioneuroblastoma in childhood and adolescence. Better prognosis with multimodal treatment? Strahlenther Onkol 181 (6): 378-84, 2005.  [PUBMED Abstract]

  25. Loy AH, Reibel JF, Read PW, et al.: Esthesioneuroblastoma: continued follow-up of a single institution's experience. Arch Otolaryngol Head Neck Surg 132 (2): 134-8, 2006.  [PUBMED Abstract]

  26. Kim DW, Jo YH, Kim JH, et al.: Neoadjuvant etoposide, ifosfamide, and cisplatin for the treatment of olfactory neuroblastoma. Cancer 101 (10): 2257-60, 2004.  [PUBMED Abstract]

  27. Dinauer C, Francis GL: Thyroid cancer in children. Endocrinol Metab Clin North Am 36 (3): 779-806, vii, 2007.  [PUBMED Abstract]

  28. Vasko V, Bauer AJ, Tuttle RM, et al.: Papillary and follicular thyroid cancers in children. Endocr Dev 10: 140-72, 2007.  [PUBMED Abstract]

  29. Grigsby PW, Gal-or A, Michalski JM, et al.: Childhood and adolescent thyroid carcinoma. Cancer 95 (4): 724-9, 2002.  [PUBMED Abstract]

  30. Skinner MA: Cancer of the thyroid gland in infants and children. Semin Pediatr Surg 10 (3): 119-26, 2001.  [PUBMED Abstract]

  31. Halac I, Zimmerman D: Thyroid nodules and cancers in children. Endocrinol Metab Clin North Am 34 (3): 725-44, x, 2005.  [PUBMED Abstract]

  32. Shapiro NL, Bhattacharyya N: Population-based outcomes for pediatric thyroid carcinoma. Laryngoscope 115 (2): 337-40, 2005.  [PUBMED Abstract]

  33. Thompson GB, Hay ID: Current strategies for surgical management and adjuvant treatment of childhood papillary thyroid carcinoma. World J Surg 28 (12): 1187-98, 2004.  [PUBMED Abstract]

  34. Harness JK, Sahar DE, et al.: Childhood thyroid carcinoma. In: Clark O, Duh Q-Y, Kebebew E, eds.: Textbook of Endocrine Surgery. 2nd ed. Philadelphia, PA: Elsevier Saunders Company, 2005., pp 93-101. 

  35. Rachmiel M, Charron M, Gupta A, et al.: Evidence-based review of treatment and follow up of pediatric patients with differentiated thyroid carcinoma. J Pediatr Endocrinol Metab 19 (12): 1377-93, 2006.  [PUBMED Abstract]

  36. Cotterill SJ, Pearce MS, Parker L: Thyroid cancer in children and young adults in the North of England. Is increasing incidence related to the Chernobyl accident? Eur J Cancer 37 (8): 1020-6, 2001.  [PUBMED Abstract]

  37. Kaplan MM, Garnick MB, Gelber R, et al.: Risk factors for thyroid abnormalities after neck irradiation for childhood cancer. Am J Med 74 (2): 272-80, 1983.  [PUBMED Abstract]

  38. Cooper DS, Doherty GM, Haugen BR, et al.: Management guidelines for patients with thyroid nodules and differentiated thyroid cancer. Thyroid 16 (2): 109-42, 2006.  [PUBMED Abstract]

  39. Flannery TK, Kirkland JL, Copeland KC, et al.: Papillary thyroid cancer: a pediatric perspective. Pediatrics 98 (3 Pt 1): 464-6, 1996.  [PUBMED Abstract]

  40. Willgerodt H, Keller E, Bennek J, et al.: Diagnostic value of fine-needle aspiration biopsy of thyroid nodules in children and adolescents. J Pediatr Endocrinol Metab 19 (4): 507-15, 2006.  [PUBMED Abstract]

  41. Feinmesser R, Lubin E, Segal K, et al.: Carcinoma of the thyroid in children--a review. J Pediatr Endocrinol Metab 10 (6): 561-8, 1997 Nov-Dec.  [PUBMED Abstract]

  42. Skinner MA: Management of hereditary thyroid cancer in children. Surg Oncol 12 (2): 101-4, 2003.  [PUBMED Abstract]

  43. Hung W, Sarlis NJ: Current controversies in the management of pediatric patients with well-differentiated nonmedullary thyroid cancer: a review. Thyroid 12 (8): 683-702, 2002.  [PUBMED Abstract]

  44. Hill CS, Ibanez ML, Samaan NA, et al.: Medullary (solid) carcinoma of the thyroid gland: an analysis of the M.D. Anderson Hospital experience with patients with the tumor, its special features, and its histogenesis. Medicine 52(2): 141-171, 1973. 

  45. Krueger JE, Maitra A, Albores-Saavedra J: Inherited medullary microcarcinoma of the thyroid: a study of 11 cases. Am J Surg Pathol 24 (6): 853-8, 2000.  [PUBMED Abstract]

  46. Newman KD, Black T, Heller G, et al.: Differentiated thyroid cancer: determinants of disease progression in patients <21 years of age at diagnosis: a report from the Surgical Discipline Committee of the Children's Cancer Group. Ann Surg 227 (4): 533-41, 1998.  [PUBMED Abstract]

  47. Luster M, Lassmann M, Freudenberg LS, et al.: Thyroid cancer in childhood: management strategy, including dosimetry and long-term results. Hormones (Athens) 6 (4): 269-78, 2007 Oct-Dec.  [PUBMED Abstract]

  48. Parisi MT, Mankoff D: Differentiated pediatric thyroid cancer: correlates with adult disease, controversies in treatment. Semin Nucl Med 37 (5): 340-56, 2007.  [PUBMED Abstract]

  49. Yeh SD, La Quaglia MP: 131I therapy for pediatric thyroid cancer. Semin Pediatr Surg 6 (3): 128-33, 1997.  [PUBMED Abstract]

  50. Powers PA, Dinauer CA, Tuttle RM, et al.: Tumor size and extent of disease at diagnosis predict the response to initial therapy for papillary thyroid carcinoma in children and adolescents. J Pediatr Endocrinol Metab 16 (5): 693-702, 2003.  [PUBMED Abstract]

  51. Vassilopoulou-Sellin R, Goepfert H, Raney B, et al.: Differentiated thyroid cancer in children and adolescents: clinical outcome and mortality after long-term follow-up. Head Neck 20 (6): 549-55, 1998.  [PUBMED Abstract]

  52. Wiersinga WM: Thyroid cancer in children and adolescents--consequences in later life. J Pediatr Endocrinol Metab 14 (Suppl 5): 1289-96; discussion 1297-8, 2001.  [PUBMED Abstract]

  53. Jarzab B, Handkiewicz-Junak D, Wloch J: Juvenile differentiated thyroid carcinoma and the role of radioiodine in its treatment: a qualitative review. Endocr Relat Cancer 12 (4): 773-803, 2005.  [PUBMED Abstract]

  54. Alessandri AJ, Goddard KJ, Blair GK, et al.: Age is the major determinant of recurrence in pediatric differentiated thyroid carcinoma. Med Pediatr Oncol 35 (1): 41-6, 2000.  [PUBMED Abstract]

  55. Borson-Chazot F, Causeret S, Lifante JC, et al.: Predictive factors for recurrence from a series of 74 children and adolescents with differentiated thyroid cancer. World J Surg 28 (11): 1088-92, 2004.  [PUBMED Abstract]

  56. Patel A, Jhiang S, Dogra S, et al.: Differentiated thyroid carcinoma that express sodium-iodide symporter have a lower risk of recurrence for children and adolescents. Pediatr Res 52 (5): 737-44, 2002.  [PUBMED Abstract]

  57. Powers PA, Dinauer CA, Tuttle RM, et al.: Treatment of recurrent papillary thyroid carcinoma in children and adolescents. J Pediatr Endocrinol Metab 16 (7): 1033-40, 2003.  [PUBMED Abstract]

  58. Berstein L, Gurney JG: Carcinomas and other malignant epithelial neoplasms. In: Ries LA, Smith MA, Gurney JG, et al., eds.: Cancer incidence and survival among children and adolescents: United States SEER Program 1975-1995. Bethesda, Md: National Cancer Institute, SEER Program, 1999. NIH Pub.No. 99-4649., Chapter 11, pp 139-148. Also available online. 16 Last accessed April 19, 2007. 

  59. Das S, Das AK: A review of pediatric oral biopsies from a surgical pathology service in a dental school. Pediatr Dent 15 (3): 208-11, 1993 May-Jun.  [PUBMED Abstract]

  60. Ulmansky M, Lustmann J, Balkin N: Tumors and tumor-like lesions of the oral cavity and related structures in Israeli children. Int J Oral Maxillofac Surg 28 (4): 291-4, 1999.  [PUBMED Abstract]

  61. Tröbs RB, Mader E, Friedrich T, et al.: Oral tumors and tumor-like lesions in infants and children. Pediatr Surg Int 19 (9-10): 639-45, 2003.  [PUBMED Abstract]

  62. Tanaka N, Murata A, Yamaguchi A, et al.: Clinical features and management of oral and maxillofacial tumors in children. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 88 (1): 11-5, 1999.  [PUBMED Abstract]

  63. Torossian JM, Beziat JL, Philip T, et al.: Squamous cell carcinoma of the tongue in a 13-year-old boy. J Oral Maxillofac Surg 58 (12): 1407-10, 2000.  [PUBMED Abstract]

  64. Bill TJ, Reddy VR, Ries KL, et al.: Adolescent gingival squamous cell carcinoma: Report of a case and review of the literature. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 91 (6): 682-5, 2001.  [PUBMED Abstract]

  65. Oksüzoğlu B, Yalçin S: Squamous cell carcinoma of the tongue in a patient with Fanconi's anemia: a case report and review of the literature. Ann Hematol 81 (5): 294-8, 2002.  [PUBMED Abstract]

  66. Reinhard H, Peters I, Gottschling S, et al.: Squamous cell carcinoma of the tongue in a 13-year-old girl with Fanconi anemia. J Pediatr Hematol Oncol 29 (7): 488-91, 2007.  [PUBMED Abstract]

  67. Sturgis EM, Moore BA, Glisson BS, et al.: Neoadjuvant chemotherapy for squamous cell carcinoma of the oral tongue in young adults: a case series. Head Neck 27 (9): 748-56, 2005.  [PUBMED Abstract]

  68. Johns ME, Goldsmith MM: Incidence, diagnosis, and classification of salivary gland tumors. Part 1. Oncology (Huntingt) 3 (2): 47-56; discussion 56, 58, 62, 1989.  [PUBMED Abstract]

  69. Ethunandan M, Ethunandan A, Macpherson D, et al.: Parotid neoplasms in children: experience of diagnosis and management in a district general hospital. Int J Oral Maxillofac Surg 32 (4): 373-7, 2003.  [PUBMED Abstract]

  70. da Cruz Perez DE, Pires FR, Alves FA, et al.: Salivary gland tumors in children and adolescents: a clinicopathologic and immunohistochemical study of fifty-three cases. Int J Pediatr Otorhinolaryngol 68 (7): 895-902, 2004.  [PUBMED Abstract]

  71. Shapiro NL, Bhattacharyya N: Clinical characteristics and survival for major salivary gland malignancies in children. Otolaryngol Head Neck Surg 134 (4): 631-4, 2006.  [PUBMED Abstract]

  72. Ellies M, Schaffranietz F, Arglebe C, et al.: Tumors of the salivary glands in childhood and adolescence. J Oral Maxillofac Surg 64 (7): 1049-58, 2006.  [PUBMED Abstract]

  73. Laikui L, Hongwei L, Hongbing J, et al.: Epithelial salivary gland tumors of children and adolescents in west China population: a clinicopathologic study of 79 cases. J Oral Pathol Med 37 (4): 201-5, 2008.  [PUBMED Abstract]

  74. Williams SB, Ellis GL, Warnock GR: Sialoblastoma: a clinicopathologic and immunohistochemical study of 7 cases. Ann Diagn Pathol 10 (6): 320-6, 2006.  [PUBMED Abstract]

  75. Scott JX, Krishnan S, Bourne AJ, et al.: Treatment of metastatic sialoblastoma with chemotherapy and surgery. Pediatr Blood Cancer 50 (1): 134-7, 2008.  [PUBMED Abstract]

  76. Rahbar R, Grimmer JF, Vargas SO, et al.: Mucoepidermoid carcinoma of the parotid gland in children: A 10-year experience. Arch Otolaryngol Head Neck Surg 132 (4): 375-80, 2006.  [PUBMED Abstract]

  77. Kaste SC, Hedlund G, Pratt CB: Malignant parotid tumors in patients previously treated for childhood cancer: clinical and imaging findings in eight cases. AJR Am J Roentgenol 162 (3): 655-9, 1994.  [PUBMED Abstract]

  78. Whatley WS, Thompson JW, Rao B: Salivary gland tumors in survivors of childhood cancer. Otolaryngol Head Neck Surg 134 (3): 385-8, 2006.  [PUBMED Abstract]

  79. Kamal SA, Othman EO: Diagnosis and treatment of parotid tumours. J Laryngol Otol 111 (4): 316-21, 1997.  [PUBMED Abstract]

  80. Rogers DA, Rao BN, Bowman L, et al.: Primary malignancy of the salivary gland in children. J Pediatr Surg 29 (1): 44-7, 1994.  [PUBMED Abstract]

  81. Bentz BG, Hughes CA, Lüdemann JP, et al.: Masses of the salivary gland region in children. Arch Otolaryngol Head Neck Surg 126 (12): 1435-9, 2000.  [PUBMED Abstract]

  82. Ribeiro Kde C, Kowalski LP, Saba LM, et al.: Epithelial salivary glands neoplasms in children and adolescents: a forty-four-year experience. Med Pediatr Oncol 39 (6): 594-600, 2002.  [PUBMED Abstract]

  83. Guzzo M, Ferrari A, Marcon I, et al.: Salivary gland neoplasms in children: the experience of the Istituto Nazionale Tumori of Milan. Pediatr Blood Cancer 47 (6): 806-10, 2006.  [PUBMED Abstract]

  84. McGuirt WF Jr, Little JP: Laryngeal cancer in children and adolescents. Otolaryngol Clin North Am 30 (2): 207-14, 1997.  [PUBMED Abstract]

  85. Bauman NM, Smith RJ: Recurrent respiratory papillomatosis. Pediatr Clin North Am 43 (6): 1385-401, 1996.  [PUBMED Abstract]

  86. Siddiqui F, Sarin R, Agarwal JP, et al.: Squamous carcinoma of the larynx and hypopharynx in children: a distinct clinical entity? Med Pediatr Oncol 40 (5): 322-4, 2003.  [PUBMED Abstract]

  87. Kashima HK, Mounts P, Shah K: Recurrent respiratory papillomatosis. Obstet Gynecol Clin North Am 23 (3): 699-706, 1996.  [PUBMED Abstract]

  88. Maloney EM, Unger ER, Tucker RA, et al.: Longitudinal measures of human papillomavirus 6 and 11 viral loads and antibody response in children with recurrent respiratory papillomatosis. Arch Otolaryngol Head Neck Surg 132 (7): 711-5, 2006.  [PUBMED Abstract]

  89. Andrus JG, Shapshay SM: Contemporary management of laryngeal papilloma in adults and children. Otolaryngol Clin North Am 39 (1): 135-58, 2006.  [PUBMED Abstract]

  90. Avidano MA, Singleton GT: Adjuvant drug strategies in the treatment of recurrent respiratory papillomatosis. Otolaryngol Head Neck Surg 112 (2): 197-202, 1995.  [PUBMED Abstract]

  91. Derkay CS, Smith RJ, McClay J, et al.: HspE7 treatment of pediatric recurrent respiratory papillomatosis: final results of an open-label trial. Ann Otol Rhinol Laryngol 114 (9): 730-7, 2005.  [PUBMED Abstract]

  92. French CA, Kutok JL, Faquin WC, et al.: Midline carcinoma of children and young adults with NUT rearrangement. J Clin Oncol 22 (20): 4135-9, 2004.  [PUBMED Abstract]

Thoracic Cancers

Note: Some citations in the text of this section are followed by a level of evidence. The PDQ editorial boards use a formal ranking system to help the reader judge the strength of evidence linked to the reported results of a therapeutic strategy. (Refer to the PDQ summary on Levels of Evidence 2 for more information.)

Thoracic cancers include breast cancer, bronchial adenomas, bronchial carcinoid tumors, pleuropulmonary blastoma, esophageal tumors, thymomas, thymic carcinomas, tumors of the heart, and mesothelioma. The prognosis, diagnosis, classification, and treatment of these thoracic cancers are discussed below.

Breast Cancer

The most frequent breast tumor seen in children is a fibroadenoma.[1] These tumors can be observed and many will regress without a need for biopsy. However, rare malignant transformation leading to phyllodes tumors has been reported.[2] Sudden rapid enlargement of a suspected fibroadenoma is an indication for needle biopsy or excision. Phyllodes tumors can be managed by wide local excision without mastectomy.[2]

Carcinomas have been reported in both males and females younger than 21 years.[3-8] There is an increased lifetime risk of breast cancer in female survivors of Hodgkin lymphoma who were treated with radiation to the chest area, however, breast cancer is also seen in patients who were treated for any cancer that was treated with chest irradiation.[7,9-11] (Refer to the PDQ summary on the Late Effects of Treatment for Childhood Cancer 7 for more information about secondary breast cancers.) Carcinomas are more frequent than sarcomas. Mammograms should start at age 25 years or 10 years postexposure to radiation therapy (whichever came last). Treatment options for children and adolescents with breast cancer include radiation, chemotherapy, and surgery. Breast tumors may also occur as metastatic deposits from leukemia, rhabdomyosarcoma, other sarcomas, or lymphoma (particularly in patients who are infected with the human immunodeficiency virus). (Refer to the PDQ summary on adult Breast Cancer Treatment 17 for more information.)

Bronchial Tumors

Bronchial tumors are a heterogeneous group of primary endobronchial lesions, and though adenoma implies a benign process, all varieties of bronchial tumors on occasion display a malignant behavior. There are three histologic types. The most frequent type is a carcinoid tumor; this is followed by mucoepidermoid carcinoma and adenoid cystic carcinoma. Carcinoid tumors account for 80% to 85% of all bronchial tumors in children.[12-16] The presenting symptoms are usually because of an incomplete bronchial obstruction with a cough, recurrent pneumonitis, and hemoptysis. Because of difficulties in diagnosis, symptoms are frequently present for months and occasionally children with wheezing have been treated for asthma with delays in diagnosis as long as 4 to 5 years. Metastatic lesions are reported in approximately 6% of cases and recurrences occur in 2% of cases. Atypical carcinoid tumors are rare but more aggressive with 50% of patients presenting with metastatic disease at diagnosis.[17] There is a single report of a child with a carcinoid tumor and metastatic disease who developed the classic carcinoid syndrome.[18] Octreotide nuclear scans may demonstrate uptake of radioactivity by the tumor or lymph nodes, suggesting metastatic spread. Bronchial tumors of all histologic types are associated with an excellent prognosis in children, even in the presence of local invasion.[19,20] The management of bronchial tumors is somewhat controversial because all bronchial tumors are usually visible endoscopically. Biopsy in these lesions may be hazardous because of hemorrhage, and endoscopic resection is not recommended. Bronchography or computed tomography scan may be helpful to determine the degree of bronchiectasis distal to the obstruction since the degree of pulmonary destruction may influence surgical therapy.[21] Epithelial cancers of the lung are rare in children. When they do occur, they tend to be of advanced stage with prognosis dependent on both histology and stage.[17]

Conservative pulmonary resection with the removal of the involved lymphatics is the treatment of choice. Sleeve segmental bronchial resection is possible in children and when feasible, is the treatment of choice.[22,23] Adenoid cystic carcinomas (cylindroma) have a tendency to spread submucosally, and late local recurrence or dissemination has been reported. In addition to en bloc resection with hilar lymphadenectomy, a frozen section examination of the bronchial margins should be carried out in children with this lesion. Neither chemotherapy nor radiation therapy is indicated for bronchial tumors, unless evidence of metastasis is documented.

Pleuropulmonary Blastoma

Pleuropulmonary blastoma is a rare and highly aggressive pulmonary malignancy in children. Pleuropulmonary blastoma appears to progress through stages with the earliest stage (type I) being a purely lung cystic neoplasm with subtle malignant changes, typically occurring in the first 2 years of life with a good prognosis,[24][Level of evidence: 3iiiA] followed by the more aggressive stages: type II (cystic and solid neoplasm) and type III (purely solid neoplasm).[25,26] There have been reports of type I transitioning directly to type III.[27] Cerebral metastasis occurs in up to 50% of patients with type III tumors.[28] An independent group of researchers has established a registry and resource Web site for this rare tumor.[29] An association between congenital lung cysts and pleuropulmonary blastoma has been reported, although cytogenetic and molecular studies can help distinguish the nonneoplastic congenital cystic adenomatoid malformation from pleuropulmonary blastoma.[30-34] Comparative genomic and fluorescent in situ hybridization methods have identified gain of chromosome 8q as the main recurrent chromosomal abnormality in pleuropulmonary blastoma.[[35]][[36]] The tumor is usually located in the lung periphery, but it may be extrapulmonary with involvement of the mediastinum, diaphragm, and/or pleura.[33,37] The tumors may recur or metastasize, in spite of primary resection.[[24]][26] Responses to chemotherapy have been reported with agents similar to those used for the treatment of rhabdomyosarcoma, and adjuvant chemotherapy may benefit patients with type I pleuropulmonary blastoma by reducing the risk of recurrence.[25,38] Achieving total resection of the tumor at any time during treatment is associated with improved prognosis.[33] Chemotherapeutic agents may include vincristine, cyclophosphamide, dactinomycin, and doxorubicin. High-dose chemotherapy with stem cell rescue has been used without success.[39] Radiation, either external beam or P-32, may be used when the tumor cannot be surgically removed. A family history of cancer in close relatives has been noted for many young patients affected by this tumor.[40] In addition, there has been a reported association between pleuropulmonary blastoma and cystic nephroma.[41,42] Data from the International Pleuropulmonary Blastoma Registry suggest that adjuvant chemotherapy may reduce the risk of recurrence.[25]

Esophageal Tumors

Esophageal cancer is rare in the pediatric age group, though it is relatively common in older adults.[43] Symptoms are related to difficulty in swallowing and associated weight loss. Most of these tumors are squamous cell carcinomas, though sarcomas can also arise in the esophagus. The most common benign tumor is leiomyoma. Diagnosis is made by histologic examination of biopsy tissue.

Treatment options for esophageal carcinoma include either external-beam intracavitary radiation therapy or chemotherapy agents commonly used to treat carcinomas: platinum derivatives, paclitaxel, and etoposide. Prognosis generally is poor for this cancer, which rarely can be completely resected. (Refer to the PDQ summary on adult Esophageal Cancer Treatment 18 for more information.)

Thymoma and Thymic Carcinoma

A cancer of the thymus is not considered a thymoma or a thymic carcinoma unless there are neoplastic changes of the epithelial cells that cover the organ.[44,45] The term thymoma is customarily used to describe neoplasms that show no overt atypia of the epithelial component. A thymic epithelial tumor that exhibits clear-cut cytologic atypia and histologic features no longer specific to the thymus is known as thymic carcinoma, also known as type C thymoma. Other tumors that involve the thymus gland include lymphomas, germ cell tumors, carcinomas, carcinoids, and thymomas. Hodgkin lymphoma and non-Hodgkin lymphoma may also involve the thymus and must be differentiated from true thymomas and thymic carcinomas.

Thymoma and thymic carcinomas are rare in adults and children.[46,47] Various diseases and syndromes are associated with thymoma, including myasthenia gravis, polymyositis, systemic lupus erythematosus, rheumatoid arthritis, thyroiditis, Isaacs syndrome or neuromyotonia (continuous muscle stiffness resulting from persistent muscle activity as a consequence of antibodies against voltage-gated potassium channels), and pure red-cell aplasia.[48,49] Endocrine (hormonal) disorders including hyperthyroidism, Addison disease, and panhypopituitarism can also be associated with a diagnosis of thymoma.[50]

These neoplasms are usually located in the front part of the chest and are usually discovered during a routine chest x-ray. Symptoms can include cough, difficulty with swallowing, tightness of the chest, chest pain, and shortness of breath, though nonspecific symptoms may occur. These tumors generally are slow growing but are potentially invasive, with metastases to distant organs or lymph nodes. Staging is related to invasiveness. Surgery is performed with the goal of a complete resection.

Radiation therapy is necessary for patients with invasive thymoma or thymic carcinoma, even with a complete resection.[50] Chemotherapy is usually reserved for patients with advanced-stage disease who have not responded to radiation therapy or corticosteroids. Agents that have been effective include doxorubicin, cisplatin, and paclitaxel.[50-52] The prognosis for patients with invasive thymoma or thymic carcinoma usually is poor, though significantly higher rates of survival have been reported for patients with tumors that are not locally invasive. (Refer to the PDQ summary on adult Thymoma and Thymic Carcinoma Treatment 19 for more information.)

Researchers have described a group of young patients with midline carcinomas with a very poor prognosis. The tumors arise in midline epithelial structures including the thymus, mediastinum, airway structures, and bladder. They exhibit squamous differentiation. Tumors from 8 of 11 patients exhibited a balanced chromosomal translocation t(15;19) involving the BRD4 and NUT genes. These patients had no response to chemotherapy and died very quickly. Tumors from the remaining three patients had a chromosomal break in the NUT gene on chromosome 15 but had normal chromosome 19. These patients were older and had a slightly longer survival than the eight patients exhibiting t(15;19).[53]

Tumors of the Heart

The most common tumors of the heart are benign and include myxomas, rhabdomyomas, and neurofibromas (i.e., tumors of the nerves that innervate the muscles).[54-56] Primary tumors of the heart may include benign and malignant teratomas, rhabdomyosarcomas, hemangiomas, and chondrosarcomas. Multiple cardiac tumors noted in the fetal or neonatal period are highly associated with a diagnosis of tuberous sclerosis.[54] In a retrospective review of 94 patients with cardiac tumors detected by prenatal or neonatal echocardiography, 68% of the patients exhibited features of tuberous sclerosis.[57] In another study, 79% (15out of 19) of patients with rhabdomyomas discovered prenatally had tuberous sclerosis, while 96% of those diagnosed postnatally had tuberous sclerosis. Most rhabdomyomas, whether diagnosed prenatally or postnatally, will spontaneously regress.[58] Other tumors of the heart can include metastatic spread of rhabdomyosarcoma, melanoma, leukemia, and carcinoma of other sites. Symptoms include abnormalities of heart rhythm, enlargement of the heart, fluid in the pericardial sac, and congestive heart failure. Successful treatment may require surgery, which may include transplantation, and chemotherapy appropriate for the type of cancer that is present.[59,60]

Mesothelioma

Mesothelioma is extremely rare in childhood with only 2% to 5% of patients presenting during the first two decades of life.[61]

This tumor can involve the membranous coverings of the lung, the heart, or the abdominal organs.[62] These tumors can spread over the surface of organs, without invading far into the underlying tissue, and may spread to regional or distant lymph nodes. Mesothelioma may develop after successful treatment of an earlier cancer, especially after treatment with radiation.[63,64] In adults, these tumors have been associated with exposure to asbestos, which was used as building insulation.[65] The amount of exposure required to develop cancer is unknown, and there is no information about the risk for children exposed to asbestos.

Benign and malignant mesotheliomas cannot be differentiated using histologic criteria. A poor prognosis is associated with lesions that are diffuse and invasive or for those that recur. In general, the course of the disease is slow, and long-term survival is common. Diagnostic thoracoscopy should be considered in suspicious cases to confirm diagnosis.[61] Radical surgical resection has been attempted with mixed results.[66] Treatment with various chemotherapeutic agents used for carcinomas or sarcomas may result in partial responses.[67] Pain is an infrequent symptom; however, radiation therapy may be used for palliation of pain.

Papillary serous carcinoma of the peritoneum is sometimes mistaken for mesothelioma.[68] This tumor generally involves all surfaces lining the abdominal organs, including the surfaces of the ovary. Treatment includes surgical resection whenever possible and use of chemotherapy with agents such as cisplatin, carboplatin, and paclitaxel. (Refer to the PDQ summary on adult Malignant Mesothelioma Treatment 20 for more information.)

References

  1. Santen RJ, Mansel R: Benign breast disorders. N Engl J Med 353 (3): 275-85, 2005.  [PUBMED Abstract]

  2. Valdes EK, Boolbol SK, Cohen JM, et al.: Malignant transformation of a breast fibroadenoma to cystosarcoma phyllodes: case report and review of the literature. Am Surg 71 (4): 348-53, 2005.  [PUBMED Abstract]

  3. Serour F, Gilad A, Kopolovic J, et al.: Secretory breast cancer in childhood and adolescence: report of a case and review of the literature. Med Pediatr Oncol 20 (4): 341-4, 1992.  [PUBMED Abstract]

  4. Drukker BH: Breast disease: a primer on diagnosis and management. Int J Fertil Womens Med 42 (5): 278-87, 1997 Sep-Oct.  [PUBMED Abstract]

  5. Rogers DA, Lobe TE, Rao BN, et al.: Breast malignancy in children. J Pediatr Surg 29 (1): 48-51, 1994.  [PUBMED Abstract]

  6. Rivera-Hueto F, Hevia-Vázquez A, Utrilla-Alcolea JC, et al.: Long-term prognosis of teenagers with breast cancer. Int J Surg Pathol 10 (4): 273-9, 2002.  [PUBMED Abstract]

  7. Kaste SC, Hudson MM, Jones DJ, et al.: Breast masses in women treated for childhood cancer: incidence and screening guidelines. Cancer 82 (4): 784-92, 1998.  [PUBMED Abstract]

  8. Costa NM, Rodrigues H, Pereira H, et al.: Secretory breast carcinoma--case report and review of the medical literature. Breast 13 (4): 353-5, 2004.  [PUBMED Abstract]

  9. Metayer C, Lynch CF, Clarke EA, et al.: Second cancers among long-term survivors of Hodgkin's disease diagnosed in childhood and adolescence. J Clin Oncol 18 (12): 2435-43, 2000.  [PUBMED Abstract]

  10. Swerdlow AJ, Barber JA, Hudson GV, et al.: Risk of second malignancy after Hodgkin's disease in a collaborative British cohort: the relation to age at treatment. J Clin Oncol 18 (3): 498-509, 2000.  [PUBMED Abstract]

  11. van Leeuwen FE, Klokman WJ, Veer MB, et al.: Long-term risk of second malignancy in survivors of Hodgkin's disease treated during adolescence or young adulthood. J Clin Oncol 18 (3): 487-97, 2000.  [PUBMED Abstract]

  12. Vadasz P, Palffy G, Egervary M, et al.: Diagnosis and treatment of bronchial carcinoid tumors: clinical and pathological review of 120 operated patients. Eur J Cardiothorac Surg 7 (1): 8-11, 1993.  [PUBMED Abstract]

  13. Kulke MH, Mayer RJ: Carcinoid tumors. N Engl J Med 340 (11): 858-68, 1999.  [PUBMED Abstract]

  14. Oliaro A, Filosso PL, Donati G, et al.: Atypical bronchial carcinoids. Review of 46 patients. J Cardiovasc Surg (Torino) 41 (1): 131-5, 2000.  [PUBMED Abstract]

  15. Moraes TJ, Langer JC, Forte V, et al.: Pediatric pulmonary carcinoid: a case report and review of the literature. Pediatr Pulmonol 35 (4): 318-22, 2003.  [PUBMED Abstract]

  16. Al-Qahtani AR, Di Lorenzo M, Yazbeck S: Endobronchial tumors in children: Institutional experience and literature review. J Pediatr Surg 38 (5): 733-6, 2003.  [PUBMED Abstract]

  17. Lal DR, Clark I, Shalkow J, et al.: Primary epithelial lung malignancies in the pediatric population. Pediatr Blood Cancer 45 (5): 683-6, 2005.  [PUBMED Abstract]

  18. Lack EE, Harris GB, Eraklis AJ, et al.: Primary bronchial tumors in childhood. A clinicopathologic study of six cases. Cancer 51 (3): 492-7, 1983.  [PUBMED Abstract]

  19. Soga J, Yakuwa Y: Bronchopulmonary carcinoids: An analysis of 1,875 reported cases with special reference to a comparison between typical carcinoids and atypical varieties. Ann Thorac Cardiovasc Surg 5 (4): 211-9, 1999.  [PUBMED Abstract]

  20. Fauroux B, Aynie V, Larroquet M, et al.: Carcinoid and mucoepidermoid bronchial tumours in children. Eur J Pediatr 164 (12): 748-52, 2005.  [PUBMED Abstract]

  21. Ahel V, Zubovic I, Rozmanic V: Bronchial adenoid cystic carcinoma with saccular bronchiectasis as a cause of recurrent pneumonia in children. Pediatr Pulmonol 12 (4): 260-2, 1992.  [PUBMED Abstract]

  22. Gaissert HA, Mathisen DJ, Grillo HC, et al.: Tracheobronchial sleeve resection in children and adolescents. J Pediatr Surg 29 (2): 192-7; discussion 197-8, 1994.  [PUBMED Abstract]

  23. Jalal A, Jeyasingham K: Bronchoplasty for malignant and benign conditions: a retrospective study of 44 cases. Eur J Cardiothorac Surg 17 (4): 370-6, 2000.  [PUBMED Abstract]

  24. Hill DA, Jarzembowski JA, Priest JR, et al.: Type I pleuropulmonary blastoma: pathology and biology study of 51 cases from the international pleuropulmonary blastoma registry. Am J Surg Pathol 32 (2): 282-95, 2008.  [PUBMED Abstract]

  25. Priest JR, Hill DA, Williams GM, et al.: Type I pleuropulmonary blastoma: a report from the International Pleuropulmonary Blastoma Registry. J Clin Oncol 24 (27): 4492-8, 2006.  [PUBMED Abstract]

  26. Miniati DN, Chintagumpala M, Langston C, et al.: Prenatal presentation and outcome of children with pleuropulmonary blastoma. J Pediatr Surg 41 (1): 66-71, 2006.  [PUBMED Abstract]

  27. Shivastava R, Saha A, Mehera B, et al.: Pleuropulmonary blastoma: transition from type I (cystic) to type III (solid). Singapore Med J 48 (7): e190-2, 2007.  [PUBMED Abstract]

  28. Priest JR, Magnuson J, Williams GM, et al.: Cerebral metastasis and other central nervous system complications of pleuropulmonary blastoma. Pediatr Blood Cancer 49 (3): 266-73, 2007.  [PUBMED Abstract]

  29. Pleuropulmonary Blastoma Registry. St. Paul, Minn: Children's Hospitals and Clinics of St. Paul. Available online 21. Last accessed November 6, 2008. 

  30. Hasiotou M, Polyviou P, Strantzia CM, et al.: Pleuropulmonary blastoma in the area of a previously diagnosed congenital lung cyst: report of two cases. Acta Radiol 45 (3): 289-92, 2004.  [PUBMED Abstract]

  31. Dosios T, Stinios J, Nicolaides P, et al.: Pleuropulmonary blastoma in childhood. A malignant degeneration of pulmonary cysts. Pediatr Surg Int 20 (11-12): 863-5, 2004.  [PUBMED Abstract]

  32. Vargas SO, Korpershoek E, Kozakewich HP, et al.: Cytogenetic and p53 profiles in congenital cystic adenomatoid malformation: insights into its relationship with pleuropulmonary blastoma. Pediatr Dev Pathol 9 (3): 190-5, 2006 May-Jun.  [PUBMED Abstract]

  33. Indolfi P, Bisogno G, Casale F, et al.: Prognostic factors in pleuro-pulmonary blastoma. Pediatr Blood Cancer 48 (3): 318-23, 2007.  [PUBMED Abstract]

  34. Taube JM, Griffin CA, Yonescu R, et al.: Pleuropulmonary blastoma: cytogenetic and spectral karyotype analysis. Pediatr Dev Pathol 9 (6): 453-61, 2006 Nov-Dec.  [PUBMED Abstract]

  35. de Krijger RR, Claessen SM, van der Ham F, et al.: Gain of chromosome 8q is a frequent finding in pleuropulmonary blastoma. Mod Pathol 20 (11): 1191-9, 2007.  [PUBMED Abstract]

  36. Quilichini B, Andre N, Bouvier C, et al.: Hidden chromosomal abnormalities in pleuropulmonary blastomas identified by multiplex FISH. BMC Cancer 6: 4, 2006.  [PUBMED Abstract]

  37. Indolfi P, Casale F, Carli M, et al.: Pleuropulmonary blastoma: management and prognosis of 11 cases. Cancer 89 (6): 1396-401, 2000.  [PUBMED Abstract]

  38. Schmaltz C, Sauter S, Opitz O, et al.: Pleuro-pulmonary blastoma: a case report and review of the literature. Med Pediatr Oncol 25 (6): 479-84, 1995.  [PUBMED Abstract]

  39. de Castro CG Jr, de Almeida SG, Gregianin LJ, et al.: High-dose chemotherapy and autologous peripheral blood stem cell rescue in a patient with pleuropulmonary blastoma. J Pediatr Hematol Oncol 25 (1): 78-81, 2003.  [PUBMED Abstract]

  40. Priest JR, McDermott MB, Bhatia S, et al.: Pleuropulmonary blastoma: a clinicopathologic study of 50 cases. Cancer 80 (1): 147-61, 1997.  [PUBMED Abstract]

  41. Bouron-Dal Soglio D, Harvey I, Yazbeck S, et al.: An association of pleuropulmonary blastoma and cystic nephroma: possible genetic association. Pediatr Dev Pathol 9 (1): 61-4, 2006 Jan-Feb.  [PUBMED Abstract]

  42. Boman F, Hill DA, Williams GM, et al.: Familial association of pleuropulmonary blastoma with cystic nephroma and other renal tumors: a report from the International Pleuropulmonary Blastoma Registry. J Pediatr 149 (6): 850-854, 2006.  [PUBMED Abstract]

  43. Gangopadhyay AN, Mohanty PK, Gopal SC, et al.: Adenocarcinoma of the esophagus in an 8-year-old boy. J Pediatr Surg 32 (8): 1259-60, 1997.  [PUBMED Abstract]

  44. Verley JM, Hollmann KH: Thymoma. A comparative study of clinical stages, histologic features, and survival in 200 cases. Cancer 55 (5): 1074-86, 1985.  [PUBMED Abstract]

  45. Hsueh C, Kuo TT, Tsang NM, et al.: Thymic lymphoepitheliomalike carcinoma in children: clinicopathologic features and molecular analysis. J Pediatr Hematol Oncol 28 (12): 785-90, 2006.  [PUBMED Abstract]

  46. Furman WL, Buckley PJ, Green AA, et al.: Thymoma and myasthenia gravis in a 4-year-old child. Case report and review of the literature. Cancer 56 (11): 2703-6, 1985.  [PUBMED Abstract]

  47. Yaris N, Nas Y, Cobanoglu U, et al.: Thymic carcinoma in children. Pediatr Blood Cancer 47 (2): 224-7, 2006.  [PUBMED Abstract]

  48. Souadjian JV, Enriquez P, Silverstein MN, et al.: The spectrum of diseases associated with thymoma. Coincidence or syndrome? Arch Intern Med 134 (2): 374-9, 1974.  [PUBMED Abstract]

  49. Coulter D, Gold S: Thymoma in the offspring of a patient with Isaacs syndrome. J Pediatr Hematol Oncol 29 (11): 797-8, 2007.  [PUBMED Abstract]

  50. Cowen D, Richaud P, Mornex F, et al.: Thymoma: results of a multicentric retrospective series of 149 non-metastatic irradiated patients and review of the literature. FNCLCC trialists. Fédération Nationale des Centres de Lutte Contre le Cancer. Radiother Oncol 34 (1): 9-16, 1995.  [PUBMED Abstract]

  51. Carlson RW, Dorfman RF, Sikic BI: Successful treatment of metastatic thymic carcinoma with cisplatin, vinblastine, bleomycin, and etoposide chemotherapy. Cancer 66 (10): 2092-4, 1990.  [PUBMED Abstract]

  52. Niehues T, Harms D, Jürgens H, et al.: Treatment of pediatric malignant thymoma: long-term remission in a 14-year-old boy with EBV-associated thymic carcinoma by aggressive, combined modality treatment. Med Pediatr Oncol 26 (6): 419-24, 1996.  [PUBMED Abstract]

  53. French CA, Kutok JL, Faquin WC, et al.: Midline carcinoma of children and young adults with NUT rearrangement. J Clin Oncol 22 (20): 4135-9, 2004.  [PUBMED Abstract]

  54. Isaacs H Jr: Fetal and neonatal cardiac tumors. Pediatr Cardiol 25 (3): 252-73, 2004 May-Jun.  [PUBMED Abstract]

  55. Elderkin RA, Radford DJ: Primary cardiac tumours in a paediatric population. J Paediatr Child Health 38 (2): 173-7, 2002.  [PUBMED Abstract]

  56. Uzun O, Wilson DG, Vujanic GM, et al.: Cardiac tumours in children. Orphanet J Rare Dis 2: 11, 2007.  [PUBMED Abstract]

  57. Tworetzky W, McElhinney DB, Margossian R, et al.: Association between cardiac tumors and tuberous sclerosis in the fetus and neonate. Am J Cardiol 92 (4): 487-9, 2003.  [PUBMED Abstract]

  58. Bader RS, Chitayat D, Kelly E, et al.: Fetal rhabdomyoma: prenatal diagnosis, clinical outcome, and incidence of associated tuberous sclerosis complex. J Pediatr 143 (5): 620-4, 2003.  [PUBMED Abstract]

  59. Michler RE, Goldstein DJ: Treatment of cardiac tumors by orthotopic cardiac transplantation. Semin Oncol 24 (5): 534-9, 1997.  [PUBMED Abstract]

  60. Stiller B, Hetzer R, Meyer R, et al.: Primary cardiac tumours: when is surgery necessary? Eur J Cardiothorac Surg 20 (5): 1002-6, 2001.  [PUBMED Abstract]

  61. Nagata S, Nakanishi R: Malignant pleural mesothelioma with cavity formation in a 16-year-old boy. Chest 127 (2): 655-7, 2005.  [PUBMED Abstract]

  62. Kelsey A: Mesothelioma in childhood. Pediatr Hematol Oncol 11 (5): 461-2, 1994 Sep-Oct.  [PUBMED Abstract]

  63. Hofmann J, Mintzer D, Warhol MJ: Malignant mesothelioma following radiation therapy. Am J Med 97 (4): 379-82, 1994.  [PUBMED Abstract]

  64. Pappo AS, Santana VM, Furman WL, et al.: Post-irradiation malignant mesothelioma. Cancer 79 (1): 192-3, 1997.  [PUBMED Abstract]

  65. Hyers TM, Ohar JM, Crim C: Clinical controversies in asbestos-induced lung diseases. Semin Diagn Pathol 9 (2): 97-101, 1992.  [PUBMED Abstract]

  66. Maziak DE, Gagliardi A, Haynes AE, et al.: Surgical management of malignant pleural mesothelioma: a systematic review and evidence summary. Lung Cancer 48 (2): 157-69, 2005.  [PUBMED Abstract]

  67. Milano E, Pourroy B, Rome A, et al.: Efficacy of a combination of pemetrexed and multiple redo-surgery in an 11-year-old girl with a recurrent multifocal abdominal mesothelioma. Anticancer Drugs 17 (10): 1231-4, 2006.  [PUBMED Abstract]

  68. Wall JE, Mandrell BN, Jenkins JJ 3rd, et al.: Effectiveness of paclitaxel in treating papillary serous carcinoma of the peritoneum in an adolescent. Am J Obstet Gynecol 172 (3): 1049-52, 1995.  [PUBMED Abstract]

Abdominal Cancers

Note: Some citations in the text of this section are followed by a level of evidence. The PDQ editorial boards use a formal ranking system to help the reader judge the strength of evidence linked to the reported results of a therapeutic strategy. (Refer to the PDQ summary on Levels of Evidence 2 for more information.)

Abdominal cancers include adrenocortical tumors, carcinomas of the stomach, cancer of the pancreas, colorectal carcinomas, carcinoid tumors, and gastrointestinal stromal cell tumors. The prognosis, diagnosis, classification, and treatment of these abdominal cancers are discussed below.  [Note: Refer to the Renal Cell Carcinoma section in the PDQ summary on Wilms Tumor and Other Childhood Kidney Tumors 22 for more information.]

Carcinoma of the Adrenal Cortex

Adrenocortical tumors are classified as carcinomas and adenomas.[1-5] Adrenocortical tumors may be hormonally active or inactive. Adenomas are generally benign, whereas adrenocortical carcinomas frequently secrete hormones and may cause the patient to develop masculine traits, irrespective of the patient’s gender. Pediatric patients with adrenocortical carcinoma often have Li-Fraumeni syndrome, which is an inherited condition that predisposes family members to multiple cancers, including breast cancer, rhabdomyosarcoma, and osteosarcoma.[6] A variety of p53 mutations associated with Li-Fraumeni syndrome have been observed in North American children with adrenocortical carcinoma, whereas in a southern Brazilian population, a distinctive p53 mutation predisposes to this disease.[1,7] Children with Beckwith-Wiedemann syndrome [8] or hemihypertrophy [9] are at an increased risk of developing carcinoma of the adrenal cortex (as well as Wilms tumor, hepatoblastoma, and other rare cancers) in the first several years of life.

These tumors spread locally to the lymph nodes and can also involve the kidneys, lungs, bones, and brain.[10] Surgical removal should be attempted but may not always be possible if the tumor has spread widely. Additional treatment may include the use of an artificial hormone that blocks the masculinizing effects of the tumor[11] or chemotherapy using cisplatin, 5-fluorouracil (5-FU), and etoposide.[4,12] A retrospective analysis in Italy and Germany identified 177 patients with adrenocortical carcinoma. Recurrence-free survival was significantly prolonged by the use of adjuvant mitotane. Benefit was present with 1 to 3 grams per day of mitotane and was associated with fewer toxic side effects than doses of 3 to 5 grams per day.[13,14] The prognosis for patients who have small, completely resected tumors generally is excellent, but prognosis can be poor for patients who have large primary tumors or metastatic disease at diagnosis.[3,15] Tumor stage has been identified as a significant prognostic factor in children with adrenocortical tumors. When possible, surgical reexcision should be attempted for local tumor recurrences and for inferior vena caval tumor invasion.[16] Adrenal tumors can present as incidental findings (incidentalomas), and these tumors should be thoroughly evaluated.[17] (Refer to the PDQ summary on adult Adrenocortical Carcinoma Treatment 23 for more information.)

Treatment options under clinical evaluation
  • ARAR0332 24: This Children's Oncology Group trial is evaluating the treatment of adrenocortical tumors with surgery and lymph node dissection. Patients with advanced disease will receive multiagent chemotherapy. Patients with stage I or stage II disease will have resection and retroperitoneal lymph node sampling (I) or dissection (II). Patients with stage III and stage IV disease will receive chemotherapy before resection. The chemotherapy is cisplatin, doxorubicin, etoposide, and oral mitotane.
Carcinoma of the Stomach

The frequency and death rate from stomach cancer has declined worldwide for the past 50 years with the introduction of food preservation practices such as refrigeration.[18] The tumor must be distinguished from other conditions such as non-Hodgkin lymphoma, malignant carcinoid, leiomyosarcoma, and various benign conditions or tumors of the stomach. Symptoms include vague upper abdominal pain, which can be associated with poor appetite, and weight loss. Many individuals become anemic but otherwise show no symptoms before the development of metastatic spread. Other symptoms may include nausea, vomiting, change in bowel habits, poor appetite, weakness, and Helicobacter pylori infection.[19] Fiberoptic endoscopy can be used to visualize the tumor or to take a biopsy sample to confirm the diagnosis. Confirmation can also involve an x-ray examination of the upper gastrointestinal tract.

Treatment should include surgical excision with wide margins. For individuals who cannot have a complete surgical resection, radiation therapy may be used along with chemotherapeutic agents such as 5-FU and irinotecan.[20] Other agents that may be of value are the nitrosoureas with or without cisplatin, etoposide, doxorubicin, or mitomycin C.

Prognosis depends on the extent of the disease at the time of diagnosis and the success of treatment that is appropriate for the clinical situation.[21] Because of the rarity of stomach cancer in the pediatric age group, little information exists regarding the treatment outcomes of children. (Refer to the PDQ summary on adult Gastric Cancer Treatment 25 for more information.)

Cancer of the Pancreas

Pancreatic tumors are rare in children and adolescents.[22] Tumors included in this general category can arise at any site within the pancreas. Cancers of the pancreas may be classified as adenocarcinomas, squamous cell carcinomas, acinic cell carcinomas, liposarcomas, lymphomas, papillary-cystic carcinomas, pancreatoblastomas, malignant insulinomas, glucagonomas, and gastrinomas.[23-25] Several cases of primitive neuroectodermal tumor of the pancreas have been reported in children and young adults.[26] Most pancreatic tumors do not secrete hormones, though some tumors secrete insulin, which can lead to symptoms of weakness, fatigue, hypoglycemia, and coma.[23,27] If the tumor interferes with the normal function of the islet cells, patients may have watery diarrhea or abnormalities of salt balance. Both carcinoma of the pancreas and pancreatoblastoma can produce active hormones and can be associated with abdominal mass, wasting, and pain.[28-30] At times, there is obstruction of the head of the pancreas, which is associated with jaundice and gastrointestinal bleeding. Elevation of alpha-fetoprotein has been seen in pancreatoblastoma.[31,32] Pancreatoblastoma is reported to be associated with Beckwith-Wiedemann syndrome and Cushing syndrome.[33,34]

Solid pseudopapillary neoplasm of the pancreas is a rare tumor of borderline malignancy that has been reported in children but more commonly occurs in young women.[35,36] Treatment consists of complete tumor resection (ideally without biopsy). Metastases may occur, but in general, prognosis is good following surgery alone.[37,38]

Diagnosis of pancreatic tumors is usually established by biopsy, using laparotomy or a minimally invasive surgery (e.g., laparoscopy). A diagnosis can be achieved only after ruling out various benign and cancerous lesions. Treatment includes various surgical procedures to remove the pancreas and duodenum or removal of part of the pancreas. Complete resection is usually possible and long-term survival is likely, though pancreatoblastoma has a high recurrence rate.[24,31] For pediatric patients, the effectiveness of radiation therapy is not known. Chemotherapy may be useful for treatment of localized or metastatic pancreatic carcinoma. The combination of cisplatin and doxorubicin has produced responses in pancreatoblastoma prior to tumor resection.[39,40] Postoperative treatment with cisplatin, doxorubicin, ifosfamide, and etoposide has also produced responses in patients with pancreatoblastoma.[41][Level of evidence: 3iiiA] Other agents that may be of value include 5-FU, streptozotocin, mitomycin C, carboplatin, gemcitabine, and irinotecan. Response rates and survival rates generally are not good. (Refer to the PDQ summary on adult Pancreatic Cancer Treatment 26 for more information.)

Colorectal Carcinoma

Carcinoma of the large bowel is rare in the pediatric age group; it is seen in only one per 1 million persons younger than 20 years in the United States annually.[42] Forty percent to 60% of the tumors arise on the right side in children in contrast to adults who have a prevalence of tumors on the left side.[43] Colon cancer in children is often associated with a family cancer syndrome.[44,45] There is an increasing risk of colorectal carcinoma in members of families with a family history of intestinal polyposis, which can lead to the development of multiple adenomatous polyps.[46] Juvenile polyps are not associated with an increased incidence or risk of cancer.

Familial polyposis is inherited as a dominant trait, which confers a high degree of risk. Early diagnosis and surgical removal of the colon eliminate the risk of developing carcinomas of the large bowel.[47] Some colorectal carcinomas in young people, however, may be associated with a mutation of the adenomatous polyposis coli (APC) gene, which also is associated with an increased risk of brain tumors and hepatoblastoma.[48] The familial APC syndrome is caused by mutation of a gene on chromosome 5q, which normally suppresses proliferation of cells lining the intestine and later development of polyps.[49] Another tumor suppressor gene on chromosome 18 is associated with progression of polyps to malignant form. Multiple colon carcinomas have also been associated with progression of polyps to a malignant form. Multiple colon carcinomas have been associated with neurofibromatosis type I and several other rarer syndromes.[50]

The histologic types of colorectal cancer include adenocarcinomas, mucinous or colloid adenocarcinomas, signet ring adenocarcinomas, and scirrhous tumors. Most tumors in the pediatric age group are mucin-producing carcinomas,[51][Level of evidence: 3iii][52][Level of evidence: 3iiA] whereas only about 15% of adult lesions are of this histology. The tumors of younger patients with this histologic variant may be less responsive to chemotherapy. These tumors arise from the surface of the bowel, usually at the site of an adenomatous polyp. The tumor may extend into the muscle layer surrounding the bowel, or the tumor may perforate the bowel entirely and seed through the spaces around the bowel, including intra-abdominal fat, lymph nodes, liver, ovaries, and the surface of other loops of bowel. A high incidence of metastasis involving the pelvis, ovaries, or both may be present in girls.[53]

Colorectal carcinoma usually presents with symptoms related to the site of the tumor.[51][Level of evidence: 3iii] Changes in bowel habits are associated with tumors of the rectum or lower colon. Tumors of the right colon may cause more subtle symptoms but are often associated with an abdominal mass, weight loss, decreased appetite, and blood in the stool. Any tumor that causes complete obstruction of the large bowel can cause bowel perforation and spread of the tumor cells within the abdominal cavity.

Because of its rarity, colorectal carcinoma is rarely diagnosed in a pediatric patient; however, vague gastrointestinal symptoms should alert the physician to investigate this possibility. Diagnostic studies that may be of value include examination of the stool for blood, studies of liver and kidney function, measurement of carcinoembryonic antigen, and various medical imaging studies, including direct examination using colonoscopy to detect polyps in the large bowel. Other conventional radiographic studies include barium enema followed by computed tomography of the chest and bone scans.[53,54]

Most patients present with evidence of metastatic disease,[51][Level of evidence: 3iii] either as gross tumor or as microscopic deposits in lymph nodes, on the surface of the bowel, or on intra-abdominal organs.[55] Complete surgical excision should be the primary aim of the surgeon, but in most instances this is impossible; removal of large portions of tumor provides little benefit for the individuals with extensive metastatic disease. Most patients with microscopic metastatic disease generally develop gross metastatic disease, and few individuals with metastatic disease at diagnosis become long-term survivors.

Current therapy includes the use of radiation for rectal and lower-colon tumors, in conjunction with chemotherapy using 5-FU with leucovorin.[56] Other agents that may be of value include irinotecan.[51][Level of evidence: 3iiiA] No significant benefit has been determined for interferon-alpha given in conjunction with 5-FU/leucovorin.[57] (Refer to the PDQ summaries on adult Colon Cancer Treatment 27 and Rectal Cancer Treatment 28 for more information.)

Carcinoid Tumors

These tumors, like bronchial adenomas, may be benign or malignant and can involve the lining of the lung or the large or small bowel.[58-62] Most lung lesions are benign; however, some metastasize.[63] Most carcinoid tumors of the appendix are discovered incidentally at the time of appendectomy, and are small, localized tumors; simple appendectomy is the therapy of choice.[64] For larger (>2 cm) tumors or tumors that have spread to local nodes, cecectomy or rarely, right hemicolectomy, is the usual treatment. It has become accepted practice to remove the entire right colon in patients with large carcinoid tumors of the appendix (>2 cm in diameter) or with tumors that have spread to the nodes; however, this practice remains controversial.[65] Treatment of metastatic carcinoid tumors of the large bowel or stomach becomes more complicated and requires treatment similar to that given for colorectal carcinoma. The carcinoid syndrome of excessive excretion of somatostatin is characterized by flushing, labile blood pressure, and metastatic spread of the tumor to the liver.[63] Symptoms may be lessened by giving somatostatin analogs, which are available in short-acting and long-acting forms.[66] (Refer to the PDQ summary on adult Gastrointestinal Carcinoid Tumors 29 for more information.)

Gastrointestinal Stromal Cell Tumor

Gastrointestinal stromal cell tumor (GIST) is a mesenchymal tumor of the intestinal tract that typically occurs in adults older than 40 years. These tumors are rare in children. Only 1.4% of all patients with GISTs are children and young adults.[67] Many of these tumors were previously diagnosed as leiomyomas, leiomyosarcomas, and leiomyoblastomas. Younger patients with GISTs are usually female and commonly present in the second decade of life with anemia-related gastrointestinal bleeding. In children, most tumors are in the stomach and may be localized or multifocal. Carney triad is associated with gastric GIST in a small number of children and in an association with extra-adrenal paraganglioma and pulmonary chondroma. Some patients may present with GIST as the first manifestation of Carney triad and for this reason, a diagnosis of Carney triad should be considered in pediatric patients presenting with GIST.[68] A familial variant of pediatric GIST has not been established. The association of KIT mutations in adults with GIST is as high as 90%; imatinib mesylate has been found to be effective therapy in these patients. GIST in adolescents and young adults has a heterogeneous presentation; some patients have tumors with mutations in KIT or PDGFA, which suggests that these tumors may respond to imatinib mesylate.[69,70] Pediatric GIST is probably biologically different from adult GIST, since KIT and PDGFA mutations are rarely detected in pediatric GIST tumors.[71] Clinical response to targeted therapies such as imatinib mesylate has not been proven. Complete surgical resection of localized disease should be the initial treatment in pediatric GIST. No chemotherapy regimen has been effective in the treatment of GIST, and in the absence of mutations in KIT, imatinib mesylate is not recommended as adjuvant therapy. The clinical course of GIST is variable, with an excellent prognosis in children with localized disease and an indolent, slowly progressive course in children with liver and lymph node metastatic disease. Because of the variable expression of mutations in KIT or PDGFA, all patients with GISTs should have their tumors examined for these mutations.[67,69,70]

References

  1. Rodriguez-Galindo C, Figueiredo BC, Zambetti GP, et al.: Biology, clinical characteristics, and management of adrenocortical tumors in children. Pediatr Blood Cancer 45 (3): 265-73, 2005.  [PUBMED Abstract]

  2. Wieneke JA, Thompson LD, Heffess CS: Adrenal cortical neoplasms in the pediatric population: a clinicopathologic and immunophenotypic analysis of 83 patients. Am J Surg Pathol 27 (7): 867-81, 2003.  [PUBMED Abstract]

  3. Michalkiewicz E, Sandrini R, Figueiredo B, et al.: Clinical and outcome characteristics of children with adrenocortical tumors: a report from the International Pediatric Adrenocortical Tumor Registry. J Clin Oncol 22 (5): 838-45, 2004.  [PUBMED Abstract]

  4. Ribeiro RC, Figueiredo B: Childhood adrenocortical tumours. Eur J Cancer 40 (8): 1117-26, 2004.  [PUBMED Abstract]

  5. Rescorla FJ: Malignant adrenal tumors. Semin Pediatr Surg 15 (1): 48-56, 2006.  [PUBMED Abstract]

  6. Li FP, Fraumeni JF Jr, Mulvihill JJ, et al.: A cancer family syndrome in twenty-four kindreds. Cancer Res 48 (18): 5358-62, 1988.  [PUBMED Abstract]

  7. Figueiredo BC, Sandrini R, Zambetti GP, et al.: Penetrance of adrenocortical tumours associated with the germline TP53 R337H mutation. J Med Genet 43 (1): 91-6, 2006.  [PUBMED Abstract]

  8. DeBaun MR, Tucker MA: Risk of cancer during the first four years of life in children from The Beckwith-Wiedemann Syndrome Registry. J Pediatr 132 (3 Pt 1): 398-400, 1998.  [PUBMED Abstract]

  9. Hoyme HE, Seaver LH, Jones KL, et al.: Isolated hemihyperplasia (hemihypertrophy): report of a prospective multicenter study of the incidence of neoplasia and review. Am J Med Genet 79 (4): 274-8, 1998.  [PUBMED Abstract]

  10. Wagner AS, Fleitz JM, Kleinschmidt-Demasters BK: Pediatric adrenal cortical carcinoma: brain metastases and relationship to NF-1, case reports and review of the literature. J Neurooncol 75 (2): 127-33, 2005.  [PUBMED Abstract]

  11. Bugg MF, Ribeiro RC, Roberson PK, et al.: Correlation of pathologic features with clinical outcome in pediatric adrenocortical neoplasia. A study of a Brazilian population. Brazilian Group for Treatment of Childhood Adrenocortical Tumors. Am J Clin Pathol 101 (5): 625-9, 1994.  [PUBMED Abstract]

  12. Hovi L, Wikström S, Vettenranta K, et al.: Adrenocortical carcinoma in children: a role for etoposide and cisplatin adjuvant therapy? Preliminary report. Med Pediatr Oncol 40 (5): 324-6, 2003.  [PUBMED Abstract]

  13. Terzolo M, Angeli A, Fassnacht M, et al.: Adjuvant mitotane treatment for adrenocortical carcinoma. N Engl J Med 356 (23): 2372-80, 2007.  [PUBMED Abstract]

  14. Schteingart DE: Adjuvant mitotane therapy of adrenal cancer - use and controversy. N Engl J Med 356 (23): 2415-8, 2007.  [PUBMED Abstract]

  15. Mayer SK, Oligny LL, Deal C, et al.: Childhood adrenocortical tumors: case series and reevaluation of prognosis--a 24-year experience. J Pediatr Surg 32 (6): 911-5, 1997.  [PUBMED Abstract]

  16. Tucci S Jr, Martins AC, Suaid HJ, et al.: The impact of tumor stage on prognosis in children with adrenocortical carcinoma. J Urol 174 (6): 2338-42, discussion 2342, 2005.  [PUBMED Abstract]

  17. Shen WT, Sturgeon C, Duh QY: From incidentaloma to adrenocortical carcinoma: the surgical management of adrenal tumors. J Surg Oncol 89 (3): 186-92, 2005.  [PUBMED Abstract]

  18. American Cancer Society.: Cancer Facts and Figures-2000. Atlanta, Ga: American Cancer Society, 2000. 

  19. Rowland M, Drumm B: Helicobacter pylori infection and peptic ulcer disease in children. Curr Opin Pediatr 7 (5): 553-9, 1995.  [PUBMED Abstract]

  20. Ajani JA: Current status of therapy for advanced gastric carcinoma. Oncology (Huntingt) 12 (8 Suppl 6): 99-102, 1998.  [PUBMED Abstract]

  21. Schwartz MG, Sgaglione NA: Gastric carcinoma in the young: overview of the literature. Mt Sinai J Med 51 (6): 720-3, 1984.  [PUBMED Abstract]

  22. Chung EM, Travis MD, Conran RM: Pancreatic tumors in children: radiologic-pathologic correlation. Radiographics 26 (4): 1211-38, 2006 Jul-Aug.  [PUBMED Abstract]

  23. Vossen S, Goretzki PE, Goebel U, et al.: Therapeutic management of rare malignant pancreatic tumors in children. World J Surg 22 (8): 879-82, 1998.  [PUBMED Abstract]

  24. Shorter NA, Glick RD, Klimstra DS, et al.: Malignant pancreatic tumors in childhood and adolescence: The Memorial Sloan-Kettering experience, 1967 to present. J Pediatr Surg 37 (6): 887-92, 2002.  [PUBMED Abstract]

  25. Raffel A, Cupisti K, Krausch M, et al.: Therapeutic strategy of papillary cystic and solid neoplasm (PCSN): a rare non-endocrine tumor of the pancreas in children. Surg Oncol 13 (1): 1-6, 2004.  [PUBMED Abstract]

  26. Movahedi-Lankarani S, Hruban RH, Westra WH, et al.: Primitive neuroectodermal tumors of the pancreas: a report of seven cases of a rare neoplasm. Am J Surg Pathol 26 (8): 1040-7, 2002.  [PUBMED Abstract]

  27. Karachaliou F, Vlachopapadopoulou E, Kaldrymidis P, et al.: Malignant insulinoma in childhood. J Pediatr Endocrinol Metab 19 (5): 757-60, 2006.  [PUBMED Abstract]

  28. Schwartz MZ: Unusual peptide-secreting tumors in adolescents and children. Semin Pediatr Surg 6 (3): 141-6, 1997.  [PUBMED Abstract]

  29. Murakami T, Ueki K, Kawakami H, et al.: Pancreatoblastoma: case report and review of treatment in the literature. Med Pediatr Oncol 27 (3): 193-7, 1996.  [PUBMED Abstract]

  30. Imamura A, Nakagawa A, Okuno M, et al.: Pancreatoblastoma in an adolescent girl: case report and review of 26 Japanese cases. Eur J Surg 164 (4): 309-12, 1998.  [PUBMED Abstract]

  31. Dhebri AR, Connor S, Campbell F, et al.: Diagnosis, treatment and outcome of pancreatoblastoma. Pancreatology 4 (5): 441-51; discussion 452-3, 2004.  [PUBMED Abstract]

  32. Bendell JC, Lauwers GY, Willett C, et al.: Pancreatoblastoma in a teenage patient. Clin Adv Hematol Oncol 4 (2): 150-3; discussion 154, 2006.  [PUBMED Abstract]

  33. Muguerza R, Rodriguez A, Formigo E, et al.: Pancreatoblastoma associated with incomplete Beckwith-Wiedemann syndrome: case report and review of the literature. J Pediatr Surg 40 (8): 1341-4, 2005.  [PUBMED Abstract]

  34. Kletter GB, Sweetser DA, Wallace SF, et al.: Adrenocorticotropin-secreting pancreatoblastoma. J Pediatr Endocrinol Metab 20 (5): 639-42, 2007.  [PUBMED Abstract]

  35. Papavramidis T, Papavramidis S: Solid pseudopapillary tumors of the pancreas: review of 718 patients reported in English literature. J Am Coll Surg 200 (6): 965-72, 2005.  [PUBMED Abstract]

  36. Choi SH, Kim SM, Oh JT, et al.: Solid pseudopapillary tumor of the pancreas: a multicenter study of 23 pediatric cases. J Pediatr Surg 41 (12): 1992-5, 2006.  [PUBMED Abstract]

  37. Moholkar S, Sebire NJ, Roebuck DJ: Solid-pseudopapillary neoplasm of the pancreas: radiological-pathological correlation. Pediatr Radiol 35 (8): 819-22, 2005.  [PUBMED Abstract]

  38. Peng CH, Chen DF, Zhou GW, et al.: The solid-pseudopapillary tumor of pancreas: the clinical characteristics and surgical treatment. J Surg Res 131 (2): 276-82, 2006.  [PUBMED Abstract]

  39. Défachelles AS, Martin De Lassalle E, Boutard P, et al.: Pancreatoblastoma in childhood: clinical course and therapeutic management of seven patients. Med Pediatr Oncol 37 (1): 47-52, 2001.  [PUBMED Abstract]

  40. Yonekura T, Kosumi T, Hokim M, et al.: Aggressive surgical and chemotherapeutic treatment of advanced pancreatoblastoma associated with tumor thrombus in portal vein. J Pediatr Surg 41 (3): 596-8, 2006.  [PUBMED Abstract]

  41. Lee YJ, Hah JO: Long-term survival of pancreatoblastoma in children. J Pediatr Hematol Oncol 29 (12): 845-7, 2007.  [PUBMED Abstract]

  42. Correa P, Haenszel W: The epidemiology of large-bowel cancer. Adv Cancer Res 26: 1-141, 1978.  [PUBMED Abstract]

  43. Sharma AK, Gupta CR: Colorectal cancer in children: case report and review of literature. Trop Gastroenterol 22 (1): 36-9, 2001 Jan-Mar.  [PUBMED Abstract]

  44. Half E E, Bresalier RS: Clinical management of hereditary colorectal cancer syndromes. Curr Opin Gastroenterol 20: 32-42, 2003. 

  45. Durno C, Aronson M, Bapat B, et al.: Family history and molecular features of children, adolescents, and young adults with colorectal carcinoma. Gut 54 (8): 1146-50, 2005.  [PUBMED Abstract]

  46. Lynch HT, Fusaro RM, Lynch JF: Cancer genetics in the new era of molecular biology. Ann N Y Acad Sci 833: 1-28, 1997.  [PUBMED Abstract]

  47. Erdman SH: Pediatric adenomatous polyposis syndromes: an update. Curr Gastroenterol Rep 9 (3): 237-44, 2007.  [PUBMED Abstract]

  48. Turcot J, Despres JP, St. Pierre F: Malignant tumors of the central nervous system associated with familial polyposis of the colon: Report of two cases. Dis Colon Rectum 2: 465-468, 1959. 

  49. Vogelstein B, Fearon ER, Hamilton SR, et al.: Genetic alterations during colorectal-tumor development. N Engl J Med 319 (9): 525-32, 1988.  [PUBMED Abstract]

  50. Pratt CB, Jane JA: Multiple colorectal carcinomas, polyposis coli, and neurofibromatosis, followed by multiple glioblastoma multiforme. J Natl Cancer Inst 83 (12): 880-1, 1991.  [PUBMED Abstract]

  51. Hill DA, Furman WL, Billups CA, et al.: Colorectal carcinoma in childhood and adolescence: a clinicopathologic review. J Clin Oncol 25 (36): 5808-14, 2007.  [PUBMED Abstract]

  52. Ferrari A, Rognone A, Casanova M, et al.: Colorectal carcinoma in children and adolescents: the experience of the Istituto Nazionale Tumori of Milan, Italy. Pediatr Blood Cancer 50 (3): 588-93, 2008.  [PUBMED Abstract]

  53. Kauffman WM, Jenkins JJ 3rd, Helton K, et al.: Imaging features of ovarian metastases from colonic adenocarcinoma in adolescents. Pediatr Radiol 25 (4): 286-8, 1995.  [PUBMED Abstract]

  54. Pratt CB, Rao BN, Merchant TE, et al.: Treatment of colorectal carcinoma in adolescents and young adults with surgery, 5-fluorouracil/leucovorin/interferon-alpha 2a and radiation therapy. Med Pediatr Oncol 32 (6): 459-60, 1999.  [PUBMED Abstract]

  55. Chantada GL, Perelli VB, Lombardi MG, et al.: Colorectal carcinoma in children, adolescents, and young adults. J Pediatr Hematol Oncol 27 (1): 39-41, 2005.  [PUBMED Abstract]

  56. Madajewicz S, Petrelli N, Rustum YM, et al.: Phase I-II trial of high-dose calcium leucovorin and 5-fluorouracil in advanced colorectal cancer. Cancer Res 44 (10): 4667-9, 1984.  [PUBMED Abstract]

  57. Wolmark N, Bryant J, Smith R, et al.: Adjuvant 5-fluorouracil and leucovorin with or without interferon alfa-2a in colon carcinoma: National Surgical Adjuvant Breast and Bowel Project protocol C-05. J Natl Cancer Inst 90 (23): 1810-6, 1998.  [PUBMED Abstract]

  58. Modlin IM, Sandor A: An analysis of 8305 cases of carcinoid tumors. Cancer 79 (4): 813-29, 1997.  [PUBMED Abstract]

  59. Deans GT, Spence RA: Neoplastic lesions of the appendix. Br J Surg 82 (3): 299-306, 1995.  [PUBMED Abstract]

  60. Doede T, Foss HD, Waldschmidt J: Carcinoid tumors of the appendix in children--epidemiology, clinical aspects and procedure. Eur J Pediatr Surg 10 (6): 372-7, 2000.  [PUBMED Abstract]

  61. Quaedvlieg PF, Visser O, Lamers CB, et al.: Epidemiology and survival in patients with carcinoid disease in The Netherlands. An epidemiological study with 2391 patients. Ann Oncol 12 (9): 1295-300, 2001.  [PUBMED Abstract]

  62. Broaddus RR, Herzog CE, Hicks MJ: Neuroendocrine tumors (carcinoid and neuroendocrine carcinoma) presenting at extra-appendiceal sites in childhood and adolescence. Arch Pathol Lab Med 127 (9): 1200-3, 2003.  [PUBMED Abstract]

  63. Tormey WP, FitzGerald RJ: The clinical and laboratory correlates of an increased urinary 5-hydroxyindoleacetic acid. Postgrad Med J 71 (839): 542-5, 1995.  [PUBMED Abstract]

  64. Pelizzo G, La Riccia A, Bouvier R, et al.: Carcinoid tumors of the appendix in children. Pediatr Surg Int 17 (5-6): 399-402, 2001.  [PUBMED Abstract]

  65. Dall'Igna P, Ferrari A, Luzzatto C, et al.: Carcinoid tumor of the appendix in childhood: the experience of two Italian institutions. J Pediatr Gastroenterol Nutr 40 (2): 216-9, 2005.  [PUBMED Abstract]

  66. Delaunoit T, Rubin J, Neczyporenko F, et al.: Somatostatin analogues in the treatment of gastroenteropancreatic neuroendocrine tumors. Mayo Clin Proc 80 (4): 502-6, 2005.  [PUBMED Abstract]

  67. Prakash S, Sarran L, Socci N, et al.: Gastrointestinal stromal tumors in children and young adults: a clinicopathologic, molecular, and genomic study of 15 cases and review of the literature. J Pediatr Hematol Oncol 27 (4): 179-87, 2005.  [PUBMED Abstract]

  68. Agaram NP, Laquaglia MP, Ustun B, et al.: Molecular characterization of pediatric gastrointestinal stromal tumors. Clin Cancer Res 14 (10): 3204-15, 2008.  [PUBMED Abstract]

  69. Price VE, Zielenska M, Chilton-MacNeill S, et al.: Clinical and molecular characteristics of pediatric gastrointestinal stromal tumors (GISTs). Pediatr Blood Cancer 45 (1): 20-4, 2005.  [PUBMED Abstract]

  70. Miettinen M, Lasota J, Sobin LH: Gastrointestinal stromal tumors of the stomach in children and young adults: a clinicopathologic, immunohistochemical, and molecular genetic study of 44 cases with long-term follow-up and review of the literature. Am J Surg Pathol 29 (10): 1373-81, 2005.  [PUBMED Abstract]

  71. Janeway KA, Liegl B, Harlow A, et al.: Pediatric KIT wild-type and platelet-derived growth factor receptor alpha-wild-type gastrointestinal stromal tumors share KIT activation but not mechanisms of genetic progression with adult gastrointestinal stromal tumors. Cancer Res 67 (19): 9084-8, 2007.  [PUBMED Abstract]

Genital/Urinary Tumors

Genital/urinary tumors include carcinoma of the bladder, ovarian cancer, and carcinoma of the cervix and vagina. The prognosis, diagnosis, classification, and treatment of these genital/urinary tumors are discussed below.

Carcinoma of the Bladder

Carcinoma of the bladder is extremely rare in children. The most common carcinoma to involve the bladder is transitional cell carcinoma, which generally presents with hematuria.[1] In contrast to adults, most pediatric bladder carcinomas are low grade, superficial, and have a good prognosis following transurethral resection.[1-3] Squamous cell and more aggressive carcinomas, however, have been reported.[4,5] Bladder cancer in adolescents may develop as a consequence of alkylating-agent chemotherapy given for other childhood tumors or leukemia.[6,7] The association between cyclophosphamide and bladder cancer is the only established relationship between a specific anticancer drug and a solid tumor.[6] One of the most important risk factors for bladder cancer in adults is cigarette smoking, which may be associated with as many as 50% of these cancers in men and 33% in women.[7] (Refer to the PDQ summary on adult Bladder Cancer Treatment 30 for more information.)

Ovarian Cancer

The majority of ovarian masses in children are not neoplastic. The most common neoplasms are germ cell tumors, followed by epithelial tumors, stromal tumors, and then miscellaneous tumors such as Burkitt lymphoma.[8,9] Ovarian tumors derived from malignant epithelial elements include: adenocarcinomas,[10] cystadenocarcinomas, endometrioid tumors, clear cell tumors, and undifferentiated carcinomas. Treatment is stage-related and may include surgery, radiation, and chemotherapy with cisplatin, carboplatin, etoposide, topotecan, paclitaxel, and other agents. In one series of 19 patients younger than 21 years with epithelial ovarian neoplasms, the average age at diagnosis was 19.7 years. Dysmenorrhea and abdominal pain were the most common presenting symptoms; 79% of the patients had stage I disease with a 100% survival rate, and only those who had small cell anaplastic carcinoma died. Girls with ovarian carcinoma (epithelial ovarian neoplasia) fare better than adults with similar histology, probably because girls usually present with low-stage disease.[11] (Refer to the PDQ summaries on Childhood Extracranial Germ Cell Tumors 31, adult Ovarian Epithelial Cancer Treatment 32, and Ovarian Low Malignant Potential Tumor Cancer Treatment 33 for more information.)

Ovarian sex cord-stromal tumors are a heterogeneous group of rare tumors that derive from the gonadal nongerm cell component.[12] Histologic subtypes display some areas of gonadal differentiation and include juvenile granulosa cell tumors (JGCT), Sertoli-Leydig cell tumors, and sclerosing stromal tumors. The most common type in girls younger than 18 years is JGCT (median age, 7.6 years; range, 6 months to 17.5 years in one study).[13] JGCT represent about 5% of ovarian tumors in children and adolescents and are distinct from the granulosa cell tumors seen in adults.[12,14-16] Most patients present with precocious puberty.[17] Other presenting symptoms include abdominal pain, abdominal mass, and ascites. JGCT has been reported in children with Ollier disease and Maffucci syndrome.[18] As many as 90% of children will have low-stage disease (International Federation of Gynecology and Obstetrics [FIGO] stage I) and are usually curable with unilateral salpingo-oophorectomy alone. Patients with advanced disease (FIGO stage II–IV) and those with high mitotic activity tumors have a poorer prognosis. Use of a cisplatin-based chemotherapy regimen has been reported in both the adjuvant and recurrent disease settings with some success.[13,16,19-21]

Carcinoma of the Cervix and Vagina

Adenocarcinoma of the cervix and vagina is rare in childhood and adolescence with fewer than 50 reported cases.[22] Two-thirds of the cases are related to the exposure of diethylstilbestrol in utero. The median age at presentation is 15 years, with a range of 7 months to 18 years, and with most patients presenting with vaginal bleeding. Adults with adenocarcinoma of the cervix or vagina will present with stage I or stage II disease 90% of the time. In children and adolescents, there is a high incidence of stage III and stage IV disease (24%). This difference may be explained by the practice of routine pelvic examinations in adults and the hesitancy to perform pelvic exams in children. The treatment of choice is surgical resection [23] followed by radiation therapy for residual microscopic disease or lymphatic metastases. The role of chemotherapy in management is unknown, though drugs commonly used in the treatment of gynecologic malignancy, carboplatin and paclitaxel, have been used. The 3-year event-free survival (EFS) for all stages is 71% ± 11%; for stage I and stage II EFS is 82% ± 11%, and for stage III and stage IV EFS is 57% ± 22%.[22]

References

  1. Hoenig DM, McRae S, Chen SC, et al.: Transitional cell carcinoma of the bladder in the pediatric patient. J Urol 156 (1): 203-5, 1996.  [PUBMED Abstract]

  2. Serrano-Durbá A, Domínguez-Hinarejos C, Reig-Ruiz C, et al.: Transitional cell carcinoma of the bladder in children. Scand J Urol Nephrol 33 (1): 73-6, 1999.  [PUBMED Abstract]

  3. Fine SW, Humphrey PA, Dehner LP, et al.: Urothelial neoplasms in patients 20 years or younger: a clinicopathological analysis using the world health organization 2004 bladder consensus classification. J Urol 174 (5): 1976-80, 2005.  [PUBMED Abstract]

  4. Sung JD, Koyle MA: Squamous cell carcinoma of the bladder in a pediatric patient. J Pediatr Surg 35 (12): 1838-9, 2000.  [PUBMED Abstract]

  5. Lezama-del Valle P, Jerkins GR, Rao BN, et al.: Aggressive bladder carcinoma in a child. Pediatr Blood Cancer 43 (3): 285-8, 2004.  [PUBMED Abstract]

  6. Johansson SL, Cohen SM: Epidemiology and etiology of bladder cancer. Semin Surg Oncol 13 (5): 291-8, 1997 Sep-Oct.  [PUBMED Abstract]

  7. IARC Working Group on the Evaluation of Carcinogenic Risks to Humans. International Agency for Research on Cancer.: Overall evaluations of carcinogenicity: an updating of IARC monographs, volumes 1 to 42. IARC Monographs on the Evaluation of Carcinogenic Risks to Humans, Supplement 7. Lyon, France: International Agency for Research on Cancer, 1987. 

  8. Morowitz M, Huff D, von Allmen D: Epithelial ovarian tumors in children: a retrospective analysis. J Pediatr Surg 38 (3): 331-5; discussion 331-5, 2003.  [PUBMED Abstract]

  9. Schultz KA, Sencer SF, Messinger Y, et al.: Pediatric ovarian tumors: a review of 67 cases. Pediatr Blood Cancer 44 (2): 167-73, 2005.  [PUBMED Abstract]

  10. Lovvorn HN 3rd, Tucci LA, Stafford PW: Ovarian masses in the pediatric patient. AORN J 67 (3): 568-76; quiz 577, 580-84, 1998.  [PUBMED Abstract]

  11. Tsai JY, Saigo PE, Brown C, et al.: Diagnosis, pathology, staging, treatment, and outcome of epithelial ovarian neoplasia in patients age < 21 years. Cancer 91 (11): 2065-70, 2001.  [PUBMED Abstract]

  12. Schneider DT, Jänig U, Calaminus G, et al.: Ovarian sex cord-stromal tumors--a clinicopathological study of 72 cases from the Kiel Pediatric Tumor Registry. Virchows Arch 443 (4): 549-60, 2003.  [PUBMED Abstract]

  13. Calaminus G, Wessalowski R, Harms D, et al.: Juvenile granulosa cell tumors of the ovary in children and adolescents: results from 33 patients registered in a prospective cooperative study. Gynecol Oncol 65 (3): 447-52, 1997.  [PUBMED Abstract]

  14. Bouffet E, Basset T, Chetail N, et al.: Juvenile granulosa cell tumor of the ovary in infants: a clinicopathologic study of three cases and review of the literature. J Pediatr Surg 32 (5): 762-5, 1997.  [PUBMED Abstract]

  15. Zaloudek C, Norris HJ: Granulosa tumors of the ovary in children: a clinical and pathologic study of 32 cases. Am J Surg Pathol 6 (6): 503-12, 1982.  [PUBMED Abstract]

  16. Vassal G, Flamant F, Caillaud JM, et al.: Juvenile granulosa cell tumor of the ovary in children: a clinical study of 15 cases. J Clin Oncol 6 (6): 990-5, 1988.  [PUBMED Abstract]

  17. Kalfa N, Patte C, Orbach D, et al.: A nationwide study of granulosa cell tumors in pre- and postpubertal girls: missed diagnosis of endocrine manifestations worsens prognosis. J Pediatr Endocrinol Metab 18 (1): 25-31, 2005.  [PUBMED Abstract]

  18. Gell JS, Stannard MW, Ramnani DM, et al.: Juvenile granulosa cell tumor in a 13-year-old girl with enchondromatosis (Ollier's disease): a case report. J Pediatr Adolesc Gynecol 11 (3): 147-50, 1998.  [PUBMED Abstract]

  19. Powell JL, Connor GP, Henderson GS: Management of recurrent juvenile granulosa cell tumor of the ovary. Gynecol Oncol 81 (1): 113-6, 2001.  [PUBMED Abstract]

  20. Schneider DT, Calaminus G, Wessalowski R, et al.: Therapy of advanced ovarian juvenile granulosa cell tumors. Klin Padiatr 214 (4): 173-8, 2002 Jul-Aug.  [PUBMED Abstract]

  21. Schneider DT, Calaminus G, Harms D, et al.: Ovarian sex cord-stromal tumors in children and adolescents. J Reprod Med 50 (6): 439-46, 2005.  [PUBMED Abstract]

  22. McNall RY, Nowicki PD, Miller B, et al.: Adenocarcinoma of the cervix and vagina in pediatric patients. Pediatr Blood Cancer 43 (3): 289-94, 2004.  [PUBMED Abstract]

  23. Abu-Rustum NR, Su W, Levine DA, et al.: Pediatric radical abdominal trachelectomy for cervical clear cell carcinoma: a novel surgical approach. Gynecol Oncol 97 (1): 296-300, 2005.  [PUBMED Abstract]

Other Rare Childhood Cancers

Other rare childhood cancers include multiple endocrine neoplasia syndrome, skin cancer, chordoma, and cancer of unknown primary site. The prognosis, diagnosis, classification, and treatment of these other rare childhood cancers are discussed below.

Multiple Endocrine Neoplasia Syndrome

These syndromes are familial disorders that are characterized by neoplastic changes in more than one endocrine organ.[1] Changes may include hyperplasia, benign adenomas, and carcinomas. There are distinct genetic disorders with characteristic clinical presentations referred to as multiple endocrine neoplasia (MEN) 1, MEN 2a, and MEN 2b. An additional complex is referred to as the Carney complex, which is an association of MEN with heart and skin tumors.[2]

The MEN 1 syndrome, also referred to as Werner syndrome, may involve tumors of the pituitary gland, the parathyroid, and adrenal, gastric, and pancreatic structures, which may secrete hormones such as insulin. The gene for this syndrome is located on chromosome 11q13. Mutation testing should be combined with clinical screening for patients and family members with proven "at risk" MEN 1.[3] The MEN 2a syndrome (Sipple syndrome) is associated with medullary thyroid carcinoma, parathyroid hyperplasia, adenomas, and pheochromocytoma. The MEN 2b syndrome is associated with medullary thyroid carcinomas, parathyroid hyperplasias, adenomas, and pheochromocytomas, mucosal neuromas, and ganglioneuromas.[4,5] Patients with the MEN 2b syndrome may have a slender body build, long and thin extremities, a high arch palate, and pectus excavatum or pes cavus. The face may be characterized by thick lips because of mucosal neuromas. Such patients can also be identified by performing a pentagastrin stimulation test or by genetic screening in families known to be affected.

A germline mutation in the ret oncogene (tyrosine-kinase receptor) on chromosome 10q11.2 is responsible for the uncontrolled growth of cells in medullary thyroid carcinoma associated with MEN 2a and MEN 2b syndromes.[6-8] The current management of medullary thyroid cancer in children from families having the MEN 2 syndromes relies on presymptomatic detection of the ret proto-oncogene mutation responsible for the disease. Children with MEN 2a should undergo prophylactic total thyroidectomy between the ages of 5 and 8 years.[8-12] Relatives of patients with MEN 2a should undergo genetic testing in early childhood, before the age of 5 years. Carriers should undergo total thyroidectomy with autotransplantation of one parathyroid gland by a certain age, depending on the type of mutation found.[13-15] Because of the increased virulence of medullary thyroid carcinoma in children with MEN 2b, it is recommended that these children undergo prophylactic thyroidectomy in infancy.[10,16] Complete removal of the thyroid gland is the recommended procedure for surgical management of medullary thyroid cancer in children, since there is a high incidence of bilateral disease.

Hirschsprung disease has been associated with the development of neuroendocrine tumors such as medullary thyroid carcinoma. RET germline inactivating mutations have been detected in up to 50% of patients with familial Hirschsprung disease and less often in the sporadic form. Cosegregation of Hirschsprung disease and medullary thyroid carcinoma phenotype is infrequently reported, but these individuals usually have a mutation in RET exon 10. It has been recommended that patients with Hirschsprung disease be screened for mutations in RET exon 10 and consideration be given to prophylactic thyroidectomy if such a mutation is discovered.[17]

The Carney complex includes the association of primary pigmented nodular adrenocortical disease with blue nevi of the skin and mucosa and a variety of additional endocrine or nonendocrine tumors. There may be myxomas of the heart, skin, or breast and tumors of peripheral nerve sheath origin.[2,18] Head and neck manifestations are not uncommon.[18]

The outcome of patients with the MEN 1 syndrome is generally good provided adequate treatment can be obtained for parathyroid, pancreatic, and pituitary tumors. The outcome for patients with the MEN 2a syndrome is also generally good, yet the possibility exists for recurrence of medullary thyroid carcinoma and pheochromocytoma.[19-21] Patients who have the MEN 2b syndrome have a worse outcome primarily due to more aggressive medullary thyroid carcinoma. Prophylactic thyroidectomy has the potential to improve the outcome in MEN 2b, but there are no long-term outcome reports published to date. For patients with the Carney complex, prognosis depends on the frequency of recurrences of cardiac and skin myxomas and other tumors.

Treatment options under clinical evaluation
  • NCI-07-C-0189 34: This phase I/II NCI trial is investigating vandetanib, an orally available tyrosine kinase receptor inhibitor, for patients aged 5 years to 18 years, with hereditary thyroid medullary carcinoma.[22,23]
Skin Cancer (Melanoma, Basal Cell, and Squamous Cell Carcinoma)

Melanoma is thought to be the most common skin cancer in children, followed by basal cell and squamous cell carcinomas (SCCs).[24-31] The incidence of melanoma in children and adolescents represents approximately 1% of the new cases of melanoma that are diagnosed annually in this country. In most instances, melanoma in the pediatric population is similar to that of adults in relation to site of presentation, symptoms, description, spread, and prognosis, but thickness does not appear to have prognostic significance.[30,32] Melanoma may grow more rapidly in prepubescent children than in older individuals and may present with more atypical clinical features in children than in adults.[33,34] Analysis of the National Cancer Data Base identified 3,159 patients aged 1 year to 19 years with melanoma. Most of the patients (90%) were age 10 years or older. More girls (56%) were seen than boys. Younger age and higher stage conferred a worse prognosis.[32]

The greatest cause of skin cancer of any type is exposure to the ultraviolet portion of sunlight.[35-38] Other causes may be related to chemical carcinogenesis, radiation exposure, immunodeficiency, or immunosuppression. The person who is most likely to develop a melanoma is easily sunburned, has poor tanning ability, and generally has light hair, blue eyes, and pale skin. Worldwide, there is an increasing incidence of both melanoma and nonmelanoma skin cancers. Melanoma presents as a relatively flat, dark-colored lesion, which may enlarge, penetrate the skin, or metastasize.

Melanomas may be congenital.[27] They are sometimes associated with large congenital black spots known as melanocytic nevi,[39] which may cover the trunk and thigh.[40-42] Melanomas can also develop in individuals with xeroderma pigmentosum, a rare recessive disorder characterized by extreme sensitivity to sunlight, keratosis, and various neurologic manifestations. Individuals with xeroderma pigmentosum may also develop other skin cancers, including SCCs and basal cell carcinomas.[28] Children with hereditary immunodeficiencies have an increased lifetime risk of developing melanoma.

Neurocutaneous melanosis is an unusual condition associated with large or multiple congenital nevi of the skin and melanin deposits within the central nervous system. These deposits may be detected by magnetic resonance imaging of the brain or spinal cord. Dysplastic nevi occur in about 5% of the U.S. population and are potential precursors of melanoma.[28] Individuals with atypical moles, which include raised lesions that may bleed and various color hues (e.g., brown, tan, pink, black), are at an increased risk of having melanoma and of having children affected by these premalignant lesions.

Basal cell carcinomas generally appear as raised lumps or ulcerated lesions, usually in areas with previous sun exposure. These tumors may be multiple and exacerbated by radiation therapy.[43] Nevoid basal cell carcinoma syndrome (Gorlin syndrome) is a rare disorder with a predisposition to the development of early-onset neoplasms, including basal cell carcinoma, ovarian fibroma, and desmoplastic medulloblastoma.[44-47] SCCs are usually reddened lesions with varying degrees of scaling or crusting, and they have an appearance similar to eczema, infections, trauma, or psoriasis.

Biopsy or excision is necessary to determine the diagnosis of any skin cancer. Diagnosis is necessary for decisions regarding additional treatment. Basal and squamous cell carcinomas are generally curable with surgery alone, but the treatment of melanoma requires greater consideration because of its potential for metastasis. Surgery for melanoma depends on the size, site, level of invasion, and metastatic extent or stage of the tumor.[28] Wide excision with skin grafting may become necessary. The current recommendation is that surgical resection include a 2-cm-deep margin for melanoma lesions, with examination of the regional lymph nodes draining the site of the melanoma. This procedure may require the injection of a radioisotope, following its distribution, and then performing excision of the associated regional lymph nodes (sentinel lymph node [SLN] biopsy technique).[48,49] This requires injection of a vital blue stain and radioisotope into the skin to characterize the pattern of lymph node drainage. Lymph node dissection is necessary if sentinel nodes are involved with the tumor; however, if there is no spread of the disease beyond the lymph nodes, adjuvant therapy with interferon-alpha-2b alone may be recommended for a period of 1 year.[28,50,51] Finding the tumor-involved regional lymph node (via SLN biopsy) may clarify the suspicion of melanoma in diagnostically difficult situations such as Spitz nevus with significant atypia at the primary site.[48] However, on the basis of reports of clinically benign melanocytic lesions involving regional lymph nodes, the prognostic value of SLN biopsy is unclear.[52] SLN biopsies have shown an increased frequency of benign nodal nevi, which might mimic metastasis of melanoma and, therefore, may raise potential diagnostic and therapeutic issues.[53] For individuals with metastatic disease, a combination of cisplatin, vinblastine, imidazole carboxamide, interleukin-2 (IL-2), and interferon-alpha-2b has been proposed.[28]

Overall 5-year survival of children and adolescents with melanoma is approximately 91%.[54] Most children and adolescents present with localized disease (68%–78%) and have an excellent outcome (96% 5-year survival). In a Surveillance, Epidemiology, and End Results (SEER) study, the 5-year survival for those with nodal or distant metastases was 77% and 57%, respectively.[54] The prognosis of children and adolescents with melanoma has been previously reported to be similar to that of adults with similar stage disease, with the prognosis depending on the tumor thickness and the extent of spread at the time of diagnosis.[55,56] However, the outcomes for children and adolescents noted in the SEER study are substantially better for those with nodal or distant metastases than the outcomes for adults in similar stages. One study compared young patients (≤20 years) with adult patients and found a higher incidence of lymph node metastases in the younger patients versus case-matched adult patients (18% vs. 10%), but, the 10-year cause-specific survival rates were similar between the two groups (89% and 79%, respectively).[57] The reason for this difference is unknown, though it is speculated that in children some cases were classified as stage III on the basis of what may have been an intranodal nevus.[52] Factors associated with worse survival from melanoma include male gender, unfavorable location of primary tumor, and regional or distant metastases.[54] Refer to the PDQ summary on adult Skin Cancer Treatment 35 for more information.

Treatment options under clinical evaluation

There are two melanoma trials available to patients aged 10 years or older. Both of these trials are combination adult/pediatric trials.

  • E1697 (ECOG):[58] Phase III trial of 4 weeks of high-dose interferon-alpha-2b in stages T3 to T4 or N1 melanoma.


  • S0008 (SWOG):[59] Phase III trial of high-dose interferon-alpha-2b versus cisplatin, vinblastine, and dacarbazine plus IL-2 in patients with high-risk melanoma.


Chordoma

Chordoma is a rare tumor that arises from remnants of the notochord within the clivus, spinal vertebrae, or sacrum. The incidence in the United States is approximately one case per 1 million people per year. In American children and adolescents, chordomas are more likely to arise in the clivus, especially in females, rather than in the sacrum, which is more common among adult males.[60] Patients usually present with pain, with or without neurologic deficits such as cranial or other nerve impairment. Diagnosis is straightforward when the typical physaliferous (soap-bubble-bearing) cells are present. Differential diagnosis is sometimes difficult and includes dedifferentiated chordoma and chondrosarcoma. Standard treatment includes surgery, which is not commonly curative because of difficulty in obtaining clear margins, and external radiation therapy. The best results have been obtained using proton-beam therapy,[61,62] but this is currently available only in Loma Linda, California, and Boston, Massachusetts. Recurrences are usually local but can include distant metastases to lungs or bone. Children younger than 5 years may have a worse outlook than older patients.[63,64] The survival rate in children and adolescents is about 50% at 4 years from diagnosis.[64] There is no known effective cytotoxic agent or combination chemotherapy for this disease, but there is a report of temporary regression after ifosfamide and doxorubicin therapy in a 19-month-old child.[65]

Cancer of Unknown Primary Site

These cancers present as a metastatic cancer for which a precise primary tumor site cannot be determined.[66] As an example, lymph nodes at the base of the skull may enlarge in relationship to a tumor that may be on the face or the scalp but is not evident by physical examination or by radiographic imaging. Thus, modern imaging techniques may indicate the extent of the disease but not a primary site. Tumors such as adenocarcinomas, melanomas, and embryonal tumors such as rhabdomyosarcomas and neuroblastomas may have such a presentation. Because of the age-related incidence of tumor types, embryonal histologies are more common in children.

For all patients who present with tumors from an unknown primary site, the treatment should be considered in relation to the pathology of the tumor and should be appropriate for the general type of cancer initiated, irrespective of the site or sites of involvement.[66] Chemotherapy and radiation therapy treatments appropriate and relevant for the general category of carcinoma or sarcoma (depending upon the histologic findings, symptoms, and extent of tumor) should be initiated as early as possible.

References

  1. de Krijger RR: Endocrine tumor syndromes in infancy and childhood. Endocr Pathol 15 (3): 223-6, 2004.  [PUBMED Abstract]

  2. Carney JA, Young WF: Primary pigmented nodular adrenocortical disease and its associated conditions. Endocrinologist 2: 6-21, 1992. 

  3. Field M, Shanley S, Kirk J: Inherited cancer susceptibility syndromes in paediatric practice. J Paediatr Child Health 43 (4): 219-29, 2007.  [PUBMED Abstract]

  4. Skinner MA, DeBenedetti MK, Moley JF, et al.: Medullary thyroid carcinoma in children with multiple endocrine neoplasia types 2A and 2B. J Pediatr Surg 31 (1): 177-81; discussion 181-2, 1996.  [PUBMED Abstract]

  5. Brauckhoff M, Gimm O, Weiss CL, et al.: Multiple endocrine neoplasia 2B syndrome due to codon 918 mutation: clinical manifestation and course in early and late onset disease. World J Surg 28 (12): 1305-11, 2004.  [PUBMED Abstract]

  6. Sanso GE, Domene HM, Garcia R, et al.: Very early detection of RET proto-oncogene mutation is crucial for preventive thyroidectomy in multiple endocrine neoplasia type 2 children: presence of C-cell malignant disease in asymptomatic carriers. Cancer 94 (2): 323-30, 2002.  [PUBMED Abstract]

  7. Alsanea O, Clark OH: Familial thyroid cancer. Curr Opin Oncol 13 (1): 44-51, 2001.  [PUBMED Abstract]

  8. Fitze G: Management of patients with hereditary medullary thyroid carcinoma. Eur J Pediatr Surg 14 (6): 375-83, 2004.  [PUBMED Abstract]

  9. Skinner MA, Moley JA, Dilley WG, et al.: Prophylactic thyroidectomy in multiple endocrine neoplasia type 2A. N Engl J Med 353 (11): 1105-13, 2005.  [PUBMED Abstract]

  10. Skinner MA: Management of hereditary thyroid cancer in children. Surg Oncol 12 (2): 101-4, 2003.  [PUBMED Abstract]

  11. Learoyd DL, Gosnell J, Elston MS, et al.: Experience of prophylactic thyroidectomy in multiple endocrine neoplasia type 2A kindreds with RET codon 804 mutations. Clin Endocrinol (Oxf) 63 (6): 636-41, 2005.  [PUBMED Abstract]

  12. Guillem JG, Wood WC, Moley JF, et al.: ASCO/SSO review of current role of risk-reducing surgery in common hereditary cancer syndromes. J Clin Oncol 24 (28): 4642-60, 2006.  [PUBMED Abstract]

  13. Heizmann O, Haecker FM, Zumsteg U, et al.: Presymptomatic thyroidectomy in multiple endocrine neoplasia 2a. Eur J Surg Oncol 32 (1): 98-102, 2006.  [PUBMED Abstract]

  14. Frank-Raue K, Buhr H, Dralle H, et al.: Long-term outcome in 46 gene carriers of hereditary medullary thyroid carcinoma after prophylactic thyroidectomy: impact of individual RET genotype. Eur J Endocrinol 155 (2): 229-36, 2006.  [PUBMED Abstract]

  15. Piolat C, Dyon JF, Sturm N, et al.: Very early prophylactic thyroid surgery for infants with a mutation of the RET proto-oncogene at codon 634: evaluation of the implementation of international guidelines for MEN type 2 in a single centre. Clin Endocrinol (Oxf) 65 (1): 118-24, 2006.  [PUBMED Abstract]

  16. Leboulleux S, Travagli JP, Caillou B, et al.: Medullary thyroid carcinoma as part of a multiple endocrine neoplasia type 2B syndrome: influence of the stage on the clinical course. Cancer 94 (1): 44-50, 2002.  [PUBMED Abstract]

  17. Skába R, Dvoráková S, Václavíková E, et al.: The risk of medullary thyroid carcinoma in patients with Hirschsprung's disease. Pediatr Surg Int 22 (12): 991-5, 2006.  [PUBMED Abstract]

  18. Ryan MW, Cunningham S, Xiao SY: Maxillary sinus melanoma as the presenting feature of Carney complex. Int J Pediatr Otorhinolaryngol 72 (3): 405-8, 2008.  [PUBMED Abstract]

  19. Lallier M, St-Vil D, Giroux M, et al.: Prophylactic thyroidectomy for medullary thyroid carcinoma in gene carriers of MEN2 syndrome. J Pediatr Surg 33 (6): 846-8, 1998.  [PUBMED Abstract]

  20. Dralle H, Gimm O, Simon D, et al.: Prophylactic thyroidectomy in 75 children and adolescents with hereditary medullary thyroid carcinoma: German and Austrian experience. World J Surg 22 (7): 744-50; discussion 750-1, 1998.  [PUBMED Abstract]

  21. Skinner MA, Wells SA Jr: Medullary carcinoma of the thyroid gland and the MEN 2 syndromes. Semin Pediatr Surg 6 (3): 134-40, 1997.  [PUBMED Abstract]

  22. Herbst RS, Heymach JV, O'Reilly MS, et al.: Vandetanib (ZD6474): an orally available receptor tyrosine kinase inhibitor that selectively targets pathways critical for tumor growth and angiogenesis. Expert Opin Investig Drugs 16 (2): 239-49, 2007.  [PUBMED Abstract]

  23. Vidal M, Wells S, Ryan A, et al.: ZD6474 suppresses oncogenic RET isoforms in a Drosophila model for type 2 multiple endocrine neoplasia syndromes and papillary thyroid carcinoma. Cancer Res 65 (9): 3538-41, 2005.  [PUBMED Abstract]

  24. Sasson M, Mallory SB: Malignant primary skin tumors in children. Curr Opin Pediatr 8 (4): 372-7, 1996.  [PUBMED Abstract]

  25. Barnhill RL: Childhood melanoma. Semin Diagn Pathol 15 (3): 189-94, 1998.  [PUBMED Abstract]

  26. Fishman C, Mihm MC Jr, Sober AJ: Diagnosis and management of nevi and cutaneous melanoma in infants and children. Clin Dermatol 20 (1): 44-50, 2002 Jan-Feb.  [PUBMED Abstract]

  27. Hamre MR, Chuba P, Bakhshi S, et al.: Cutaneous melanoma in childhood and adolescence. Pediatr Hematol Oncol 19 (5): 309-17, 2002 Jul-Aug.  [PUBMED Abstract]

  28. Ceballos PI, Ruiz-Maldonado R, Mihm MC Jr: Melanoma in children. N Engl J Med 332 (10): 656-62, 1995.  [PUBMED Abstract]

  29. Schmid-Wendtner MH, Berking C, Baumert J, et al.: Cutaneous melanoma in childhood and adolescence: an analysis of 36 patients. J Am Acad Dermatol 46 (6): 874-9, 2002.  [PUBMED Abstract]

  30. Pappo AS: Melanoma in children and adolescents. Eur J Cancer 39 (18): 2651-61, 2003.  [PUBMED Abstract]

  31. Huynh PM, Grant-Kels JM, Grin CM: Childhood melanoma: update and treatment. Int J Dermatol 44 (9): 715-23, 2005.  [PUBMED Abstract]

  32. Lange JR, Palis BE, Chang DC, et al.: Melanoma in children and teenagers: an analysis of patients from the National Cancer Data Base. J Clin Oncol 25 (11): 1363-8, 2007.  [PUBMED Abstract]

  33. Mones JM, Ackerman AB: Melanomas in prepubescent children: review comprehensively, critique historically, criteria diagnostically, and course biologically. Am J Dermatopathol 25 (3): 223-38, 2003.  [PUBMED Abstract]

  34. Ferrari A, Bono A, Baldi M, et al.: Does melanoma behave differently in younger children than in adults? A retrospective study of 33 cases of childhood melanoma from a single institution. Pediatrics 115 (3): 649-54, 2005.  [PUBMED Abstract]

  35. Pappo AS, Kaste SC, Rao BN, et al.: Childhood melanoma. In: Balch CM, Houghton AN, Sober AJ, et al., eds.: Cutaneous Melanoma. 3rd ed., St. Louis, Mo: Quality Medical Publishing Inc., 1998, pp 175-186. 

  36. Heffernan AE, O'Sullivan A: Pediatric sun exposure. Nurse Pract 23 (7): 67-8, 71-8, 83-6, 1998.  [PUBMED Abstract]

  37. Berg P, Lindelöf B: Differences in malignant melanoma between children and adolescents. A 35-year epidemiological study. Arch Dermatol 133 (3): 295-7, 1997.  [PUBMED Abstract]

  38. Elwood JM, Jopson J: Melanoma and sun exposure: an overview of published studies. Int J Cancer 73 (2): 198-203, 1997.  [PUBMED Abstract]

  39. Krengel S, Hauschild A, Schäfer T: Melanoma risk in congenital melanocytic naevi: a systematic review. Br J Dermatol 155 (1): 1-8, 2006.  [PUBMED Abstract]

  40. Ka VS, Dusza SW, Halpern AC, et al.: The association between large congenital melanocytic naevi and cutaneous melanoma: preliminary findings from an Internet-based registry of 379 patients. Melanoma Res 15 (1): 61-7, 2005.  [PUBMED Abstract]

  41. Zaal LH, Mooi WJ, Klip H, et al.: Risk of malignant transformation of congenital melanocytic nevi: a retrospective nationwide study from The Netherlands. Plast Reconstr Surg 116 (7): 1902-9, 2005.  [PUBMED Abstract]

  42. Bett BJ: Large or multiple congenital melanocytic nevi: occurrence of cutaneous melanoma in 1008 persons. J Am Acad Dermatol 52 (5): 793-7, 2005.  [PUBMED Abstract]

  43. Griffin JR, Cohen PR, Tschen JA, et al.: Basal cell carcinoma in childhood: case report and literature review. J Am Acad Dermatol 57 (5 Suppl): S97-102, 2007.  [PUBMED Abstract]

  44. Gorlin RJ: Nevoid basal cell carcinoma syndrome. Dermatol Clin 13 (1): 113-25, 1995.  [PUBMED Abstract]

  45. Kimonis VE, Goldstein AM, Pastakia B, et al.: Clinical manifestations in 105 persons with nevoid basal cell carcinoma syndrome. Am J Med Genet 69 (3): 299-308, 1997.  [PUBMED Abstract]

  46. Amlashi SF, Riffaud L, Brassier G, et al.: Nevoid basal cell carcinoma syndrome: relation with desmoplastic medulloblastoma in infancy. A population-based study and review of the literature. Cancer 98 (3): 618-24, 2003.  [PUBMED Abstract]

  47. Veenstra-Knol HE, Scheewe JH, van der Vlist GJ, et al.: Early recognition of basal cell naevus syndrome. Eur J Pediatr 164 (3): 126-30, 2005.  [PUBMED Abstract]

  48. Pacella SJ, Lowe L, Bradford C, et al.: The utility of sentinel lymph node biopsy in head and neck melanoma in the pediatric population. Plast Reconstr Surg 112 (5): 1257-65, 2003.  [PUBMED Abstract]

  49. Shah NC, Gerstle JT, Stuart M, et al.: Use of sentinel lymph node biopsy and high-dose interferon in pediatric patients with high-risk melanoma: the Hospital for Sick Children experience. J Pediatr Hematol Oncol 28 (8): 496-500, 2006.  [PUBMED Abstract]

  50. Navid F, Furman WL, Fleming M, et al.: The feasibility of adjuvant interferon alpha-2b in children with high-risk melanoma. Cancer 103 (4): 780-7, 2005.  [PUBMED Abstract]

  51. Chao MM, Schwartz JL, Wechsler DS, et al.: High-risk surgically resected pediatric melanoma and adjuvant interferon therapy. Pediatr Blood Cancer 44 (5): 441-8, 2005.  [PUBMED Abstract]

  52. Roaten JB, Partrick DA, Bensard D, et al.: Survival in sentinel lymph node-positive pediatric melanoma. J Pediatr Surg 40 (6): 988-92; discussion 992, 2005.  [PUBMED Abstract]

  53. Holt JB, Sangueza OP, Levine EA, et al.: Nodal melanocytic nevi in sentinel lymph nodes. Correlation with melanoma-associated cutaneous nevi. Am J Clin Pathol 121 (1): 58-63, 2004.  [PUBMED Abstract]

  54. Strouse JJ, Fears TR, Tucker MA, et al.: Pediatric melanoma: risk factor and survival analysis of the surveillance, epidemiology and end results database. J Clin Oncol 23 (21): 4735-41, 2005.  [PUBMED Abstract]

  55. Gibbs P, Moore A, Robinson W, et al.: Pediatric melanoma: are recent advances in the management of adult melanoma relevant to the pediatric population. J Pediatr Hematol Oncol 22 (5): 428-32, 2000 Sep-Oct.  [PUBMED Abstract]

  56. Conti EM, Cercato MC, Gatta G, et al.: Childhood melanoma in Europe since 1978: a population-based survival study. Eur J Cancer 37 (6): 780-4, 2001.  [PUBMED Abstract]

  57. Livestro DP, Kaine EM, Michaelson JS, et al.: Melanoma in the young: differences and similarities with adult melanoma: a case-matched controlled analysis. Cancer 110 (3): 614-24, 2007.  [PUBMED Abstract]

  58. Agarwala SS, Eastern Cooperative Oncology Group: Phase III Randomized Adjuvant Study of High-Dose Interferon alfa-2b Therapy in Patients With Stage II or III Melanoma, ECOG-1697, Clinical trial, Active.  [PDQ Clinical Trial] 36

  59. Flaherty LE, Southwest Oncology Group: Phase III Randomized Study of Interferon alfa Versus Cisplatin, Vinblastine, and Dacarbazine Plus Interferon alfa and Interleukin-2 in Patients With High-Risk Melanoma, SWOG-S0008, Clinical trial, Closed.  [PDQ Clinical Trial] 37

  60. McMaster ML, Goldstein AM, Bromley CM, et al.: Chordoma: incidence and survival patterns in the United States, 1973-1995. Cancer Causes Control 12 (1): 1-11, 2001.  [PUBMED Abstract]

  61. Hug EB, Sweeney RA, Nurre PM, et al.: Proton radiotherapy in management of pediatric base of skull tumors. Int J Radiat Oncol Biol Phys 52 (4): 1017-24, 2002.  [PUBMED Abstract]

  62. Noël G, Habrand JL, Jauffret E, et al.: Radiation therapy for chordoma and chondrosarcoma of the skull base and the cervical spine. Prognostic factors and patterns of failure. Strahlenther Onkol 179 (4): 241-8, 2003.  [PUBMED Abstract]

  63. Coffin CM, Swanson PE, Wick MR, et al.: Chordoma in childhood and adolescence. A clinicopathologic analysis of 12 cases. Arch Pathol Lab Med 117 (9): 927-33, 1993.  [PUBMED Abstract]

  64. Borba LA, Al-Mefty O, Mrak RE, et al.: Cranial chordomas in children and adolescents. J Neurosurg 84 (4): 584-91, 1996.  [PUBMED Abstract]

  65. Scimeca PG, James-Herry AG, Black KS, et al.: Chemotherapeutic treatment of malignant chordoma in children. J Pediatr Hematol Oncol 18 (2): 237-40, 1996.  [PUBMED Abstract]

  66. Kuttesch JF Jr, Parham DM, Kaste SC, et al.: Embryonal malignancies of unknown primary origin in children. Cancer 75 (1): 115-21, 1995.  [PUBMED Abstract]

Get More Information From NCI

Call 1-800-4-CANCER

For more information, U.S. residents may call the National Cancer Institute's (NCI's) Cancer Information Service toll-free at 1-800-4-CANCER (1-800-422-6237) Monday through Friday from 9:00 a.m. to 4:30 p.m. Deaf and hard-of-hearing callers with TTY equipment may call 1-800-332-8615. The call is free and a trained Cancer Information Specialist is available to answer your questions.

Chat online

The NCI's LiveHelp® 38 online chat service provides Internet users with the ability to chat online with an Information Specialist. The service is available from 9:00 a.m. to 11:00 p.m. Eastern time, Monday through Friday. Information Specialists can help Internet users find information on NCI Web sites and answer questions about cancer.

Write to us

For more information from the NCI, please write to this address:

NCI Public Inquiries Office
Suite 3036A
6116 Executive Boulevard, MSC8322
Bethesda, MD 20892-8322

Search the NCI Web site

The NCI Web site 39 provides online access to information on cancer, clinical trials, and other Web sites and organizations that offer support and resources for cancer patients and their families. For a quick search, use our “Best Bets” search box in the upper right hand corner of each Web page. The results that are most closely related to your search term will be listed as Best Bets at the top of the list of search results.

There are also many other places to get materials and information about cancer treatment and services. Hospitals in your area may have information about local and regional agencies that have information on finances, getting to and from treatment, receiving care at home, and dealing with problems related to cancer treatment.

Find Publications

The NCI has booklets and other materials for patients, health professionals, and the public. These publications discuss types of cancer, methods of cancer treatment, coping with cancer, and clinical trials. Some publications provide information on tests for cancer, cancer causes and prevention, cancer statistics, and NCI research activities. NCI materials on these and other topics may be ordered online or printed directly from the NCI Publications Locator 40. These materials can also be ordered by telephone from the Cancer Information Service toll-free at 1-800-4-CANCER (1-800-422-6237), TTY at 1-800-332-8615.

Changes to This Summary (12/02/2008)

The PDQ cancer information summaries are reviewed regularly and updated as new information becomes available. This section describes the latest changes made to this summary as of the date above.

Head and Neck Cancers 41

Added Ventkitaraman et al. as reference 12 42.

Added text 43 about adjustment of the I-131 dose for weight and other age-dependent safety factors (cited Luster et al. as reference 47 and Parisi et al. as reference 48).

Added Laikui et al. as reference 73 44.

Thoracic Cancers 45

Added Hill et al. as reference 24 46 and level of evidence 3iiiA.

Added text 46 about comparative genomic and fluorescent in situ hybridization methods related to gain of chromosome 8q as the main recurrent chromosomal abnormality in pleuropulmonary blastoma (cited deKrijger et al. as reference 35 and Quilichini et al. as reference 36).

Added text 47 to include neuromyotonia as another name associated with Isaacs syndrome.

Abdominal Cancers 48

Added Ferrari et al. as reference 52 49 and level of evidence 3iiA.

More Information

About PDQ

Additional PDQ Summaries

Important:

This information is intended mainly for use by doctors and other health care professionals. If you have questions about this topic, you can ask your doctor, or call the Cancer Information Service at 1-800-4-CANCER (1-800-422-6237).



Glossary Terms

Level of evidence 3iiA
Consecutive case series (not population-based) with total mortality as an endpoint. See Levels of Evidence for Adult and Pediatric Cancer Treatment Studies (PDQ®) for more information.
Level of evidence 3iii
Nonconsecutive case series. See Levels of Evidence for Adult and Pediatric Cancer Treatment Studies (PDQ®) for more information.
Level of evidence 3iiiA
Nonconsecutive case series with total mortality as an endpoint. See Levels of Evidence for Adult and Pediatric Cancer Treatment Studies (PDQ®) for more information.
Level of evidence 3iiiDiv
Nonconsecutive case series with tumor response rate as an endpoint. See Levels of Evidence for Adult and Pediatric Cancer Treatment Studies (PDQ®) for more information.


Table of Links

1http://www.cancer.gov/cancerinfo/pdq/pediatric-treatment-board
2http://www.cancer.gov/cancertopics/pdq/levels-evidence-adult-treatment/HealthPr
ofessional
3http://www.cancer.gov/cancertopics/pdq/treatment/unusual-cancers-childhood/Pati
ent
4http://www.cancer.gov/espanol/pdq/tratamiento/canceres-infantiles-poco-comunes/
HealthProfessional
5http://cancer.gov/cancerinfo/pdq/supportivecare
6http://cancer.gov/clinicaltrials
7http://www.cancer.gov/cancertopics/pdq/treatment/lateeffects/HealthProfessional
8http://www.cancer.gov/cancertopics/pdq/treatment/nasopharyngeal/HealthProfessio
nal
9http://www.cancer.gov/search/viewclinicaltrials.aspx?version= heal
thprofessional &cdrid=454849
10http://www.cancer.gov/cancertopics/pdq/treatment/unusual-cancers-childhood/Heal
thProfessional/84.cdr#Section_84
11http://www.cancer.gov/cancertopics/pdq/treatment/thyroid/HealthProfessional
12http://www.cancer.gov/cancertopics/pdq/treatment/oropharyngeal/HealthProfession
al
13http://www.cancer.gov/cancertopics/pdq/treatment/lip-and-oral-cavity/HealthProf
essional
14http://www.cancer.gov/cancertopics/pdq/treatment/salivarygland/HealthProfession
al
15http://www.cancer.gov/cancertopics/pdq/treatment/laryngeal/HealthProfessional
16http://seer.cancer.gov/publications/childhood/carcinomas.pdf
17http://www.cancer.gov/cancertopics/pdq/treatment/breast/healthprofessional
18http://www.cancer.gov/cancertopics/pdq/treatment/esophageal/HealthProfessional
19http://www.cancer.gov/cancertopics/pdq/treatment/thymoma/HealthProfessional
20http://www.cancer.gov/cancertopics/pdq/treatment/malignantmesothelioma/HealthPr
ofessional
21http://www.ppbregistry.org
22http://www.cancer.gov/cancertopics/pdq/treatment/wilms/HealthProfessional
23http://www.cancer.gov/cancertopics/pdq/treatment/adrenocortical/HealthProfessio
nal
24http://www.cancer.gov/search/viewclinicaltrials.aspx?version= heal
thprofessional &cdrid=467191
25http://www.cancer.gov/cancertopics/pdq/treatment/gastric/HealthProfessional
26http://www.cancer.gov/cancertopics/pdq/treatment/pancreatic/HealthProfessional
27http://www.cancer.gov/cancertopics/pdq/treatment/colon/HealthProfessional
28http://www.cancer.gov/cancertopics/pdq/treatment/rectal/HealthProfessional
29http://www.cancer.gov/cancertopics/pdq/treatment/gastrointestinalcarcinoid/Heal
thProfessional
30http://www.cancer.gov/cancertopics/pdq/treatment/bladder/HealthProfessional
31http://www.cancer.gov/cancertopics/pdq/treatment/extracranial-germ-cell/HealthP
rofessional
32http://www.cancer.gov/cancertopics/pdq/treatment/ovarianepithelial/HealthProfes
sional
33http://www.cancer.gov/cancertopics/pdq/treatment/ovarian-low-malignant-potentia
l/HealthProfessional
34http://www.cancer.gov/search/viewclinicaltrials.aspx?version= heal
thprofessional &cdrid=559838
35http://www.cancer.gov/cancertopics/pdq/treatment/skin/HealthProfessional
36http://www.cancer.gov/search/viewclinicaltrials.aspx?version= h
ealthprofessional &cdrid=66727
37http://www.cancer.gov/search/viewclinicaltrials.aspx?version= h
ealthprofessional &cdrid=68162
38https://cissecure.nci.nih.gov/livehelp/welcome.asp
39http://cancer.gov
40https://cissecure.nci.nih.gov/ncipubs
41http://www.cancer.gov/cancertopics/pdq/treatment/unusual-cancers-childhood/Heal
thProfessional/106.cdr#Section_106
42http://www.cancer.gov/cancertopics/pdq/treatment/unusual-cancers-childhood/Heal
thProfessional/206.cdr#Section_206
43http://www.cancer.gov/cancertopics/pdq/treatment/unusual-cancers-childhood/Heal
thProfessional/19.cdr#Section_19
44http://www.cancer.gov/cancertopics/pdq/treatment/unusual-cancers-childhood/Heal
thProfessional/23.cdr#Section_23
45http://www.cancer.gov/cancertopics/pdq/treatment/unusual-cancers-childhood/Heal
thProfessional/112.cdr#Section_112
46http://www.cancer.gov/cancertopics/pdq/treatment/unusual-cancers-childhood/Heal
thProfessional/34.cdr#Section_34
47http://www.cancer.gov/cancertopics/pdq/treatment/unusual-cancers-childhood/Heal
thProfessional/40.cdr#Section_40
48http://www.cancer.gov/cancertopics/pdq/treatment/unusual-cancers-childhood/Heal
thProfessional/113.cdr#Section_113
49http://www.cancer.gov/cancertopics/pdq/treatment/unusual-cancers-childhood/Heal
thProfessional/68.cdr#Section_68
50http://cancer.gov/cancerinfo/pdq/cancerdatabase
51http://cancer.gov/cancerinfo/pdq/adulttreatment
52http://cancer.gov/cancerinfo/pdq/pediatrictreatment
53http://cancer.gov/cancerinfo/pdq/screening
54http://cancer.gov/cancerinfo/pdq/prevention
55http://cancer.gov/cancerinfo/pdq/genetics
56http://cancer.gov/cancerinfo/pdq/cam