Principles of Adjuvant Therapy in Childhood Cancer

Published on 26/02/2015 by admin

Filed under Pediatrics

Last modified 22/04/2025

Print this page

rate 1 star rate 2 star rate 3 star rate 4 star rate 5 star
Your rating: none, Average: 0 (0 votes)

This article have been viewed 1491 times

Chapter 64

Principles of Adjuvant Therapy in Childhood Cancer

Historical Overview

Early strides in improving oncologic outcomes through the use of single chemotherapeutic agents were first reported by Farber in 19481 and Li in 1956.2 As additional chemotherapeutic agents were developed, they were combined in multidrug regimens that demonstrated both significantly improved response rates and response duration.3,4 By the late 1970s, multimodal therapy was shown to improve cure rates in children with Wilms tumor5 and was being adopted for the treatment of rhabdomyosarcoma, Ewing sarcoma, lymphoma, and other solid tumors.6 The close collaboration of multidisciplinary cooperative groups and the development of improved supportive care measures added impetus to progress. During the 1990s, dose-intensive chemotherapy programs were shown to be successful in improving outcome for patients with Burkitt lymphoma, neuroblastoma, and other advanced-stage solid tumors.79 In addition, improvements in outcome were achieved either by alternating effective groups of chemotherapeutic agents to overcome or prevent resistance,10 or by administering agents by continuous infusion rather than bolus.11

By 2001, noncytotoxic biologic therapies (e.g., signal transduction inhibitors, various tissue growth factor receptor inhibitors, antiangiogenesis agents, tumor-targeted antibodies, and adoptive immunotherapy techniques) were developed to target specific biologic pathways.12 In addition, improvements in radiation therapy have led to the development of intraoperative radiation therapy and radiosurgery techniques. Collectively, these advancements have resulted in the continually increasing survival of children with solid tumors and a profound improvement in their quality of life.

The aim of this chapter is to present the key aspects of contemporary multimodal therapy, touching on treatment rationales, the impact of tumor biology on specific treatment approaches, and future trends.

Incidence and Survival Rates

Although childhood cancers account for only 2% of all reported cancer cases in the general population, they account for 10% of all deaths in children.13 On average, 1 to 2 of every 10,000 children in the USA develop cancer each year.14 The distribution of the types of cancer in childhood is different than in adults. Whereas most cancers in adults have an epithelial cell origin, <10% of childhood cancers fall into this category.

The incidence of specific cancers varies by age, gender, and race. Overall, however, it rose from 11.5 cases per 100,000 children in 1975 to 14.8 per 100,000 in 2004.14 The peak incidence (>200 cases per million) is in children younger than 2 years of age. The incidence then decreases to a low of 82.5 cases per million by age 9 years, at which point it begins to rise again through adolescence. In children younger than age 2, central nervous system malignancies, neuroblastoma, acute myeloid leukemia (AML), Wilms tumor, and retinoblastoma account for the majority of diagnoses. In children 2 to 4 years of age, acute lymphoblastic leukemia (ALL) is the most common childhood cancer. After age 9 years, the incidence of Hodgkin disease, osteosarcoma, and Ewing sarcoma begins to increase sharply.13

Over the past several decades, the mortality from childhood cancers has declined dramatically (40%),15 while the 5-year survival rate has risen from 58% in 1975–1977 to close to 80% in 1996–2003.14

Tumor Pathology and Chemotherapeutic Regimens

Although the spectrum of malignancies in children is more limited than that in adults, the exact diagnosis is often more difficult to establish due to the common histologic appearance of small round blue cells. As these primitive malignancies often lack morphologically distinguishing characteristics, Ewing sarcoma, neuroblastoma, lymphoma, small-cell osteosarcoma, and primitive neuroectodermal tumors may appear quite similar by light microscopy. Given that chemotherapy regimens must be carefully tailored to each specific tumor type, pediatric subspecialists have continued to better define prognostic subgroups for many tumors. These subgroups help to determine the best therapy and the dose intensity required for optimal outcomes. The initial step in the accurate diagnosis of a tumor is the availability of adequate tissue with which to make the diagnosis. The quality and quantity of this tissue should be discussed with the surgeon, the pathologist, and the pediatric oncologist before the procedure to ensure the proper handling of the specimen (e.g., the need for fresh tissue, frozen samples, and fixed specimens for histologic and biologic diagnostic use). Whereas light microscopy remains the primary tool of pathologists, immunohistology, electron microscopy, DNA content of tumor, cytogenetic abnormalities, and specific tumor gene expression are now used to determine specific tumor subgroups.

Tumor Biology

Genetic alterations within a single cell, such as the activation of an oncogene or the loss of a tumor suppressor gene, can lead to the accumulation of cells lacking the ability to respond to growth-regulating signals and the subsequent development of cancer.

Understanding normal cell growth and regulation is a prerequisite to understanding both the genetic basis for the development of childhood cancer and the mechanisms of action of chemotherapeutic agents designed to kill rapidly proliferating cancer cells (Fig. 64-1). Normal cell growth occurs by the regulated progression of the cell through the cell cycle of DNA replication and mitosis. This cycle is separated by two intervening growth phases, referred to as G1 and G2. Cells can temporarily leave the cell cycle and enter a resting state referred to as G0. They are programmed to proceed through the cell cycle by a series of external and internal stimuli. Binding of proteins (growth factors) to cell surface receptors stimulates a cascade of cytoplasmic signaling proteins (membrane kinases and signal transducers) that carries the stimuli to the nucleus. Other proteins (transcription factors) then bind to the DNA, resulting in the expression of growth-regulating genes. When functioning normally, these genes promote or prevent cell division, direct the cell to differentiate, or initiate apoptosis, the process of programmed cell death.

Alterations in one or several of these signaling proteins can lead to the unregulated cell growth characteristics of cancer cells. Oncogenes result from mutation or overexpression of the normal growth-promoting proto-oncogenes. Tumor suppressor genes are normally present in cells and function as negative regulators to slow the process of proliferation and allow time for cellular repair. When oncogenes become activated or tumor suppressor gene function is lost, cells lose their ability to respond to the usual regulatory protein stimuli and proliferate rapidly. Rapid cell proliferation leads to accumulation of more genetic defects, activation of additional oncogenes, and loss of more negative regulators as the cells become increasingly more malignant. Through the study of chromosomal aberrations, more than 100 oncogenes and 25 tumor suppressor genes have now been identified.

Cancer Cytogenetics

With the discovery of chromosomal banding techniques in the 1970s, cancer cytogeneticists were able to identify sub-chromosomal deletions, inversions, and translocations occurring in cancer cells. Investigation of these aberrant regions led to the identification of oncogenes and tumor suppressor genes, a process that continues today.

The presence of consistent cytogenetic abnormalities associated with a specific childhood leukemia or solid tumor is useful in both cancer diagnosis and prognosis. Specific cytogenetic aberrations have been identified in rhabdomyosarcoma, Ewing sarcoma, synovial sarcoma, germ cell tumors, medulloblastoma, neuroblastoma, retinoblastoma, and Wilms tumor.16 Chromosomal aberrations can also be helpful in predicting prognosis. For example, the finding of a chromosome 1q deletion, the presence of double-minute chromatin bodies, or the presence of homogeneous staining regions in neuroblastoma confer a poor prognosis.17

The treatment of Wilms tumor based on risk stratification determined by multiple tumor characteristics that impact prognosis (including loss of heterozygosity at 1p and 16q) is a topic of ongoing study. In the future, specific tumors may be identified by a specific ‘fingerprint’ determined by microarray analysis that can simultaneously analyze expression of thousands of genes on a single chip.18 The ability to tailor therapy to individual patients based on the genetic characteristics of their particular tumor is quickly becoming a reality.

Clincial Trials

The goal of chemotherapy in children is to maximize tumor kill while maintaining acceptable side effects. Clinical trials have led to the development of combination chemotherapy regimens for most childhood cancers. Adjuvant therapy (initiated after local control measures) has remained the mainstay of treatment, though neoadjuvant chemotherapy (initiated before definitive local control measures) has proved to be effective in patients with metastatic disease as well as in those with large primary tumors. In the latter clinical setting, chemotherapy is used to decrease the size of the tumor, making it more amenable to complete resection.

The first step in the clinical development of an anticancer agent is to define a tolerable dose. Phase I clinical trials are designed as dose-escalation studies to determine the maximally tolerated dose of a new drug given as a single agent. In a phase II trial, a consistent dose of the agent is tested for efficacy in a variety of tumor types to establish the spectrum of activity of the agent. Once an agent has demonstrated activity toward a specific cancer, this agent is tested for efficacy when combined with other known active agents for that tumor system.

Standard therapy for a specific tumor type is established through phase III clinical trials. These trials use a prospective randomized design to compare two chemotherapy combinations. At the conclusion of the trial, the chemotherapy regimen with the greatest efficacy and the least toxicity is selected as the standard regimen for that tumor. Subsequent phase III trials compare new regimens with this newly established standard. It is through the development of phase I through III clinical trials within national cooperative groups that treatment advances are made.

Combination chemotherapy remains the mainstay of treatment. The likelihood of cure is maximized when all available active agents are administered simultaneously after local control measures have been undertaken and the tumor burden is as low as possible.19 This approach has led to increased survival rates in children with neuroblastoma, Ewing sarcoma, anaplastic Wilms tumor, and osteosarcoma.

Adjuvant and Neoadjuvant Chemotherapy

The use of adjuvant chemotherapy is supported by the finding that less than 20% of sarcoma and lymphoma patients with initially nonmetastatic solid tumors can be cured by operative or radiation therapy alone or combined.20 Tumor recurrence is generally at a distant site, lending support to the hypothesis that micrometastatic disease exists at the time of presentation for most patients with clinically nonmetastatic disease. As many as 40% of patients with Wilms tumor can be cured with resection or radiation therapy alone. However, survival increases to 90% with the addition of combination chemotherapy.21

As the goal of adjuvant chemotherapy is to prevent the growth of metastatic disease, it is vital that chemotherapy begins as soon as possible after local control measures are employed. For this reason, most current chemotherapy protocols for childhood solid tumors advise that chemotherapy is initiated within 2 weeks of operation.22 In children with Wilms tumor, rapid assessment of tumor biology and risk stratification is important for determining the appropriate chemotherapy regimen.

Neoadjuvant chemotherapy has become standard in the treatment of Ewing sarcoma and osteosarcoma, and has the theoretical advantage of minimizing resistance to chemotherapy.10,23,24 Delayed surgical intervention may allow a more complete or less morbid resection, as well as histologic assessment of tumor responsiveness to the chemotherapy agents. Neoadjuvant chemotherapy is beneficial only in tumors for which a known highly effective combination chemotherapy program limits the risk of tumor progression at the primary site. For example, diagnostic biopsy followed by neoadjuvant chemotherapy and delayed resection of the primary tumor for complex neuroblastoma reduces the operative complication rate without compromising survival.25

Dose Intensity and Duration of Chemotherapy

Advances in supportive care have enabled increased dose intensity of active chemotherapy agents in pediatric clinical trials. These advances decrease or minimize the toxic effects of higher-dose chemotherapy on normal tissues. The use of cytokines (granulocyte colony-stimulating factor [G-CSF] and interleukin-11 [IL-11]) to speed recovery of white blood cells and platelets,26,27 and the use of cardioprotectant agents to allow use of a higher cumulative dose of doxorubicin have helped in the development of new dose-intensive therapy for solid tumors.28 Similarly, progress in bone marrow and stem cell transplantation have allowed dose intensities to be pushed to the upper limits.2932

The positive impact of increasing the dose intensity on improving response rate and survival has been demonstrated for Burkitt lymphoma, osteosarcoma, Ewing sarcoma, testicular cancer, and advanced ovarian cancer.7–9,3336 The duration of chemotherapy programs for most pediatric solid tumors has been 1 year. However, as the dose intensity of chemotherapy programs has increased, the duration of therapy has concomitantly decreased. This downward trend is likely to continue.

Chemotherapeutic Agents

Chemotherapy drugs are divided into classes by their mechanism of action. These classes include alkylating agents (cisplatin and its analogs), antimetabolites, topoisomerase inhibitors, antimicrotubule agents, differentiation agents, miscellaneous nonclassified agents, and biologic agents. Understanding the mechanism of action for each of these agents helps in establishing combination therapies with synergistic antitumor effects. The most common agents from each class, their mechanism of action, common side effects, and tumors in which they are active are listed in Table 64-1.

TABLE 64-1

Chemotherapeutic Agents

image

image

A, alopecia; ALL, acute lymphoblastic leukemia; APML, acute promyelocytic leukemia; BMT, bone marrow transplant; BP, blood pressure; CNS tumors, central nervous system tumors; HD, high dose; myelo, myelosuppression; NB, neuroblastoma; neuro, neurologic toxicity; N/V, nausea and vomiting; PEG, polyethylene glycol; rhabdo, rhabdomyosarcoma; SIADH, syndrome of inappropriate antidiuretic hormone; VOD, veno-occlusive disease.

Data from Balis FM, Holcenberg JS, Poplack DG. General principles of chemotherapy. In: Pizzo P, Poplack D, editors. Principles and Practice of Pediatric Oncology. 3rd ed. Philadelphia: Lippincott-Raven; 1997. p. 215–72; Ratain M, Teicher B, O’Dwyer P, et al. Pharmacology of cancer chemotherapy. In: DeVita V, Hellman S, Rosenberg S, editors. Cancer: Principles and Practice of Oncology. Philadelphia: Lippincott-Raven; 1997. p. 375–85; Dorr R, Von Hoff D. Drug monographs. In: Dorr R, Von Hoff D, editors. Cancer Chemotherapy Handbook. Norwalk, CT: Appleton & Lange; 1994. p. 129.

Acute Chemotherapy Toxicity and Supportive Care

Most acute toxicities in childhood solid tumor therapy are reversible. Because toxicity is greatest in normal cells that have the highest rate of turnover, normal bone marrow cells, mucosal lining cells, liver cells, and hair cells are frequently affected. The most common side effects from combination chemotherapy include nausea and vomiting, myelosuppression, hair loss, mucositis, diarrhea, liver function abnormalities, and allergic reactions.

Myelosuppression is an expected side effect of almost all treatment protocols for solid tumors. Transfusions of packed cells and platelets are frequently needed. Of greatest concern is the risk of severe life-threatening bacterial or fungal infections that occur during episodes of neutropenia. In dose-intensive regimens, more than 75% of chemotherapy courses result in hospitalization for fever, with the incidence of bacteremia ranging from 10– 20% per course.37

Several chemotherapeutic agents have specific toxicities. For example, vincristine and doxorubicin are vesicants and can cause severe skin and tissue necrosis if the drug extravasates into the subcutaneous tissue. Because doxorubicin and related anthracyclines have cumulative cardiotoxic effects, the total lifetime anthracycline dose must be limited for each patient to minimize the risk of developing congestive heart failure. Cisplatin has toxic renal effects and is often combined with another nephrotoxic agent, ifosfamide, in the treatment of osteosarcoma and neuroblastoma. Cisplatin also can cause hearing loss, especially in patients who receive high doses. Vincristine and vinblastine can cause cumulative peripheral neuropathies, and drug doses frequently must be altered to prevent significant morbidity. All of these toxicities must be considered when designing therapeutic programs.

Success in improving treatment outcomes is partially attributed to advances in supportive care. The routine use of hematopoietic growth factors, specifically G-CSF, results in more rapid granulocyte recovery and shorter hospitalizations for fever and neutropenia.26 In addition, IL-11 enhances platelet recovery, decreases the depth of the platelet nadir, and decreases platelet transfusion requirements.27,38,39 It is well tolerated and is beneficial in combination regimens that induce severe thrombocytopenia.40,41

The gastrointestinal tract is injured by cytarabine, anthracyclines, and high-dose methotrexate; however, the folate derivative Leucovorin can be given to rescue normal mucosal and bone marrow cells from the effects of the latter. No rescue is known for the mucositis and diarrhea which occur with other agents. In addition to enhancing platelet production, IL-11 may help speed recovery from gastrointestinal injury after chemotherapy.42

Renal toxicity can occur from the use of cisplatin, ifosfamide, and high-dose methotrexate. Cisplatin causes renal tubular damage, leading to elevation of levels of blood urea nitrogen and creatinine; this effect is generally reversible. Both ifosfamide and cisplatin cause renal electrolyte wasting in which hypokalemia, hypocalcemia, hypophosphatemia, and hypomagnesemia can occur. Renal injury from these agents can be improved by hyperhydration and forced diuresis. Mesna can prevent hemorrhagic cystitis resulting from cyclophosphamide and ifosfamide by binding to the bladder-toxic acrolein metabolites.40

Additionally, a recent study demonstrates that amifostine administered prior to and during cisplatin infusion significantly reduces the risk of severe ototoxicity in children with average risk medulloblastoma who are undergoing treatment with dose-intense chemotherapy.43

Targeting Biologic Pathways

Over the past several decades, many of the key genetic events that control carcinogenesis have been identified. More recently, progress has been made in the development of agents designed to target specific biologic pathways. These agents include signal transduction inhibitors (tissue growth factor receptor inhibitors, antiangiogenesis agents, and biologic response modifiers), individual cytokines, tumor-targeted antibody therapies, and adoptive immunotherapy techniques. The development of these agents differs from the development of agents used in standard cytotoxic therapy. Whereas standard phase I studies for cytotoxic agents are designed to define the maximal tolerated dose, in biologic targeted therapy the optimal therapeutic dose is well below the maximal tolerated dose. The challenge in evaluating these newer agents is how to select the optimal dose and schedule, combine tumor-targeted agents with classic cytotoxic therapy, and validate the intended effect on the selected target for these biologic compounds designed to treat minimal residual disease.

Biologic Response Modifiers

The goal of biologic response modifiers is to stimulate the immune system to help eradicate tumors. The human immune system is designed to identify and destroy foreign cells. One of the mysteries of oncology is why a patient’s immune system is often unable to eliminate malignant cells. The development of biologic response modifiers is a relatively new branch of cancer therapy. It is designed to enhance or stimulate the natural products of the immune system (lymphocytes, antibodies, and cytokines) to better recognize and destroy cancer cells.

T-lymphocytes directly interact with specific cell surface antigens on a target cell, causing cell lysis through cytotoxic granule release or programmed cell death. These cytotoxic lymphocytes are involved with the killing of tumor cells. To initiate this response, antigens must be presented to the T cell by antigen-presenting cells (APCs) that express the antigens bound to major histocompatibility complex proteins in the presence of stimulatory cytokines. Cytokines or interleukins (e.g., IL-2, interferon-α, tumor necrosis factor) are proteins produced by helper T-lymphocytes and monocytes that help recruit other effector cells, including APCs, as well as regulate antibody production. The effector cells of the immune system (e.g., granulocytes, monocytes, macrophages, eosinophils, dendritic cells) can become tumor selective when activated by a specific antibody, a process called antibody-dependent cell-mediated cytotoxicity.

Immunotherapy with biologic response modifiers takes advantage of all these immune functions. The goal of this therapy is to improve the immunogenicity of a tumor and allow it to be recognized and targeted for destruction by the immune system. Immunotherapy can be divided into two major categories: adoptive immunotherapy and tumor-targeted antibody therapy.

Tumor-Targeted Antibody Therapy

Passive immunity involves the use of monoclonal antibodies (mAbs) or cytotoxic effector cells produced in vitro and infused into the patient. mAbs have been tested in patients with neuroblastoma. A drawback of murine mAb therapy is that when these antibodies are repeatedly infused into humans, most patients will ultimately produce a human/anti-mouse antibody, which renders further antibody treatment useless.

Chimeric human/mouse antibodies have been produced that decrease the risk of human/anti-mouse antibody generation.48 Recombinant chimeric antibodies are produced by linking the constant region of human antibodies to the variable combining region of a mouse mAb. These chimeric antibodies have been successfully used in the treatment of children with high-risk neuroblastoma.49

Although this field of targeted biologic therapy is still in its infancy, the future looks bright for the continued development of new targeted therapies. These therapies are the outgrowth of the rapid advances being made in the understanding of cellular signaling pathways and immune mechanisms.

Local Tumor Control

As metastatic disease in young patients is more readily eradicated than in adults, control of local disease is critical to favorable outcomes in children. Advances in techniques used to obtain local control (other than complete surgical excision) are continually changing as new technology offers more and more opportunity to effectively treat disease locally while minimizing morbidity.

Radiation Oncology

Given the important role played by radiation therapy in the treatment of numerous pediatric tumors, it is important for clinicians to understand its biologic principles. Radiation impacts tumor growth through two primary mechanisms of action. Radiation can have a direct impact on the cellular DNA, resulting in impaired cell division. Alternatively, it can result in the production of reactive free radicals that indirectly damage genetic material and interfere with the reproductive capacity of normal or malignant tissues. In most cases, the radiation effect is through production of these free radicals.

The sensitivity of normal and malignant cells to radiation varies widely between cell populations. Ionizing radiation initially results in sublethal damage to cells. The therapeutic effect of radiation therapy exploits the differences between the ability of a normal cell to repair this sublethal damage and the slower response of radiosensitive tumor cells. Fractionated dosing allows normal cells to recover while having a cumulative effect on tumor cells. The effect of ionizing radiation on tumors depends on the number of actively reproducing cells at the time of exposure and the length of the cellular regeneration cycle. As most of the damage is indirect and focused on reproduction, malignant lesions usually show a delayed effect to radiation therapy. The tumor may begin to shrink or eventually disappear weeks to months after treatment.

Acute reactions to ionizing radiation depend on the balance between replication and cell death. These reactions are affected by increased intervals between dose fractions that allow enhanced cellular repopulation. The radiation fraction size has a small impact on what volume of cells are immediately destroyed. Conversely, the long-term effects of therapy depend primarily on the total exposure dose and the size of each treatment fraction. The therapeutic ratio may be enhanced by exploiting the difference between the early and late radiation effects. Techniques can be used to reduce the late effects by lowering the dose per fraction and increasing the number of fractions delivered over the conventional treatment time.

Radiation therapy may be combined with operation in a strategic manner to deliver the highest effective dose to a well-defined site while minimizing the dose to surrounding normal structures.50 Preoperative radiation may permit a smaller treatment area because the operative bed has not been manipulated. In larger tumors, preoperative radiation may reduce the lesion volume sufficiently to allow a subsequent resection. In addition, potential tumor seeding during operative removal may be reduced because cells that may be surgically disseminated have been rendered incapable of reproducing. On the other hand, preoperative radiation may delay the surgical procedure and alter the staging information obtained at operation.

For these reasons, many combined strategies use postoperative radiation, which allows the treatment fields and doses to be determined after operative resection and histologic assessment.51 Higher doses can be delivered postoperatively once the target volumes have been more accurately defined. Doses to the periphery of the tumor can be fine-tuned, depending on the presence of gross, microscopic, or no residual disease. However, postoperative delivery may require a wider treatment area after extensive surgical manipulation.

Soft tissue sarcoma provides a model for the adjunctive role of radiation therapy. Resection is the primary method of obtaining local control; however, radiation therapy can be effective when clear margins are not possible.52 Combined therapy has resulted in dramatically improved survival.53 In extremity lesions, radiation also allows more conservative resection with limb sparing. Although local tumor control rates of 75–98% have been achieved with limb salvage rates greater than 80%, wound complications occur in as many as 40% of patients. Neoadjuvant radiation at more modest doses (30 Gy total) has decreased the complication rate while maintaining excellent (>95%) five-year local control, and ultimately, limb salvage.52 Postoperative adjuvant radiation therapy also may be advantageous.54

Several aspects of radiation treatment in pediatric patients warrant special consideration. Attention should be paid to immobilizing or sedating children so that ionizing doses can be targeted to the desired area without inappropriate exposure of surrounding tissues. Pediatric radiation oncologists may use lower treatment doses and accept a higher recurrence rate to ensure lower toxicity, especially in important developing organs such as the brain. The normal tolerance of organs or tissues may be adversely affected when chemotherapeutic agents are also used. The long-term effects of combined-modality therapy must be considered in regard to musculoskeletal and dental tissues, central nervous system (CNS) and neuropsychological sequelae, and endocrine and gonadal dysfunction, as well as direct effects on the heart, lungs, or kidneys.55 The following sections describe techniques that allow safe, efficacious doses of radiation to be delivered, often in combination with surgical excision, to produce the maximal therapeutic benefit.

Brachytherapy

Brachytherapy is radiation treatment in which the ionizing source is in contact with the lesion, usually within the primary tumor. Catheters are placed in the tumor during surgery and may be loaded with temporary or permanent implant sources. Remote afterloading decreases radiation exposure to personnel and family members, and can be performed in the patient’s room or on an outpatient basis. Low-dose-rate sources such as cesium provide about 1 cGy/min, whereas high-dose-rate sources such as iridium provide about 100 cGy/min.

As interstitial implants allow continuous-dose delivery over a much shorter time, they offer a radiobiologic advantage in high-grade tumors with rapid cell growth kinetics. Close cooperation between surgeon and radiation oncologist during the implant procedure is critical to ensure the most effective mapping of the tumor bed target.

Children with soft tissue sarcomas can benefit from specialized radiation treatment strategies. For children who have microscopic residual disease after operation, radiation provides excellent local tumor control.56 High-dose-rate brachytherapy can be delivered in only a few minutes, which is particularly helpful in young children. In contrast, low-dose-rate brachytherapy provided by low energy source techniques requires sedation, immobilization, long exposure times, and hospitalization. The short therapy duration associated with high-dose-rate therapy also allows rapid reinstitution of chemotherapy. Morbidity is usually related to skin or mucosal reactions, which may progress as a ‘recall’ phenomenon in patients treated with radiosensitizing agents such as anthracyclines.57 Brachytherapy, alone or in combination with external-beam irradiation, has been shown to provide a high rate of local tumor control in pediatric soft tissue sarcomas.5860

Intraoperative Radiation

Intraoperative radiation therapy (IORT) allows the radiation dose to be directly applied to the target area while shielding adjacent structures. Whenever disease remains in surgically inaccessible areas, IORT may be an effective adjunct. Phase I and II studies have demonstrated that IORT can be performed safely in children.61 It is used for patients in whom unresectable disease is present at diagnosis or for delayed primary or second-look procedures, residual lesions, or local tumor recurrence.

In children with retroperitoneal tumors, IORT may, however, lead to urologic complications. In a study reported in 1990, 3 of 6 patients treated with IORT and external-beam therapy required intervention for fibrotic ureteral strictures or renal artery stenosis; in two cases, the injured structures were within the supplemental external-beam treatment field. Also, neuropathies developed in two other patients. Nevertheless, all patients were survivors for up to 42 months’ follow-up.62 In a later study, the complications were minimized through more extensive dissection of normal structures and avoidance of overlapping radiation fields.63

Intensity-Modulated Radiation Therapy

Techniques continue to evolve to improve the impact of radiation therapy on tumor response while minimizing the dose of radiation imparted to surrounding normal tissues. Stereotactic radiation therapy is sometimes used for CNS tumors.6467 Imaging systems, treatment-planning software, and delivery systems have undergone dramatic advancements that allow sophisticated delivery of more precise courses of radiation. Intensity-modulated radiation therapy (IMRT) is an advanced form of three-dimensional conformal therapy that uses non-uniform radiation beam intensities determined by using various computer-based optimization techniques. Experience with IMRT in children is growing. Reports of mixed tumor cases, including pediatric patients, suggest that IMRT will be effective in reducing treatment-related morbidity and allow dose escalation to the target volume.68,69 Significant reductions in radiation exposure to critical structures has been shown for intracranial, cervical, and abdominopelvic lesions.70

Proton Therapy

The driving principle in the evolution of radiation therapy techniques is to provide high doses of radiation to the tumor mass while reducing the dose delivered to surrounding tissues susceptible to the early and late complications associated with ionizing radiation. Proton therapy relies on the same mechanism of action as other forms of radiation therapy, but takes advantage of the physical properties of the energy transfer achieved to increase the precision of the radiation doses delivered. The physics of proton therapy result in less energy deposited in the tissue between the skin and the tumor, and increased energy delivered to the tumor when compared to photon therapy. This yields a biological effectiveness 10% greater than photons at the tumor site and the potential for significantly less impact on the surrounding tissue.

Proton therapy is very costly and there are currently fewer than 50 centers in the world capable of providing this treatment. Although most patients who have undergone therapy to date are adults, limited experience with pediatric CNS tumors and sarcomas has been reported.71,72 Researchers at the Massachusetts General Hospital have described 30 patients with Ewing sarcoma who received proton therapy as a component of multimodal management. Three-year local control in 86% of patients was achieved with few adverse events.71 The utility of proton therapy in other childhood tumors, including neuroblastoma and rhabdomyosarcoma, is also being explored.72

Innovative Adjunctive Techniques

Radiofrequency Ablation

Radiofrequency ablation (RFA) is a technique that applies thermal energy via a probe that results in coagulation necrosis of the target tissue. The technique involves image-guided application of the probe, primarily using ultrasound. The probe can be introduced percutaneously, laparoscopically, or via an open exposure. In adults, the most common applications are for primary or metastatic hepatic lesions, renal tumors, and pulmonary lesions.7377 Treatment of pediatric tumors with RFA is largely anecdotal. A small series reported the use of percutaneous RFA on fetuses with sacrococcygeal teratoma, but found a 50% fetal mortality rate.78 Another report described severe soft tissue injury and sciatic nerve destruction at birth in an infant treated prenatally.79

Transcatheter Arterial Chemoembolization

Transcatheter arterial chemoembolization (TACE) is a technique used to directly instill chemotherapeutic agents into a tumor, thereby minimizing systemic toxicity. TACE is most commonly used in the management of liver lesions, taking advantage of the preferential arterial blood supply to these tumors.80,81

Although the pediatric experience with TACE is limited, there is some evidence to support its efficacy.82,83 In one series, a suspension of cisplatin, doxorubicin, and mitomycin-C mixed with bovine collagen and radiopaque contrast material was used in 11 children with unresectable or recurrent lesions.82 Six hepatoblastoma patients had an initial partial response, as measured by imaging and α-fetoprotein levels. Of these patients, three underwent subsequent surgical resection; one had progressive disease and died, and two survived for more than 15 months. The other three patients eventually also died of known progressive disease. In three children with hepatocellular carcinoma, one underwent surgical resection and was a long-term survivor for more than 65 months, one was alive with disease for more than 36 months, and one died of progressive liver failure with no evidence of malignancy. In another relatively recent report documenting the outcomes of 16 infants and children with unresectable hepatoblastoma, Li et al reported that TACE facilitated safe and complete surgical resection in 12 cases.83 Three patients underwent partial resection. One patient underwent successful orthotopic liver transplantation after receiving TACE therapy.

Overall, chemoembolization is feasible in young patients and is associated with tolerable toxicity. This option represents a possible therapeutic alternative in persistent, unresectable, or recurrent hepatoblastoma, or in nonmetastatic hepatocellular carcinoma. Its use has also been proposed in unresectable Wilms tumor.84

Sentinel Lymph Node Biopsy

The utility of sentinel lymph node biopsy in children is evolving. Although it is standard practice for patients with intermediate thickness melanomas,85 its value in soft tissue sarcomas remains unclear.86,87

The initial draining lymph node (sentinel node) is predictive of regional nodal metastases in a variety of tumors.88 It is most commonly used to predict nodal status of patients with melanoma or breast cancer. In most cases, a combination of technetium-labeled sulfur colloid with either Lymphazurin or methylene blue dye is used to localize the sentinel node. Preoperative lymphoscintigraphy provides information regarding the location of the primary lymph node draining the tumor site. Lymphatic mapping is accomplished by injecting the technetium-labeled sulfur colloid at the primary tumor site. The affected region is then imaged to identify the sentinel lymph node. Just before incision, the blue dye is injected at the primary tumor site and a gamma probe is used to identify areas of high counts. The underlying tissue is then examined for lymph nodes containing the blue dye. The lymph nodes are removed and sent for histopathologic examination. If the sentinel node shows no evidence of metastases, the related regional lymphatic bed is highly likely to be tumor free. In these patients, the morbidity of lymphadenectomy can be avoided.

Late Effects of Cancer Therapy

According to the most recent National Cancer Institute statistics: (1) there are more than 328,000 survivors of childhood cancer in the USA and (2) 1 in 900 adults is now a cancer survivor.89 The remarkable increase in survival has created a new and growing population who are at increased risk for late adverse effects of treatment and a diminished quality of life. All aspects of combined-modality therapy can contribute to the late effects. A number of chemotherapy agents have been associated with late toxicities. In addition, radiation therapy can significantly inhibit further growth of bone, muscle, heart, and kidney within the radiation field, and also can affect fertility.

Tissues with the highest cell-turnover rate are generally the most susceptible to the acute toxicities of chemotherapy, whereas tissues that replicate slowly or that can no longer regenerate may be susceptible to the late effects of therapy. Children are more susceptible to certain late effects of therapy than are adults because their tissues are still growing. Damage to these tissues may affect growth, fertility, and neuropsychological development.

Growth retardation is a late effect unique to children. The degree of impairment depends on the dose of chemotherapy or radiation and the age of the child at the time of therapy. The younger the child is at the time of the insult, the more severe the sequelae. More than 50% of childhood brain tumor patients treated with 3000 cGy or more to the whole brain will have severe growth retardation, with adult height being lower than the 5th percentile.90 Cranial irradiation can lead to growth hormone deficiency, which results in poor linear growth, unless growth hormone replacement is given. In addition, patients who have received total-body irradiation or spinal radiation may not be able to achieve their full height potential because the irradiated bones have limited growth potential, even with growth hormone stimulation.91

Adjuvant therapy can also cause musculoskeletal problems, including scoliosis, avascular necrosis, osteoporosis, and atrophy or hypoplasia of tissues. Radiation to the head and neck results in hypoplasia of the jaw, orbit, or neck, with associated atrophy of the soft tissues. Associated endocrine, dental, and psychological consequences can occur.92 Osteoporosis occurs as a result of corticosteroid treatment and from high-dose irradiation, as used for sarcomas.

Specific organs are often affected by chemotherapy. Heart, liver, lung, thyroid, and gonadal function are often impaired. Gonadal dysfunction (azoospermia, amenorrhea) frequently results from alkylator treatment. Therapy with mechlorethamine, vincristine, prednisone, and procarbazine has resulted in azoospermia in 80–100% of all male patients.93 Combination chemotherapy programs for childhood Hodgkin disease have been adjusted to replace mechlorethamine with cyclophosphamide and eliminate dacarbazine from standard treatments in an attempt to decrease the infertility risk. It should be noted that the children of childhood cancer survivors are not at an increased risk for congenital anomalies.94

Cardiotoxicity from anthracycline antibiotics has been a problem in the treatment of Ewing sarcoma, osteosarcoma, and lymphomas. However, the use of continuous infusion anthracycline can decrease the risk of cardiac muscle damage and subsequent congestive heart failure.95 Another strategy is the use of dexrazoxane to prevent anthracycline-induced cardiotoxicity.28 Survivors of childhood brain tumor therapy treated with a combination of chemotherapy, irradiation, and surgery had a significantly increased risk of stroke, blood clots, and angina-like symptoms compared with their siblings.96

Pulmonary toxicity is a source of significant late toxicity of cancer therapy. Many alkylating agents and radiation therapy contribute to pulmonary fibrosis, resulting in decreased lung volume, lung compliance, and diffusing capacity. Nitrosoureas and bleomycin are the most common agents to cause pulmonary fibrosis.

Other significant organ-related late effects include chronic renal insufficiency from cisplatin therapy, chronic cystitis from cyclophosphamide or ifosfamide treatment, and prolonged hypogammaglobulinemia and T-lymphocyte dysfunction after multiple high-dose alkylators for bone marrow transplant.97

The most significant late effect of cancer therapy is the risk of secondary malignancy. This risk is highest in patients who have received both chemotherapy and radiation therapy. In 2001, Neglia and colleagues reported that the estimated cumulative incidence of all subsequent malignant neoplasms in a cohort of 13,581 childhood cancer survivors was 3.2% at 20 years after the primary diagnosis.98 Hodgkin disease survivors have the highest secondary malignancy rates. Breast cancer is the most common solid tumor, with an estimated actuarial incidence in women of 35% by age 40 years. These patients are also at risk of developing leukemia, non-Hodgkin lymphoma, and thyroid carcinoma.99 In survivors of childhood Hodgkin disease, the cumulative estimated incidence of second malignancies at 30 years ranges from 18% to 31%.100,101 Patients who have received additional multimodality therapy for recurrent Hodgkin disease have the highest risk of second tumors. Patients with soft tissue sarcomas, retinoblastoma, and Ewing sarcoma who receive high-dose radiation to the primary lesion are at increased risk for secondary osteosarcoma within the radiation field.102 Etoposide has been recognized as causing secondary AML.

References

1. Farber, S, Diamond, LK. Temporary remissions in acute leukemia in children produced by folic acid antagonist, 4-aminopteroyl-glutamic acid (aminopterin). N Engl J Med. 1948; 238:787–793.

2. Li, MC, Hertz, R, Spencer, D, et al. Effect of methotrexate upon choriocarcinoma and chorioadenoma. Proc Soc Exp Biol Med. 1956; 93:361–366.

3. Frei, E, III., Freireich, EJ, Gehan, E, et al. Studies of sequential and combination antimetabolite therapy in acute leukemia: 6-Mercaptopurine and methotrexate. Blood. 1961; 18:431–454.

4. Green, DM, Jaffe, N. Wilms’ tumor: Model of a curable pediatric malignant solid tumor. Cancer Treat Rev. 1978; 5:143–172.

5. D’Angio, GJ, Evans, AE, Breslow, N, et al. The treatment of Wilms’ tumor: Results of the National Wilms’ Tumor Study. Cancer. 1976; 38:647.

6. Link, MP, Goorin, AM, Miser, AW, et al. The effect of adjuvant chemotherapy on relapse-free survival in patients with osteosarcoma of the extremity. N Engl J Med. 1986; 314:1600–1606.

7. Schwenn, MR, Blattner, SR, Lynch, E, et al. HiC-COM: A 2-month intensive chemotherapy regimen for children with stage III and IV Burkitt’s lymphoma and B-cell acute lymphoblastic leukemia. J Clin Oncol. 1991; 9:133–138.

8. Cheung, NK, Heller, G. Chemotherapy dose intensity correlates strongly with response, median survival and median progression-free survival in metastatic neuroblastoma. J Clin Oncol. 1991; 9:1050–1058.

9. Antman, K, Ayash, L, Elias, A, et al. A phase II study of high dose cyclophosphamide, thiotepa, and carboplatin with autologous marrow support in women with measurable advanced breast cancer responding to standard therapy. J Clin Oncol. 1992; 10:102–110.

10. Grier, H, Krailo, M, Link, M, et al. Improved outcome in non-metastatic Ewing’s sarcoma and PNET of bone with the addition of ifosfamide and etoposide to vincristine, Adriamycin, cyclophosphamide, and actinomycin: A Children’s Cancer Group and Pediatric Oncology Group report. J Clin Oncol. 1994; 13(Suppl):421.

11. Clark, PI, Slevin, ML, Joel, SP, et al. A randomized trial of two etoposide schedules in small-cell lung cancer: The influence of pharmacokinetics on efficacy and toxicity. J Clin Oncol. 1994; 12:1427–1435.

12. Worth, L, Jeha, S, Kleinerman, E. Biologic response modifiers in pediatric cancer. Hematol Oncol Clin North Am. 2001; 5:723–740.

13. Parker, SL, Tong, T, Bolden, S, et al. Cancer statistics, 1997. CA Cancer J Clin. 1997; 47:5–27.

14. National Cancer Institute fact sheet [Internet]. Available at http://www.cancer.gov/cancertopics/factsheet/Sites-Types/childhood.

15. Gloeckler Ries, LA, Percy, CL, Bunin, GR. SEER Pediatric Monograph. Available at http://seer.cancer.gov/publications/childhood/introduction.

16. Kreissman, SG. Molecular genetics: Toward an understanding of childhood cancer. Semin Pediatr Surg. 1993; 2:2–10.

17. Brodeur, GM. Neuroblastoma: Clinical applications of molecular parameters. Brain Pathol. 1990; 1:47–54.

18. MacGregor, PF. Gene expression in cancer: The application of microarrays. Expert Rev Mol Diagn. 2003; 3:185–200.

19. Goldie, JH, Coldman, AJ. A mathematic model for relating the drug sensitivity of tumors to the spontaneous mutation rate. Cancer Treat Rep. 1979; 63:1727–1731.

20. Balis, FM, Holcenberg, JS, Poplack, DG. General principles of chemotherapy. In Pizzo P, Poplack D, eds.: Principles and practice of pediatric oncology, 3rd ed, Philadelphia: Lippincott-Raven, 1997.

21. D’Angio, GJ, Evans, AE, Breslow, N, et al. The treatment of Wilms’ tumor: Results of the National Wilms’ Tumor Study. Cancer. 1976; 38:633–646.

22. Green, DM, Breslow, NE, Evans, I, et al. The effect of chemotherapy dose intensity on the hematological toxicity of treatment for Wilms’ tumor: A report from the National Wilms’ Tumor Study. Am J Pediatr Hematol Oncol. 1994; 16:207–212.

23. Picci, P, Rougraff, BT, Bacci, G, et al. Prognostic significance of histopathologic response to chemotherapy in nonmetastatic Ewing’s sarcoma of the extremities. J Clin Oncol. 1993; 11:1763–1769.

24. Provisor, AJ, Ettinger, LJ, Nachman, JB, et al. Treatment of nonmetastatic osteosarcoma of the extremity with preoperative and postoperative chemotherapy: A report from the Children’s Cancer Group. J Clin Oncol. 1997; 5:76–84.

25. Shamberger, RC, Allarde-Segundo, A, Kozakewich, HP, et al. Surgical management of stage III and IV neuroblastoma: Resection before or after chemotherapy? J Pediatr Surg. 1991; 26:1113–1118.

26. Householder, SE, Rackoff, W, Goldman, J, et al. A case-control retrospective study of the efficacy of granulocyte colony stimulating factor in children with neuroblastoma. Am J Pediatr Hematol Oncol. 1994; 16:132–137.

27. Tepler, I, Elias, L, Smith, JW, et al. A randomized placebo-controlled trial of recombinant human interleukin-11 in cancer patients with severe thrombocytopenia due to chemotherapy. Blood. 1996; 87:3607–3614.

28. Lipshultz, SE. Dexrazoxane for protection against cardiotoxic effects of anthracyclines in children. J Clin Oncol. 1996; 14:328–331.

29. Strother, D, Ashley, D, Kellie, SJ, et al. Feasibility of four consecutive high-dose chemotherapy cycles with stem-cell rescue for patients with newly diagnosed medulloblastoma or supratentorial primitive neuroectodermal tumor after craniospinal radiotherapy: Results of a collaborative study. J Clin Oncol. 2001; 19:2696–2704.

30. Welte, K, Reiter, A, Mempel, K, et al. A randomized phase-III study of the efficacy of granulocyte colony-stimulating factor in children with high-risk acute lymphoblastic leukemia. Berlin-Frankfurt-Munster Study Group. Blood. 1996; 87:3143–3150.

31. Burdach, SE, Müschenich, M, Josephs, W, et al. Granulocyte-macrophage-colony stimulating factor for prevention of neutropenia and infections in children and adolescents with solid tumors. Results of a prospective randomized study. Cancer. 1995; 76:510–516.

32. Hawkins, DS, Felgenhauer, J, Park, J, et al. Peripheral blood stem cell support reduces the toxicity of intensive chemotherapy for children and adolescents with metastatic sarcomas. Cancer. 2002; 95:1354–1365.

33. Frei, E, Canellos, GP. Dose: A critical factor in cancer chemotherapy. Am J Med. 1980; 69:585–594.

34. Broun, R, Nichols, CR, Kneebone, P, et al. Long-term outcome of patients with relapsed and refractory germ cell tumors treated with high-dose chemotherapy and autologous bone marrow rescue. Ann Intern Med. 1992; 117:124–128.

35. Smith, MA, Ungerleider, RS, Horowitz, ME, et al. Influence of doxorubicin dose intensity on response and outcome for patients with osteogenic sarcoma and Ewing’s sarcoma. J Natl Cancer Inst. 1991; 83:1460–1470.

36. Kaye, SB, Lewis, CR, Dave, J, et al. A randomized study of two doses of cisplatin and cyclophosphamide in epithelial ovarian cancer. Lancet. 1992; 340:329–333.

37. Kreissman, SG, Rackoff, W, Lee, M, et al. High dose cyclophosphamide with carboplatin: A tolerable regimen suitable for dose intensification in children with solid tumors. J Pediatr Hematol Oncol. 1997; 19:309–312.

38. Gordon, MS, McCaskill-Stevens, WJ, Battiato, LA, et al. A phase I trial of recombinant human interleukin 11 (Neumega rgIL-11 growth factor) in women with breast cancer receiving chemotherapy. Blood. 1996; 87:3615–3624.

39. Ali-Nazir, A, Davenport, G, Reaman, G, et al. A phase I/II trial of rhIL-11 following ifosfamide, carboplatin, and etoposide (ICE) chemotherapy in pediatric patients with solid tumors or lymphoma. J Clin Oncol. 1996; 15(Suppl):274.

40. Shaw, IC, Graham, MI. Mesna: A short review. Cancer Treat Rev. 1987; 14:67–86.

41. Moritz, T, MacKay, W, Glassner, B, et al. Retrovirus-mediated expression of DNA repair protein in bone marrow protects hematopoietic cells from nitrosourea-induced toxicity in vitro and in vivo. Cancer Res. 1995; 55:2608–2614.

42. Sonis, S, Muska, A, O’Brien, J, et al. Alterations in the frequency, severity, and duration of chemotherapy induced mucositis in hamsters by interleukin-11. Eur J Cancer. 1995; 31:261–266.

43. Fouladi, M, Chintagumpala, M, Ashley, D, et al. Amifostine protects against cisplatin-induced ototoxicity in children with average-risk medulloblastoma. J Clin Oncol. 2008; 26:3749–3755.

44. Corey, SJ. Targeted therapy: for kids, too. Review. Pediatr Blood Cancer. 2005; 45:623–634.

45. Levitzki, A, Klein, S. Signal transduction therapy of cancer. Mol Aspects Med. 2010; 31:287–329.

46. Restifo, NP, Snzol, M. Cancer vaccines. In: DeVita VT, Hellman S, Rosenberg SA, eds. Cancer: Principles and Practice of Oncology. 5th ed. Philadelphia: Lippincott-Raven; 1997:3033.

47. Rosenberg, SA. Principles of cancer management: Biologic therapy. In: DeVita VT, Hellman S, Rosenberg SA, eds. Cancer: Principles and Practice of Oncology. 5th ed. Philadelphia: Lippincott-Raven; 1997:364.

48. Frost, JD, Hank, JA, Reahman, GH, et al. A phase I/IB trial of murine monoclonal anti-GD2 antibody 14. G2a plus interleukin-2 in children with refractory neuroblastoma: A report of the Children’s Cancer Group. Cancer. 1997; 80:317–333.

49. Yu, AL, Gilman, AL, Ozkaynak, MF, et al. Anti-GD2 antibody with GM-CSF, interleukin-2, and isotretinoin for neuroblastoma. N Engl J Med. 2010; 363:1324–1334.

50. Eisbruch, A, Lichter, AS. What a surgeon needs to know about radiation. Ann Surg Oncol. 1997; 4:516–522.

51. Liu, L, Glicksman, AS. The role of radiation in the management of soft tissue sarcoma. Med Health RI. 1997; 80:32–36.

52. Temple, WJ, Temple, CLF, Arthur, K, et al. Prospective cohort study of neoadjuvant treatment in conservative surgery of soft tissue sarcomas. Ann Surg Oncol. 1997; 4:586–590.

53. Marcus, KC, Grier, HE, Shamberger, RC, et al. Childhood soft tissue sarcoma: A 20-year experience. J Pediatr. 1997; 131:603–607.

54. Lindberg, RD, Martin, RG, Romsdahl, MM, et al. Conservative surgery and postoperative radiotherapy in 300 adults with soft-tissue sarcomas. Cancer. 1981; 47:2391–2397.

55. Fryer, CJH. Principles of pediatric radiation oncology. In: Holland JF, Frei E, Bast RC, eds. Cancer Medicine. 4th ed. Baltimore: Williams & Wilkins; 1996:2899–2905.

56. Donaldson, S, Breneman, J, Asmar, L, et al. Hyperfractionated radiation in children with rhabdomyosarcoma. Results of an intergroup rhabdomyosarcoma pilot study. Int J Radiat Oncol Biol Phys. 1995; 32:903–911.

57. Nag, S, Grecula, J, Ruymann, FB. Aggressive chemotherapy, organ-preserving surgery, and high-dose-rate remote brachytherapy in the treatment of rhabdomyosarcoma in infants and young children. Cancer. 1993; 72:2769–2776.

58. Nag, S, Olson, T, Ruymann, F, et al. High-dose-rate brachytherapy in childhood sarcomas: A local control strategy preserving bone growth and function. Med Pediatr Oncol. 1995; 25:463–469.

59. Nag, S, Martinez-Monge, R, Ruyman, F, et al. Innovation in the management of soft tissue sarcomas in infants and young children: High dose brachytherapy. J Clin Oncol. 1997; 15:3075–3084.

60. Merchant, TE, Parsh, N, del Valle, PL, et al. Brachytherapy for pediatric soft-tissue sarcoma. Int J Radiat Oncol Biol Phys. 2000; 46:427–432.

61. Zelefsky, MJ, LaQuaglia, MP, Ghavimi, F, et al. Preliminary results of phase I/II study of high-dose-rate intraoperative radiation therapy for pediatric tumors. J Surg Oncol. 1996; 62:267–272.

62. Ritchey, ML, Gunderson, LL, Smithson, WA, et al. Pediatric urologic complications with intraoperative radiation therapy. J Urol. 1990; 143:89–91.

63. Haase, GM, Meagher, D, Jr., McNeely, LK, et al. Electron beam intraoperative radiation therapy for pediatric neoplasms. Cancer. 1994; 74:740–747.

64. Mehta, MP. The physical, biologic, and clinical basis of radiosurgery. Curr Probl Cancer. 1995; 19:265–329.

65. Shaw, E, Scott, C, Souhami, L, et al. Radiosurgery for the treatment of previously irradiated recurrent primary brain tumors and brain metastases: Initial analysis of Radiation Therapy Oncology Group protocol (RTOG) 9005. Int J Radiat Oncol Biol Phys. 1994; 30:166.

66. Smyth, MD, Sneed, PK, Ciricillo, SF, et al. Stereotactic radiosurgery for pediatric intracranial arteriovenous malformations: The University of California at San Francisco experience. J Neurosurg. 2002; 97:48–55.

67. Shrieve, DC, Alexander, E, Wen, PY, et al. Comparison of stereotactic radiosurgery and brachytherapy in the treatment of recurrent glioblastoma multiforme. Neurosurg. 1995; 36:275–284.

68. Teh, BS, Mai, WY, Grant, WH, et al. Intensity modulated radiotherapy (IMRT) decreases treatment-related morbidity and potentially enhances tumor control. Cancer Invest. 2002; 20:437–451.

69. Swift, P. Novel techniques in the delivery of radiation in pediatric oncology. Pediatr Clin North Am. 2002; 9:1107–1129.

70. Bhatnagar, A, Deutsch, M. The role for intensity modulated radiation therapy (IMRT) in pediatric population. Technol Cancer Res Treat. 2006; 5:591–595.

71. Merchant, TE. Proton beam therapy in pediatric oncology. Cancer J. 2009; 15:298–305.

72. Cotter, SE, McBride, SM, Yock, TI. Proton radiation for solid tumors of childhood. Technol Cancer ResTreat. 2012; 11:267–277.

73. Curley, SA. Radiofrequency ablation of malignant liver tumors. Ann Surg Oncol. 2003; 10:338–347.

74. Scaife, CL, Curley, SA. Complication, local recurrence, and survival rates after radiofrequency ablation for hepatic malignancies. Surg Oncol Clin North Am. 2003; 12:243–255.

75. Mayo-Smith, WW, Dupuy, DE, Parikh, PM, et al. Image-guided percutaneous radiofrequency ablation of solid renal masses: Techniques and outcomes of 38 treatment sessions in 32 consecutive patients. AJR Am J Roentgenol. 2003; 180:1503–1508.

76. Farrell, MA, Charboneau, WJ, DiMarco, DS, et al. Image-guided radiofrequency ablation of solid renal tumors. Am J Roentgenol. 2003; 180:1509–1513.

77. Herrera, LJ, Fernando, HC, Perry, Y, et al. Radiofrequency ablation of pulmonary malignant tumors in non-surgical candidates. J Thorac Cardiovasc Surg. 2003; 125:929–937.

78. Paek, BW, Jennings, RW, Harrison, MR, et al. Radiofrequency ablation of human fetal sacrococcygeal teratoma. Am J Obstet Gynecol. 2001; 184:503–507.

79. Ibrahim, D, Ho, E, Scherl, LA, et al. Newborn with an open posterior hip dislocation and sciatic nerve injury after intrauterine radiofrequency ablation of a sacrococcygeal teratoma. J Pediatr Surg. 2003; 38:248–250.

80. Gunvén, P. Liver embolizations in oncology: A review. Part I. (chemo)embolizations. Med Oncol. 2008; 25:1–11.

81. Bittles, MA, Hoffer, FA. Interventional radiology and the care of the pediatric oncology patient. Surg Oncol. 2007; 16:229–233.

82. Malogolowkin, MH, Stanley, P, Steele, DA, et al. Feasibility and toxicity of chemoembolization for children with liver tumors. J Clin Oncol. 2000; 18:1279–1284.

83. Li, J, Chu, J, Yang, J, et al. Preoperative transcatheter selective arterial chemoembolization in treatment of unresectable hepatoblastoma in infants and children. Cardiovasc Intervent Radiol. 2008; 31:1117–1123.

84. Li, M, Zhou, Y, Huang, Y, et al. A retrospective study of the preoperative treatment of advanced Wilms tumor in children with chemotherapy versus transcatheter arterial chemoembolization alone or combined with short-term chemotherapy. J Vasc Interv Radiol. 2011; 22:279–286.

85. Topar, G, Zelger, B. Assessment of value of the sentinel lymph node biopsy in melanoma in children and adolescents and applicability of subcutaneous infusion anesthesia. J Pediatr Surg. 2007; 42:1716–1720.

86. DeCorti, F, Dall’Igna, P, Bisogno, G, et al. Sentinel node biopsy in pediatric soft tissue sarcomas of extremities. Pediatr Blood Cancer. 2009; 52:51–54.

87. 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. 2003; 112:1257–1265.

88. Morton, DL, Wend, D, Wong, JH, et al. Technical details of intraoperative lymphatic mapping for early stage melanoma. Arch Surg. 1992; 127:392–399.

89. Mariotto, AB, Rowland, JH, Yabroff, KR, et al. Long-term survivors of childhood cancers in the United States. Cancer Epidemiol Biomarkers Prev. 2009; 18:1033–1040.

90. Oberfield, SE, Allen, JC, Pollack, J, et al. A long-term endocrine sequelae after treatment of medulloblastoma: Prospective study of growth and thyroid function. J Pediatr. 1986; 108:219–223.

91. Huma, Z, Boulad, F, Black, P, et al. Growth in children after bone marrow transplantation for acute leukemia. Blood. 1995; 86:819–824.

92. Larson, DL, Kroll, S, Jaffe, N, et al. Long-term effects of radiotherapy in childhood and adolescence. Am J Surg. 1990; 160:348–350.

93. Whitehead, E, Shalet, SM, Morris-Jones, PH, et al. Gonadal function after combination chemotherapy for Hodgkin’s disease in childhood. Arch Dis Child. 1982; 57:287–291.

94. Green, DM, Zevon, MA, Lowrie, G, et al. Congenital anomalies in children of patients who received chemotherapy for cancer in childhood or adolescence. N Engl J Med. 1991; 325:141–146.

95. Legha, SS, Benjamin, RS, Mackay, B, et al. Reduction of doxorubicin cardiotoxicity by prolonged continuous infusion. Ann Intern Med. 1982; 96:133–139.

96. Gurney, J, Kaden-Lottick, N, Packer, R, et al. Endocrine and cardiovascular late effects among adult survivors of childhood brain tumors: Childhood Cancer Survivors Study. Cancer. 2003; 97:663–673.

97. Blatt, J, Copeland, DR, Bleyer, WA. Late effects of childhood cancer and its treatment. In: Pizzo PA, Poplack DG, eds. Principles and Practice of Pediatric Oncology. 3rd ed. Philadelphia: Lippincott-Raven; 1997:1303.

98. Neglia, JP, Friedman, DL, Yasui, Y, et al. Second malignant neoplasms in five-year survivors of childhood cancer; childhood cancer survivor study. J Natl Cancer Inst. 2001; 93:618–629.

99. Bhatia, S, Robison, LL, Oberlin, O, et al. Breast cancer and other second neoplasm after childhood Hodgkin’s disease. N Engl J Med. 1996; 334:745–751.

100. Jenkin, D, Greenberg, M, Fitzgerald, A. Second malignant tumours in childhood Hodgkin’s disease. Med Pediatr Oncol. 1996; 26:373–379.

101. Sankila, R, Garwicz, S, Olsen, JH, et al. Risk of subsequent malignant neoplasms among 1,641 Hodgkin’s disease patients diagnosed in childhood and adolescence: A population-based cohort study in the five Nordic countries. J Clin Oncol. 1996; 14:1442–1446.

102. Smith, MB, Xue, H, Strong, L, et al. Forty year experience with second malignancies after treatment of childhood cancer: Analysis of outcome following the development of the second malignancy. J Pediatr Surg. 1993; 28:1342–1348.