Dramatic improvements in survival have been achieved for children and adolescents with cancer.[1] Between 1975 and 2020, childhood cancer mortality decreased by more than 50%.[1-3] Between 2013 and 2019, the 5-year relative survival rate was 90% for children and adolescents younger than 20 years with NHL.[3] In 2020, there were an estimated 30,500 survivors of childhood and adolescent NHL in the United States.[4]
Childhood and adolescent cancer survivors require close monitoring because cancer therapy side effects may persist or develop months or years after treatment. For information about the incidence, type, and monitoring of late effects in childhood and adolescent cancer survivors, see Late Effects of Treatment for Childhood Cancer.
On the basis of immunophenotype, molecular biology, and clinical response to treatment, most NHL cases occurring in childhood and adolescence fall into three categories:
Other rare types of pediatric NHL include the following:
Lymphoma (Hodgkin lymphoma and NHL) is the third most common childhood malignancy, and NHL accounts for approximately 7% of cancers in children younger than 20 years in the United States.[3]
The following factors affect the incidence of NHL in children and adolescents:[2,3]
In sub-Saharan Africa, the incidence of Epstein-Barr virus (EBV)–induced Burkitt lymphoma is tenfold to twentyfold higher than the incidence in the United States, resulting in a much higher incidence of NHL.[6]
The incidence of NHL is increasing overall because of a slight increase in the incidence for patients aged 15 to 19 years. Conversely, the incidence of NHL in children younger than 15 years has remained constant over the past several decades.[3]
The incidence and age distribution of histological types of NHL according to sex is described in Table 1.
Incidence of NHL per Million Person-Years | ||||||||
---|---|---|---|---|---|---|---|---|
Males | Females | |||||||
ALCL = anaplastic large cell lymphoma; DLBCL = diffuse large B-cell lymphoma; NHL = non-Hodgkin lymphoma. | ||||||||
aAdapted from Percy et al.[9] | ||||||||
bIndolent and aggressive histologies (more commonly seen in adult patients) are mostly found in older adolescents. | ||||||||
Age (y) | <5 | 5–9 | 10–14 | 15–19 | <5 | 5–9 | 10–14 | 15–19 |
Burkitt | 3.2 | 6 | 6.1 | 2.8 | 0.8 | 1.1 | 0.8 | 1.2 |
Lymphoblastic | 1.6 | 2.2 | 2.8 | 2.2 | 0.9 | 1.0 | 0.7 | 0.9 |
DLBCL | 0.5 | 1.2 | 2.5 | 6.1 | 0.6 | 0.7 | 1.4 | 4.9 |
Other (mostly ALCL) | 2.3 | 3.3 | 4.3 | 7.8b | 1.5 | 1.6 | 2.8 | 3.4b |
Relatively little data on the epidemiology of childhood NHL have been published. However, known risk factors include the following:
Unlike adults with NHL who present most often with nodal disease, children typically have extranodal disease involving the mediastinum, abdomen, and/or head and neck, as well as the bone marrow or CNS. In high-income countries, Burkitt lymphoma occurs in the abdomen in approximately 60% of cases, with 15% to 20% of cases arising in the head and neck.[15,16] This high incidence of extranodal disease substantiates the use of the Murphy staging system for pediatric NHL, instead of the Ann Arbor staging system.
The following tests and procedures are used to diagnose childhood NHL:
In high-income countries and with current treatments, more than 80% of children and adolescents with NHL survive at least 5 years, although outcome depends on a number of factors, including clinical stage and histology.[17]
Prognostic factors for childhood NHL include the following:
For more information about the tumor biology and genomic alterations associated with each type of NHL, some of which are being evaluated as potential prognostic biomarkers, see the following sections:
Regardless of histology, pediatric patients with NHL that is refractory to first-line therapy have a very poor prognosis,[18-22] with the exception of patients with anaplastic large cell lymphoma.[18,23] As opposed to other hematologic malignancies, it has been difficult to demonstrate the prognostic value of early response to therapy in pediatric NHL.
International pediatric NHL response criteria have been proposed but require prospective evaluation. The clinical utility of these new criteria are under investigation.[28]
In contrast to the prognostic value of minimal residual disease (MRD) in patients with acute leukemia, the prognostic value of MRD after therapy is initiated remains uncertain and requires further investigation in pediatric patients with NHL.
In general, patients with low-stage disease (i.e., single extra-abdominal/extrathoracic tumor or totally resected intra-abdominal tumor) have an excellent prognosis (5-year survival rate of approximately 90%), regardless of histology.[24,27,33-35] Apart from this finding, the outcome by clinical stage, using appropriate therapy on the basis of risk stratification, does not differ significantly.
A surrogate for tumor burden, specifically elevated levels of LDH, has been shown to be prognostic in many studies.[24,33,36]
Patients with morphologically involved bone marrow with more than 5% lymphoma cells are considered to have stage IV disease. MDD is generally defined as submicroscopic bone marrow involvement that is present at diagnosis. MDD is generally detected by sensitive methods such as flow cytometry or reverse transcription–polymerase chain reaction (RT-PCR).
An Associazione Italiana Ematologia Oncologia Pediatrica (AIEOP) study used an MDD cutoff level of 3% by flow cytometry. The study observed a 5-year EFS rate of 60% for patients with MDD greater than 3% versus 83% for the remaining patients (P = .04).[40]
The largest experience with MDD for T-cell lymphoblastic lymphoma (n = 273) is from the COG AALL0434 (NCT00408005) study, in which MDD had no prognostic impact. Patients with bone marrow MDD levels of less than 1% had an EFS rate of 82.4% compared with 89.5% for those with MDD levels of 1% or higher (P = .3084).[41]
The presence of MDD is significantly associated with uncommon histological subtypes containing small cell and/or lymphohistiocytic components.[42]
In pediatric NHL, some sites of disease appear to have prognostic value, including the following:
NHL in infants is rare (1% in BFM trials from 1986 to 2002).[8] In this retrospective review, the outcome for infants was inferior compared with the outcome for older patients with NHL.[8]
Adolescents have also been reported to have outcomes inferior to those of younger children.[15,17,59] This adverse effect of age appears to be most pronounced for adolescents with diffuse large B-cell lymphoma and, to a lesser degree, T-cell lymphoblastic lymphoma.[17,59] Conversely, for patients with Burkitt lymphoma, adolescent age (≥15 years) was not an independent risk factor for inferior outcome.[26,36] Adolescents with mature B-cell lymphoma who are treated using pediatric protocols have a superior outcome compared with those treated with adult regimens (EFS rates, 88% vs. 66%).[60][Level of evidence C2]
An immune response against the ALK protein (i.e., anti-ALK antibody titer) may correlate with lower clinical stage and predicted relapse risk but not OS.[61] A study by the EICNHL, which combined the level of anti-ALK antibody with MDD, demonstrated that patients with newly diagnosed anaplastic large cell lymphoma could be stratified into three risk groups, with the following PFS rates:[53]
In a cohort of Japanese patients with anaplastic large cell lymphoma who were treated on the ALCL99 (NCT00006455) study, comparable results were obtained for a three-category risk classification algorithm.[44] For a cohort of 180 patients with anaplastic large cell lymphoma who were treated on several European studies, low anti-ALK antibody titer retained prognostic significance in a multivariate analysis, along with MDD, MRD, and uncommon histology (small cell and others).[32][Level of evidence B4]
In children, non-Hodgkin lymphoma (NHL) is distinct from the more common forms of lymphoma observed in adults. While lymphomas in adults are more commonly low or intermediate grade, almost all NHL that occurs in children is high grade.[1,2] The World Health Organization (WHO) classifies NHL according to the following features:[2,3]
On the basis of the WHO classification, most NHL cases in childhood and adolescence fall into the following three categories:
Compared with treatments for adults, aggressive Burkitt regimens in pediatrics have been used to treat patients with both Burkitt lymphoma and large B-cell histologies, resulting in no difference in outcome based on histology.[4-8] The exception is for patients with primary mediastinal B-cell lymphoma, who have had inferior outcomes with these regimens.[4-7,9]
Historically, for patients with pediatric Burkitt lymphoma, secondary cytogenetic abnormalities, other than MYC rearrangement, have been associated with an inferior outcome,[10,11] and cytogenetic abnormalities involving gain of 7q or deletion of 13q may be associated with an inferior outcome on the FAB/LMB-96 chemotherapy protocol.[11,12] For pediatric patients with diffuse large B-cell lymphoma and chromosomal rearrangement at MYC (8q24), outcomes may be worse.[11]
Results from the Inter-B-NHL Ritux 2010 (NCT01516580) phase III trial showed that the addition of rituximab to chemotherapy for patients with aggressive mature B-cell NHL improved event-free survival (EFS) rates, from 82% to 94%. The small number of treatment failures, resulting from a high EFS rate, make it challenging to confirm these previously identified candidate prognostic biomarkers.[13]
Large B-cell lymphoma with IRF4 rearrangement is included in the 5th edition of the WHO Classification of Hematolymphoid Tumors.[3,14] Large B-cell lymphoma with IRF4 cases have a translocation that juxtaposes the IRF4 oncogene next to one of the immunoglobulin loci and has been associated with a favorable prognosis compared with diffuse large B-cell lymphoma cases lacking this finding.[15,16]
For more information about the tumor biology and genomic alterations, see the sections on Tumor biology (Genomics of Burkitt lymphoma), Tumor biology (Genomics of diffuse large B-cell lymphoma), and Tumor biology (Genomics of primary mediastinal B-cell lymphoma).
For more information about the tumor biology and genomic alterations, see the Tumor Biology (Genomics of lymphoblastic lymphoma) section.
In adults, patients with ALK-negative disease have an inferior outcome. However, in children, the difference in outcome between patients with ALK-positive and ALK-negative disease has not been demonstrated.[17-19] In a series of 375 children and adolescents with systemic ALK-positive anaplastic large cell lymphoma enrolled on the ALCL99 (NCT00006455) study, the presence of a small cell or lymphohistiocytic component was observed in 32% of patients. This finding was significantly associated with a high risk of failure in the multivariate analysis, controlling for clinical characteristics.[20] With longer follow-up, presence of the small cell/lymphohistiocytic pattern maintained its prognostic significance on multivariate analysis.[21]
In the COG-ANHL0131 (NCT00059839) study, despite a different chemotherapy backbone, patients with the small cell variant of anaplastic large cell lymphoma, as well as other histological variants, had a significantly increased risk of failure.[19]
For more information about the tumor biology and genomic alterations, see the Tumor Biology (Genomics of anaplastic large cell lymphoma) section.
The WHO classification is the most widely used NHL classification and is shown in Table 2, with immunophenotype and common clinical and molecular findings in pediatric NHL.[1-3]
WHO Classification | Immunophenotype | Clinical Presentation | Chromosome Abnormalities | Genes Affected | |
---|---|---|---|---|---|
CNS = central nervous system; TdT = terminal deoxynucleotidyl transferase; WHO = World Health Organization; + = positive. | |||||
aAdapted from Percy et al.[1] | |||||
Burkitt lymphoma | Mature B cell | Intra-abdominal (sporadic), head and neck (non-jaw, sporadic), jaw (endemic), bone marrow, CNS | t(8;14)(q24;q32), t(2;8)(p11;q24), t(8;22)(q24;q11) | MYC, TCF3, ID3, CCND3, TP53 | |
High-grade B-cell lymphoma with 11q aberrations | Mature B cell | Nodal | 11q alteration, no MYC rearrangement | ||
Large B-cell lymphoma with IRF4 rearrangement | Mature B cell | Nodal (typically head and neck) | Cryptic IRF1 rearrangement with IGH locus | IRF4 | |
Diffuse large B-cell lymphoma | Mature B cell | Nodal, abdominal, bone, primary CNS (when associated with immunodeficiency), mediastinal | No consistent cytogenetic abnormality identified | ||
Primary mediastinal (thymic) large B-cell lymphoma | Mature B cell, often CD30+ | Mediastinal, but may also have other nodal or extranodal disease (i.e., abdominal, often kidney) | 9p and 2p gains | CIITA, TNFAIP3, SOCS1, PTPN11, STAT6 | |
ALK-positive large B-cell lymphoma | Generalized lymphadenopathy, bone marrow in 25% | t(2;5)(p23;q35); less common variant translocations involving ALK | ALK, NPM | ||
T-cell lymphoblastic leukemia/lymphoma | T lymphoblasts (TdT, CD2, CD3, CD7, CD4, CD8) | Mediastinal mass, bone marrow | |||
B-cell lymphoblastic leukemia/lymphoma | B lymphoblasts (CD19, CD79a, CD22, CD10, TdT) | Skin, soft tissue, bone, lymph nodes, bone marrow | |||
Pediatric-type follicular lymphoma | Mature B cell | Nodal (typically head and neck) | TNFRSF14, MAP2K1 | ||
Pediatric nodal marginal zone lymphoma | Mature B cell | Nodal (typically head and neck) |
Other types of lymphoma, such as the nonanaplastic large cell peripheral T-cell lymphomas (including T/NK lymphomas), cutaneous lymphomas, and indolent B-cell lymphomas (e.g., follicular lymphoma and marginal zone lymphoma), are more commonly seen in adults and rarely occur in children. The WHO classification has designated pediatric-type follicular lymphoma and pediatric nodal marginal zone lymphoma as distinct entities from the counterparts observed in adults.[3]
For more information about the treatment of NHL in adult patients, see the following summaries:
The Ann Arbor staging system is used for all lymphomas in adults and for Hodgkin lymphoma in pediatrics. However, the Ann Arbor staging system has less prognostic value in pediatric non-Hodgkin lymphoma (NHL), primarily because of the high incidence of extranodal disease. Therefore, the most widely used staging schema for childhood NHL is that of the St. Jude Children’s Research Hospital (Murphy Staging).[1] A new staging system defines bone marrow and central nervous system (CNS) involvement using modern techniques to document the presence of malignant cells. However, the basic definitions of bone marrow and CNS disease are essentially the same. The clinical utility of this staging system is under investigation.[2]
Radiographic imaging is essential in the staging of patients with NHL. Ultrasonography may be the preferred method for assessment of an abdominal mass, but computed tomography (CT) scan and magnetic resonance imaging (MRI) have been used for staging.
The role of functional imaging in pediatric NHL is evolving and still being refined. Gallium scans have been replaced by fluorine F 18-fludeoxyglucose positron emission tomography (PET) scanning, which is now routinely performed at many centers.[3] A review of the revised International Workshop Criteria comparing CT imaging alone or CT together with PET imaging demonstrated that the combination of CT and PET imaging was more accurate than CT imaging alone.[4,5]
While the International Working Group (formerly called the International Harmonization Project for PET) response criteria have been attempted in adults, the prognostic value of PET scanning for staging pediatric NHL remains under investigation.[3,6,7] Data support that PET identifies more abnormalities than does CT scanning,[8] but it is unclear whether this should be used to upstage pediatric patients and change therapy. The International Working Group has updated their response criteria for malignant lymphoma to include PET, immunohistochemistry, and flow cytometry data.[5,9]
In stage I childhood NHL, a single tumor or nodal area is involved, excluding the abdomen and mediastinum.
In stage II childhood NHL, disease extent is limited to a single tumor with regional node involvement, two or more tumors or nodal areas involved on one side of the diaphragm, or a primary gastrointestinal tract tumor (completely resected) with or without regional node involvement.
In stage III childhood NHL, tumors or involved lymph node areas occur on both sides of the diaphragm. Stage III NHL also includes any primary intrathoracic (mediastinal, pleural, or thymic) disease, extensive primary intra-abdominal disease, or any paraspinal or epidural tumors.
In stage IV childhood NHL, tumors involve the bone marrow and/or CNS, regardless of other sites of involvement.
Bone marrow involvement has been defined as 5% or more malignant cells in an otherwise normal bone marrow, with normal peripheral blood counts and smears. Patients with lymphoblastic lymphoma who have more than 25% malignant cells in the bone marrow are usually considered to have leukemia and may be appropriately treated on leukemia clinical trials.
CNS disease in lymphoblastic lymphoma is defined by criteria similar to that used for acute lymphocytic leukemia (i.e., white blood cell count of at least 5/μL and malignant cells in the cerebrospinal fluid [CSF]). For other types of NHL, the definition of CNS disease is any malignant cell present in the CSF regardless of cell count. The Berlin-Frankfurt-Münster group analyzed the prevalence of CNS involvement in more than 2,300 pediatric patients with NHL. Overall, CNS involvement was diagnosed in 6% of patients. CNS involvement (percentage of patients) according to NHL subtype was as follows:[10]
Many of the advancements in childhood cancer survival have been made by using combinations of known and/or new agents to improve the best available, accepted therapy. Clinical trials in pediatrics are designed to compare potentially better therapy with currently accepted standard therapy. This comparison may be done in a randomized study of two treatment arms or by evaluating a single new treatment and comparing the results with those previously obtained with standard therapy.
All children with non-Hodgkin lymphoma (NHL) should consider enrolling in a clinical trial. Treatment planning by a multidisciplinary team of cancer specialists with experience treating tumors of childhood is strongly recommended to determine, coordinate, and implement treatment to achieve optimal survival. Children with NHL should be referred for treatment by a multidisciplinary team of pediatric oncologists at an institution with experience in treating pediatric cancers. Information about ongoing National Cancer Institute (NCI)–supported clinical trials is available from the NCI website.
NHL in children is generally considered to be widely disseminated at diagnosis, even when the tumor is apparently localized. As a result, combination chemotherapy is recommended for most patients.[1] Exceptions to this treatment strategy include the following:
In contrast to the treatment of adults with NHL, the use of radiation therapy is limited in children with NHL. Study results include the following:
Radiation therapy may have a role in treating patients who have not had a complete response to chemotherapy. Data to support limiting the use of radiation therapy in the treatment of pediatric NHL come from the Childhood Cancer Survivor Study.[7] This analysis demonstrated that radiation exposure was a significant risk factor for subsequent neoplasms and death in long-term survivors.
The treatment of NHL in childhood and adolescence has historically been based on the histological subtype of the disease. A study by the Children’s Cancer Group demonstrated that the outcomes for patients with lymphoblastic lymphoma were superior with longer acute lymphoblastic leukemia–like therapy, while patients with nonlymphoblastic NHL (Burkitt lymphoma) had superior outcomes with short, intensive, pulsed therapy. The outcomes for patients with large cell lymphoma were similar with either approach.[8]
Outcomes for children and adolescents with recurrent NHL remain very poor, with the exception of patients with anaplastic large cell lymphoma.[9-14] Patients or families who desire additional disease-directed therapy should consider entering trials of novel therapeutic approaches. Regardless of whether a decision is made to pursue disease-directed therapy at the time of progression, palliative care remains a central focus of management. This ensures that quality of life is maximized while attempting to reduce symptoms and stress related to terminal illness.
Table 3 describes the treatment options for newly diagnosed and recurrent childhood NHL.
The most common potentially life-threatening clinical situations, seen in patients with lymphoblastic lymphoma and Burkitt lymphoma, are the following:
Patients with large mediastinal masses are at risk of tracheal compression, superior vena caval compression, large pleural and pericardial effusions, and right and left ventricular outflow compression. Thus, cardiac or respiratory arrest is a significant risk, particularly if the patient is placed in a supine position for procedures such as computed tomography (CT) or echocardiography scans.[15] Most of these procedures can be performed with patients on their side or prone.
Because of the risk of complications from general anesthesia or heavy sedation, a careful physiological and radiographic evaluation of the patient should be completed, and the least invasive procedure should be used to establish the diagnosis of lymphoma.[16,17] The following procedures may be used:
In situations when the above procedures do not yield a diagnosis, the use of a CT-guided core-needle biopsy should be considered. This procedure can frequently be performed using light sedation and local anesthesia before more invasive procedures are undertaken. Care should be taken to keep patients out of a supine position. Mediastinoscopy, anterior mediastinotomy, or thoracoscopy are the procedures of choice when other diagnostic modalities fail to establish the diagnosis. A formal thoracotomy is rarely, if ever, indicated for the diagnosis or treatment of childhood lymphoma.
Occasionally, it will not be possible to perform a diagnostic operative procedure because of the risk of complications from general anesthesia or heavy sedation. In these situations, preoperative treatment with steroids or, less commonly, localized radiation therapy should be considered. Because preoperative treatment may affect the ability to obtain an accurate tissue diagnosis, a diagnostic biopsy should be done as soon as the risk of complications from general anesthesia or heavy sedation is reduced.
Tumor lysis syndrome results from rapid breakdown of malignant cells, causing several metabolic abnormalities, most notably hyperuricemia, hyperkalemia, and hyperphosphatemia. Patients may present with tumor lysis syndrome before the start of therapy.
Hyperhydration and allopurinol or rasburicase (urate oxidase) are essential components of therapy in all patients, except those with the most limited disease.[19-24] In patients with G6PD deficiency, rasburicase may cause hemolysis or methemoglobinuria and should be avoided. An initial prephase consisting of low-dose cyclophosphamide and vincristine does not obviate the need for allopurinol or rasburicase and hydration.
Hyperuricemia and tumor lysis syndrome, particularly when associated with ureteral obstruction, frequently result in life-threatening complications.
Although the use of positron emission tomography (PET) to assess rapidity of response to therapy appears to have prognostic value in Hodgkin lymphoma and some types of NHL observed in adult patients, it remains under investigation in pediatric NHL. To date, there are insufficient data for pediatric NHL to support a finding that early response to therapy assessed by PET has prognostic value.
Diagnosing relapsed disease solely based on imaging requires caution because false-positive results are common.[25-28] Data also demonstrate that PET scanning can produce false-negative results.[29] A study of young adults with primary mediastinal B-cell lymphoma demonstrated that 9 of 12 patients who had residual mediastinal masses at the end of therapy had positive PET scans. Seven of these nine patients had the masses resected, but no viable tumor was found.[30] Before changes in therapy are undertaken based on residual masses noted by imaging, even if the PET scan is positive, a biopsy to prove residual disease is warranted.[28]
Cancer in children and adolescents is rare, although the overall incidence has slowly increased since 1975.[1] 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 following pediatric specialists and others to ensure that children receive treatment, supportive care, and rehabilitation to achieve optimal survival and quality of life:
For specific information about supportive care for children and adolescents with cancer, see the summaries on Supportive and Palliative Care.
The American Academy of Pediatrics has outlined guidelines for pediatric cancer centers and their role in the treatment of children and adolescents with cancer.[2] At these centers, clinical trials are available for most types of cancer that occur in children and adolescents, and the opportunity to participate is offered to most patients and their families. Clinical trials for children and adolescents diagnosed with cancer are generally designed to compare potentially better therapy with current standard therapy. Other types of clinical trials test novel therapies when there is no standard therapy for a cancer diagnosis. Most of the progress in identifying curative therapies for childhood cancers has been achieved through clinical trials. Information about ongoing clinical trials is available from the NCI website.
In the United States, Burkitt lymphoma accounts for about 40% of childhood non-Hodgkin lymphoma (NHL) cases and exhibits a consistent, aggressive clinical behavior.[1] The overall incidence of Burkitt lymphoma in the United States is 2.4 cases per 1 million person-years and is higher among boys than girls (3.8 vs. 0.9).[2,3] For more information about the incidence of Burkitt lymphoma by age and sex distribution, see Table 1.
The most common primary sites of disease are the abdomen and the lymphatic tissue of Waldeyer ring.[4] Other sites of involvement include testes, bone, skin, bone marrow, and central nervous system (CNS). While lung involvement does not tend to occur, pleural and peritoneal spread are seen.[4]
The malignant cells of Burkitt lymphoma show a mature B-cell phenotype and are negative for the enzyme terminal deoxynucleotidyl transferase. These malignant cells usually express surface immunoglobulin (Ig), most bearing a clonal surface IgM with either kappa or lambda light chains. A variety of additional B-cell markers (e.g., CD19, CD20, CD22) are usually present, and most childhood Burkitt lymphomas express CD10.[1]
Burkitt lymphoma expresses a characteristic chromosomal translocation, usually t(8;14) and more rarely t(8;22) or t(2;8). Each of these translocations juxtaposes the MYC oncogene and the immunoglobulin locus (IG, mostly the IGH locus) regulatory elements, resulting in the inappropriate expression of MYC, a gene involved in cellular proliferation.[5,6] The presence of one of the variant translocations t(2;8) or t(8;22) does not appear to affect response or outcome.[7,8]
Mapping of IGH-translocation breakpoints demonstrated that IG::MYC translocations in sporadic Burkitt lymphoma most commonly occur through aberrant class-switch recombination and less commonly through somatic hypervariant. Translocations resulting from aberrant variable, diversity, and joining (VDJ) gene segment recombinations are rare.[9] These findings are consistent with a germinal center derivation of Burkitt lymphoma.
While MYC translocations are present in all Burkitt lymphoma, cooperating genomic alterations appear to be required for lymphoma development. Some of the more commonly observed recurring variants that have been identified in Burkitt lymphoma in pediatric and adult cases are listed below. The clinical significance of these variants for pediatric Burkitt lymphoma remains to be elucidated.
A study that compared the genomic landscape of endemic Burkitt lymphoma with the genomics of sporadic Burkitt lymphoma found the expected high rate of Epstein-Barr virus (EBV) positivity in endemic cases, with much lower rates in sporadic cases. There was general similarity between the patterns of variants for endemic and sporadic cases and for EBV-positive and EBV-negative cases. However, EBV-positive cases showed significantly lower variant rates for selected genes/pathways, including SMARCA4, CCND3, TP53, and apoptosis.[8]
Cytogenetic evidence of MYC rearrangement is the gold standard for diagnosis of Burkitt lymphoma. For cases in which cytogenetic analysis is not available, the World Health Organization (WHO) has recommended that the Burkitt-like diagnosis be reserved for lymphoma resembling Burkitt lymphoma or with more pleomorphism, large cells, and a proliferation fraction (i.e., MIB-1 or Ki-67 immunostaining) of 99% or greater.[1] BCL2 staining by immunohistochemistry is variable. The absence of a translocation involving the BCL2 gene does not preclude the diagnosis of Burkitt lymphoma and has no clinical implications.[15]
Burkitt-like lymphoma with 11q aberration was added as a provisional entity in the 2017 revised WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues.[16] In the 5th edition of the WHO classification, this entity was renamed high-grade B-cell lymphoma with 11q aberrations.[17] In this entity, MYC rearrangement is absent, and the characteristic chromosome 11q finding (detected cytogenetically and/or with copy-number DNA arrays) is 11q23.2-q23.3 gain/amplification and 11q24.1-qter loss.[18,19]
For information about prognostic factors for Burkitt lymphoma, see the Prognosis and Prognostic Factors for Childhood NHL section.
The treatment of Burkitt lymphoma is the same as treatment for diffuse large B-cell lymphoma, and the following discussion is pertinent to both types of childhood NHL.
Unlike mature B-lineage NHL seen in adult patients, there is no difference in outcome based on histology in pediatric patients (Burkitt lymphoma or diffuse large B-cell lymphoma). Pediatric Burkitt lymphoma and diffuse large B-cell lymphoma are clinically very aggressive, and patients are treated with very intensive regimens.[21-26]
Tumor lysis syndrome is often present at diagnosis or after initiation of treatment. This emergent clinical situation should be anticipated and addressed before treatment is started. For more information, see the Tumor lysis syndrome section.
Current treatment strategies are based on risk stratification, as described in Table 4. Involvement of the bone marrow may lead to confusion about whether the patient has lymphoma or leukemia. Traditionally, patients with more than 25% marrow blasts are classified as having mature B-cell leukemia, and those with fewer than 25% marrow blasts are classified as having lymphoma. It is not clear whether these arbitrary definitions are biologically distinct, but there is no question that patients with leukemic involvement should be treated with protocols designed for Burkitt lymphoma.[21,23,26]
Stratum | Disease Manifestation | |
---|---|---|
ALL = acute lymphoblastic leukemia; BFM = Berlin-Frankfurt-Münster; CNS= central nervous system; FAB = French-American-British; LDH = lactate dehydrogenase; LMB = Lymphome Malin de Burkitt; NHL = non-Hodgkin lymphoma. | ||
aBased on results of the FAB/LMB-96 study, a serum LDH level more than twice the upper limit of normal has been used to define a group B high-risk group in the international B-cell NHL study ANHL1131 (NCT01516567).[22] | ||
COG-C5961 (FAB/LMB-96);[22,23,27] COG-ANHL1131 (Inter-B-NHL Ritux 2010) [26] | A | Completely resected stage I and abdominal stage II |
Ba | Multiple extra-abdominal sites | |
Nonresected stage I and II, III, IV (marrow <25% blasts, no CNS disease); epidural masses (stage III Murphy staging) are treated as group B unless there is evidence of dural invasion | ||
C | Mature B-cell ALL (>25% blasts in marrow) and/or CNS disease | |
BFM Group [28] | R1 | Completely resected stage I and abdominal stage II |
R2 | Nonresected stage I or II and stage III with LDH <500 IU/L | |
R3 | Stage III with LDH 500–999 IU/L | |
Stage IV, B-ALL (>25% blasts), no CNS disease, and LDH <1,000 IU/L | ||
R4 | Stage III, IV, B-cell ALL with LDH >1,000 IU/L | |
Any CNS disease |
The following studies have contributed to the development of current treatment regimens for pediatric patients with Burkitt lymphoma and diffuse large B-cell lymphoma.
Evidence (chemotherapy):
Both the BFM and FAB/LMB studies demonstrated that omission of craniospinal irradiation, even in patients presenting with CNS disease, does not affect outcome (COG-C5961 [FAB/LMB-96] and NHL-BFM-90 [GER-GPOH-NHL-BFM-90]).[21-23,28]
Evidence (rituximab):
Standard treatment options for Burkitt lymphoma and diffuse large B-cell lymphoma are described in Table 5.
Trial | Stratum | Disease Manifestations | Treatment |
---|---|---|---|
ALL = acute lymphoblastic leukemia; BFM = Berlin-Frankfurt-Münster; CNS = central nervous system; COG = Children's Oncology Group; FAB = French-American-British; LDH = lactate dehydrogenase; LMB = Lymphome Malin de Burkitt; NHL = non-Hodgkin lymphoma; POG = Pediatric Oncology Group. | |||
COG-C5961 (FAB/LMB-96) [22,27] COG-ANHL01 [31,32]; [24][Level of evidence C1] COG-ANHL1131 (Inter-B-NHL Ritux 2010) [26] | A | Completely resected stage I and abdominal stage II | Two cycles of chemotherapy [27] |
B | Multiple extra-abdominal sites | Prephase + four cycles of chemotherapy (reduced-intensity arm) [22,34] | |
Nonresected stage I and II, III (normal LDH) | |||
Stage III (elevated LDH), marrow <25% blasts, no stage IV CNS disease | Prephase + four cycles of chemotherapy (reduced-intensity arm) + six doses of rituximab [26] | ||
C | Mature B-cell ALL (>25% blasts in marrow) and/or stage IV CNS disease | Prephase + six cycles of chemotherapy (full-intensity arm) and only two maintenance cycles + six doses of rituximab [26] | |
GER-GPOH-NHL-BFM-95 [21] | R1 | Completely resected stage I and abdominal stage II | Two cycles of chemotherapy |
R2 | Nonresected stage I/II and stage III with LDH <500 IU/L | Prephase + four cycles of chemotherapy (4-hour methotrexate infusion) |
There is no standard treatment option for patients with recurrent or progressive disease. For patients with recurrent or refractory aggressive mature B-cell NHL, survival rates range between 10% and 50%. In the largest series, the survival rate was about 20%.[23,35,36]; [37][Level of evidence C1] Three large retrospective multivariable analyses identified the following prognostic factors:
Treatment options for recurrent or refractory Burkitt lymphoma and diffuse large B-cell lymphoma include the following:
Chemoresistance makes remission difficult to achieve.
Evidence (treatment of recurrent or refractory Burkitt lymphoma):
If remission can be achieved, high-dose therapy plus HSCT remains the best option for survival. Patients not in remission at the time of transplant fare significantly worse.[38,41,46,48-50] The very poor outcome of patients whose disease is refractory to salvage chemotherapy suggests that a nonexperimental transplant option should not be pursued in these patients.[41,49,50] If a complete remission was reported, survival ranges between 30% to 75%, albeit all series have a small number of patients (i.e., fewer than 40).[41,46,47,49,51] The benefit of autologous versus allogeneic HSCT remains unclear.[36,41,51,52]; [48][Level of evidence B4]; [53][Level of evidence C2]
For more information about transplant, see Pediatric Autologous Hematopoietic Stem Cell Transplant, Pediatric Allogeneic Hematopoietic Stem Cell Transplant, and Pediatric Hematopoietic Stem Cell Transplant and Cellular Therapy for Cancer.
Evidence (HSCT therapy):
Information about National Cancer Institute (NCI)–supported clinical trials can be found on the NCI website. For information about clinical trials sponsored by other organizations, see the ClinicalTrials.gov website.
Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.
Primary mediastinal B-cell lymphoma, previously considered a subtype of diffuse large B-cell lymphoma, is now a separate entity in the WHO classification. For more information, see the Primary Mediastinal B-cell Lymphoma section.
Diffuse large B-cell lymphoma is an aggressive mature B-cell neoplasm that represents 10% to 20% of pediatric NHL cases.[54,55] Diffuse large B-cell lymphoma occurs more frequently during the second decade of life than during the first decade.[55,56] For more information on the incidence of diffuse large B-cell lymphoma by age and sex distribution, see Table 1.
Pediatric diffuse large B-cell lymphoma may present in a manner clinically similar to that of Burkitt lymphoma, although more often it is localized, and less often it involves the bone marrow or CNS.[54,56] For more information, see the Clinical presentation section in the Burkitt lymphoma section.
Gene expression profiling of diffuse large B-cell lymphoma in adults has defined molecular subtypes. These subtypes are based on the suspected cell of origin, including germinal center B cell (GCB), activated B cell (ABC), and 10% to 15% of cases that remain unclassifiable. Current comprehensive molecular profiling of diffuse large B-cell lymphoma in adults has led to the proposal of additional subclassification beyond the cell of origin. This additional subclassification is based on genetic variants and copy number variations.[57,58] Diffuse large B-cell lymphoma in children and adolescents differs biologically from diffuse large B-cell lymphoma in adults in the following ways:
Large B-cell lymphoma with IRF4 rearrangement (LBCL-IRF4) is a distinct entity in the 5th edition of the WHO classification of lymphoid neoplasms.[67]
High-grade B-cell lymphoma, NOS, is defined as a clinically aggressive B-cell lymphoma that lacks MYC plus BCL2 and/or BCL6 rearrangements. In addition, this entity does not meet criteria for diffuse large B-cell lymphoma, NOS, or Burkitt lymphoma.[72]
For information about prognostic factors for diffuse large B-cell lymphoma, see the Prognosis and Prognostic Factors for Childhood NHL section.
As with Burkitt lymphoma, current treatment strategies are based on risk stratification, as described in Table 5. The treatment of diffuse large B-cell lymphoma is the same as the treatment of Burkitt lymphoma. For information about the treatment of diffuse large B-cell lymphoma, see the Standard treatment options for Burkitt lymphoma section.
Radiation therapy can be considered for patients who are unresponsive to salvage therapies or as a consolidation therapy for select patients.[73]
The treatment of recurrent diffuse large B-cell lymphoma is the same as the treatment of recurrent Burkitt lymphoma. For more information, see the Treatment options for recurrent or refractory Burkitt lymphoma section.
Information about NCI-supported clinical trials can be found on the NCI website. For information about clinical trials sponsored by other organizations, see the ClinicalTrials.gov website.
Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.
In the pediatric population, primary mediastinal B-cell lymphoma is predominantly seen in older adolescents, accounting for 1% to 2% of all pediatric NHL cases.[56,74-76]
As the name suggests, primary mediastinal B-cell lymphoma occurs in the mediastinum. The tumor can be locally invasive (e.g., pericardial and lung extension), and it can be associated with superior vena cava syndrome. The tumor can disseminate outside the thoracic cavity with nodal and extranodal involvement, with predilection to the kidneys. However, CNS and marrow involvement are exceedingly rare.[77]
Primary mediastinal B-cell lymphoma was previously considered a subtype of diffuse large B-cell lymphoma, but is now a separate entity in the World Health Organization (WHO) classification.[17] These tumors arise in the mediastinum from thymic B cells and show a diffuse large cell proliferation with sclerosis that compartmentalizes neoplastic cells.
Primary mediastinal B-cell lymphoma can be very difficult to distinguish morphologically from the following types of lymphoma:
Primary mediastinal B-cell lymphoma has distinctive gene expression and variant profiles compared with diffuse large B-cell lymphoma. However, its gene expression and variant profiles have features similar to those seen in Hodgkin lymphoma.[79-81] Primary mediastinal B-cell lymphoma is also associated with a distinctive constellation of chromosomal aberrations compared with other NHL subtypes. Because primary mediastinal B-cell lymphoma is primarily a cancer of adolescents and young adults, the genomic findings are presented without regard to age.
For information on prognostic factors for primary mediastinal B-cell lymphoma, see the Prognosis and Prognostic Factors for Childhood NHL section.
There are limited studies to evaluate prognostic factors in children with primary mediastinal B-cell lymphoma.
Treatment options for primary mediastinal B-cell lymphoma include the following:
Pediatric and adolescent patients with stage III primary mediastinal large B-cell lymphoma fared significantly worse on the FAB/LMB-96 (NCT00002757) study, with a 5-year EFS rate of 66%, compared with 85% for adolescents with nonmediastinal diffuse large B-cell lymphoma.[92][Level of evidence B4] Similarly, in the NHL-BFM-95 trial, patients with primary mediastinal B-cell lymphoma had an EFS rate of 50% at 3 years.[21] However, a study of young adults treated with DA-EPOCH-R showed excellent disease-free survival rates.[93]
Evidence (DA-EPOCH-R):
Evidence (LMB-based chemotherapy plus rituximab):
Primary mediastinal B-cell lymphoma in adults is currently and primarily treated with a combination of chemotherapy and the monoclonal antibody rituximab (chemoimmunotherapy). Following chemoimmunotherapy, adult patients receive radiation therapy if they have a residual abnormality that is concerning for active tumor. Although most patients with primary mediastinal B-cell lymphoma demonstrate residual tissue abnormalities at the end of chemoimmunotherapy, this does not definitively indicate active tumor. Positron emission tomography–computed tomography (PET-CT) scans are useful to differentiate active tumor from fibrotic tissue in patients treated for mediastinal lymphoma.
Although lymphoma is responsive to radiation therapy, the role of radiation therapy has not been clearly determined in the up-front setting of primary mediastinal B-cell lymphoma. The results from a prospective randomized trial in adults with primary mediastinal B-cell lymphoma who were treated with R-CHOP with or without radiation therapy demonstrated that patients assigned to radiation therapy had a superior EFS, with no differences in PFS and OS.[97] The role of radiation therapy is less clear in the setting of more dose-intensive regimens that contain rituximab, such as DA-EPOCH-R.
Pediatric data are limited on the use of radiation therapy in the initial management of primary mediastinal B-cell lymphoma. Prospective pediatric studies that did not include radiation therapy have been conducted. In a retrospective analysis on the use of DA-EPOCH-R, radiation therapy was only administered in a small subset of pediatric patients (4 of 36 patients), highlighting the limited use of radiation therapy among pediatric patients treated outside of clinical trials.[94] The management of primary mediastinal B-cell lymphoma in pediatric patients, as with other childhood cancers, requires considering the efficacy and the long-term toxicity of the treatment. In particular, the potential for cardiac and pulmonary toxicities and secondary malignancies must be considered. More intensive chemotherapy regimens may allow for omitting radiation therapy but may also increase cardiac toxicity.
The U.S. Food and Drug Administration granted accelerated approval of pembrolizumab for the treatment of adult and pediatric patients with refractory primary mediastinal large B-cell lymphoma or who have relapsed after two or more previous lines of therapy. The approval was based on data from 53 patients (median age, 33 years; range, 20–61 years). The overall response rate was 41%, which included 12% complete responses and 29% partial responses.[98]
Information about NCI-supported clinical trials can be found on the NCI website. For information about clinical trials sponsored by other organizations, see the ClinicalTrials.gov website.
The following is an example of a national and/or institutional clinical trial that is currently being conducted:
Lymphoblastic lymphoma comprises approximately 20% of childhood non-Hodgkin lymphoma (NHL) cases.[1,2] For more information about the incidence of lymphoblastic lymphoma by age and sex distribution, see Table 1.
As many as 75% of patients with T-cell lymphoblastic lymphoma will present with an anterior mediastinal mass, which may manifest as dyspnea, wheezing, stridor, dysphagia, or swelling of the head and neck.
Pleural and/or pericardial effusions may be present. Involvement of lymph nodes, usually above the diaphragm, may be a prominent feature. There may also be involvement of bone, skin, bone marrow, central nervous system (CNS), abdominal organs (but rarely bowel), and, occasionally, other sites such as lymphoid tissue of Waldeyer ring, testes, or subcutaneous tissue. Abdominal involvement is less common in T-cell lymphoblastic lymphoma than in Burkitt lymphoma.
Involvement of the bone marrow may lead to confusion about whether the patient has lymphoma with bone marrow involvement or leukemia with extramedullary disease. Traditionally, patients with more than 25% marrow blasts are considered to have T-cell acute lymphoblastic leukemia (T-ALL), and those with fewer than 25% marrow blasts are considered to have stage IV T-cell lymphoblastic lymphoma. The World Health Organization (WHO) classifies lymphoblastic lymphoma as the same disease as ALL.[3] The debate centers on whether they truly represent the same disease.[4] It is not yet clear whether these arbitrary definitions are biologically distinct or relevant for treatment design.
B-cell lymphoblastic lymphoma is more often localized nodal disease, but it can present with extranodal disease (e.g., isolated testicular or cutaneous disease).[5,6]
Lymphoblastic lymphomas are usually positive for terminal deoxynucleotidyl transferase. More than 75% of cases have a T-cell immunophenotype and the remaining cases have a precursor B-cell phenotype.[5]
As opposed to pediatric T-cell acute lymphoblastic leukemia (T-ALL), the molecular biology and chromosomal abnormalities of pediatric lymphoblastic lymphoma are not as well characterized. Many genomic alterations that occur in T-ALL also occur in T-cell lymphoblastic lymphoma. Examples include the following:
For the genomic alterations described above, NOTCH1 and FBXW7 variants may confer a more favorable prognosis for patients with T-cell lymphoblastic lymphoma. In contrast, loss of heterozygosity at chromosome 6q, PTEN variants, and KMT2D variants may be associated with an inferior prognosis.[8-12] For example, one study noted that the presence of a KMT2D and/or PTEN variant was associated with a high risk of relapse in patients with wild-type NOTCH1 or FBXW7, but these variants were not associated with an increased risk of relapse in patients with variants in NOTCH1 or FBXW7.[11] Studies with larger numbers of patients are needed to better define the critical genomic determinants of outcome for patients with T-cell lymphoblastic lymphoma.
There have been few studies of the genomic characteristics of B-cell lymphoblastic lymphoma. One report described copy number alterations for pediatric B-cell lymphoblastic lymphoma cases. The study noted that some gene deletions that are common in B-ALL (e.g., CDKN2A, IKZF1, and PAX5) appeared to occur with appreciable frequency in B-cell lymphoblastic lymphoma.[4]
The morphology and immunophenotype of B-cell lymphoblastic lymphoma are known to overlap with those of B-ALL, but few studies have examined the genomic landscape of B-cell lymphoblastic lymphoma, partially due to the lack of sufficient material for genomic analysis.[4] One study has better evaluated the genomic alterations associated with pediatric B-cell lymphoblastic lymphoma.[13] The study analyzed 97 cases of B-cell lymphoblastic lymphoma using a combination of targeted DNA, whole-exome, and RNA sequencing. Overall, the results showed remarkable similarities in the variant and transcriptional landscape between B-cell lymphoblastic lymphoma and B-ALL.
For information about prognostic factors for lymphoblastic lymphoma, see the Prognosis and Prognostic Factors for Childhood NHL section.
Low-stage (stage I or stage II) lymphoblastic lymphoma is primarily a B-cell disease. Treatment with short, pulsed chemotherapy (i.e., doxorubicin, cyclophosphamide, vincristine, and prednisone [CHOP]), followed by 6 months of maintenance therapy, produces a disease-free survival (DFS) rate of about 60% and an overall survival (OS) rate exceeding 90%.[14,15] However, the use of an ALL treatment approach, consisting of induction, consolidation, and maintenance therapy for a total of 24 months, has produced DFS rates higher than 90% in children with low-stage lymphoblastic lymphoma.[6,16,17]
Patients with high-stage (stage III or stage IV) lymphoblastic lymphoma, most often T-cell disease, have DFS rates higher than 80%.[16-18] Mediastinal involvement is common, but radiation therapy is not necessary for patients with mediastinal masses, except in the emergency treatment of symptomatic superior vena cava obstruction or airway obstruction. In these cases, either corticosteroid therapy or low-dose radiation therapy is usually given. For more information, see the Mediastinal masses section.
The following studies have contributed to the development of current treatment regimens for pediatric patients with lymphoblastic lymphoma.
The Pediatric Oncology Group conducted a trial to test the effectiveness of high-dose methotrexate in the treatment of patients with T-ALL and T-cell lymphoblastic lymphoma. In the lymphoma patients (n = 66), high-dose methotrexate did not demonstrate a benefit, with a 5-year event-free survival (EFS) rate of 88%.[19][Level of evidence A1] Of note, all of these patients received prophylactic cranial radiation therapy, even though other studies have shown that it is not required for patients with T-cell lymphoblastic lymphoma.[17,18] In this study, the benefit of adding the cardioprotectant dexrazoxane was tested in a randomized fashion. The addition of dexrazoxane did not affect patient outcomes, and it provided cardioprotective benefits, as demonstrated by echocardiographic and laboratory assessments.[20][Level of evidence B4]
In the NHL-BFM-90 study, the 5-year DFS rate was 90%, and there was no difference in outcome between patients with stage III and stage IV disease.[16] Patients with precursor B-cell lymphoblastic lymphoma appeared to have similar results using the same therapy.[2] All patients received prophylactic cranial radiation therapy. In the NHL-BFM-95 study, the amount of daunorubicin and asparaginase in induction was reduced and patients did not receive prophylactic cranial radiation therapy.[17] The DFS rate in this study was similar to the rate in the NHL-BFM-90 study. However, the EFS rate was lower, at 82%, because of a higher incidence of subsequent neoplasms.[17] A single-center study reported that patients treated for lymphoblastic lymphoma had a higher incidence of subsequent neoplasms than did patients treated for other pediatric NHL.[21] However, studies from the Children's Oncology Group (COG) and the Childhood Cancer Survivor Study Group did not support this finding.[18,22-24]
Evidence for chemotherapy (low-stage treatment regimens for lymphoblastic lymphoma):
Evidence for chemotherapy (high-stage treatment regimens for lymphoblastic lymphoma):
Equivalent outcomes were observed for patients treated on arms A1, B1, A2, and B2. The 5-year EFS rates were 81%, 80%, 84%, and 80%, respectively. The OS rates were 84%, 88%, 85%, and 85%, respectively. Patients with CNS disease at diagnosis had a 5-year EFS rate of 63% and an OS rate of 81%.
For patients with recurrent or refractory lymphoblastic lymphoma, survival rates range from 10% to 40%.[22,26]; [27][Level of evidence B4]; [28,29][Level of evidence C1] As in patients with Burkitt lymphoma, chemoresistant disease is common.
There are no standard treatment options for patients with recurrent or refractory disease.
Treatment options for recurrent or refractory lymphoblastic lymphoma include the following:
Evidence (treatment of recurrent or refractory lymphoblastic lymphoma):
Information about National Cancer Institute (NCI)–supported clinical trials can be found on the NCI website. For information about clinical trials sponsored by other organizations, see the ClinicalTrials.gov website.
Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.
Anaplastic large cell lymphoma accounts for approximately 10% of childhood non-Hodgkin lymphoma (NHL) cases.[1] For more information about the incidence of anaplastic large cell lymphoma by age and sex distribution, see Table 1.
Clinically, systemic anaplastic large cell lymphoma has a broad range of presentations. These include involvement of lymph nodes and a variety of extranodal sites, particularly skin and bone and, less often, gastrointestinal tract, lung, pleura, and muscle. Involvement of the central nervous system (CNS) and bone marrow is uncommon.
Anaplastic large cell lymphoma is often associated with systemic symptoms (e.g., fever, weight loss) and a prolonged waxing and waning course, making diagnosis difficult and often delayed. Patients with anaplastic large cell lymphoma may present with signs and symptoms consistent with hemophagocytic lymphohistiocytosis.[2]
There is a subgroup of patients with anaplastic large cell lymphoma who have leukemic peripheral blood involvement. These patients usually exhibit significant respiratory distress with diffuse lung infiltrates or pleural effusions and have hepatosplenomegaly.[3,4]
While mature T cell is the predominant immunophenotype of anaplastic large cell lymphoma, null-cell disease (i.e., no T-cell, B-cell, or natural killer-cell surface antigen expression) does occur. The World Health Organization (WHO) classifies anaplastic large cell lymphoma as a subtype of peripheral T-cell lymphoma.[5,6]
All anaplastic large cell lymphoma cases are CD30-positive. More than 90% of pediatric anaplastic large cell lymphoma cases have a chromosomal rearrangement involving the ALK gene. About 85% of these chromosomal rearrangements will be t(2;5)(p23;q35), leading to the expression of the NPM::ALK fusion protein. The other 15% of cases are composed of variant ALK translocations.[7] The anti-ALK immunohistochemical staining pattern is quite specific for the type of ALK translocation. Cytoplasm and nuclear ALK staining is associated with NPM::ALK fusion proteins, whereas cytoplasmic staining of ALK is only associated with the variant ALK translocations, as shown in Table 6.[8]
Gene Fusion | Partner Chromosome Location | Frequency of Gene Fusion |
---|---|---|
aAdapted from Tsuyama et al.[8] | ||
NPM::ALK | 5q36.1 | Approximately 80% |
TPM3::ALK | 1p23 | Approximately 15% |
ALO17::ALK | 17q25.3 | Rare |
ATIC::ALK | 2q35 | Rare |
CLTC::ALK | 17q23 | Rare |
MSN::ALK | Xp11.1 | Rare |
MYH9::ALK | 22q13.1 | Rare |
TFG::ALK | 3q12.2 | Rare |
TPM4::ALK | 19p13 | Rare |
TRAF1::ALK | 9q33.2 | Rare |
In adults, ALK-positive anaplastic large cell lymphoma is viewed differently from other peripheral T-cell lymphomas because prognosis tends to be superior.[9] Also, adult patients with ALK-negative anaplastic large cell lymphoma have an inferior outcome compared with patients who have ALK-positive disease.[10] In children, however, this difference in outcome between ALK-positive and ALK-negative disease has not been demonstrated. In addition, no correlation has been found between outcome and the specific ALK-translocation type.[11-13]
One European series included 375 children and adolescents with systemic ALK-positive anaplastic large cell lymphoma. The presence of a small cell or lymphohistiocytic component was observed in 32% of patients, and it was significantly associated with a high risk of failure in the multivariate analysis, controlling for clinical characteristics (hazard ratio, 2.0; P = .002).[12] The prognostic implication of the small cell variant of anaplastic large cell lymphoma was also shown in the COG-ANHL0131 (NCT00059839) study, despite using a different chemotherapy backbone.[13]
For information on prognostic factors for anaplastic large cell lymphoma, see the Prognosis and Prognostic Factors for Childhood NHL section.
Children and adolescents with high-stage (stage III or IV) anaplastic large cell lymphoma have a disease-free survival rate of approximately 60% to 75%.[14-19]
It is unclear which treatment strategy is best for patients with anaplastic large cell lymphoma. Current data do not suggest superiority of one treatment regimen over another for these standard treatment options.
Commonly used treatment regimens include the following:
Evidence (treatment of anaplastic large cell lymphoma):
CNS involvement in patients with anaplastic large cell lymphoma is rare at diagnosis. In an international study of systemic childhood anaplastic large cell lymphoma, 12 of 463 patients (2.6%) had CNS involvement, 3 of whom had isolated CNS disease (primary CNS lymphoma). For the CNS-positive group who received multiagent chemotherapy, including high-dose methotrexate, cytarabine, and intrathecal treatment, the EFS rate was 50% (95% CI, 25%–75%), and the OS rate was 74% (95% CI, 45%–91%) at a median follow-up of 4.1 years. The role of cranial radiation therapy has been difficult to assess.[27]
Unlike mature B-cell or lymphoblastic lymphoma, the survival rates for patients with recurrent or refractory anaplastic large cell lymphoma are 40% to 60%.[28-31]
There is no standard approach for the treatment of recurrent or refractory anaplastic large cell lymphoma.
Treatment options for recurrent or refractory anaplastic large cell lymphoma include the following:
Although remissions can be achieved with single-agent therapy (e.g., vinblastine, brentuximab vedotin, or crizotinib), CNS progressions after therapy have been observed in patients with recurrent anaplastic large cell lymphoma. A large retrospective review of a study database found that the incidence of CNS involvement at relapse is about 4%. The median time to relapse with CNS involvement was 8 months for 26 patients. The 3-year OS rate after relapse was about 50%.[42]
Chemotherapy, followed by autologous or allogeneic HSCT, if remission can be achieved, has been used in this setting.[29,30,39,40,43]
Evidence (chemotherapy and targeted therapy):
Evidence (autologous vs. allogeneic HSCT):
Information about National Cancer Institute (NCI)–supported clinical trials can be found on the NCI website. For information about clinical trials sponsored by other organizations, see the ClinicalTrials.gov website.
The following are examples of national and/or institutional clinical trials that are currently being conducted:
Patients with tumors that have molecular variants addressed by treatment arms included in the trial will be offered treatment on Pediatric MATCH. Additional information can be obtained on the NCI website and ClinicalTrials.gov website.
Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.
The incidence of lymphoproliferative disease or lymphoma is 100-fold higher in immunocompromised children than in the general population. The causes of such immune deficiencies include the following:
Non-Hodgkin lymphoma (NHL) associated with immunodeficiency is usually aggressive. Most cases occur in extralymphatic sites and have a higher incidence of primary central nervous system (CNS) involvement.[1-4]
Lymphoproliferative disease observed in primary immunodeficiency usually shows an aggressive mature B-cell phenotype and large cell histology.[2] Mature T-cell lymphoma and anaplastic large cell lymphoma have been observed.[2] Children with primary immunodeficiency and NHL are more likely to have high-stage (stage III or stage IV) disease and present with symptoms related to extranodal disease, particularly in the gastrointestinal tract and CNS.[2]
Treatment options for lymphoproliferative disease associated with primary immunodeficiency include the following:
Patients with primary immunodeficiency can achieve complete and durable remissions with standard chemotherapy regimens for NHL, although toxicity is increased.[2]; [5][Level of evidence C1] Recurrences in these patients are common and may not represent the same clonal disease.[6] Immunologic correction through allogeneic HSCT is often required to prevent recurrences.
The incidence of NHL is increased in patients with DNA repair syndromes, including ataxia-telangiectasia, Nijmegen breakage syndrome, and constitutional mismatch repair deficiency. Aggressive mature B-cell NHL accounts for most NHL seen in patients with ataxia-telangiectasia (84%) and Nijmegen breakage syndrome (46%), while T-cell lymphoblastic lymphoma (81%) is observed in patients with constitutional mismatch repair deficiency.[5]
Patients with DNA repair defects are particularly difficult to treat.[7,8] Overall survival (OS) rates at 5 and 10 years are poor, at 40% to 60%.[5,9]
Treatment options for NHL associated with DNA repair defect syndromes include the following:
NHL in children with HIV often presents with fever, weight loss, and symptoms related to extranodal disease, such as abdominal pain or CNS symptoms.[1] Most childhood HIV-related NHL is of mature B-cell phenotype but with a spectrum, including primary effusion lymphoma, primary CNS lymphoma, mucosa-associated lymphoid tissue (MALT), Burkitt lymphoma, and diffuse large B-cell lymphoma.[10,11]
HIV-associated NHL can be broadly grouped into the following three subcategories:
Highly active antiretroviral therapy has decreased the incidence of NHL in HIV-positive individuals, particularly for primary CNS lymphoma cases.[13,14]
Treatment options for HIV-associated NHL include the following:
In the era of highly active antiretroviral therapy, children with HIV and NHL are treated with standard chemotherapy regimens for NHL. However, the prevention (using prophylaxis) and early detection of infection is warranted.[1,13,14] Although the number of pediatric patients with HIV-associated NHL is too small to perform meaningful clinical trials, studies of adult patients support the addition of rituximab to standard treatment regimens.[15] Treatment of recurrent disease is based on histology using standard approaches.
PTLD represents a spectrum of clinically and morphologically heterogeneous lymphoid proliferations. Essentially all PTLDs after HSCT are associated with EBV, but EBV-negative PTLD can be seen after solid organ transplant.[3] While most PTLDs are of B-cell phenotype, approximately 10% are mature (peripheral) T-cell lymphomas.[16] The B-cell stimulation by EBV may result in multiple clones of proliferating B cells. Both polymorphic and monomorphic histologies may be present in a patient, even within the same lesion of PTLD.[17] Thus, histology of a single biopsied site may not be representative of the entire disease process.
The World Health Organization (WHO) has classified PTLD into the following three subtypes:[16]
EBV lymphoproliferative disease posttransplant may manifest as isolated hepatitis, lymphoid interstitial pneumonitis, meningoencephalitis, or an infectious mononucleosis-like syndrome. The definition of PTLD is frequently limited to lymphomatous lesions (low stage or high stage), which are often extranodal (frequently in the allograft).[3] PTLD may less commonly present as a rapidly progressive, high-stage disease that clinically resembles septic shock, and these patients have a poor prognosis. However, the use of rituximab and low-dose chemotherapy may improve outcomes in these patients.[18,19] U.S. transplant and cancer registries show that PTLD accounts for about 3% of all pediatric NHL diagnoses; 65% of PTLDs have diffuse large B-cell lymphoma histology, and 9% of PTLDs have Burkitt histology.[20]
PTLD represents a broad spectrum of disorders. The variant profile was evaluated in 31 pediatric patients with PTLDs, including 7 PTLD cases with Burkitt lymphoma histology (PTLD-BL) and 24 PTLD cases with diffuse large B-cell lymphoma histology (PTLD-DLBC).[21] While both groups were generally EBV positive, PTLD-BL cases expressed an EBV latency type 1 pattern and had variants in MYC, ID3, DDXC3, ARID1A, or CCND3, resembling Burkitt lymphoma in immunocompetent children. In contrast, the PTLD-DLBC cases were more heterogenous and appeared to be a molecularly distinct group. In general, pediatric PTLD-DLBC cases were genetically less complex than cases of adult PTLD-DLBC and diffuse large B-cell lymphoma in immunocompetent pediatric patients.
Treatment options for PTLD include the following:
First-line therapy for patients with PTLD is to reduce immunosuppressive therapy as much as possible.[27,28] However, this may not be possible because of the increased risk of organ rejection or graft-versus-host disease (GVHD).
Rituximab, an anti-CD20 antibody, has been used in the posttransplant setting. Rituximab as a single agent to treat PTLD after organ transplant has demonstrated efficacy in adult patients, but data are lacking in pediatric patients.
Evidence (rituximab):
For more information, see the Polymorphic Posttransplant Lymphoproliferative Disorder section in B-Cell Non-Hodgkin Lymphoma Treatment.
Low-intensity chemotherapy has been effective in patients with EBV-positive, CD20-positive B-lineage PTLD.[19,29] An event-free survival (EFS) rate of 67% was demonstrated in a Children's Oncology Group study using rituximab plus cyclophosphamide and prednisone in children with PTLD after solid organ transplant in whom immune suppression was reduced.[19][Level of evidence B4]
Some studies suggest that modified conventional lymphoma therapy is effective for patients who have PTLD with MYC translocations and Burkitt lymphoma histology.[24,25][Level of evidence C2] A multicenter retrospective review summarized the treatments and outcomes of 35 patients with PTLD-BL. Fluorescence in situ hybridization (FISH) detected the MYC translocation in 95% of cases. Treatments ranged from rituximab only to FAB/LMB therapy. The 3-year EFS and OS rates for all patients were 66.2% and 88.0%, respectively. The most commonly used therapy was a low-dose chemotherapy approach that is similar to the COG regimen (cyclophosphamide, prednisone, and rituximab [CPR]; n = 13). Using this approach, the EFS rate was 52.7%, and the OS rate was 84.6%.[26]
Patients with T-cell or Hodgkin-like PTLD are usually treated with standard lymphoma-specific chemotherapy regimens.[30-33]
Antirejection therapy is usually decreased or discontinued when chemotherapy is given to avoid excessive toxicity. There are no data to guide the re-initiation of immunosuppressive therapy after chemotherapy treatment. There is little evidence of benefit for chemotherapy after HSCT.
Adoptive immunotherapy with either donor lymphocytes or ex vivo–generated EBV-specific cytotoxic T lymphocytes (EBV-CTLs) has been effective in treating patients with PTLD after blood or bone marrow transplant.[34,35] To make this approach more broadly applicable, banks of off-the-shelf, third-party, allogeneic EBV-CTLs derived from healthy donors have been developed.[36,37] EBV-CTLs were evaluated in 46 patients with PTLD that had either progressed during rituximab treatment, not fully responded to rituximab treatment, or had recurred after a previous response. The following results were observed:[38]
Information about National Cancer Institute (NCI)–supported clinical trials can be found on the NCI website. For information about clinical trials sponsored by other organizations, see the ClinicalTrials.gov website.
The following is an example of a national and/or institutional clinical trial that is currently being conducted:
An international collaboration identified 95 cases of lymphoid neoplasms after the diagnosis of ALL.[39] Of these cases, 52 were characteristic of EBV-associated lymphoproliferative disease in the setting of immunodeficiency. These 52 cases were analyzed, along with 14 additional cases identified from the literature (n = 66). All cases occurred in the maintenance phase or within 6 months of completing maintenance (median, 14 months into maintenance therapy). Treatment strategies varied, but two-thirds of the patients were event-free survivors at 5 years.
Low-grade or intermediate-grade mature B-cell lymphomas—such as small lymphocytic lymphoma, mucosa-associated lymphoid tissue (MALT) lymphoma, mantle cell lymphoma, myeloma, or follicular cell lymphoma—are rarely seen in children. The World Health Organization (WHO) classification has identified pediatric-type follicular lymphoma and pediatric nodal marginal zone lymphoma as entities separate from their adult counterparts.[1]
The Children's Oncology Group (COG) opened a registry study (COG-ANHL04B1) to learn more about the clinical and pathological features of these rare types of pediatric non-Hodgkin lymphoma (NHL). This study banks tissue for pathobiology studies and collects limited data on clinical presentation and outcome of therapy.[2]
Gray zone lymphomas represent a hybrid malignancy, with an unclassifiable B-cell lymphoma and classical Hodgkin lymphoma, which may present together in an initial biopsy or sequentially as a relapse.[3]
A retrospective case series study assessed the clinical characteristics and outcomes of six patients with gray zone lymphomas from Austria. The three male and three female patients ranged in age from 15 to 17 years. Two of the six patients had B symptoms and high lactate dehydrogenase (LDH) levels. All patients had mediastinal masses, and five of six patients had positive cervical/supraclavicular lymph nodes. Extranodal involvement of the pleura and lung was common. Initial therapy with B-cell NHL treatments in five patients led to a complete response (CR) in one patient and progressive disease and death in one patient. The other three patients relapsed with primarily classical Hodgkin lymphoma histology and required treatment with salvage therapy. All of these patients survived after high-dose therapies and hematopoietic stem cell transplants (HSCT). One patient who initially received Hodgkin lymphoma therapy achieved a CR and survived.[4]
Pediatric-type follicular lymphoma is a disease that genetically and clinically differs from its adult counterpart and is recognized by the WHO classification as a separate entity from follicular lymphoma observed commonly in adults.[5] The genetic hallmark of adult follicular lymphoma is t(14;18)(q32;q21) involving BCL2. However, this translocation must be excluded to diagnose pediatric-type follicular lymphoma.[5-8]
Pediatric-type follicular lymphoma predominantly occurs in males, is associated with a high proliferation rate, and is more likely to be localized disease.[6,9,10] In pediatric-type follicular lymphoma, a high-grade component (i.e., grade 3 with high proliferative index such as Ki-67 expression of >30%) resembling diffuse large B-cell lymphoma can frequently be detected at initial diagnosis but does not indicate a more aggressive clinical course in children. Unlike follicular lymphoma in adults, pediatric-type follicular lymphoma does not transform to diffuse large B-cell lymphoma.[5,6,8,10,11] Limited-stage disease is observed with pediatric-type follicular lymphoma. Cervical lymph nodes and tonsils are common sites, but disease has also occurred in extranodal sites such as the testis, kidney, gastrointestinal tract, and parotid gland.[6-8,11-14]
Pediatric-type follicular lymphoma and nodal marginal zone lymphoma are rare indolent B-cell lymphomas that are clinically and molecularly distinct from these tumor types in adults.
Pediatric-type follicular lymphoma is rare in children, with only case reports and small case series to guide therapy. The outcomes of patients with pediatric-type follicular lymphoma are excellent, with an event-free survival (EFS) rate of about 95%.[6,8-11,13] Unlike in adult follicular lymphoma, the clinical course in pediatric patients is not dominated by relapses.[6,8,11,12]
Treatment options for pediatric-type follicular lymphoma include the following:
Studies suggest that for children with stage I disease who had a complete resection, a watch-and-wait approach without chemotherapy may be indicated. Patients with higher-stage disease also have a favorable outcome with low-intensity and intermediate-intensity chemotherapy, with an EFS rate of 94% and an overall survival (OS) rate of 100% (2-year median follow-up).[2,6,9,10] Although the number of pediatric patients with pediatric follicular-type lymphoma is too small to perform meaningful clinical trials, studies of adult patients with follicular lymphoma support the addition of rituximab to standard treatment regimens.
For patients with BCL2-rearranged tumors, treatment similar to that of adult patients with follicular lymphoma is administered.
For more information, see the Follicular Lymphoma (Grades 1–3a) section in B-Cell Non-Hodgkin Lymphoma Treatment.
Marginal zone lymphoma is a type of indolent lymphoma that is rare in pediatric patients. Marginal zone lymphoma can present as nodal or extranodal disease and almost always as low-stage (stage I or stage II) disease. It is unclear whether the marginal zone lymphoma that is observed in pediatric patients is clinicopathologically different from the disease that is observed in adults. Most extranodal marginal zone lymphoma in pediatrics presents as MALT lymphoma and may be associated with Helicobacter pylori (gastrointestinal) or Chlamydophila psittaci (conjunctival), previously called Chlamydia psittaci.[22,23]
Treatment options for marginal zone lymphoma (including MALT lymphoma) include the following:
Most pediatric patients with marginal zone lymphomas require no more than local therapy involving curative surgery and/or radiation therapy.[22,24] Treatment of patients with MALT lymphoma of the gastric mucosa may also include antibiotic therapy, which is considered standard treatment in adults. However, the use of antibiotic therapy in children has not been well studied because there are so few cases.
Evidence (treatment of marginal zone lymphoma):
Although the number of pediatric patients with MALT lymphoma is too small to perform meaningful clinical trials, studies of adult patients support the use of rituximab with or without chemotherapy. For more information, see the Marginal Zone Lymphoma section in B-Cell Non-Hodgkin Lymphoma Treatment.
Intralesional interferon-alpha for conjunctival MALT lymphoma has been studied in trials.[26]
Other types of NHL that are rare in adults and are exceedingly rare in pediatric patients include primary CNS lymphomas. Because of the small numbers of patients, it is difficult to ascertain whether the disease observed in children is the same as the disease observed in adults.
Reports suggest that the outcome of pediatric patients with primary CNS lymphoma (OS rate, 70%–80%) may be superior to that of adults with primary CNS lymphoma.[27-30]
Most children have diffuse large B-cell lymphoma, although other histologies have been observed.
Treatment options for primary CNS lymphoma include the following:
Therapy with high-dose intravenous methotrexate and cytosine arabinoside is the most successful, and intrathecal chemotherapy may be needed only when malignant cells are present in the cerebrospinal fluid.[31]
There are case reports describing the administration of repeated doses of intraventricular rituximab in patients with refractory primary CNS lymphoma, with excellent results reported.[32,33] This apparently good outcome needs to be confirmed, and similar results have not been observed in adults. It is generally believed that rituximab does not cross the blood-brain barrier.
Among patients who have a partial response to induction therapy, and particularly those who are not eligible for transplant, reduced-dose whole-brain radiation therapy with a boost to residual disease may be a viable treatment approach that merits further investigation.[34,35]
For more information about treatment options for non–AIDS-related primary CNS lymphoma, see Primary CNS Lymphoma Treatment.
Peripheral T-cell lymphoma, excluding anaplastic large cell lymphoma, is rare in children.
Mature T-cell/natural killer (NK)–cell lymphoma or peripheral T-cell lymphoma has a postthymic phenotype (e.g., terminal deoxynucleotidyl transferase negative), usually expresses CD4 or CD8, and has rearrangement of T-cell receptor genes, either alpha-beta and/or gamma-delta chains. The most common phenotype observed in children is peripheral T-cell lymphoma, not otherwise specified (NOS), although angioimmunoblastic lymphoma, enteropathy-associated lymphoma (associated with celiac disease), subcutaneous panniculitis-like lymphoma, angiocentric lymphoma, and extranodal NK/T-cell peripheral T-cell lymphoma have been reported.[36-40]
Extranodal NK/T-cell lymphoma is a rare subtype of NHL, constitutes between 0.2% and 1.6% of newly diagnosed cases of NHL in children and adolescents, and is closely associated with the Epstein-Barr virus (EBV).[41] The incidence varies by region. The incidence is between 3% and 10% in Asian countries and 1% in western countries.[42] The common primary tumor sites are the nasal cavity and paranasal sinuses.[43] A standard treatment for pediatric patients has not been established. A series of 34 patients were treated with chemotherapy with or without asparaginase. At a median follow-up of 54 months, patients with lower-stage (I/II) disease had 5-year EFS and OS rates of 66.2% and 94.7%, respectively, compared with 26.0% and 42.3% for patients with stage III/IV disease. For all patients, there was no statistically significant difference in outcomes between patients who received asparaginase-containing regimens and those who did not. All patients with stage I/II disease received radiation therapy, whereas only 4 of 13 patients with higher-stage disease received radiation therapy. The 5-year EFS rate was 66.7% for stage III/IV patients who received hematopoietic stem cell transplant (HSCT) and 11.1% for patients who did not receive HSCT (P = .054).[44][Level of evidence C1]
Although very rare, gamma-delta hepatosplenic T-cell lymphoma may be seen in children.[39] This tumor has also been associated with children and adolescents who have Crohn disease and have been treated with immunosuppressive therapy. This lymphoma has been fatal in all cases.[45]
Optimal therapy for peripheral T-cell lymphoma is unclear for both pediatric and adult patients.
Treatment options for peripheral T-cell lymphoma include the following:
There have been four retrospective analyses of treatment and outcome for pediatric patients with peripheral T-cell lymphoma.
Evidence (treatment of peripheral T-cell lymphoma):
For more information about the treatment of adults, see Peripheral T-Cell Non-Hodgkin Lymphoma Treatment.
Primary cutaneous lymphomas are very rare in pediatric patients (1 case per 1 million person-years), but the incidence increases in adolescents and young adults. All histologies of NHL have been observed to involve the skin. More than 80% of cutaneous lymphomas are the T-cell or NK-cell phenotype.[48]
Subcutaneous panniculitic T-cell lymphomas (SPTCL) are very rare lymphomas with panniculitis-like infiltration of subcutaneous tissue by cytotoxic T-cells. SPTCL account for less than 1% of all peripheral T-cell lymphomas.[49-51] SPTCL can be observed with malignant T cells, expressing alpha-beta chain T-cell receptor or gamma-delta T-cell receptor rearrangements.
In adults, the gamma-delta subtype of SPTCL is associated with a more aggressive course and a worse prognosis than the alpha-beta subtype of SPTCL.[52] Morbidity and mortality are frequently related to the development of hemophagocytic syndrome, which was reported in one series of adults to occur in 17% of patients with alpha-beta SPTCL and in 45% of patients with gamma-delta SPTCL. The 5-year OS rate is 82% for patients with alpha-beta SPTCL and 11% for patients with gamma-delta SPTCL.[52]
SPTCL is heterogeneous in the pediatric age group and does not necessarily follow the course observed in adults. In a retrospective series of 18 children (median age, 11.1 years; range, 0.52–14.7 years, with 3 children aged <1 year), most presented with single or multiple subcutaneous nodules or patchy skin lesions on the limbs and/or trunk. Most of the patients also had fever, asthenia, and weight loss. Four out of five patients screened were positive for the HAVCR2 gene variant in the T-cell immunoglobulin domain and mucin domain 3 (TIM-3) lineages.[53][Level of evidence C3] Seven cases were associated with hemophagocytic syndrome, similar to 7 of 11 pediatric cases in another series.[54]; [53][Level of evidence C3]
The diagnosis of primary cutaneous anaplastic large cell lymphoma can be difficult to distinguish pathologically from more benign diseases such as lymphomatoid papulosis.[55] Primary cutaneous lymphomas are now thought to represent a spectrum of disorders, distinguished by clinical presentation.
Because of the rarity of cutaneous T-cell lymphoma, no standard treatments have been established. Management and treatment of patients with cutaneous T-cell lymphoma should be individualized and, in some cases, watchful waiting may be appropriate. Treatment may only be necessary if hemophagocytic syndrome develops.[56]
There is no standard treatment regimen for SPTCL. Spontaneous remissions have been observed, particularly in younger children. Older children, however, may have a course of disease that is complicated by hemophagocytic syndrome. First-line treatment consists of either chemotherapy or immunomodulatory drugs. Chemotherapy was the mainstay of treatment before 2019. Immunomodulatory therapy or observational follow-up became the mainstay of treatment after this time. Immunomodulatory agents include steroids combined with cyclosporine A or ruxolitinib. In a series of 18 patients, the CR rate was 71.4% for patients treated with these immunomodulatory agents.[53][Level of evidence C3]
An oral retinoid (bexarotene) has been reported to be active against SPTCL in a series of 15 patients from three institutions.[57] In a series of 11 pediatric patients, aggressive polychemotherapy was used in all patients. Nine of 11 patients sustained clinical remission, with a median follow-up of 3.5 years.[54] Additional treatment options include high-dose steroids, bexarotene, denileukin diftitox, multiagent chemotherapy, and HSCT.[51,56-61]
Primary cutaneous anaplastic large cell lymphoma usually does not express ALK and may be treated successfully with surgical resection and/or local radiation therapy without systemic chemotherapy.[62] There are reports of surgery alone also being curative for patients with ALK-positive cutaneous anaplastic large cell lymphoma, but extensive staging and vigilant follow-up is required.[63,64]
Mycosis fungoides is rarely reported in children and adolescents,[65-68] accounting for about 0.5% to 7% of all cases. In a systematic review of 571 children and adolescents with mycosis fungoides, the mean age of diagnosis was 12.2 years, and the mean age at onset was 8.6 years.
Compared with adults, pediatric patients are diagnosed with an earlier stage of mycosis fungoides and have a higher rate of atypical presentations, specifically the hypopigmented variant.[67] One of the largest series of pediatric patients with mycosis fungoides (n = 71; diagnosed aged <18 years) was followed for a mean of 9.2 years (range, 1–24 years).[67]
Factors associated with worse overall 10-year survival were delay in establishing the correct diagnosis, granulomatous slack skin, granulomatous mycosis fungoides, history of organ transplant, and stage 2 disease at the time of diagnosis.[69][Level of evidence C3] For information about the treatment of adults, see Mycosis Fungoides and Other Cutaneous T-Cell Lymphomas Treatment.
Mycosis fungoides in pediatric patients may respond to various therapies, including topical steroids, retinoids, radiation therapy, or phototherapy (e.g., narrowband UVB treatment [NBUVB]), but remission may not be durable.[70-73] In a retrospective series of 71 pediatric patients with mycosis fungoides, the overall response rate (CR + partial remission) was 88%. However, CR was achieved in only 40% of patients initially.[67][Level of evidence C3]
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.
General Information About Childhood Non-Hodgkin Lymphoma (NHL)
Added spinal cord as a site of disease that may have prognostic value. Added text to state that in a review of the Berlin-Frankfurt-Münster (BFM) database, 1.2% of children with NHL presented with symptoms of spinal cord compression. These cases were comprised of Burkitt lymphoma, B-cell lymphoblastic lymphoma, diffuse large B-cell lymphoma, anaplastic large cell lymphoma, and T-cell lymphoblastic lymphoma. The 5-year event-free survival and overall survival rates of patients with spinal cord compression did not differ from those of patients without spinal cord compression at diagnosis. Approximately one-third of long-term survivors had persistent neurological symptoms (cited Riquelme et al. as reference 58).
Treatment Option Overview for Childhood NHL
Revised Table 3 to include radiation therapy as a treatment option for primary mediastinal B-cell lymphoma.
Added text about the results of a large-scale retrospective study that assessed the spectrum of MYC-rearranged B-cell lymphomas and the fluorescence in situ hybridization results for MYC, BCL2, and BCL6 rearrangements and MYC immunoglobulin rearrangement partners in pediatric and young adult patients (cited Gagnon et al. as reference 66).
Added text to state that radiation therapy can be considered for patients who are unresponsive to salvage therapies or as a consolidation therapy for select patients (cited Saifi et al. as reference 73).
Added radiation therapy as a treatment option for primary mediastinal B-cell lymphoma.
Added Radiation therapy as a new subsection.
Added text to state that the morphology and immunophenotype of B-cell lymphoblastic lymphoma are known to overlap with those of B-cell acute lymphoblastic leukemia, but few studies have examined the genomic landscape of B-cell lymphoblastic lymphoma, partially due to the lack of sufficient material for genomic analysis. Also added text about the results of one study that has better evaluated the genomic alterations associated with pediatric B-cell lymphoblastic lymphoma (cited Kroeze et al. as reference 13).
Rare NHL Occurring in Children
Added Moser et al. as reference 46.
This summary is written and maintained by the PDQ Pediatric Treatment Editorial Board, which is editorially independent of NCI. The summary reflects an independent review of the literature and does not represent a policy statement of NCI or NIH. More information about summary policies and the role of the PDQ Editorial Boards in maintaining the PDQ summaries can be found on the About This PDQ Summary and PDQ® Cancer Information for Health Professionals pages.
This PDQ cancer information summary for health professionals provides comprehensive, peer-reviewed, evidence-based information about the treatment of childhood non-Hodgkin lymphoma. It is intended as a resource to inform and assist clinicians in the care of their patients. It does not provide formal guidelines or recommendations for making health care decisions.
This summary is reviewed regularly and updated as necessary by the PDQ Pediatric Treatment Editorial Board, which is editorially independent of the National Cancer Institute (NCI). The summary reflects an independent review of the literature and does not represent a policy statement of NCI or the National Institutes of Health (NIH).
Board members review recently published articles each month to determine whether an article should:
Changes to the summaries are made through a consensus process in which Board members evaluate the strength of the evidence in the published articles and determine how the article should be included in the summary.
The lead reviewers for Childhood Non-Hodgkin Lymphoma Treatment are:
Any comments or questions about the summary content should be submitted to Cancer.gov through the NCI website's Email Us. Do not contact the individual Board Members with questions or comments about the summaries. Board members will not respond to individual inquiries.
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 Treatment Editorial Board uses a formal evidence ranking system in developing its level-of-evidence designations.
PDQ is a registered trademark. Although the content of PDQ documents can be used freely as text, it cannot be identified as an NCI PDQ cancer information summary unless it is presented in its entirety and is regularly updated. However, an author would be permitted to write a sentence such as “NCI’s PDQ cancer information summary about breast cancer prevention states the risks succinctly: [include excerpt from the summary].”
The preferred citation for this PDQ summary is:
PDQ® Pediatric Treatment Editorial Board. PDQ Childhood Non-Hodgkin Lymphoma Treatment. Bethesda, MD: National Cancer Institute. Updated <MM/DD/YYYY>. Available at: https://www.cancer.gov/types/lymphoma/hp/child-nhl-treatment-pdq. Accessed <MM/DD/YYYY>. [PMID: 26389181]
Images in this summary are used with permission of the author(s), artist, and/or publisher for use within the PDQ summaries only. Permission to use images outside the context of PDQ information must be obtained from the owner(s) and cannot be granted by the National Cancer Institute. Information about using the illustrations in this summary, along with many other cancer-related images, is available in Visuals Online, a collection of over 2,000 scientific images.
Based on the strength of the available evidence, treatment options may be described as either “standard” or “under clinical evaluation.” These classifications should not be used as a basis for insurance reimbursement determinations. More information on insurance coverage is available on Cancer.gov on the Managing Cancer Care page.
More information about contacting us or receiving help with the Cancer.gov website can be found on our Contact Us for Help page. Questions can also be submitted to Cancer.gov through the website’s Email Us.