The SEER (Surveillance, Epidemiology, and End Results) data for the years 1975–1998 show that children with non-Hodgkin lymphoma (NHL) have a better treatment outcome than do adults. Many factors may contribute to this age-related difference. Some factors are related to the patient (e.g., drug distribution and clearance, performance status, compliance, sex) whereas others pertain to tumor histology and biology. The spectrum of NHL subtypes is well known to differ in children and adults. From ages 5 through 14 years, Burkitt lymphoma is the predominant histologic subtype, whereas diffuse large B-cell lymphoma is most common in the 15- to 29-year age range. Because different treatment strategies are often used in children and adults with NHL, the choice of therapy for adolescents and young adults (ages 15 through 29 years) is challenging and somewhat controversial. It is reasonable to consider pediatric strategies for some adolescents and very young adults with NHL, and pediatric strategies are currently used to treat adults with certain subtypes of NHL (Burkitt lymphoma, lymphoblastic lymphoma). However, the use of pediatric strategies in adults does not guarantee a comparable outcome, as illustrated by trials for adult lymphoblastic lymphoma. There is clearly a need for further biologic study of NHL in children, adolescents, and young adults. Age-related differences in tumor biology have been demonstrated in anaplastic large-cell lymphoma (ALCL) and diffuse large B-cell lymphoma (DLBCL). Additional biologic data will not only improve prognosis and treatment stratification but, more important, will lead to the identification of specific molecular targets for therapy.

Children with non-Hodgkin lymphoma (NHL) reportedly have an outcome superior to that of adults.1,2 Within the pediatric population, there are reports of a poorer outcome for some adolescents than for younger children.2,3 These differences among pediatric patients with NHL have been reported to vary according to gender and histologic subtype. The SEER (Surveillance, Epidemiology, and End Results) data covering the years 1975–1998 indicate that the probability of survival in patients with NHL declines with increasing age: when survival was analyzed according to age in 5-year increments from birth through 44 years, survival progressively declined as age increased (Figure 1; see Color Figures, page 517).2 Many factors may contribute to these age-related differences in treatment outcome. Some factors are related to the host, whereas others pertain to tumor histology and biology.

Drug distribution and clearance are known to vary with age,4 and the maximum tolerated dose (MTD) of many anticancer agents is higher in children than in adults.4 Concomitant drug administration, which may also differ with age, may also influence drug clearance and metabolism. For example, certain anticonvulsants induce the hepatic cytochrome P450 enzymes, thereby enhancing the metabolism (i.e., clearance) of various chemotherapeutic agents (e.g., vincristine, etoposide). A study of children with ALL showed that those receiving anticonvulsants had a poorer treatment outcome.5 

Performance status, including the underlying status of organ function, also influences treatment outcome. This factor is of concern in the elderly but is less likely to affect adolescents and adults in their twenties. Among adults, the international IPI score, which incorporates performance status, is used for screening and prognosis.6 This score is not generally used in the management of NHL in children and adolescents.

The patient’s compliance with the treatment plan is another critical factor in treatment outcome. In the pediatric population, adolescents may be less likely than others to remain compliant. Lack of compliance is often difficult to document, although certain studies, such as the measurement of red blood cell 6TGN in the case of oral 6-mercaptopurine therapy, may give some indication.7 Compliance may be improved if rigorous adherence to protocol guidelines is emphasized and optimal supportive care is provided in the clinic and at home.

Among children and adolescents, gender is reported to be associated with age-related outcome for certain histologic subtypes of NHL. For example, a large Berlin-Frankfurt-Münster (BFM) study showed that among patients with T-lymphoblastic lymphoma and DLBCL, the outcome of female patients was significantly inferior, particularly among adolescents.3 The reasons for these differences have yet to be elucidated, although the potential role of pubertal changes has been suggested.

Outcome is influenced by age-related differences in the distribution of NHL histologic subtypes and by the selection of appropriate histology-directed treatment.2,8 Further complicating the picture, age-related biologic differences within specific histologic subtypes may also influence treatment results. It is therefore important to identify the relative proportion of histologic subtypes and associated biologic features within various age groups and to examine what treatment strategies have been reported to be the most effective for these groups. A common and somewhat controversial challenge centers on the selection of therapy for adolescents and young adults (ages 15 to 29 years) with NHL. The remainder of this review will focus on age-related differences in tumor histology, tumor biology, and treatment selection.

The annual incidence of NHL steadily increases throughout life, in contrast to the bimodal age distribution of Hodgkin lymphoma, which peaks at ages 20 to 25 years and has a smaller peak at ages 75 to 80 years.2 In the year 2000, the estimated incidence of NHL per million in the United States (SEER) was 2.8 (66 patients) for ages < 5 years, 5.5 (110 patients) for ages 5–9 years, 8.8 (147 patients) for ages 10–14 years, 14.3 (290 patients) for ages 15–19, 21.8 (413 patients) for ages 20–24 years, and 39.3 (762 patients) for ages 25–29 years.2 

The spectrum of NHL subtypes in children has long been known to differ from that observed in adults (Figure 2; see Color Figures, page 517). The NHL subtypes in children are predominantly extranodal high-grade tumors, whereas those that occur in adults are more commonly low-and intermediate-grade tumors. In a large single-institution review of childhood NHL, the spectrum of subtypes included Burkitt lymphoma (38.8%), lymphoblastic lymphoma (28.1%), large-cell lymphoma (26.3%), and rare subtypes, including follicular lymphoma (6.8%).9 In striking contrast, adults in North America most commonly have follicular lymphoma and DLBCL; Burkitt lymphoma and lymphoblastic lymphoma occur relatively infrequently.

The SEER data provide insights about the specific age ranges in which these differences begin to emerge.2,8 From ages 5 to 14 years, Burkitt lymphoma is the predominant histologic subtype, whereas diffuse large B-cell lymphomas are the most common subtype in the 15- to 19-year age group.2,8 More specifically, the diffuse large B-cell subtype is the predominant histologic diagnosis in the 15–19, 20–24 and 25–29 year age groups.2 The proportion of cases of follicular lymphoma is also greater in the 25- through 29-year age group than in younger age groups.2 Thus, age 15 to 29 years appears to coincide with the transition from pediatric to adult subtypes of lymphoma (Figure 3; see Color Figures, page 517).2 

Stage- and histology-directed multiagent chemotherapy is the foundation of modern therapy for NHL. Involved-field radiation is not widely used for newly diagnosed disease, because it did not appear to improve treatment outcome if appropriately intensive chemotherapy was used in children with limited10 or advanced-stage11 NHL and in adults with localized aggressive disease.12 Debulking surgery is not indicated except in the case of a localized terminal ileocecal mass with mesenteric nodal involvement only, which can be completely resected. Complete resection alters the tumor stage and the indicated therapy.

Pediatric regimens and results

The Children’s Cancer Group (CCG) trial, which compared the cyclophosphamide-based COMP regimen with the LSA2L2 regimen designed for ALL, demonstrated that the COMP regimen was superior for the treatment of advanced- stage Burkitt lymphoma, whereas the LSA2L2 regimen was superior for the management of advanced-stage lymphoblastic lymphoma.13 Subsequent refinement, including the addition of high-dose methotrexate and high-dose cytarabine to a cyclophosphamide-based backbone14,16 and the further intensification of therapy,17,19 have resulted in cure rates in excess of 80%. In the NCI protocol, results were improved by adding the IVAC regimen (ifosfamide, etoposide, high-dose cytarabine) to the previously studied CODOX-M backbone (cyclophosphamide, vincristine, high-dose methotrexate, and intrathecal therapy).19 Excellent results were also achieved with the BFM90 regimen, which featured three risk groups in which the intensity of therapy was tailored to the degree of surgical resection, serum LDH activity, and tumor stage.17 The 6-year event-free survival (EFS) for stages III and IV NHL and B-ALL were 86%, 73% and 74%, respectively. The French LMB-89 regimen also features a risk-based 3-arm regimen with intensification based on degree of resection and disease stage.18 The 5-year EFS for stages III and IV NHL and BALL were 91%, 87% and 87%, respectively. An international collaborative trial (LMB-96) confirmed these excellent results.20,21 Unifying features of these highly effective pediatric regimens include risk-based treatment, brief but intensive multiagent cyclophosphamide-based chemotherapy, and aggressive central nervous system (CNS) prophylaxis and treatment.

Adult regimens and results

Adults with Burkitt lymphoma can be successfully treated with regimens designed for children. For example, the NCI CODOX-M/IVAC regimen resulted in an equally excellent result for adults and children with Burkitt lymphoma.19 Similarly, French investigators have demonstrated that an excellent result can be achieved in adults with Burkitt lymphoma by using pediatric LMB-based regimens.22 In their study, the 3-year overall survival rate was 80% for patients with stage III NHL and 57% for those with stage IV NHL or B-ALL. Other effective regimens designed for adults share the features of pediatric regimens described above.23,24 For example, the hyper-CVAD regimen features courses of cyclophosphamide, vincristine, doxorubicin, and prednisone alternating with courses of high-dose methotrexate and cytarabine given in a dose-intensive fashion.23 The outcome of treatment with hyper-CVAD23 appeared to be improved by the addition of rituximab, particularly in elderly patients.24 

Biology and prognostic factor

Gene-expression profiling has recently advanced our understanding of the molecular features of Burkitt lymphoma.25,27 Studies that focus specifically on children are under way and may lead to a greater understanding of the potential biologic differences of the Burkitt lymphomas in children and in adults. A study comparing the karyotypic abnormalities in Burkitt lymphoma in children versus adults found no significant difference in the karyotype complexity or the nature of the chromosomal abnormalities; however, the prognostic significance of specific chromosomal abnormalities differed in the two groups.28 In children, abnormalities of chromosome arms 22q and 13q were associated with a poorer outcome, whereas in adults, abnormalities of chromosome 17 appeared to be associated with risk.28 

Recommendations and conclusions

Contemporary treatment for Burkitt lymphoma in children and adults is very similar. Thus, in the case of this histologic subtype, there is little controversy about using a dose-intensive therapeutic approach that emphasizes alkyators (e.g., cyclophosphamide, ifosfamide), high-dose methotrexate, and cytarabine. Continued study of both pediatric and adult Burkitt lymphoma is needed to identify potential prognostic markers, and more importantly, potential targets for novel therapeutic agents.

Pediatric regimens and results

Strategies to improve the cure rate for children with lymphoblastic lymphoma have generally built on the results of the CCG trial demonstrating that an ALL-directed approach (induction, extended maintenance, and CNS prophylaxis components) is superior to the cyclophosphamide-based COMP regimen.13 The best result to date (a 5-year EFS of 90%) was achieved with the BFM90 regimen, which features an induction phase, a consolidation phase of 4 courses of high-dose methotrexate (5 g/m2), and a maintenance phase of 6-mercaptopurine and low-dose methotrexate.29 Components of this and other successful regimens that are thought to contribute to improved treatment outcome include high-dose methotrexate, asparaginase, anthracyclines, a re-intensification (i.e., re-induction) phase, and a prolonged maintenance phase (approximately 2 years of therapy).30 

Adult regimens and results

As in children, treatment outcome in adults with lymphoblastic lymphoma has been improved with regimens derived from ALL protocols.1,31,32 Nevertheless, the outcomes in adults with lymphoblastic NHL appear to be inferior to those in children. In sequential GMALL studies that featured multiagent induction, consolidation, and re-induction phases delivered over 6 to 12 months, overall survival, continuous complete remission (CR), and disease-free survival estimates at 7 years were 51%, 65%, and 62%, respectively.31 The MD Anderson study of hyper-CVAD resulted in 3-year progression-free survival and overall survival estimates of 66% and 70%, respectively.32 A recent study in Vancouver suggested that outcome was improved by incorporation of an intensification phase featuring hematopoietic stem cell support for patients in partial remission (PR) or CR after induction therapy;1 however, others have observed no benefit from addition of such an intensification phase.33 The apparent worse outcome in adults than in children treated with ALL-derived regimens has yet to be explained but may reflect age-related differences in the biologic features of these tumors.

Biology

The majority of pediatric lymphoblastic lymphomas have a precursor T-cell immunophenotype; the precursor B-cell immunophenotype is less common. There is very little information available about the biologic features of pediatric lymphoblastic lymphoma. This lack probably reflects the limited tissue obtained from non-invasive biopsy samples of anterior mediastinal masses with associated airway compression. Recently, the cytogenetic analysis of 13 pediatric patients with advanced-stage lymphoblastic lymphoma revealed a high (85%) frequency of chromosomal abnormalities.34 A larger cohort of children must be studied to determine the frequency and prognostic significance of genetic abnormalities in these tumors. Data from gene array studies may provide insight into the potential differences between lymphoblastic NHL in adults and children. These studies may also identify genes whose expression is associated with a poorer prognosis, as has been done in T-cell lymphoblastic leukemia. Specifically, HOX11 activation is reportedly associated with a superior outcome, and TAL1 or LYL1 expression with a poorer outcome, in patients with T-ALL.35 The expression of these genes has yet to be studied in large numbers of patients with lymphoblastic lymphoma; however, preliminary array data from children have been reported,36 and additional studies are under way in the current Children’s Oncology Group (COG) trial.

Conclusions and recommendations

A multiagent regimen derived from successful strategies to treat T-cell ALL is indicated for children and young adults; the current COG trial for lymphoblastic lymphoma is open to patients up to 35 years of age. Moreover, the demonstration that young adults with ALL have a superior outcome when treated with more intensive pediatric regimens37 provides a strong rationale for a similar approach in the treatment of young adults with lymphoblastic lymphoma. There is clearly a need for biologic studies of both pediatric and adult lymphoblastic NHL to identify patients with a poor prognosis for whom early intervention is indicated and to identify possible targets for novel therapeutic interventions.

Pediatric regimens and results

Successful therapies for anaplastic large-cell lymphoma (ALCL) in children are more diverse than those for other histologic subtypes of childhood NHL. In Europe, an immunophenotype-directed approach has historically been used in the treatment of large cell lymphomas, including ALCL.38,39 The BFM have reported an excellent result using a B-cell approach (i.e., a strategy like that used to treat Burkitt lymphoma), achieving a 5-year EFS of 76% for all stages combined.38 In the United States, a histology-based approach has historically been used to treat children with large cell lymphoma. Many of these regimens are CHOP-based.40 One of the most successful (a 70% EFS for advanced-stage disease) is the anthracycline-containing APO regimen, which features an induction phase followed by sequential maintenance phases delivered over one year.41 Current cooperative group trials in Europe and the United States are building on the BFM B-cell and APO strategies, respectively. Both studies are examining the benefit of adding vinblastine, which is active as a single agent against multiply relapsed ALCL.42 

Adult regimens and results

Adults with ALCL are also generally treated with a multiagent anthracycline-containing regimen.43,48 However, the treatment outcome is inferior to that achieved in children. Age-related biologic differences in the lymphomas may explain this disparity.

Biologic markers

In a number of adult trials, patients with anaplastic lymphoma kinase (ALK)+ ALCL had an overall survival rate superior to that in ALK patients;45,47 however, some studies have not observed this survival advantage.49 In those that do, the younger age of patients with ALK+ disease is suggested to be responsible for the advantage. The majority of pediatric patients with ALCL are ALK+.43 Among adult patients with ALK+ ALCL, the 5-year overall survival rates have approached those achieved in children. It is important that ALK ALCL not be confused with lymphomatoid papulosis and primary cutaneous ALCL, which are also ALK and do not require intensive chemotherapy.

Other biologic markers have been thought to have prognostic significance in ALCL.50 In a large study of 143 cases of adult and pediatric ALCL, CD56 expression was also identified as an independent predictor of outcome, regardless of ALK expression status.50 Although children were included in this study, their number was not defined. A subsequent case report and review of the literature did not identify a uniformly aggressive clinical course for children with CD56+ ALCL but indicated the need to study a larger cohort of patients with this phenotype.51 

A recent COG study of 40 pediatric patients with ALCL identified ALK expression and activation of STAT3 in most tumors; however, CD56 expression was infrequent, as was cytoplasmic localization of survivin and TIMP1.52 The authors suggested that although STAT3 is activated, the downstream anti-apoptotic effect of activated STAT3 is likely to be limited by other mechanisms, and that these effects may contribute to the relatively good prognosis of ALCL in children.

Conclusions and recommendation

Although the EFS rates of 70% to 80% achieved with the pediatric regimens described above38,41 suggest that they offer an advantage to adolescents and very young adults, they have not been proven in randomized trials to be superior to the most commonly used adult regimens with respect to either age group or ALK status. However, the outcome of the small number of ALK children treated with the BFM approach was not substantially inferior to that of ALK+ patients.38 It is therefore reasonable to use the BFM or APO regimen for adolescents and very young adults with ALCL; however, outcomes may still worsen with increasing age because of the known and unknown biologic differences between ALCL in adults and children. Future advances may be achieved by including an immunotherapeutic agent such as anti-CD30.53,54 Clinical trials of anti-CD30 in combination with conventional chemotherapy are currently under way in both children and adults. The success of allogeneic bone marrow transplantation (BMT) for refractory or high-risk pediatric ALCL also suggests the potential immunotherapeutic benefit of a graft-versus-lymphoma effect.55 

Pediatric regimens and results

The outcome of children with DLBCL is excellent. The approach most commonly used is that described earlier for Burkitt lymphoma. For example, in the international LMB-96 trial, 4-year EFS for patients with DLBCL (excluding primary mediastinal disease) was 92%, compared with 93% for those with Burkitt lymphoma.20 Those with primary mediastinal tumor had a slightly inferior 4-year EFS of 71%. The APO regimen has also been shown to have activity against DLBCL, but the 4-year EFS of 63.8% was inferior to that achieved with the Burkitt lymphoma regimen LMB-96.41 

Adult regimens and results

The prognosis for adults with DLBCL is inferior to that for children. CHOP has been the most widely used combination for more than 30 years for DLBCL in adults: the addition of other agents to the CHOP backbone (e.g., CHOP-BLEO, M-BACOD, MACOP-B, ProMACE-CytaBOM) failed to improve on the outcome achieved with CHOP.56 Subsequent trials focused on intensification of therapy, but the addition of rituximab to the CHOP regimen has had the greatest effect on outcomes for adults with DLBCL. The treatment of adults with DLBCL is covered in detail by Dr. Habermann in this volume.

Biologic markers

Gene expression profiling studies of adult DLBCL have identified two molecularly distinct subtypes: those with a germinal center (GC) B-like phenotype and those with an activated B-cell (ABC)–like phenotype.57 Among adults, DLBCL with a GC phenotype has a better outcome than DLBCL with an ABC phenotype.57 Most cases of DLBCL in children were recently found to have a GC phenotype.58 The authors suggest that this finding may explain the superior treatment outcome observed in children. The high frequency of the GC phenotype in pediatric patients was confirmed in a subsequent international study.59 That study also showed that pediatric patients had higher proliferation rates, greater c-MYC protein expression, and less BCL2 protein expression than is reported in adults.59 The authors reported that pediatric DLBCL, whose phenotype is similar to Burkitt lymphoma, appear to be characterized by c-Myc–driven proliferation, whereas adult DLBCL is characterized by BCL2 protein expression and associated inhibition of apoptosis.59 

Gene-expression profiling of the mediastinal large B-cell lymphoma (MLBCL) differentiates it from other DLBCLs and suggests that it is related to Hodgkin lymphoma. Adults with MLBCL have a better outcome than adults with other DLBCLs.

Conclusions and recommendations

The excellent outcome achieved in pediatric DLBCL using treatment designed for children with Burkitt lymphoma suggests that it is worthy of consideration for older adolescents and very young adults whose tumor is characterized by a pediatric phenotype. In contrast, for those whose tumor is characterized by an adult phenotype (i.e., BCL2+), a strategy that incorporates rituximab may be preferable. Clearly, additional clinical trials are needed to definitively answer these questions.

Pediatric treatment

The follicular lymphomas occur very infrequently in children and adolescents;60 however, when they do occur, questions often arise about the optimal treatment approach. Historically, many have been successfully treated with low-intensity CHOP-based regimens.61 Follicular lymphomas in children were found to differ biologically from those in adults. Those occurring in children are generally BCL2-negative; when BCL2-positive, they tend to be more advanced in stage and have a less successful treatment outcome.61 In some studies, children with completely resected limited-stage disease have been observed rather than being given systemic chemotherapy.62 

Adult regimens

The spectrum of treatment approaches for managing follicular lymphoma in adults is addressed in detail in the manuscript by Salles in this publication. Options include observation, involved-field irradiation, conventional-dose chemotherapy, high-dose chemotherapy with stem cell support, immunotherapy with rituximab, other new antibodies or radioimmunotherapy, alone and in combination with chemotherapy, depending on clinical stage and disease status.

Conclusions and recommendations

Close observation only may be considered for adolescents and very young adults with completely resected, BCL2, limited-stage disease, although mild systemic chemotherapy has often been given. For patients with BCL2 but incompletely resected limited-stage disease or BCL2+ disease, low-dose systemic chemotherapy is usually given. Involved-field irradiation is generally avoided in children. The role of rituximab in this age group has not been determined, but the agent is worthy of consideration for bulky, BCL2+ disease.

The non-ALCL mature T-cell lymphomas occur very infrequently in children.60 Therefore, there is very little information about their clinical and biologic features in various age groups. Historically, children and adolescents with these lymphomas have been treated on large-cell lymphoma regimens. Current treatment strategies are derived from those that have been successful in adults. Diagnoses that carry a particularly poor prognosis, such as hepatosplenic T-cell lymphoma, may require more intensive or novel frontline therapy. Additional clinical and biologic data on these tumors in both adults and children are needed before age-specific treatment recommendations can be made.

Age-related differences in the outcome of treatment for NHL reflect both host factors and tumor histology/biology factors. It is reasonable to consider pediatric strategies for some adolescents and very young adults with NHL. In fact, pediatric strategies are currently used to treat adults with certain subtypes of NHL (e.g., Burkitt lymphoma, lymphoblastic lymphoma). However, the use of pediatric strategies in the treatment of adults does not guarantee a comparable outcome, as illustrated above in the discusssion of lymphoblastic lymphoma. There is clearly a need for further biologic study of NHL in children, adolescents, and young adults. The tumor biology of ALCL and DLBCL has been shown to differ with age. Additional biologic data will not only help to improve prognosis and treatment stratification but, more importantly, will allow the identification of specific molecular targets for therapy.

Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN, and the University of Tennessee, Memphis, College of Medicine, Memphis, TN

Acknowledgments

Supported in part by a Grant from the National Cancer Institute (CA 21765) and by the American Lebanese Syrian Associated Charities (ALSAC).

1
Song K, Barnett M, Gascoyne R, et al. Primary therapy for adults with T-cell lymphoblastic lymphoma with hematopoietic stem-cell transplantation results in favorable outcomes.
Ann Oncol
.
2007
;
18
:
535
–540.
2
O’Leary M, Sheaffer, J, Keller, F, Shu, X-O, Cheson, B. Lymphomas and reticuloendothelial neoplasms. In: Bleyer A OLM, Barr R, Ries LAG, ed. Cancer Epidemiology in Older Children and Young Adults 15 to 29 Years of Age, Including SEER Incidence and Survival: 1975–2000. NIH pub. No. 06-5767. Bethesda, MD: National Cancer Institute;
2006
:25–38.
3
Burkhardt B, Zimmermann M, Oschlies I, et al. The impact of age and gender on biology, clinical features and treatment outcome of non-Hodgkin lymphoma in childhood and adolescence.
Br J Haematol
.
2005
;
131
:
39
–49.
4
Crom WR. Pharmacokinetics in the child.
Environ Health Perspect
.
1994
;
102
:
111
–117.
5
Relling MV, Pui CH, Sandlund JT, et al. Adverse effect of anticonvulsants on efficacy of chemotherapy for acute lymphoblastic leukaemia.
Lancet
.
2000
;
356
:
285
–290.
6
A predictive model for aggressive non-Hodgkin’s lymphoma. The International Non-Hodgkin’s Lymphoma Prognostic Factors Project.
N Engl J Med
.
1993
;
329
:
987
–994.
7
Relling MV, Hancock ML, Rivera GK, et al. Mercaptopurine therapy intolerance and heterozygosity at the thiopurine S-methyltransferase gene locus.
J Natl Cancer Inst
.
1999
;
91
:
2001
–2008.
8
Percy CL, Smith MA, Linet M, Gloeckler Ries LA, Friedman DL. Lymphomas and reticuloendothelial neoplasms. In: Ries LAG, Smith MA, Gurney JG, et al, eds. Cancer Incidence and Survival among Children and Adolescents: United States SEER Program 1975–1995. Bethesda, MD: National Cancer Institute, SEER Program. NIH Pub. No. 99-4649;
1999
:35–49.
9
Murphy SB, Fairclough DL, Hutchison RE, Berard CW. Non-Hodgkin’s lymphomas of childhood: an analysis of the histology, staging, and response to treatment of 338 cases at a single institution.
J Clin Oncol
.
1989
;
7
:
186
–193.
10
Link MP, Shuster JJ, Donaldson SS, Berard CW, Murphy SB. Treatment of children and young adults with early-stage non-Hodgkin’s lymphoma.
N Engl J Med
.
1997
;
337
:
1259
–1266.
11
Murphy SB, Hustu HO. A randomized trial of combined modality therapy of childhood non- Hodgkin’s lymphoma.
Cancer
.
1980
;
45
:
630
–637.
12
Reyes F, Lepage E, Ganem G, et al. ACVBP versus CHOP plus radiotherapy for localized aggressive lymphoma.
N Engl J Med
.
2005
;
352
:
1197
–1205.
13
Anderson JR, Jenkin RD, Wilson JF, et al. Long-term follow-up of patients treated with COMP or LSA2L2 therapy for childhood non-Hodgkin’s lymphoma: a report of CCG-551 from the Childrens Cancer Group.
J Clin Oncol
.
1993
;
11
:
1024
–1032.
14
Murphy SB, Bowman WP, Abromowitch M, et al. Results of treatment of advanced-stage Burkitt’s lymphoma and B cell (SIg+) acute lymphoblastic leukemia with high-dose fractionated cyclophosphamide and coordinated high-dose methotrexate and cytarabine.
J Clin Oncol
.
1986
;
4
:
1732
–1739.
15
Bowman WP, Shuster JJ, Cook B, et al. Improved survival for children with B-cell acute lymphoblastic leukemia and stage IV small noncleaved-cell lymphoma: a pediatric oncology group study.
J Clin Oncol
.
1996
;
14
:
1252
–1261.
16
Patte C, Philip T, Rodary C, et al. High survival rate in advanced-stage B-cell lymphomas and leukemias without CNS involvement with a short intensive polychemotherapy: results from the French Pediatric Oncology Society of a randomized trial of 216 children.
J Clin Oncol
.
1991
;
9
:
123
–132.
17
Reiter A, Schrappe M, Tiemann M, et al. Improved treatment results in childhood B-cell neoplasms with tailored intensification of therapy: a report of the Berlin-Frankfurt-Munster Group Trial NHL-BFM 90.
Blood
.
1999
;
94
:
3294
–3306.
18
Patte C, Auperin A, Michon J, et al. The Societe Francaise d’Oncologie Pediatrique LMB89 protocol: highly effective multiagent chemotherapy tailored to the tumor burden and initial response in 561 unselected children with B-cell lymphomas and L3 leukemia.
Blood
.
2001
;
97
:
3370
–3379.
19
Magrath I, Adde M, Shad A, et al. Adults and children with small non-cleaved-cell lymphoma have a similar excellent outcome when treated with the same chemotherapy regimen.
J Clin Oncol
.
1996
;
14
:
925
–934.
20
Patte C, Auperin A, Gerrard M, et al. Results of the randomized international FAB/LMB96 trial for intermediate risk B-cell non-Hodgkin lymphoma in children and adolescents: it is possible to reduce treatment for the early responding patients.
Blood
.
2007
;
109
:
2773
–2780.
21
Cairo MS, Gerrard M, Sposto R, et al. Results of a randomized international study of high-risk central nervous system B non-Hodgkin lymphoma and B acute lymphoblastic leukemia in children and adolescents.
Blood
.
2007
;
109
:
2736
–2743.
22
Soussain C, Patte C, Ostronoff M, et al. Small noncleaved cell lymphoma and leukemia in adults. A retrospective study of 65 adults treated with the LMB pediatric protocols.
Blood
.
1995
;
85
:
664
–674.
23
Thomas DA, Cortes J, O’Brien S, et al. Hyper-CVAD program in Burkitt’s-type adult acute lymphoblastic leukemia.
J Clin Oncol
.
1999
;
17
:
2461
–2470.
24
Thomas DA, Faderl S, O’Brien S, et al. Chemoimmunotherapy with hyper-CVAD plus rituximab for the treatment of adult Burkitt and Burkitt-type lymphoma or acute lymphoblastic leukemia.
Cancer
.
2006
;
106
:
1569
–1580.
25
Hummel M, Bentink S, Berger H, et al. A biologic definition of Burkitt’s lymphoma from transcriptional and genomic profiling.
N Engl J Med
.
2006
;
354
:
2419
–2430.
26
Dave SS, Fu K, Wright GW, et al. Molecular diagnosis of Burkitt’s lymphoma.
N Engl J Med
.
2006
;
354
:
2431
–2442.
27
Harris NL, Horning SJ. Burkitt’s lymphoma—the message from microarrays.
N Engl J Med
.
2006
;
354
:
2495
–2498.
28
Onciu M, Schlette E, Zhou Y, et al. Secondary chromosomal abnormalities predict outcome in pediatric and adult high-stage Burkitt lymphoma.
Cancer
.
2006
;
107
:
1084
–1092.
29
Reiter A, Schrappe M, Ludwig WD, et al. Intensive ALL-type therapy without local radiotherapy provides a 90% event-free survival for children with T-cell lymphoblastic lymphoma: a BFM group report.
Blood
.
2000
;
95
:
416
–421.
30
Amylon MD, Shuster J, Pullen J, et al. Intensive high-dose asparaginase consolidation improves survival for pediatric patients with T cell acute lymphoblastic leukemia and advanced stage lymphoblastic lymphoma: a Pediatric Oncology Group study.
Leukemia
.
1999
;
13
:
335
–342.
31
Hoelzer D, Gokbuget N, Digel W, et al. Outcome of adult patients with T-lymphoblastic lymphoma treated according to protocols for acute lymphoblastic leukemia.
Blood
.
2002
;
99
:
4379
–4385.
32
Thomas DA, O’Brien S, Cortes J, et al. Outcome with the hyper-CVAD regimens in lymphoblastic lymphoma.
Blood
.
2004
;
104
:
1624
–1630.
33
Le Gouill S, Lepretre S, Briere J, et al. Adult lymphoblastic lymphoma: a retrospective analysis of 92 patients under 61 years included in the LNH87/93 trials.
Leukemia
.
2003
;
17
:
2220
–2224.
34
Lones MA, Heerema NA, Le Beau MM, et al. Chromosome abnormalities in advanced stage lymphoblastic lymphoma of children and adolescents: a report from CCG-E08.
Cancer Genet Cytogenet
.
2007
;
172
:
1
–11.
35
Ferrando AA, Neuberg DS, Staunton J, et al. Gene expression signatures define novel oncogenic pathways in T cell acute lymphoblastic leukemia.
Cancer Cell
.
2002
;
1
:
75
–87.
36
Raetz EA, Perkins SL, Bhojwani D, et al. Gene expression profiling reveals intrinsic differences between T-cell acute lymphoblastic leukemia and T-cell lymphoblastic lymphoma.
Pediatr Blood Cancer
.
2006
;
47
:
130
–140.
37
Deangelo DJ. The treatment of adolescents and young adults with acute lymphoblastic leukemia.
Hematology Am Soc Hematol Educ Program
.
2005
:
123
–130.
38
Seidemann K, Tiemann M, Schrappe M, et al. Short-pulse B-non-Hodgkin lymphoma-type chemotherapy is efficacious treatment for pediatric anaplastic large cell lymphoma: a report of the Berlin-Frankfurt-Munster Group Trial NHL-BFM 90.
Blood
.
2001
;
97
:
3699
–3706.
39
Brugieres L, Deley MC, Pacquement H, et al. CD30(+) anaplastic large-cell lymphoma in children: analysis of 82 patients enrolled in two consecutive studies of the French Society of Pediatric Oncology.
Blood
.
1998
;
92
:
3591
–3598.
40
Murphy SB. Pediatric lymphomas: recent advances and commentary on Ki-1-positive anaplastic large-cell lymphomas of childhood.
Ann Oncol
.
1994
;
5
:
31
–33.
41
Laver JH, Kraveka JM, Hutchison RE, et al. Advanced-stage large-cell lymphoma in children and adolescents: results of a randomized trial incorporating intermediate-dose methotrexate and high-dose cytarabine in the maintenance phase of the APO regimen: a Pediatric Oncology Group phase III trial.
J Clin Oncol
.
2005
;
23
:
541
–547.
42
Brugieres L, Quartier P, Le Deley MC, et al. Relapses of childhood anaplastic large-cell lymphoma: treatment results in a series of 41 children—a report from the French Society of Pediatric Oncology.
Ann Oncol
.
2000
;
11
:
53
–58.
43
Morris SW, Xue L, Ma Z, Kinney MC. Alk+ CD30+ lymphomas: a distinct molecular genetic subtype of non-Hodgkin’s lymphoma.
Br J Haematol
.
2001
;
113
:
275
–295.
44
Tilly H, Gaulard P, Lepage E, et al. Primary anaplastic large-cell lymphoma in adults: clinical presentation, immunopheno-type, and outcome.
Blood
.
1997
;
90
:
3727
–3734.
45
Falini B, Pileri S, Zinzani PL, et al. ALK+ lymphoma: clinicopathological findings and outcome.
Blood
.
1999
;
93
:
2697
–2706.
46
Gascoyne RD, Aoun P, Wu D, et al. Prognostic significance of anaplastic lymphoma kinase (ALK) protein expression in adults with anaplastic large cell lymphoma.
Blood
.
1999
;
93
:
3913
–3921.
47
Hoskins PJ, Le N, Gascoyne RD, et al. Advanced diffuse large-cell lymphoma treated with 12-week combination chemotherapy: natural history of relapse after initial complete response and prognostic variables defining outcome after relapse.
Ann Oncol
.
1997
;
8
:
1125
–1132.
48
Shiota M, Nakamura S, Ichinohasama R, et al. Anaplastic large cell lymphomas expressing the novel chimeric protein p80NPM/ALK: a distinct clinicopathologic entity.
Blood
.
1995
;
86
:
1954
–1960.
49
Greer J P FJ, Kallianpur AR, Goodman SA, Stein RS, Whitlock JM, Wolff SN, Kinney MC. Ki-1 anaplastic large cell lymphoma (ALCL): correlation of clinical features with p80(NPM/ALK) expression and histology [abstract].
Blood
.
1996
;
88
:
224a
.
50
Suzuki R, Kagami Y, Takeuchi K, et al. Prognostic significance of CD56 expression for ALK-positive and ALK-negative anaplastic large-cell lymphoma of T/null cell phenotype.
Blood
.
2000
;
96
:
2993
–3000.
51
Dunphy CH, DeMello DE, Gale GB. Pediatric CD56+ anaplastic large cell lymphoma: a review of the literature.
Arch Pathol Lab Med
.
2006
;
130
:
1859
–1864.
52
Nasr MR, Laver JH, Chang M, Hutchison RE. Expression of anaplastic lymphoma kinase, tyrosine-phosphorylated STAT3, and associated factors in pediatric anaplastic large cell lymphoma: a report from the children’s oncology group.
Am J Clin Pathol
.
2007
;
127
:
770
–778.
53
Wahl AF, Klussman K, Thompson JD, et al. The anti-CD30 monoclonal antibody SGN-30 promotes growth arrest and DNA fragmentation in vitro and affects antitumor activity in models of Hodgkin’s disease.
Cancer Res
.
2002
;
62
:
3736
–3742.
54
Cerveny CG, Law CL, McCormick RS, et al. Signaling via the anti-CD30 mAb SGN-30 sensitizes Hodgkin’s disease cells to conventional chemotherapeutics.
Leukemia
.
2005
;
19
:
1648
–1655.
55
Woessmann W, Peters C, Lenhard M, et al. Allogeneic haematopoietic stem cell transplantation in relapsed or refractory anaplastic large cell lymphoma of children and adolescents—a Berlin-Frankfurt-Munster group report.
Br J Haematol
.
2006
;
133
:
176
–182.
56
Fisher RI, Gaynor ER, Dahlberg S, et al. Comparison of a standard regimen (CHOP) with three intensive chemotherapy regimens for advanced non-Hodgkin’s lymphoma.
N Engl J Med
.
1993
;
328
:
1002
–1006.
57
Alizadeh AA, Eisen MB, Davis RE, et al. Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling.
Nature
.
2000
;
403
:
503
–511.
58
Oschlies I, Klapper W, Zimmermann M, et al. Diffuse large B-cell lymphoma in pediatric patients belongs predominantly to the germinal-center type B-cell lymphomas: a clinicopathologic analysis of cases included in the German BFM (Berlin-Frankfurt-Munster) Multicenter Trial.
Blood
.
2006
;
107
:
4047
–4052.
59
Perkins SL. Are pediatric and adult diffuse large B-cell lymphoma the same disease?
Pediatr Blood Cancer
.
2006
;
46
:
847
.
60
Sandlund JT, Downing JR, Crist WM. Non-Hodgkin’s lymphoma in childhood.
N Engl J Med
.
1996
;
334
:
1238
–1248.
61
Lorsbach RB, Shay-Seymore D, Moore J, et al. Clinicopathologic analysis of follicular lymphoma occurring in children.
Blood
.
2002
;
99
:
1959
–1964.
62
Atra A, Meller ST, Stevens RS, et al. Conservative management of follicular non-Hodgkin’s lymphoma in childhood.
Br J Haematol
.
1998
;
103
:
220
–223.