Key Points

Epidemiology and Biological Characteristics

Relative to adult Hodgkin lymphoma, pediatric Hodgkin lymphoma has (1) Distinct features in relation to geographic distribution, gender, association with Epstein-Barr virus (EBV), and histological subtype; (2) Relatively higher incidence in developing countries and in males; (3) Nodular lymphocyte-predominant and mixed cellularity subtypes are more frequent.

Staging Evaluation

(1) All patients should have a history and physical examination; complete blood count; blood chemistries; upright posteroanterior and lateral thoracic radiographs; computed tomography (CT) of the neck, chest, abdomen, and pelvis; and functional nuclear imaging studies with 18fluoro-2-deoxyglucose (FDG) positron emission tomography (PET). (2) Bone marrow biopsies should be done in selected patients with clinical stage III/IV disease or B symptoms, although this is done less frequently with the use of FDG-PET imaging. (3) Staging laparotomy is rarely appropriate, but biopsy of specific sites with equivocal findings by clinical staging should be considered when results will alter therapy. (4) Two-thirds of children are stage I or II, and one-third have B symptoms.

Primary Therapy

(1) Risk-adapted therapy based on some of the following presenting features at diagnosis: B symptoms, mediastinal and peripheral lymph-node bulk, extranodal extension of disease to contiguous tissues, number of involved nodal regions, Ann Arbor stage, gender, and erythrocyte sedimentation rate (ESR). (2) Response to chemotherapy: (a) Treatment response is a profound prognostic factor. (b) Reduction in therapy may be based on rapidity and the degree of response to chemotherapy. (c) Augmentation of therapy may be based on lack of or insufficient response to chemotherapy. (3) Toxicity: (a) Children have a relatively increased risk, compared with adults, for long-term cardiopulmonary compromise, musculoskeletal growth impairment, and subsequent malignant neoplasms. (b) Late effects experience (e.g., second cancers, cardiopulmonary effects) from the legacy of archaic extended-field high-dose radiotherapy (RT) technique have limited bearing on modern clinical practice. (4) Chemotherapy: (a) Common regimens: (i) ABVE-PC (doxorubicin, bleomycin, vincristine, etoposide, prednisone, cyclophosphamide), (ii) VAMP (vincristine, doxorubicin, methotrexate, prednisone), (iii) OEPA (vincristine, etoposide, prednisone, doxorubicin), (iv) OEPA/COPDAC (cyclophosphamide, vincristine, prednisone, and dacarbazine added to OEPA), (v) BEACOPP (bleomycin, etoposide, doxorubicin added to COPP; see below), (vi) ABVD (doxorubicin, bleomycin, vinblastine, dacarbazine), (vii) Stanford V (mechlorethamine, doxorubicin, vinblastine, vincristine, bleomycin, etoposide, prednisone). (b) Historical regimens: (i) MOPP (mechlorethamine, vincristine, procarbazine, prednisone), (ii) COPP (MOPP substituting cyclophosphamide for mechlorethamine), (iii) COPP/ABV (hybrid regimen of COPP and ABVD without dacarbazine), (iv) OPPA (OEPA, but with procarbazine substituted for etoposide—historically used in girls). (c) Early-stage favorable disease most commonly uses ABVD (based on adolescent and young adult experience) or derivative chemotherapy, VAMP, or OEPA. (d) Intermediate- or high-risk disease uses dose-dense combinations: ABVE-PC, OEPA/COPDAC, or BEACOPP. (5) Radiotherapy: (a) Use of low-dose, involved-site RT following chemotherapy is response based. RT used for slowly responding, bulk, residual, or recurrent disease is the emerging standard across all stages. However, elimination of RT in favorable early-stage Hodgkin lymphoma with low-intensity chemotherapy is not yet a standard. (b) Ongoing concerns about the incremental toxicity of RT, particularly secondary cancers and cardiovascular disease, remains a stimulus for investigations of chemotherapy alone. (c) Shift to involved-site RT from traditional involved-field volumes. (d) Compared with adult practice, more RT is used in advanced stage while less RT is used in early stage. (e) Using modern RT techniques—including deep inspiration breath hold, intensity-modulated RT, and/or proton therapy to minimize the radiation dose to the organs at risk—represents best clinical practices. (6) Results: (a) Chemotherapy alone: long-term event-free survival (EFS) ranges from 85% to 95% in patients with localized favorable disease and 70% to 90% in those with advanced or unfavorable disease. (b) Incremental improvements in EFS with addition of involved-field RT are in the 5% to 10% range. Rapid response to suitably intensive chemotherapy predicts for negligible benefit from RT. (c) The subset of patients who relapse after initial therapy have a reasonably good but variable potential for long-term disease remission with second-line therapies.

Introduction

Given the high cure rates in treating children with Hodgkin lymphoma (HL) and their susceptibility to late effects, the ongoing challenge has been the development of less toxic therapy. In this regard, the progress in HL management in children has often presaged that in adult patients. While many similarities between pediatric and adult HL exist, there are several distinctions that suggest different biological processes, though this remains controversial. Moreover, there has been an increasing biological and therapeutic distinction made between young children versus “adolescents and young adults” (AYA). Differences associated with age include gender ratio, the proportion of patients with the most common histological subtypes, the underlying biology (e.g., the role of Epstein-Barr virus [EBV]), and the potential for cure. Pediatric HL has an excellent prognosis with higher survival rates. Given the high cure rates, HL, particularly in children, has provided much of the knowledge base we have about late toxicities of radiotherapy (RT) and cytotoxic chemotherapy. Because of the increased vulnerability of children to the adverse effects of therapy, the management of pediatric HL has led the way in the evolution of treatment strategies that consider both the toxicity and efficacy of therapy.

Historically, the desire to avoid the musculoskeletal hypoplasia that occurred following high-dose extended-field RT, and leukemogenesis and infertility associated with certain alkylating agents, led to a more rational combined-modality therapy away from the historical use of higher-dose, extended-field RT. Subsequent observations of cardiovascular dysfunction and increased risk of secondary cancers have played additional roles in modifying therapeutic approaches. The first generation of combined-modality therapy regimens used cycles of chemotherapy to replace a portion of the RT in laparotomy-staged children. Second- and third-generation regimens used doxorubicin- and etoposide-containing combinations to replace or reduce offending alkylating agents. Concurrent with advances in diagnostic imaging, surgical staging with laparotomy has been abandoned after demonstrating efficacy of the combined-modality treatment approach. At the same time, fluorodeoxyglucose–positron emission tomography/computed tomography (FDG-PET/CT) imaging, given its high specificity and sensitivity, has become a standard for staging and response evaluation, allowing for further individual tailoring of the overall intensity of therapy, especially as to use of RT and what regions to target radiotherapeutically.

In time, risk-adapted trials evolved that prescribed fewer cycles of multiagent chemotherapy and lower radiation doses and treatment volumes for patients with favorable clinical presentations. This has been further refined with response-adapted algorithms in which therapy is either augmented or reduced as assessed by initial response to systemic therapy, as a rapid response is an important prognostic factor. Importantly, this has allowed for elimination or further reduction of RT when HL is seen to have an early and complete response (CR) to initial systemic therapy. The definition of risk groups for disease stratification can vary in different trials and has changed with therapeutic advances.

Because of the spectrum of prognostic factors in pediatric HL, unique developmental and gender-related predispositions to therapy effects, and variable responsiveness to chemotherapy, no single treatment method is ideal for all patients. Contemporary treatment for children and adolescents with HL uses a risk-adapted and response-adapted approach that considers presenting risk features at diagnosis as well as response to initial chemotherapy. Therapy duration and intensity are selected to optimize the cure probability with minimal treatment-related morbidity. For instance, the Children's Oncology Group (COG) intermediate-risk group Phase III trial AHOD0031 was the first HL trial to demonstrate that a rapid interim response to chemotherapy would allow for the elimination of involved-field RT.

Consequently, the evolution of therapy for pediatric HL has served as a model for other cancers. In this respect, newer agents such as brentuximab vedotin, an anti-CD30 antibody, as well as nivolumab and pembrolizumab—anti-programmed death (PD)-1 and PD-1 ligand (PD-L1) checkpoint inhibitor immunotherapies—are being investigated given that overexpression of PDL1 and PDL2 on Reed-Sternberg cells, the malignant stem cell of HL, is a hallmark of the immune evasion that occurs in HL. A major focus of ongoing cooperative group investigations aims to identify subgroups of HL that may be treated with reduced volume and dose of irradiation or with chemotherapy alone. The concerns about RT are to a certain extent the legacy of the significant late complications (second malignancies, cardiopulmonary dysfunction, and musculoskeletal hypoplasia) observed following archaic extended-field high-dose RT techniques that have limited bearing on modern clinical practice. Nevertheless, children are more susceptible to late effects of therapy. Thus, the trends in clinical trials have been to strive to reduce or omit adjuvant RT despite many individual trials showing a small improvement in disease-free survival (DFS) from involved-field RT and even a meta-analysis showing a small survival advantage in early-stage patients. With better risk- and response-adapted therapies, there has been a general decline in RT for the upfront management of pediatric HL. RT remains important, however, to (1) complement chemotherapy when there is slowly responding, residual, or possibly initial bulky disease; (2) reduce chemotherapy intensity in selected patients for whom the predicted late toxicity risk for RT is small; and (3) help manage relapsed disease.

Etiology and Epidemiology

Pediatric HL accounts for 5% to 6% of childhood cancers and exhibits distinctive epidemiological features. The childhood form, which presents in patients younger than 12 years of age, is associated with a marked male predominance, increasing family size, and decreasing socioeconomic status. In high-income countries, HL is rarely diagnosed in children younger than 5 years of age. The young adult form, which presents in patients ages 12 to 34 years, is associated with a higher socioeconomic status in industrialized countries. Overall, the incidence is highest in developed countries (North America and Europe) and very rare in Asian populations. In childhood, however, some low- and middle-income regions have relatively higher incidences. In adolescents, the incidence between males and females is roughly equal, and most older adolescent patients are white. The risk for young adult HL decreases significantly with increased sibship size and birth order. Specifically, the risk of HL in young adults is lower in individuals with multiple older, but not younger, siblings. Histological subtypes also vary by age at presentation. Mixed-cellularity subtype (more commonly associated with EBV) is more common in HL of childhood and the elderly, whereas nodular sclerosing subtype is more frequently observed in adolescents and young adults.

An uncommon familial pattern of the disease is observed, which is a clinically underappreciated fact owing to the rarity of HL. The association of HL with inherited patterns of lymphoma is based on the observed concordance in primary relatives of AYA patients. One study reported a 7-fold increased risk of HL in offspring of an affected parent and an 11-fold increased risk in patients less than 37 years old with an affected sibling. Other epidemiological studies similarly have shown that first-degree siblings of HL patients have a 3- to 5-fold increased risk for HL that may be as high as a 9-fold increased risk in same-sex siblings, with one study showing that female same-sex siblings have the highest risk. Even stronger indirect evidence for a genetic basis to HL is the reported observation of a 99-fold increased risk in identical twins, but not in dizygotic twins. The nodular lymphocyte-predominant variant—thought to be more akin to a low-grade B cell non-Hodgkin lymphoma with a lengthy time to diagnosis and time to relapse—also has a familial pattern of inheritance in some instances, linked to mutation in the ataxia telangectasia gene. Increased risk for nodular lymphocyte-predominant HL has also been observed in individuals with autoimmune lymphoproliferative syndrome (ALPS), a disorder associated with defects in fas-mediated apoptosis. Moreover, genome-wide association studies (GWASs) are providing new insights into the genetic susceptibility to develop HL.

Pediatric HL exhibits epidemiological features similar to that seen with paralytic poliomyelitis. Delayed exposure to an infectious agent might increase the risk of the young adult form of HL, whereas early and intense exposure to an infectious agent might increase the risk for the childhood form of HL. Data also indicate an association between nursery school or daycare attendance and reduced risk of HL among young adults, supporting a model in which childhood exposure to common infections promotes maturation of cellular immunity. The presence of high-antibody titers to EBV, in situ hybridization evidence of EBV genomes in Reed-Sternberg cells, and EBV early RNA1 and 2 (EBER1 and EBER2) sequences provide evidence that enhanced activation of EBV may play a role in the development of HL. The incidence of EBV-associated HL varies by age, sex, ethnicity, histological subtype, regional economic level, and underlying immune function. Approximately 30% to 40% of patients with HL have associated EBV. In one series of childhood HL, EBV early RNA1 was expressed in the malignant Reed-Sternberg cells in 58% of cases. More specifically, an association with EBV is greater in populations of lower socioeconomic status, cases of mixed cellularity HL, and cases occurring in both young children and in the elderly. Finally, there is an increased incidence of EBV-associated HL in patients with primary immunodeficiency disorders, such as common variable immunodeficiency, as well as with secondary immunodeficiency (e.g., HIV infection and immunosuppression for solid organ transplantation).

As the treatment of HL has been a clinical laboratory for the late sequelae of therapy, including secondary cancers, these GWASs are providing clues that the same predisposition to HL is a risk factor for late complications. For instance, a genetic locus at chromosome 6q21 was observed in a cohort of patients receiving RT for HL as adolescents but not as adults in association with later development of secondary cancers. This risk locus was found to upregulate a transcriptional suppressor, PRDMI, for which 30% of HL survivors with this risk haplotype developed a second cancer within 30 years compared with only 3% for those without this risk haplotype.

Pathology and Pathways of Spread

The stem cell of HL is the Reed-Sternberg cell, which is a multinucleated giant activated B cell that has lost its “B-cell identity” characteristically expressed as CD30 and CD15 positive, while most often CD20 negative. It accounts for roughly 1% of the cellular mass of HL while the bulk of HL's microenvironment is reactive, inflammatory cells consisting of a mixture of lymphocytes, histiocytes, neutrophils, eosinophils, plasma cells, and fibroblasts. The latter may give rise to a characteristic nodular pattern of fibrosis in the common nodular sclerosing subtype of classical HL. Thus, when examining or imaging HL, one is measuring inflammatory cells—a totally unique situation among the broad spectrum of cancers. Nodular lymphocyte-predominant Hodgkin lymphoma (NLPHL) is a disease related to classical HL by histology and historical patterns of similar treatment (now changing). The malignant clone of NLPHL is a Reed-Sternberg-like B cell that is commonly CD20 positive on immunohistochemistry, while up to 30% of cases may be CD30 positive.

Beyond the scope of this chapter is the biology of the Reed-Sternberg cell, which is reviewed elsewhere. Accumulating evidence points to suppression of antitumor immune surveillance and apoptotic pathways via local production of cytokine and antiapoptotic proteins. Complex infiltration of T regulatory cells, constitutive expression of nuclear factor kappa B (NFκB), mTOR and JAK/STAT signal transduction pathways, immune tolerance, and upregulation of programmed death ligands such as PD-L1 and PD-L2 have been described that represent potential new therapeutic targets to be exploited in future clinical trials.

The pathological features of HL are similar in adults and children; however, the distribution of the histological subtypes defined by the World Health Organization may vary by age at presentation. NLPHL makes up almost 10% to 15% of pediatric cases.

Nodular sclerosing HL represents the most common histological subtype in pediatric cases under the rubric of classical HL, affecting approximately 70% of adolescents and children. Nodular sclerosing HL most commonly involves the lower cervical, supraclavicular, and mediastinal lymph nodes. The bulky growth of some involved nodal regions (particularly in the mediastinum) may be associated with persistent radiographic abnormalities even when the patient has fully responded to therapy. An additional classical form, mixed-cellularity HL, is observed in approximately 15% of patients, is more common in children aged 10 years or younger, and more frequently presents as advanced disease with extranodal involvement. Lymphocyte-depleted HL is rare in children but relatively more common in patients infected with human immunodeficiency virus (HIV). Lymphocyte-depleted disease in the HIV-positive patients is often associated with EBV. Lymphocyte-rich classical Hodgkin lymphoma (LRHL) makes up approximately 5% of all HL and closely overlaps with the nodular lymphocyte-predominant subtype in presenting clinical features and prognosis. The median age at presentation for LRHL (32 years) is, however, higher than for NLPHL, and there is a slightly higher incidence of mediastinal involvement and stage III disease at presentation.

Staging, Clinical Manifestations, Patient Evaluation, and Prognostic Factors

Staging

Physical examination and diagnostic imaging evaluations are used in pediatric patients to designate a clinical stage according to the Ann Arbor staging system, while in adult practice this has been modified by the Lugano staging classification of lymphomas. This historical staging system is based on anatomical groups of regional lymph nodes as delineated at the 1970 Ann Arbor symposium. It was subsequently revised at the Cotswolds meeting in 1989, although not all recommendations are in current use. This staging system persists to date with stage I identifying disease confined to one nodal region. Stage II signifies disease confined to two or more regions, but on one side of the diaphragm. Stage III indicates nodal disease that is on both sides of the diaphragm, while stage IV is diffuse or disseminated disease that includes extranodal sites. A special subset of stages I and II disease may be designated as extralymphatic or extranodal with the “E” suffix, which historically had been interpreted to be a disease distribution that could be encompassed within a reasonable radiation treatment portal. This operational definition is currently problematic, as the role of RT is being curtailed, but still is needed for prognostic purposes to designate early stage. Largely ignored from the Cotswold recommendations in current practice is the suffix “X” to designate bulky disease (> 10 cm maximum dimension) and a category of response to therapy, unconfirmed/uncertain complete remission (CR[u]), introduced to accommodate the difficulty of persistent radiological abnormalities of uncertain significance prior to the current use of metabolic imaging. PET imaging has become important for initial staging as well as for response assessment to therapy.

Lymphoma staging continues to be unique with the inclusion of prognostically significant constitutional symptoms. The absence of symptoms is labeled with the suffix “A” while “B” symptoms included in the staging assignment are unexplained fever with temperatures taken orally that are higher than 38°C, unexplained weight loss of 10% within 6 months preceding diagnosis, and drenching night sweats. At one point, pruritus had been included as a B symptom but was dropped many decades ago.

In the past, pathological staging, based on the findings of a staging laparotomy, including splenectomy, was routinely used to assess infradiaphragmatic disease. The increasing use of systemic therapy in children and the development of more accurate diagnostic imaging modalities eventually led to the routine use of clinical staging and abandonment of surgical staging except to assess any equivocal findings. Currently, surgical staging—most typically, nodal sampling without splenectomy—is pursued only if the anticipated findings will significantly alter the treatment plan.

Clinical Manifestations

Pediatric patients most commonly present with painless cervical or supraclavicular lymphadenopathy. Mediastinal lymphadenopathy occurs in up to two-thirds of patients and may be associated with a nonproductive cough or other symptoms of tracheal or bronchial compression. Axillary or inguinal lymphadenopathy is less frequently seen as the first presenting sign. Primary infradiaphragmatic disease is rare in pediatric patients, occurring in fewer than 5% of cases. Splenic involvement occurs in 30% to 40% of pediatric patients with HL, whereas hepatic involvement is exceedingly rare. The pulmonary parenchyma, chest wall, pleura, and pericardium are the most commonly involved extranodal sites of disease. Bone marrow involvement at the time of initial presentation of HL is also uncommon in children. Approximately 65% of children have stages I and II disease and 35% have stages III and IV disease ( Table 85.1 ).

TABLE 85.1
Pediatric Hodgkin Lymphoma: Demographic and Clinical Characteristics at Presentation
Children a , b
No. (%)
N = 1985
Children c
No. (%)
N = 2836
Adults c
No. (%)
N = 18898
Adults b
No. (%)
N = 1912
Total Patients
<10 y 360 (18) 312 (11)
≥10 y 1625 (82) 2524 (89) 18,898 (100) 1912 (100)
Gender
Male 1100 (55) 1455 (51) 10,330 (55) 1147 (60)
Female 885 (45) 1381 (49) 8568 (45) 765 (40)
Histology
Lymphocyte predominant 192 (10) 177 (6.3) 1224 (6.5) 96 (5.0)
Lymphocyte depleted 8 (0.3) 321 (1.7) 115 (6.0)
Mixed cellularity 307 (16) 284 (10) 3176 (17) 325 (17)
Nodular sclerosing 1431 (72) 2142 (76) 11,583 (61) 1377 (72)
Not classified 55 (2.8) 225 (7.9) 2,594 (14)
B Symptoms
Present 564 (28) 863 (39) 6,477 (48) 612 (32)
Absent 1421 (72) 1337 (61) 7012 (52) 1300 (68)
Stage d
I 229 (12) 522 (19) 4208 (23) 210 (11)
II 1078 (54) 1337 (49) 7021 (39) 899 (47)
III 391 (20) 518 (19) 3569 (20) 593 (31)
IV 287 (15) 366 (13) 3,156 (18) 210 (11)

a Data taken from Ruhl et al. and Nachman et al.

b Data taken from Cleary et al.

c Data taken from Bazzeh et al.

d Data derived from both pathologically and clinically staged patients.

Nonspecific systemic symptoms are often associated with lymphadenopathy and may include fatigue, anorexia, mild weight loss, and pruritus. The prognostically significant B symptoms have already been defined earlier. B symptoms occur in approximately 33% of patients (see Table 85.1 ). Laboratory changes observed at presentation are nonspecific but may provide clues about the extent of disease. Hematological abnormalities may include anemia, neutrophilic leukocytosis, lymphopenia, eosinophilia, and monocytosis. Anemia may be associated with the presence of advanced disease and may result from impaired mobilization of iron stores or, less commonly from, hemolysis. Several autoimmune disorders have been reported in patients with HL, including nephrotic syndrome, autoimmune hemolytic anemia, autoimmune neutropenia, immune thrombocytopenia, and autoimmune lymphoproliferative disorder. These conditions typically remit as the lymphoma responds to therapy. Several acute-phase reactants—including erythrocyte sedimentation rate, serum copper, ferritin, and C-reactive protein levels—may be elevated at diagnosis and useful in follow-up evaluations.

Patient Evaluation

Posteroanterior and lateral thoracic radiographs should be performed as soon as HL becomes part of the differential diagnosis to assess mediastinal involvement, airway patency, and other intrathoracic structures. This is particularly important if sedation is planned for diagnostic procedures. Either an excisional lymph node biopsy or core needle biopsy is the preferred diagnostic procedure (avoiding cytological fine needle aspirates), because it permits evaluation of the malignant Hodgkin Reed-Sternberg cells within the background of characteristic architectural changes of the specific histological subtypes. All nodal regions, including Waldeyer's ring, should be assessed by careful physical examination. Historically, an upright chest radiograph was also important to assess mediastinal bulk that is defined by mediastinal lymphadenopathy measuring 33% or more of the maximum intrathoracic cavity. Whether this definition for bulk mediastinal adenopathy may be replaced by CT-based mass dimensions or volumes is currently under debate. CT is most frequently used to evaluate the nodal regions in the neck, axilla, thoracic and abdominal cavities, and pelvis. Administration of both oral and intravenous contrast agents is required for CT to accurately distinguish lymphadenopathy from other infradiaphragmatic structures. Organ size is an unreliable indicator of lymphomatous involvement in the liver or spleen because tumor deposits may be less than 1 cm in diameter and not visualized by diagnostic imaging modalities. Increased size of either organ can, of course, be caused by nonmalignant conditions. The presence of hypodense lesions by CT and/or abnormal functional avidity PET scanning provides stronger evidence of tumor infiltration in these organs.

Functional nuclear-imaging studies are appropriately used in patients with HL as a diagnostic and monitoring modality. PET scanning uses uptake of the radioactive glucose analogue, FDG, as a correlate of tumor activity. PET scanning is now widely available and has become a standard part of the staging workup and assessment of response to therapy. Fused PET/CT offers the advantage of integrating functional and anatomic tumor characteristics. Residual or persistent FDG avidity appears to be useful in predicting prognosis and the need for additional therapy in posttreatment evaluation. Moreover, PET may be useful in evaluating abnormalities that become clinically manifest or appear on imaging in order to assess recurrence. PET use for follow-up is not recommended, as other reports suggest low rates of diagnosing relapsed disease and a high degree of false-positive findings. Because extranodal disease involving the bones and bone marrow is relatively uncommon in children, these staging evaluations can be omitted in patients presenting with localized and asymptomatic disease. Bone pain should be evaluated with plain radiographs; Magnetic resonance imaging (MRI) may also be necessary in this situation for questions about focal bone involvement. PET scanning seems to be supplanting the need for technetium-99 bone scans for skeletal evaluation, which historically have also been performed when there was an elevated serum alkaline-phosphatase concentration beyond that expected for age or extranodal disease identified by other staging evaluations. Although bone marrow involvement is relatively rare in pediatric HL, bilateral bone marrow biopsy had historically been recommended in any patient with clinical stage III or IV disease or B symptoms. Yet, a meta-analysis of nine clinical studies, including both pediatric and adult patients, showed that PET/CT achieved high sensitivity (96.9%) and high specificity (99.7%) in detecting bone marrow involvement in newly diagnosed patients with HL. Thus, this standard recommendation has been revised by European investigators when PET imaging shows no bone or bone marrow involvement. Because the pattern of infiltration in the bone marrow may be diffuse or focal and is often accompanied by reversible marrow fibrosis, a bone marrow aspirate alone is inadequate to assess the marrow for disease.

Prognostic Factors

The identification of prognostic factors has taken on importance as a determinant of risk- and response-adapted treatment algorithms. Many prognostic factors have been identified from adult trials. Regardless, this effort is complicated by several concepts. First, prognostic factors are in flux as more effective or higher-intensity therapy may negate adverse risk factors previously demonstrated. Second, various risk stratification schemes have been used by different institutions and cooperative groups to allocate patients to various treatment regimens, making comparisons of patient populations challenging. Most data are based on reports that primarily include adults. While there is overlap in the biology and response to therapy of pediatric and adult forms of HL, there is divergence in prognostic groupings just as there are differences in treatment regimens. Even among pediatric protocols and cooperative groups, there is a lack of standardization in prognostic factors ( Table 85.2 ). Third, there is an evolving understanding that early response to systemic therapy may be an important prognostic factor that may be used to guide further treatment decisions. While this concept is under investigation in adults with HL, one may acknowledge that response-based therapy has recently developed firm roots in pediatric HL treatment paradigms. Thus, there is much variability in classifications of Hodgkin patients into favorable, intermediate, and unfavorable/advanced strata.

TABLE 85.2
Risk Stratification in Pediatric Hodgkin Lymphoma Cooperative Group Clinical Trials
Adapted from Kelly KM. Management of children with high-risk Hodgkin lymphoma. Br J Haematol 2012;157(1):3-13.
Trial Low Risk Intermediate Risk High Risk
Children's Oncology Group
AHOD1331 (high risk) IIB bulk, IIIB, IVA/B
AHOD0431 (low risk); AHOD0031 (intermediate risk); AHOD0831 (high risk) IA, IIA with no bulk IA bulk or E; IB; IIA bulk or E; IIB; IIIA, IVA IIIB, IVB
C5942 IA, IB, IIA with no bulk, no hilar nodes and < 4 sites IA, IB, IIA with bulk, hilar nodes or ≥ 4 sites; III IV
C59704 (high risk) IIB/IIIB with bulk, IV
P9425/P9426 (106) IA, IIA with no bulk IB, IIA, IIIA 1 with bulk; IIIA 2 IIB, IIIB, IV
German Multicenter/EuroNet
GPOH-HD 95; GPOH-HD 2002; EuroNet-PHL-C1 a ; EuroNet-PHL-C2 b , IA/B, IIA I E A/B; II E A; IIB; IIIA (risk factors: ESR ≥ 30 mm/h or bulk ≥ 200 mL) II E B; III E A/B; IIIB; IV
Stanford/St. Jude/Dana-Farber Cancer Institute Consortium
HOD08 c (low risk); HOD05 d (intermediate risk); HOD99 e (high risk) IA, IIA with no bulk, E and < 3 sites IB, IIIA, IA/IIA with E, ≥ 3 sites or bulk IIB, IIIB, IV
E, Extralymphatic; stage IIIA 1 , minimal splenic, splenic hilar, or celiac involvement; stage IIIA 2 , massive splenic or lower abdominal node involvement.

a ClinicalTrials.gov identifier: NCT00433459

b ClinicalTrials.gov identifier: NCT02797717

c ClinicalTrials.gov identifier: NCT00846742

d ClinicalTrials.gov identifier: NCT00352027

e ClinicalTrials.gov identifier: NCT00846742

Dating from the time that early-stage HL was treated with subtotal or total nodal irradiation that included splenic irradiation—often termed extended-field RT—numerous adverse prognostic factors in stage I–II disease have identified those patients who benefit from combined-modality therapy. In pediatric management, extended-field RT was abandoned in favor of primary chemotherapy and reduced-dose involved-field RT in the 1970s and 1980s in order to reduce late effects of full-dose extended-field RT. This switch in adult management of early-stage disease did not occur until the mid-1990s. Regardless, the concept of risk-adapted therapy was first developed in which the presence of poor prognostic factors drives more intensive therapy. At the same time, favorable factors identify a population appropriately treated with less intensive therapy designed to maintain high cure rates with fewer acute and late side effects. Prognostic factors identified in these analyses include the number of involved lymphoid regions, size of individual nodes, extent of mediastinal disease, patient gender and age, presence of B symptoms or pruritus, histology, ESR, and overall tumor burden as measured by number of sites and disease bulk.

Even for pediatric management, as there are efforts to develop new protocols for AYA patients, it is relevant to note that the adult cooperative groups have refined favorable versus unfavorable groupings. For instance, the European Organization for Research and Treatment of Cancer (EORTC) and Groupe d'Etudes des Lymphomes de l'Adulte (GELA) specify the following as unfavorable factors: age greater than 50 years, ESR greater than or equal to 50 in the absence of B symptoms, ESR greater than or equal to 30 with B symptoms, and greater than or equal to 4 sites of involvement or bulky mediastinal involvement. For the German Hodgkin Lymphoma Study Group (GHSG), the following are considered unfavorable factors: ESR greater than or equal to 50 in the absence of B symptoms; ESR greater than or equal to 30 with B symptoms; and greater than or equal to 3 sites of involvement, extranodal involvement, or bulky mediastinal mass.

For pediatric practice, adverse prognostic features have included advanced stage, B symptoms, extranodal extension, peripheral or mediastinal bulky disease, hilar adenopathy, and 3 or more nodal regions. Based on the most recent intermediate-risk COG protocol AHOD0031, a preliminary prognostic scoring system has been proposed, with the acronym CHIPS for Childhood Hodgkin International Prognostic Score. Based on a multivariate analysis, four predictors—stage IV, large mediastinal adenopathy, albumin less than 3.5, and fever—were identified as predictive of adverse event-free survival (EFS). This simple scoring system needs further validation, particularly as treatment algorithms change and prognostic factors may change and become more difficult to identify. For the present, several prognostic factors continue to influence the success and choice of therapy for pediatric practice:

  • 1.

    Stage of disease: Stage persists as the most important prognostic variable. Patients with advanced-stage disease, especially stage IV, have a poorer outcome than patients with early-stage disease.

  • 2.

    B symptoms: These constitutional symptoms likely result from cytokine secretion and correlate with biological aggressiveness. Thus, unexplained fevers, drenching night sweats, or significant weight loss with definitions noted earlier in the Ann Arbor staging classification system continue to have prognostic importance and, in turn, influence management decisions. The presence of B symptoms has correlated with a higher likelihood of systemic disease, including occult subdiaphragmatic disease when staging laparotomies were once performed. Evidence suggests that fevers and weight loss have more prognostic significance than night sweats alone. The statistical analysis that generated CHIPS for intermediate-risk Hodgkin lymphoma, however, suggested that only fevers had prognostic significance.

  • 3.

    Bulk : The bulk of disease combines the number of disease sites and the volume of involvement at each site. Patients with several sites of involvement, defined variably as either three or four or more sites of disease, fare less well. This prognostic factor may also influence treatment selection as to the reduction or elimination of RT after primary chemotherapy for patients with early-stage HL. Moreover, the presence of large mediastinal adenopathy or bulky disease in nonmediastinal sites has been another consistent risk factor. A variety of definitions of large mediastinal adenopathy have been reported in the literature. The most commonly used definition is based on a measurement of the maximum width of the mediastinal mass on standing posteroanterior (PA) chest radiograph compared with the maximum intrathoracic diameter. A ratio greater than one-third is defined as “bulky.” Other reports have used a ratio with the intrathoracic width at T5–6 as the denominator, while still others use absolute measurements, surface area calculations, or volume measurements. In the absence of a chest radiograph, CT-based measurements of mediastinal masses have not reached a consensus recommendation, but some commentators have suggested that a 10-cm maximal diameter may be reasonable. Bulky disease in nonmediastinal sites has similarly been classified with varying definitions. Some protocols define bulky as greater than or equal to 10 cm, while others use greater than or equal to 5 cm, greater than or equal to 6 cm, and even greater than or equal to 7 cm. In Europe, the EuroNet Pediatric Hodgkin Lymphoma Group uses a volumetric definition of bulk. This was initially assessed by a definition of 50 mL, but subsequent experience has found the 200-mL volume definition to be more prognostically discriminatory and is now used in their EuroNet PHL C2 trial to stratify patients. Moreover, volume of a given site of disease is calculated by a simple formula estimating an ellipsoid: V = (xyz)/2, where x, y, and z are the diameters of the mass in three dimensions. This becomes much more complex when measuring mediastinal masses for which adding multiple ellipsoids of disease to avoid counting normal structures is the procedure. Moreover, FDG-PET at diagnosis allows for novel three-dimensional (3D) measures of disease burden with parameters of metabolic tumor burden and total glycolysis uptake that are proving to have prognostic importance beyond traditional anatomic definitions of bulk.

  • 4.

    Laboratory studies , including the ESR, hemoglobin level, and serum albumin, have been reported to predict worse outcomes. This could reflect disease biology or bulk. A recent low-risk pediatric HL trial demonstrated that the ESR has prognostic importance. As already noted for adult cooperative group trials, the ESR has been an important stratification factor in treatment protocols. Albumin is a key component of CHIPS. Moreover, both low hemoglobin and albumin levels have been found to be important factors in an International Prognostic Score, which has been validated only for patients older than 15 years.

  • 5.

    Histological subtype has importance. As already noted, patients with NLPHL are biologically different and generally have improved DFS and overall survival (OS) relative to classical HL. Separate approaches with minimal therapy have been demonstrated for early-stage NLPHL patients. Patients with lymphocyte depleted Hodgkin lymphoma (LDHL) fare poorly. Mixed reports suggest better or poorer outcome of other histologies that may be related to other prognostic factors as well. Several reports suggest that mixed-cellularity subtype of classical HL may have better prognosis than nodular sclerosis subtype in the pediatric age group.

  • 6.

    Age is a significant factor, with survival rates for children with HL approaching 85% to 95% and higher than adults stage for stage. In a report from Stanford, the 5- and 10-year survival rates for children with HL less than or equal to 10 years of age was 94% and 92%, respectively, compared with 93% and 86% for adolescents (aged 11 to 16 years old) and 84% and 73% for adults, respectively. Differences in treatment approaches may play a role in these differential outcomes, as higher PFS rates have been observed in patients with intermediate-risk HL treated on pediatric versus adult protocols.

  • 7.

    Rapidity of response to initial chemotherapy is an important prognostic variable. Early response to therapy was initially observed in advanced-stage HL patients treated in the Pediatric Oncology Group (POG) 8725 trial, where 93% of patients who attained a CR after 3 cycles of chemotherapy remained disease free. This was also confirmed for lower-stage patients and afterwards incorporated into the latest (COG) frontline trials. Early CR to therapy has also been successfully incorporated into the German trials with low-risk patients who achieve CR after 2 cycles of OEPA not requiring further RT. Response-based therapy is currently the paradigm on which modern pediatric trials are based in North America and Europe, tailoring treatment intensity to interim response based on PET/CT.

Primary Therapy

Risk-Adapted Treatment Approach

Contemporary treatment for children and adolescents with HL involves a risk-adapted—and now, a response-adapted—approach based on the patient's presenting features at diagnosis and reevaluation after one or two cycles of chemotherapy. Factors included in the risk assessment may vary across studies, but they most often include the presence of B symptoms, mediastinal and peripheral lymph node bulk, extranodal extension of disease to contiguous structures, number of involved nodal regions, Ann Arbor stage, and gender. A favorable clinical presentation is typically characterized as localized (stage I/II) nodal involvement in the absence of B symptoms and bulky disease. While the historical definition of mediastinal bulk has been based on the ratio greater than one-third between the transverse dimension of the mediastinal mass to the intrathoracic cavity on an upright chest radiograph, some trials have moved to using a simple size criterion on cross-sectional imaging as employed for peripheral lymph node bulk. That definition, however, is highly variable across studies, as already noted, ranging from a 4- to 10-cm diameter as a minimal threshold while European trials use a more rigorous volume classification. Moreover, there is additional subjectivity in the definition of bulk when there are multiple matted or adjacent nodes, contributing to confusion on such risk stratification in practice. Fewer than three or four involved nodal regions are considered favorable. In some risk-adapted treatment protocols, patients with localized disease presenting with unfavorable features are designated intermediate in risk and treated similarly to those with advanced-stage disease; in others, a therapy intermediate in intensity is prescribed. The criteria for unfavorable clinical presentations has also differed among studies, but most often it is comprised of the presence of B symptoms, bulky lymphadenopathy, hilar lymphadenopathy, more than 3 to 4 involved nodal regions, extranodal extension to contiguous structures, or advanced-stage (IIIB-IV) disease. The results of contemporary trials indicate that children and adolescents with early-stage or favorable presentations of HL are excellent candidates for reduced therapy. Recent trials evaluated whether intensification of therapy improves outcomes in patients with intermediate- and high-risk presentations.

Although not widely used to guide therapy assignment in pediatric trials, other factors such as gender, age at diagnosis, and histology are most definitely considered in individual patients. The trials organized by the German-Austrian Pediatric Oncology Group (GPOH) and one trial by the Children's Cancer Group (now integrated into the COG) have been unique in their aims to prospectively evaluate gender-based therapy. Long-term follow-up of the GPOH 90 and 95 studies demonstrate that the substitution of etoposide for procarbazine in the vincristine, prednisone, procarbazine, and doxorubicin (OPPA) regimen does not compromise DFS and provides less potential risk for gonadal toxicity. Although age at presentation has not been used as a criterion to assign therapy in prospective trials, reports describing outcomes after treatment with chemotherapy alone stress the benefits of this approach in younger children at higher risk of radiation-related toxicity.

Prognostic groupings now dictate the intensity of therapy with the goal of matching up just enough cytotoxic therapy to yield a high chance for cure while minimizing those exposures responsible for late complications. Within a recent cycle of COG trials from 2002 to 2011, patients were allocated into a favorable group if they had stage IA or IIA disease without bulk. Unfavorable patients were those with stage IIIB or IVB disease. The heterogenous group of patients in between were deemed intermediate risk (see Table 85.2 ). The current COG trials beginning in 2014 categorize patients into early favorable (stage I/II, no bulky disease), early unfavorable (I/IIA with bulk, IB and IIB no bulk) and high-risk strata (IIB with bulk, IIIB, IVA, and IVB). In Europe, the GPOH has merged into a geographically broader EuroNet cooperative group in which patients have been categorized by early, intermediate, or advanced treatment groups (TGs) as follows :

  • 1.

    TG-1: Patients of stages IA/B and IIA without bulk (< 200 mL) and without ESR (< 30 mm/h)

  • 2.

    TG-2: Patients of stages IEA/B, IIEA, IIB or IIIA and patients of stages IA/B and IIA with bulk (≥ 200 mL) and/or ESR (≥ 30 mm/h)

  • 3.

    TG-3: Patients of stages IIEB, IIIEA/B, IIIB or IVA/B

Finally, a collaborative group of investigators from St. Jude, Stanford, and Harvard have defined a favorable early-stage cohort that may be treated with minimal therapy with stage I-IIA disease without mediastinal bulk, no extranodal disease, and less than 3 sites of disease.

A summary of recent trials and results in children with early- and intermediate/advanced-stage HL is provided in Tables 85.3 and 85.4 .

TABLE 85.3
Treatment Results for Favorable Pediatric Hodgkin Lymphoma
Group, Years of Enrollment Patients (n) Stage Chemotherapy Radiation (Gy), Field Survival (%) Follow-Up Interval, (Years)
DFS, EFS, or RFS Overall
Euronet-PHL-C1
2007-2013
787
856
I/II 2 OEPA If CR, no RT
< CR 19.8 IF ± 10 IS
86
87

3
COG AHOD0431
2006-2009
175
100
I/II 3 AV-PC If CR, no RT
If PR, 21 IF
77.5
82.8
99.6 4
Stanford-St. Jude-Dana Farber
Consortium a ,
1990-2000
49
61
I/II† 4 VAMP If CR, 15 IF
< CR, 25.5 IF
95.2
84.5
100
93
10
Stanford-St. Jude-Dana Farber
Consortium a ,
2000-2008
47
41
I/II† 4 VAMP If CR, no RT
< CR, 25.5 IF
89.4
87.5
100
100
5
CCG5942 a ,
1995-1998
109
94
IA/B, IIA 4 COPP/ABV 21 IF 97
100
100
3 (as randomized)
10 (as treated)
106
113
IA/B, IIA 4 COPP/ABV None 92
89.1
100
3 (as randomized)
10 (as treated)
GOPH-HD-2002
2002-2005
62
126
1A/B, IIA 2 OEPA (boys) or
2 OPPA (girls)
If CR, no RT
< CR, 19.8-35 IF
93.2
91.7
100
100
5
POG 9426 a ,
1996-2000
112
135
I-IIIA 2 ABVE if RER
4 ABVE if SER
25.5 IF 87.3
85.4
97.1
95.9
8
POG-9226 a ,
1992-1993
51 I-IIIA 4 DBVE 25.5 IF 91 98 6
POG-8625 a ,
1986-1992
81 I-IIIA 4 MOPP/ABVD 25.5 IF 91 98 8
ABVD, Adriamycin, bleomycin, vinblastine, and dacarbazine; CCG, Children's Cancer Group; COP(P), cyclophosphamide, Oncovin, prednisone, and procarbazine; COPP/ABV, cyclophosphamide, Oncovin, procarbazine, prednisone/Adriamycin, bleomycin, vinblastine; DBVE, doxorubicin, bleomycin, vincristine, and etoposide; DFS, disease-free survival; EF, extended-field; EFS, event-free survival; IF, involved-field; MOPP, nitrogen mustard, Oncovin, procarbazine, and prednisone; OEPA, Oncovin, etoposide, prednisone, Adriamycin; OPA, Oncovin, prednisone, Adriamycin; OPPA, Oncovin, procarbazine, prednisolone, and Adriamycin: RFS, relapse-free survival; VAMP, vinblastine, doxorubicin, methotrexate, and prednisone.

a Denotes study population results that include lymphocyte-predominant HL (NLPHL) patients.

TABLE 85.4
Treatment Results for Intermediate and Advanced Stage Pediatric Hodgkin Lymphoma
Group, Years of Enrollment Patients (n) Stage Chemotherapy Radiation (Gy), Field Survival (%) Follow-Up Interval (y)
DFS, EFS or RFS Overall
COG-AHOD0831
2009-2011
165 IB, IVB 5 ABVE-PC 21 risk-adapted IS 80.2 95.9 4
COG-AHOD0031
2002-2009
361 I/IIA with bulk, I/IIB, IIIA, IVA RER: 4 ABVE-PCSER: 4 ABVESER: 4 ABVE + 2 DECA No RT 84.3 98.8 4
355 21 IF 87.9 98.8
151 21 IF 75.2 94.3
153 79.3 96.5
GPOH-HD-2002
2002-2005
139 I E , IIB, II E A/B, 2 OEPA/OPPA + 2 or 4 COPP/COPADC 19.8 IF ± 15.2 IS 88.3 99.5 5
239 III E A/B, IIIB, IV 86.9 94.9
CCG 59704 a ,
1999-2002
99 IIB or IIIB with bulk, IV All: 4 BEACOPP then
(1) RER: Female:
4 COPP/ABV
(2) RER: Male:
2 ABVD
(3) SER:
4 BEACOPP
(1) None
(2) 21 IF
(3) 21 IF
(< CR: boost 14 Gy)
94 97 5
POG-9425
1997-2001
216 IB, IIA/IIIA 1 with bulky mediastinum or IIIA 2 , IIB/IIIB/IVB 3 ABVE-PC for RER
5 ABVE-PC for SER
21 IF
21 IF
86
83
95
95
5
CCG-5942 a ,
1995-1998
109
33
IIB, III
IV
6 COPP/ABVCOPP/ABV + CHOP + Ara-C/VP-16 21 IF
21 IF
87
84
95
3
10
81
79.9
94
3
10
GPOH HD-95
1995-2001
2114327356 I E , IIB, II E A/B,III E A/B, IIIB, IV 2 OEPA/OPPA + 2 or 4 COPP/COPADC <CR, 19.8 IF ± 15.2 IS 91.4 98.1 10
If CR, no RT 68.5 97.7
19.8 IF ± 15.2 IS 88.7 95.3
If CR, no RT 88.2 100
POG-8725 a ,
1987-1992
80 IIB, IIIA 2 , IIIB, IV 4 MOPP/4 ABVD 21 EF 80 87 5
81 4 MOPP/4 ABVD none 79 96 5
CCG 521
1986-1990
54 III/IV 6 ABVD 21 EF 87 90 4
57 12 MOPP/ABVD none 77 84
ABVD, Adriamycin, bleomycin, vinblastine, and dacarbazine; ABVE-PC, Adriamycin, bleomycin, vincristine, etoposide, prednisone, and cyclophosphamide; BEACOPP, bleomycin, etoposide, doxorubicin, cyclophosphamide, vincristine, procarbazine, and prednisone; CCG, Children's Cancer Group; COP(P), cyclophosphamide, Oncovin, prednisone, and procarbazine; COPP/ABV, cyclophosphamide, Oncovin, procarbazine, prednisone/Adriamycin, bleomycin, and vinblastine; CR, complete response; DFS, disease-free survival; EF, extended-field; EFS, event-free survival; EVAP/ABV, etoposide, vinblastine, cytarabine, cisplatin/Adriamycin, bleomycin, and vincristine; IF, involved-field; MOPP, nitrogen mustard, Oncovin, procarbazine, and prednisone; OEPA, Oncovin, etoposide, prednisone, and Adriamycin; OPA, Oncovin, prednisone, and Adriamycin; OPPA, Oncovin, procarbazine, prednisolone, and Adriamycin; POG, Pediatric Oncology Group; PR, partial response; RFS, relapse-free survival; RER, rapid early response; SER, slow early response.

a Denotes study population results that include lymphocyte-predominant HL (NLPHL) patients.

You're Reading a Preview

Become a Clinical Tree membership for Full access and enjoy Unlimited articles

Become membership

If you are a member. Log in here