Treatment of Hodgkin lymphoma is associated with 2 major types of risk: that the treatment may fail to cure the disease or that the treatment will prove unacceptably toxic. Careful assessment of the amount of the lymphoma (tumor burden), its behavior (extent of invasion or specific organ compromise), and host related factors (age; coincident systemic infection; and organ dysfunction, especially hematopoietic, cardiac, or pulmonary) is essential to optimize outcome. Elaborately assembled prognostic scoring systems, such as the International Prognostic Factors Project score, have lost their accuracy and value as increasingly effective chemotherapy and supportive care have been developed. Identification of specific biomarkers derived from sophisticated exploration of Hodgkin lymphoma biology is bringing promise of further improvement in targeted therapy in which effectiveness is increased at the same time off-target toxicity is diminished. Parallel developments in functional imaging are providing additional potential to evaluate the efficacy of treatment while it is being delivered, allowing dynamic assessment of risk during chemotherapy and adaptation of the therapy in real time. Risk assessment in Hodgkin lymphoma is continuously evolving, promising ever greater precision and clinical relevance. This article explores the past usefulness and the emerging potential of risk assessment for this imminently curable malignancy.

Over the past 60 years, continuous improvement in the management of Hodgkin lymphoma has brought clinicians and patients to an era in which the large majority of patients are cured, regardless of disease presentation.1-7  This improvement in outcome has brought a new obligation to those who wish to optimally manage this previously lethal malignancy: an obligation to maintain very high cure rates while simultaneously minimizing toxicity, especially persistent late toxicity, which may permanently reduce the quality of life of survivors or even cause their death. A fine balance must be maintained in which maximal effectiveness of treatment, which presently is built around multiagent chemotherapy and judicious use of radiation, is maintained while minimizing exposure to interventions associated with major late toxicity. In brief, clinicians must recommend just enough treatment to achieve the greatest efficacy and yet induce the least harm. Careful assessment of risk is an essential part of achieving this balance. Such risk assessment must, in turn, address multiple factors, of which some are intrinsic to the host, others are related to tumor burden and tumor biology, and lastly, several are evaluable at diagnosis and determinable as the treatment course unfolds (Figure 1). Full appreciation of important factors that increase the risk of treatment failure or the likelihood of undesirable, potentially avoidable acute or late toxicity and how these risks can be minimized is essential to optimal management of Hodgkin lymphoma today. This review examines these risk factors and identifies strategies that minimize their impact on our patients. It is necessary to acknowledge, however, that important risk-altering biological characteristics may remain undescribed at present but be identified and become important with further research.

Figure 1

Complex interaction affecting risk of treatment failure for patients with newly diagnosed Hodgkin lymphoma. Green arrows show how the malignant Hodgkin Reed-Sternberg cells interact with preexisting host factors, including cancer predisposition, pharmacogenetics, and acquired organ dysfunction, each of which may enhance malignant cell survival or interfere with effective treatment delivery. In addition, the Hodgkin Reed-Sternberg cells manipulate cells in their microenvironment, inducing release of growth-enhancing and immune-suppressing cytokines. Treatment (red arrows) reduces the risk of treatment failure by exerting direct cytotoxicity on the Hodgkin Reed-Sternberg cells, by interrupting the stimulation of tumor cell growth encouraged by microenvironmental cells, and by restoring an effective immune response. Treatment effectiveness is modulated (orange arrow) by host factors, with some (eg, good performance status, young age) increasing host tolerance for higher dose treatment and therefore effectiveness, and others (eg, organ dysfunction, coincident HIV infection) diminishing treatment effectiveness.

Figure 1

Complex interaction affecting risk of treatment failure for patients with newly diagnosed Hodgkin lymphoma. Green arrows show how the malignant Hodgkin Reed-Sternberg cells interact with preexisting host factors, including cancer predisposition, pharmacogenetics, and acquired organ dysfunction, each of which may enhance malignant cell survival or interfere with effective treatment delivery. In addition, the Hodgkin Reed-Sternberg cells manipulate cells in their microenvironment, inducing release of growth-enhancing and immune-suppressing cytokines. Treatment (red arrows) reduces the risk of treatment failure by exerting direct cytotoxicity on the Hodgkin Reed-Sternberg cells, by interrupting the stimulation of tumor cell growth encouraged by microenvironmental cells, and by restoring an effective immune response. Treatment effectiveness is modulated (orange arrow) by host factors, with some (eg, good performance status, young age) increasing host tolerance for higher dose treatment and therefore effectiveness, and others (eg, organ dysfunction, coincident HIV infection) diminishing treatment effectiveness.

Close modal

Many studies have identified patient-related risk factors that impact outcome of treatment of individuals with Hodgkin lymphoma. Table 1 lists those risk factors most relevant to current-day management, including age, gender, HIV infection, and prior organ compromise such as pulmonary disease related to smoking and cardiac dysfunction reflecting underlying coronary artery disease. Each of these factors has a profound, highly significant effect on outcome; however, not all can be altered or addressed effectively using currently available interventions. Previously acquired pulmonary compromise, usually related to cigarette smoking, may necessitate omission of bleomycin from primary treatment. Deciding when to drop bleomycin is made more challenging due to the lack of useful objective screening assessment tools. Formal pulmonary function testing, even with inclusion of carbon monoxide diffusion capacity, is quite unreliable at identifying patients at risk for significant bleomycin toxicity.8-10  The decision to omit bleomycin must be made on clinical grounds. I have found that a useful rule of thumb is to consider the potential impact of a relatively rapid loss of 30% to 40% of current respiratory reserve. If a patient appears, based on a review of current activity levels and exercise tolerance, capable of absorbing that much loss of lung function from current pulmonary reserve, bleomycin can be safely, but still carefully, included in planned chemotherapy. Such a patient has adequate reserve to tolerate pulmonary injury if it occurs. When I do not think such a loss could be endured safely, I omit bleomycin at least until pulmonary reserve improves, as may happen if the compromise was due to the Hodgkin lymphoma, perhaps reflecting a large mediastinal mass or lung involvement; or permanently if prior damage from smoking or occupational exposure appears irreversible. Concern has been expressed that coincident use of bleomycin and neutrophil growth factors may exacerbate bleomycin-related pulmonary toxicity.11  However, 2 well-conducted studies, a retrospective review12  and a prospective clinical trial,13  have failed to substantiate this suspicion. Neutrophil growth factors should be used sparingly in the management of Hodgkin lymphoma14 ; however, when they are necessary, there is no need to avoid them due to concern over coincident use of bleomycin.

Table 1

Risk factors affecting outcome of treatment of patients with Hodgkin lymphoma that are intrinsic to the patient

Risk factorFrequency, %5-y OS, %PReference
Age 27, 33, 38-42, 111-117 
 >45 y 34 96 vs 77 <.0001  
 >60 y 18 95 vs 64 <.0001  
 >79 y 91 vs 34 <.0001  
Male gender 55 91 vs 88 .018 27, 33, 39-42 
HIV infection 1.2 89 vs 41 <.0001 16, 17, 20-27 
Prior reduced lung function, major 5-10 * Not applicable BCCA “experience” 
Prior reduced cardiac function 5-10 * Not applicable BCCA “experience” 
Risk factorFrequency, %5-y OS, %PReference
Age 27, 33, 38-42, 111-117 
 >45 y 34 96 vs 77 <.0001  
 >60 y 18 95 vs 64 <.0001  
 >79 y 91 vs 34 <.0001  
Male gender 55 91 vs 88 .018 27, 33, 39-42 
HIV infection 1.2 89 vs 41 <.0001 16, 17, 20-27 
Prior reduced lung function, major 5-10 * Not applicable BCCA “experience” 
Prior reduced cardiac function 5-10 * Not applicable BCCA “experience” 

Frequency with which the risk factor was encountered and single-variable impact (5-year OS; % absent vs % present) in a large sample (n = 1443) of consecutively diagnosed, unselected patients with Hodgkin lymphoma in British Columbia between 1998 and 2013. Note: a steady improvement in overall survival has occurred across this time interval.

BCCA, British Columbia Cancer Agency; OS, overall survival.

*

Presence of these risk factors reduces OS by ∼20% (hazard rate for overall survival: ∼0.75).

Underlying cardiac disease may similarly affect the safety of chemotherapy, in this case, the use of anthracyclines. A prior history of congestive heart failure or ongoing evidence of impaired cardiac reserve such as a left ventricular ejection fraction <50% should prompt careful consideration of the risk that exposure to doxorubicin or doxorubicin plus mediastinal radiation will worsen underlying cardiomyopathy. In such cases, careful serial monitoring of ventricular function must be included in the patient’s assessments during treatment, and omission of the anthracyclines and substitution with an alternative chemotherapeutic agent such as etoposide should be considered.

Another aspect of organ function affecting risk is that of bone marrow tolerance for exposure to cytotoxic agents. This risk emerges clearly in studies focused on the relationship between gender and bone marrow function. Because myelosuppression is reflected in number and depth of episodes of neutropenia, and female gender correlates with increased sensitivity to marrow suppression, these studies typically demonstrate that women have more episodes of neutropenia and deeper and more prolonged nadirs in neutrophil counts compared with men given the same doses of chemotherapy.15  Because depth and length of myelosuppression reflect biological potency of chemotherapy agents, patients who experience greater myelosuppression (in this case, women) have better outcomes, reflecting the more effective dosing of the chemotherapy.

Coincident infection with HIV alters the behavior of Hodgkin lymphoma, leading to more frequent systemic symptoms, earlier spread to extranodal tissue, and markedly decreased failure-free survival (FFS) and OS rates.16-19  There is now ample evidence that the use of highly active antiretroviral treatment (HAART) not only reduces the incidence of HIV-associated Hodgkin lymphoma but HAART plus vigorous supportive care employing prophylactic anti-Pneumocystis, antifungal, and antiherpesvirus antibiotics; neutrophil growth factors; and comprehensive social intervention substantially improve outcome in patients with coincident Hodgkin lymphoma and HIV infection. Most studies indicate that such interventions reduce the risk of death by at least 50%.16,17,20-27 

A final factor relevant to treatment of Hodgkin lymphoma is older age, which has quite consistently been noted to have an adverse impact, although the threshold for its impact has varied across studies from age 45 years to >70 years. The challenge when considering age is that, in many ways, it is simply a proxy for physiological function. Ignoring age places the patient at exaggerated risk, but unduly emphasizing it risks undertreatment. In addition to assessment of specific organ function, such as cardiac or pulmonary function as discussed above, a reasonable and practical approach to adjusting treatment based on age is to start treatment with a modest dose reduction by 20% to 30% of the myelosuppressive chemotherapy agents and to escalate to full doses with subsequent cycles, seeking to reach the maximum that can be achieved without undue toxicity as early in overall treatment as feasible.

Knowledge of relevant patient-specific risk factors is important in the crafting of optimal treatment. Thus, treatment outcome for Hodgkin lymphoma patients with certain patient-specific risk factors indicating a diminished prognosis or signaling specific organ dysfunction can be substantially improved by employing individualized interventions.

Risk factors related to tumor burden and tumor biology can be roughly divided into 2 categories; the first being older assessments describing global clinical factors and laboratory tests, which often reflect not only disease-specific factors but also, indirectly, patient-specific characteristics; the second being newer assessments based on specific biological characteristics of the lymphoma itself.

Risk factors describing global clinical factors and laboratory tests

The most obvious clinical risk factor impacting outcome for patients with Hodgkin lymphoma is stage of disease, which is equally obviously reflective of net tumor burden. The most recent version of the staging system (the Cotswold revision of the Ann Arbor system) includes a basic measure of tumor bulk: the diameter of the largest single tumor mass.28,29  There is universal agreement that stage affects risk of treatment failure and must be considered in treatment planning. Patients are most often divided into 2 groups: those with limited-stage disease, typically including those with stage I or II disease; and those with advanced-stage (stage III or IV) disease. Often, especially in Europe, additional substaging focuses on specific risk factors such as bulk of tumor, presence of B symptoms, number of involved nodal groups, and certain laboratory measurements such as erythrocyte sedimentation rate, and assigns patients to favorable or unfavorable substages of limited disease.30,31  Because stage and substage directly determine planned duration of treatment, with limited-stage disease typically treated using 2 to 4 cycles of chemotherapy and advanced-stage disease using 6 or more cycles, this aspect of tumor burden as a distinct risk factor for treatment failure is intrinsically acknowledged in the modern treatment of Hodgkin lymphoma.

Over several decades extending from the 1970s to the 1990s, clinical factors impacting prognosis and outcome of patients with Hodgkin lymphoma were described in many studies, some of which focused on all patients, whereas others attempted to identify factors specific to stage of disease or define subsets of patients such as those with specific histologic subtypes.27,30,32-43  The extent to which these clinical factors impact patient outcomes has diminished substantially as the effectiveness of interventions has improved. Obvious and clinically important changes in the accuracy and diminished relevance of clinical risk factors can be readily seen when one examines the impact of the factors needed to assign a score using the pivotal International Prognostic Factors Project (IPFP) score.42  The IPFP was an international effort coordinated by the German Hodgkin Study Group, in which investigators assembled data on a large number of potentially relevant prognostic factors and treatment outcomes from 25 Hodgkin lymphoma treatment centers or cooperative groups for 5141 patients with advanced-stage Hodgkin lymphoma treated primarily (>75%) with doxorubicin, bleomycin, vinblastine, and dacarbazine (ABVD)44-46  as delivered in the late 1980s and early 1990s. In the final multivariable analysis, the IPFP identified 7 independently prognostic factors, shown in Table 2, each of which contributed approximately equally to the impact on freedom from progression (FFP) and OS: age, gender, stage, hemoglobin level, serum albumin level, peripheral blood white blood cell count, and degree of peripheral blood lymphopenia. Given 1 point for each factor present, patients were assigned a score from 0 to 7, which resulted in a wide range of outcomes measured as 5-year FFP and 5-year OS rates. For example, the investigators found that a patient with no adverse factors (IPFP score 0) had 5-year FFP and 5-year OS rates of 84% and 89%, respectively, whereas a patient with 4 factors (IPFP score 4) had 5-year FFP and 5-year OS rates of 51% and 61%, respectively. The strengths of the IPFP were the large number of patients examined, the international participation, ready availability in standard clinical practice of the factors identified, and the use of ABVD as the primary treatment in the large majority of patients, although the inclusion of some patients treated with regimens other than ABVD somewhat weakens the strength of this latter observation. As useful as this index has proven, there are several limitations in the original publication: 446 patients were excluded due to age outside the range from 15 to 65 years or receipt of noncurative chemotherapy; 40% of the included patients also received treatment with radiation; 25% of the patients had stage I or II disease; all 7 variables included in the final score were available for only 1618 (34%) of the patients, forcing the statisticians to interpolate the missing variables based on assumptions about their probable distribution; and, lastly and most importantly, the patients were primarily treated in the 1980s and early 1990s, before secondary treatment with high-dose chemotherapy and autologous hematopoietic stem cell transplantation (ASCT) came into wide use for patients with relapse or primary progression despite ABVD.47-55 

Table 2

Prognostic factors with independent impact on outcome for patients with advanced-stage Hodgkin lymphoma identified in the IPFP

FactorCriterionFrequency, %
Age >44 y 21 
Gender Male 61 
Stage IV 42 
Serum albumin, g/L <40 35 
Hemoglobin, g/L <105 ∼20* 
WBC count ×109/L >15 19 
Lymphocyte count ×109/L <0.6 or <8% of total WBC 21 
FactorCriterionFrequency, %
Age >44 y 21 
Gender Male 61 
Stage IV 42 
Serum albumin, g/L <40 35 
Hemoglobin, g/L <105 ∼20* 
WBC count ×109/L >15 19 
Lymphocyte count ×109/L <0.6 or <8% of total WBC 21 

WBC, white blood cell.

*

Value estimated from primary publication.

Much has changed in the management of patients with advanced-stage Hodgkin lymphoma since the analysis underlying the IPFP scoring system was conducted. Diagnostic imaging has improved, first with the introduction of faster computed tomographic scanning, with its ability to provide finer and more exact detail, and later with the introduction of functional imaging based on fluorodeoxyglucose positron emission tomography (FDG-PET). Improved imaging not only increases the accuracy of disease identification but also introduces the artifact of stage migration, which in turn improves apparent treatment outcomes.56  Chemotherapy dose delivery has improved coincident with the widespread use of neutrophil growth factors, although the necessity to employ such growth factors has frequently been questioned.14  Diagnostic accuracy has improved, reducing the modest but still important number of patients with poorer-prognosis non-Hodgkin lymphomas such as anaplastic large-cell lymphoma57,58  and T-cell/histiocyte-rich large B-cell lymphoma58-60  erroneously included in series of patients thought to have classical Hodgkin lymphoma. Lastly, and most importantly, as described above, ASCT has been universally adopted as standard secondary, potentially curative, treatment for patients who have relapse after ABVD or primary progression during ABVD. The combined impact of these mitigating factors (stage migration due to improved imaging, improved dose delivery, more accurate diagnoses, and wide use of ASCT) can be seen when one examines the outcome achieved by primary treatment with ABVD in major clinical trials and large single-institution series over the years from the late 1980s through the early 2000s (Table 3). The 5-year OS rate has increased from just over 70% to approximately 90% despite the nominal use of exactly the same chemotherapy regimen (ABVD).

Table 3

Improvement in 5-year OS rates from the late 1980s to the 2000s

5-y OS, %Year of publicationReference
73 1992 45 
78 1998 42 
82 2003 46 
83 2003 118 
86 2008 119 
84 2009 
90 2009 
91 2012 
88 2013 
5-y OS, %Year of publicationReference
73 1992 45 
78 1998 42 
82 2003 46 
83 2003 118 
86 2008 119 
84 2009 
90 2009 
91 2012 
88 2013 

Improvement in 5-year OS rates after primary treatment with ABVD or equivalent chemotherapy for advanced-stage Hodgkin lymphoma seen in serial major international clinical trials and large single-institution series.

The impact of this apparent improvement in outcome after primary treatment with ABVD for advanced-stage Hodgkin lymphoma on the usefulness of the IPFP scoring system is evident in the results we have seen at the British Columbia Cancer Agency (BCCA) (Table 4; Figure 2).7 Table 4 shows a comparison of outcomes for subgroups of patients with varying IPFP scores as seen in the original IPFP report42  and in our single-institution experience of 579 consecutive patients treated with ABVD or equivalent chemotherapy.7 Figure 2 shows updated FFP curves for 675 consecutive patients treated with ABVD or equivalent chemotherapy at the BCCA through 2009 broken down by IPFP score. These results show that the 42% spread in the 5-year FFP rate, which ranged from 84% to 42% in the original publication for patients with a score of 0 compared with those with a score ≥5, has markedly narrowed to a 17% spread, ranging from 83% to 66%, using ABVD today. Even more importantly, presently, for the 94% of patients with advanced-stage Hodgkin lymphoma who present with IPFP scores of 0 to 4, the 5-year OS rate has improved to approximately 90%. Clearly, the usefulness of the IPFP score has diminished markedly with time. The same is true for the individual factors that make up the IPFP score and those that have been described in multiple other publications addressing clinical prognostic factors. This change reflects the general principle that as overall treatment strategies improve, the impact of individual (and even aggregated) prognostic factors diminishes. With even the worst subsets of patients (such as the small group [6%] of patients with an IPFP score of ≥5) having a likelihood of cure, with ABVD exceeding 65% and 5-year OS rates >85%, prognostic models based on clinical factors such as those used in the IPFP scoring system no longer have useful clinical relevance. We must search for a different approach to estimating risk.

Table 4

Five-year FFP and 5-year OS rates according to IPFP scores

IPFP scoreNumber of patients (%)BCCA 5-y FFP, %IPFP report 5-y FFP, %BCCA 5-y OS, %IPFP report 5-y OS, %
48 (8.3) 83 ± 6 84 ± 4 98 ± 2 89 ± 2 
166 (28.7) 86 ± 3 77 ± 3 97 ± 2 90 ± 2 
157 (27.1) 80 ± 3 67 ± 2 92 ± 2 81 ± 2 
109 (18.8) 74 ± 4 60 ± 3 87 ± 4 78 ± 3 
62 (10.7) 67 ± 6 51 ± 4 85 ± 5 61 ± 4 
≥5 37 (6.4) 66 ± 8 42 ± 5 74 ± 8 56 ± 5 
Reference  42 42 
IPFP scoreNumber of patients (%)BCCA 5-y FFP, %IPFP report 5-y FFP, %BCCA 5-y OS, %IPFP report 5-y OS, %
48 (8.3) 83 ± 6 84 ± 4 98 ± 2 89 ± 2 
166 (28.7) 86 ± 3 77 ± 3 97 ± 2 90 ± 2 
157 (27.1) 80 ± 3 67 ± 2 92 ± 2 81 ± 2 
109 (18.8) 74 ± 4 60 ± 3 87 ± 4 78 ± 3 
62 (10.7) 67 ± 6 51 ± 4 85 ± 5 61 ± 4 
≥5 37 (6.4) 66 ± 8 42 ± 5 74 ± 8 56 ± 5 
Reference  42 42 

A comparison of the 5-year outcome seen in 579 consecutive patients treated with ABVD for advanced-stage Hodgkin lymphoma at the BCCA and the projected outcome seen in patients included in the IPFP.

Figure 2

Time to progression for 675 consecutive adult patients with advanced-stage Hodgkin lymphoma. Adult patients with advanced-stage Hodgkin lymphoma treated with ABVD or equivalent chemotherapy at the BCCA through 2009 by IPFP score. A score of 0 is indicated by a solid black line (n = 57); a score of 1 by a dashed purple line (n = 185); a score of 2 by a turquoise solid line (n = 186); a score of 3 by a solid gray line (n = 133); a score of 4 by a solid blue line (n = 76); and a score 5 to 7 by a solid green line (n = 38). Cum, cumulative.

Figure 2

Time to progression for 675 consecutive adult patients with advanced-stage Hodgkin lymphoma. Adult patients with advanced-stage Hodgkin lymphoma treated with ABVD or equivalent chemotherapy at the BCCA through 2009 by IPFP score. A score of 0 is indicated by a solid black line (n = 57); a score of 1 by a dashed purple line (n = 185); a score of 2 by a turquoise solid line (n = 186); a score of 3 by a solid gray line (n = 133); a score of 4 by a solid blue line (n = 76); and a score 5 to 7 by a solid green line (n = 38). Cum, cumulative.

Close modal

Risk factors reflecting specific biologic characteristics

Risk factor assessment systems for cancer based on global clinical factors and/or laboratory tests are fundamentally crude in that they reflect a mix of intrinsic host factors such as age, comorbid conditions, gender, and others, plus factors that primarily but not exclusively reflect tumor burden, such as largest mass size, stage, number of extranodal sites, and constitutional symptoms. Even specific clinical laboratory tests such as hemoglobin level, lactate dehydrogenase level, serum albumin level, erythrocyte sedimentation rate, and degree of peripheral blood lymphopenia blend tumor and patient characteristics, obscuring the contribution of intrinsic tumor biology to disease behavior and treatment outcome. It is appealing to hope that examination of specific biologic characteristics of the malignant cells themselves may be more informative and less confusing to interpret. Additionally, identification of factors unique to malignant cells may provide attractive specific targets for therapeutic intervention that promise to improve the therapeutic index of treatment by concentrating the treatment effect on the malignant cells and sparing normal cells.

A large and steadily growing number of variously specific biologic characteristics of Hodgkin lymphoma (biomarkers) have been identified to have an apparent impact on risk (Table 5).61-88  These biomarkers are of several types: antigens expressed on the Hodgkin Reed-Sternberg cells, antigens expressed on circulating lymphocytes, antigens expressed on microenvironmental cells within the tumor and associated biologically with the Hodgkin Reed-Sternberg cells, presence of Epstein-Barr virus in the Hodgkin Reed-Sternberg cells, circulating biomarkers detectable in the serum, specific gene expression and miRNA profiles obtained by analysis of biopsied tumors, and specific germline polymorphisms. All are of interest; however, different subsets are relevant to risk assessment for clinical management in different ways. Increased expression of antigens expressed by Hodgkin Reed-Sternberg cells, including aberrant T-cell antigens, FOXP3, CD20, BCL-XL, and p53, as well as loss of HLA class II markers, can be assessed at the time of diagnosis and may predict a worse outcome. In most, but not all, studies that have focused on their presence, increased numbers of macrophages within the tumor microenvironment measured by various immunohistochemical markers, especially CD68 and CD163, reproducibly identified patients with higher risk of relapse and higher risk of eventual death from Hodgkin lymphoma. Elevated levels of specific serum biomarkers, including TARC, galectin-1, CD163, IL-10, IL-10 receptor, IL-6, CD30, TNF, TNF receptor, CD4, CD8, CD25, and CD54, have been reported to be associated with a worse prognosis. A polygene gene expression profile, including approximately 15 genes, performed on tumor biopsies and, therefore, primarily reflecting microenvironmental cells, appears to identify a subset of Hodgkin lymphoma patients with a markedly higher risk of treatment resistance.62,69  Lastly, certain germline polymorphisms of IL-10, IL-6, and NPAT may be associated with poorer prognosis (IL-10 and IL-6) or risk of development of nodular lymphocyte–predominant Hodgkin lymphoma (NPAT).71  Several challenges arise, however, as we try to turn these interesting biological observations into clinically relevant biomarkers. Most problematic is the lack of wide validation of the significance of these biomarkers, which typically have been demonstrated in only small series of selected patients and have not been reproducibly shown to be significant in multiple independent series of patients. Additionally, many of these biomarkers, especially those detected in serum or those expressed as surface markers on Hodgkin Reed-Sternberg cells, are not independent in their impact on prognosis. Rather, they travel together such that elevated levels or elevated expression of one is often associated with elevation of several others.

Table 5

Biomarkers with potential impact on outcome in patients treated for Hodgkin lymphoma

FactorImpact on prognosisReference
Assessment of Hodgkin Reed-Sternberg cells   
 Aberrant T-cell antigen expression* Negative 61 
 FOXP3 expression* Negative 66 
 CD20 expression* Negative 66 
 BCL-XL* Negative 77 
 p53* Negative 77 
 HLA class II, loss* Negative 75 
 Presence of Epstein-Barr virus Negative 82-86 
Assessment of microenvironmental or circulating nonneoplastic cells, cytokines, and membrane-associated antigens   
 Fibroblast growth factor 2 Negative 67 
 Syndecan-1 Negative 67 
 Tumor-associated macrophages Negative 68, 70,72, 87, 88 
 CD68 expression Negative 66 
 Serum TARC, elevated Negative 63, 65 
 Serum galectin-1, elevated Negative 64, 79 
 Serum CD163, elevated Negative 65 
 Serum IL-10, elevated Negative 73, 76, 78 
 Serum IL-10 receptor, elevated Negative 76 
 Serum IL-6, elevated Negative 76 
 Serum CD30, elevated Negative 76, 78 
 Serum TNF, elevated Negative 76 
 Serum TNF receptor, elevated Negative 76 
 Serum CD4, elevated Negative 78 
 Serum CD8, elevated Negative 78 
 Serum CD25, elevated Negative 78 
 Serum CD54, elevated Negative 78 
Gene expression and miRNA profiling reflecting the tumor microenvironment   
 Gene expression profiling Positive or negative 62, 69 
 Global miRNA levels, including MIR21, MIR30E, MIR30D, and MIR92B Positive or negative 80, 81 
Host germline polymorphisms and mutations   
IL-10-specific polymorphism 592AA Negative 74 
IL-6-specific polymorphism 174GG Negative 74 
 Germline NPAT mutation Marker for risk of nodular lymphocyte–predominant
Hodgkin lymphoma 
71 
FactorImpact on prognosisReference
Assessment of Hodgkin Reed-Sternberg cells   
 Aberrant T-cell antigen expression* Negative 61 
 FOXP3 expression* Negative 66 
 CD20 expression* Negative 66 
 BCL-XL* Negative 77 
 p53* Negative 77 
 HLA class II, loss* Negative 75 
 Presence of Epstein-Barr virus Negative 82-86 
Assessment of microenvironmental or circulating nonneoplastic cells, cytokines, and membrane-associated antigens   
 Fibroblast growth factor 2 Negative 67 
 Syndecan-1 Negative 67 
 Tumor-associated macrophages Negative 68, 70,72, 87, 88 
 CD68 expression Negative 66 
 Serum TARC, elevated Negative 63, 65 
 Serum galectin-1, elevated Negative 64, 79 
 Serum CD163, elevated Negative 65 
 Serum IL-10, elevated Negative 73, 76, 78 
 Serum IL-10 receptor, elevated Negative 76 
 Serum IL-6, elevated Negative 76 
 Serum CD30, elevated Negative 76, 78 
 Serum TNF, elevated Negative 76 
 Serum TNF receptor, elevated Negative 76 
 Serum CD4, elevated Negative 78 
 Serum CD8, elevated Negative 78 
 Serum CD25, elevated Negative 78 
 Serum CD54, elevated Negative 78 
Gene expression and miRNA profiling reflecting the tumor microenvironment   
 Gene expression profiling Positive or negative 62, 69 
 Global miRNA levels, including MIR21, MIR30E, MIR30D, and MIR92B Positive or negative 80, 81 
Host germline polymorphisms and mutations   
IL-10-specific polymorphism 592AA Negative 74 
IL-6-specific polymorphism 174GG Negative 74 
 Germline NPAT mutation Marker for risk of nodular lymphocyte–predominant
Hodgkin lymphoma 
71 

IL, interleukin; miRNA, microRNA; TARC, thymus and activation-regulated chemokine; TNF, tumor necrosis factor.

*

Presence detected by immunohistochemistry on tumor Hodgkin Reed-Sternberg cells.

Presence detected by immunohistochemistry on circulating peripheral blood CD30-positive cells.

Presence detected by immunohistochemistry on tumor microenvironment cells.

Although there remain challenges due to lack of validation and/or cross-association among the many potentially important biomarkers for increased risk of treatment resistance in Hodgkin lymphoma, some of these biomarkers have emerged as more attractive candidates to signal higher risk. In particular, the increased presence of tissue-infiltrating macrophages, whether measured by immunohistochemistry66,68,70,72  or implied by specific gene expression profiles,62,69  appears to be a reproducible and powerful negative prognostic factor. Appropriately, the presence of increased numbers of tissue-infiltrating macrophages is now being examined in prospective clinical trials to see whether this finding can be sufficiently reproducibly demonstrated to justify its use to identify patients for either reduction of treatment because macrophage numbers are very low or escalation of treatment because they are very high. Certain serum markers also seem promising, especially serum TARC, galectin-1, and IL-10, because their prognostic impact has been validated independently63-65,73,76,78,79  and because they can be readily measured not only at diagnosis, when their prognostic importance can be assessed, but also serially during treatment to determine whether they can reliably identify patients whose treatment response is proving to be suboptimal. Finally, examining the overall gene expression profile detectable in biopsy tissue involved with Hodgkin lymphoma, which necessarily profiles the gene expression of the host reactive cells and not the malignant Hodgkin Reed-Sternberg cells, appears capable of separating a minority (29%) of patients with a sixfold worse prognosis from a majority (71%) with a much more favorable prognosis using modern chemotherapy.62 

Treatment of Hodgkin lymphoma is typically delivered over several months, and treatment of advanced-stage disease often takes at least 6 to 8 months to complete. It is therefore attractive to try to identify risk factors during treatment that signal a higher likelihood that treatment will fail so that a change in (or an addition to) treatment can be considered. Attempts to find such an assessable factor in the past have proven unreliable. Speed of response has been assessed with the hope that rapid responders would do well and that a change in treatment plan would improve outcomes for slow responders, but this has not proven reliable. Quality of response early in the delivery of multiple cycles of chemotherapy is conceptually similar and is discussed below. Lastly, the absence of a complete response at the end of planned chemotherapy may identify patients with higher risk of relapse. Unfortunately, in the past, response assessment relying on such techniques as gallium, magnetic resonance, and even computed tomographic imaging has run afoul of the tendency of Hodgkin lymphoma to be associated with residual, sometimes large fibronecrotic masses, even when viable tumor cells have been eradicated. Thus, slow or incomplete response, as previously measured with historically available imaging techniques, has not reliably identified poor-prognosis patients, nor has intensification of chemotherapy dosing or addition of radiation based on speed or quality of response proven effective at reducing treatment failures.89-96  This situation may now be changing, however, with the wide availability of functional imaging using FDG-PET. Further complicating interpretation of the available literature is the possibility that FDG-PET during treatment, so-called interim PET, may have different usefulness in the management of limited-stage Hodgkin lymphoma compared with advanced-stage disease.

Interim PET as a risk factor for limited-stage Hodgkin lymphoma

Current management of adults with limited-stage Hodgkin lymphoma (stage IA or IIA, nonbulky [greatest diameter <10 cm]), typically consisting either of brief chemotherapy followed by involved-field or involved-nodal radiation or of only brief chemotherapy, cures almost all patients, and secondary treatment rescues many of those who relapse, virtually eliminating death from Hodgkin lymphoma in this population.97,98  For that reason, the current focus of clinical research is on identifying subsets of patients with limited-stage disease for whom treatment can be de-escalated without forfeiting effectiveness, perhaps by reducing the number of chemotherapy cycles or eliminating the radiation. Substantial consistency of results is apparent in the reported experiences employing interim PET after 2 to 3 cycles of ABVD for patients with limited-stage Hodgkin lymphoma (Table 6).99-102  Approximately 80% to 85% of patients reach an FDG-PET-negative state after 2 to 3 cycles of ABVD, and such patients have an approximately 90% likelihood of remaining free of relapse, even when radiation is omitted from their management. Thus, patients with a negative FDG-PET scan after 2 cycles of ABVD are at low risk of treatment failure. The prognostic value of a positive FDG-PET scan is less clear because most investigators have chosen to change treatment modality based on it, switching to radiation (Table 7), leaving both the scan’s usefulness in indicating a need to use radiation and its impact on risk unclear.

Table 6

Selected large studies reporting interim PET scan results in patients with limited-stage Hodgkin lymphoma treated with ABVD

No. of patientsCycles of chemotherapyNegative interim PET scanReference
n%
571 426 75 102 
441 381 86 99 
221 183 83 updated from 101 
80 2-4 70 87 100 
No. of patientsCycles of chemotherapyNegative interim PET scanReference
n%
571 426 75 102 
441 381 86 99 
221 183 83 updated from 101 
80 2-4 70 87 100 
Table 7

Modifiable risk factors and effective interventions affecting outcome in the treatment of Hodgkin lymphoma

Risk factorFrequency, %Effective interventionsReference
Reduced pulmonary function, major ∼5-10 Omit bleomycin 120, 121 
Left ventricular ejection fraction <50% ∼5-10 Omit doxorubicin; consider substitution with etoposide * 
HIV infection 1.2 Vigorous supportive care with appropriate antibiotics and neutrophil growth factors 16, 17, 20-27 
Positive interim PET scan, limited-stage disease 15-20 Involved-field or involved-nodal radiation 99-102 
Risk factorFrequency, %Effective interventionsReference
Reduced pulmonary function, major ∼5-10 Omit bleomycin 120, 121 
Left ventricular ejection fraction <50% ∼5-10 Omit doxorubicin; consider substitution with etoposide * 
HIV infection 1.2 Vigorous supportive care with appropriate antibiotics and neutrophil growth factors 16, 17, 20-27 
Positive interim PET scan, limited-stage disease 15-20 Involved-field or involved-nodal radiation 99-102 

Frequency with which the risk factor was encountered in a large sample (n = 1443) of consecutively diagnosed, unselected patients with Hodgkin lymphoma in British Columbia between 1998 and 2013.

Interim PET as a risk factor for advanced-stage Hodgkin lymphoma

The accuracy and usefulness of interim PET as a risk factor in patients with advanced-stage Hodgkin lymphoma are considerably less clear than for limited-stage patients.103-110  ABVD is the only multiagent chemotherapy program for which interim PET has been evaluated extensively. Table 8 shows the outcome for patients treated with ABVD for advanced-stage Hodgkin lymphoma and compares results for those with a positive interim PET scan during chemotherapy to those whose interim PET scan had become negative. A negative interim PET scan is found in approximately 80% of patients and appears to be strongly predictive for a favorable outcome. In addition, a negative interim PET scan appears to override or at least rival the prognostic impact of the IPFP score. However, for the approximately 20% of patients with a positive interim PET scan, the positive result’s impact is much less clear, even when the chemotherapy regimen is not altered. Reported FFS rates range from zero to almost 40% (Table 8). Furthermore, it remains quite unclear whether the negative prognostic impact of a positive interim PET scan can be overcome by changing the treatment approach. Preliminary observations that a switch to an escalated regimen of bleomycin, etoposide, doxorubicin, cyclophosphamide, vincristine, procarbazine, and prednisone after a positive interim PET scan may as much as double 2- to 3-year FFS rates require confirmation.106  Once again, similarly to its value in limited-stage Hodgkin lymphoma, the primary usefulness of an interim PET scan in the assessment of patients with advanced-stage Hodgkin lymphoma appears to be in consistently identifying patients who have a low risk of eventual treatment failure with strong negative predictive impact after a negative interim PET scan; however, the usefulness of a positive interim PET scan remains problematic, with its value in assessing risk obscured by the potential impact of altering treatment midway though management. Thus, in both situations in which we can extract instructive observations, the interim PET scan seems most valuable for its ability to identify low risk of treatment failure; the value of a positive interim PET scan remains undetermined.

Table 8

Selected large studies reporting interim PET scan results in patients treated with 6 to 8 cycles of ABVD for advanced-stage Hodgkin lymphoma

No. of patientsOutcome by interim PET scan resultReference
NegativePositive
n (%)FFS, % (y)n (%)FFS, % (y)
260 215 (83) 95 (3) 45 (17) 28 (3) 103, 104 
260 210 (81) 95 (2) 50 (19) 13 (2) 105 
160 137 (86) 92 (2) 23 (14) * 106 
77 61 (79) 96 (2) 16 (21) 0 (2) 107 
91 77 (85) 73 (3) 14 (15) 38 (3) 110 
No. of patientsOutcome by interim PET scan resultReference
NegativePositive
n (%)FFS, % (y)n (%)FFS, % (y)
260 215 (83) 95 (3) 45 (17) 28 (3) 103, 104 
260 210 (81) 95 (2) 50 (19) 13 (2) 105 
160 137 (86) 92 (2) 23 (14) * 106 
77 61 (79) 96 (2) 16 (21) 0 (2) 107 
91 77 (85) 73 (3) 14 (15) 38 (3) 110 

Interim PET scans were performed after 2 cycles of chemotherapy.

*

Positive interim PET scan outcome is not interpretable due to change in chemotherapy.

The assessment of risk has 3 basic purposes in the management of serious disease such as Hodgkin lymphoma. First, risk assessment establishes reasonable assumptions in terms of prognosis, aligning the expectations of the patient, the patient’s family, and the treating physicians and, often, especially in the case of Hodgkin lymphoma, providing solid justification for optimism that the disease will be permanently eradicated. Second, identification of some risks may suggest specific additions to or alterations of the treatment plan that can meaningfully alter the risk of treatment failure. An example of such a modifiable risk factor is coincident HIV infection, with the need to add markedly enhanced supportive care and coincident HAART. Third, isolation of risk factors based on specific biological characteristics of the disease may suggest avenues for the development of targeted therapy that can focus its impact exclusively on the malignancy (or on the malignancy and the elements in the microenvironment providing a growth advantage) and avoid the negative impact of off-target toxicity, a characteristic all too often retained by conventional chemotherapeutic agents and radiation treatments. The clinical, biological, and imaging-related risk factors described in this article reach across this spectrum of purposes for assessing risk in Hodgkin lymphoma.

The author thanks his clinical colleagues at the BCCA and the physicians of British Columbia for their continued support and referral of patients; and Ms Suman Singh for help with maintenance of the BCCA Lymphoid Cancer Database.

Contribution: J.M.C. composed the entire article.

Conflict-of-interest disclosure: The author declares no competing financial interests.

Correspondence: Joseph M. Connors, Center for Lymphoid Cancer, British Columbia Cancer Agency, 600 West 10th Ave, Vancouver, BC, Canada V5Z 4E6; e-mail: jconnors@bccancer.bc.ca

1
Gordon
 
LI
Hong
 
F
Fisher
 
RI
, et al. 
Randomized phase III trial of ABVD versus Stanford V with or without radiation therapy in locally extensive and advanced-stage Hodgkin lymphoma: an intergroup study coordinated by the Eastern Cooperative Oncology Group (E2496).
J Clin Oncol
2013
, vol. 
31
 
6
(pg. 
684
-
691
)
2
Carde
 
PP
Karrasch
 
M
Fortpied
 
C
, et al. 
ABVD (8 cycles) versus BEACOPP (4 escalated cycles ≥ 4 baseline) in stage III-IV high-risk Hodgkin lymphoma (HL): first results of EORTC 20012 Intergroup randomized phase III clinical trial [abstract].
J Clin Oncol
2012
, vol. 
30
 
suppl
 
510s. [Abstract 8002]
3
Viviani
 
S
Zinzani
 
PL
Rambaldi
 
A
, et al. 
Michelangelo Foundation
Gruppo Italiano di Terapie Innovative nei Linfomi; Intergruppo Italiano Linfomi
ABVD versus BEACOPP for Hodgkin’s lymphoma when high-dose salvage is planned.
N Engl J Med
2011
, vol. 
365
 
3
(pg. 
203
-
212
)
4
Connors
 
JM
Hodgkin’s lymphoma—the great teacher.
N Engl J Med
2011
, vol. 
365
 
3
(pg. 
264
-
265
)
5
Hoskin
 
PJ
Lowry
 
L
Horwich
 
A
, et al. 
Randomized comparison of the stanford V regimen and ABVD in the treatment of advanced Hodgkin’s lymphoma: United Kingdom National Cancer Research Institute Lymphoma Group Study ISRCTN 64141244.
J Clin Oncol
2009
, vol. 
27
 
32
(pg. 
5390
-
5396
)
6
Federico
 
M
Luminari
 
S
Iannitto
 
E
, et al. 
HD2000 Gruppo Italiano per lo Studio dei Linfomi Trial
ABVD compared with BEACOPP compared with CEC for the initial treatment of patients with advanced Hodgkin’s lymphoma: results from the HD2000 Gruppo Italiano per lo Studio dei Linfomi Trial.
J Clin Oncol
2009
, vol. 
27
 
5
(pg. 
805
-
811
)
7
Moccia
 
AA
Donaldson
 
J
Chhanabhai
 
M
, et al. 
International Prognostic Score in advanced-stage Hodgkin’s lymphoma: altered utility in the modern era.
J Clin Oncol
2012
, vol. 
30
 
27
(pg. 
3383
-
3388
)
8
O’Sullivan
 
JM
Huddart
 
RA
Norman
 
AR
Nicholls
 
J
Dearnaley
 
DP
Horwich
 
A
Predicting the risk of bleomycin lung toxicity in patients with germ-cell tumours.
Ann Oncol
2003
, vol. 
14
 
1
(pg. 
91
-
96
)
9
McKeage
 
MJ
Evans
 
BD
Atkinson
 
C
Perez
 
D
Forgeson
 
GV
Dady
 
PJ
New Zealand Clinical Oncology Group
Carbon monoxide diffusing capacity is a poor predictor of clinically significant bleomycin lung.
J Clin Oncol
1990
, vol. 
8
 
5
(pg. 
779
-
783
)
10
Lucraft
 
HH
Wilkinson
 
PM
Stretton
 
TB
Read
 
G
Role of pulmonary function tests in the prevention of bleomycin pulmonary toxicity during chemotherapy for metastatic testicular teratoma.
Eur J Cancer Clin Oncol
1982
, vol. 
18
 
2
(pg. 
133
-
139
)
11
Bastion
 
Y
Reyes
 
F
Bosly
 
A
, et al. 
Possible toxicity with the association of G-CSF and bleomycin.
Lancet
1994
, vol. 
343
 
8907
(pg. 
1221
-
1222
)
12
Saxman
 
SB
Nichols
 
CR
Einhorn
 
LH
Pulmonary toxicity in patients with advanced-stage germ cell tumors receiving bleomycin with and without granulocyte colony stimulating factor.
Chest
1997
, vol. 
111
 
3
(pg. 
657
-
660
)
13
Younes
 
A
Fayad
 
L
Romaguera
 
J
Pro
 
B
Goy
 
A
Wang
 
M
Safety and efficacy of once-per-cycle pegfilgrastim in support of ABVD chemotherapy in patients with Hodgkin lymphoma.
Eur J Cancer
2006
, vol. 
42
 
17
(pg. 
2976
-
2981
)
14
Evens
 
AM
Cilley
 
J
Ortiz
 
T
, et al. 
G-CSF is not necessary to maintain over 99% dose-intensity with ABVD in the treatment of Hodgkin lymphoma: low toxicity and excellent outcomes in a 10-year analysis.
Br J Haematol
2007
, vol. 
137
 
6
(pg. 
545
-
552
)
15
Klimm
 
B
Reineke
 
T
Haverkamp
 
H
, et al. 
German Hodgkin Study Group
Role of hematotoxicity and sex in patients with Hodgkin’s lymphoma: an analysis from the German Hodgkin Study Group.
J Clin Oncol
2005
, vol. 
23
 
31
(pg. 
8003
-
8011
)
16
Berenguer
 
J
Miralles
 
P
Ribera
 
JM
, et al. 
Characteristics and outcome of AIDS-related Hodgkin lymphoma before and after the introduction of highly active antiretroviral therapy.
J Acquir Immune Defic Syndr
2008
, vol. 
47
 
4
(pg. 
422
-
428
)
17
Tanaka
 
PY
Pessoa
 
VP
Pracchia
 
LF
Buccheri
 
V
Chamone
 
DA
Calore
 
EE
Hodgkin lymphoma among patients infected with HIV in post-HAART era.
Clin Lymphoma Myeloma
2007
, vol. 
7
 
5
(pg. 
364
-
368
)
18
McNeil
 
C
HIV infection with Hodgkin’s disease: the virus makes a difference.
J Natl Cancer Inst
1997
, vol. 
89
 
11
(pg. 
754
-
755
)
19
Tirelli
 
U
Errante
 
D
Dolcetti
 
R
, et al. 
Hodgkin’s disease and human immunodeficiency virus infection: clinicopathologic and virologic features of 114 patients from the Italian Cooperative Group on AIDS and Tumors.
J Clin Oncol
1995
, vol. 
13
 
7
(pg. 
1758
-
1767
)
20
Montoto
 
S
Shaw
 
K
Okosun
 
J
, et al. 
HIV status does not influence outcome in patients with classical Hodgkin lymphoma treated with chemotherapy using doxorubicin, bleomycin, vinblastine, and dacarbazine in the highly active antiretroviral therapy era.
J Clin Oncol
2012
, vol. 
30
 
33
(pg. 
4111
-
4116
)
21
Hentrich
 
M
Berger
 
M
Wyen
 
C
, et al. 
Stage-adapted treatment of HIV-associated Hodgkin lymphoma: results of a prospective multicenter study.
J Clin Oncol
2012
, vol. 
30
 
33
(pg. 
4117
-
4123
)
22
Bohlius
 
J
Schmidlin
 
K
Boué
 
F
, et al. 
Collaboration of Observational HIV Epidemiological Research Europe
HIV-1-related Hodgkin lymphoma in the era of combination antiretroviral therapy: incidence and evolution of CD4⁺ T-cell lymphocytes.
Blood
2011
, vol. 
117
 
23
(pg. 
6100
-
6108
)
23
Clifford
 
GM
Rickenbach
 
M
Lise
 
M
, et al. 
Swiss HIV Cohort Study
Hodgkin lymphoma in the Swiss HIV Cohort Study.
Blood
2009
, vol. 
113
 
23
(pg. 
5737
-
5742
)
24
Xicoy
 
B
Ribera
 
JM
Miralles
 
P
, et al. 
GESIDA Group; GELCAB Group
Results of treatment with doxorubicin, bleomycin, vinblastine and dacarbazine and highly active antiretroviral therapy in advanced stage, human immunodeficiency virus-related Hodgkin’s lymphoma.
Haematologica
2007
, vol. 
92
 
2
(pg. 
191
-
198
)
25
Hentrich
 
M
Maretta
 
L
Chow
 
KU
, et al. 
Highly active antiretroviral therapy (HAART) improves survival in HIV-associated Hodgkin’s disease: results of a multicenter study.
Ann Oncol
2006
, vol. 
17
 
6
(pg. 
914
-
919
)
26
Biggar
 
RJ
Jaffe
 
ES
Goedert
 
JJ
Chaturvedi
 
A
Pfeiffer
 
R
Engels
 
EA
Hodgkin lymphoma and immunodeficiency in persons with HIV/AIDS.
Blood
2006
, vol. 
108
 
12
(pg. 
3786
-
3791
)
27
Specht
 
L
Hasenclever
 
D
Engert
 
A
Horning
 
SJ
Prognostic factors.
Hodgkin Lymphoma
2011
Heidelberg, Germany
Springer
(pg. 
97
-
116
)
28
Carbone
 
PP
Kaplan
 
HS
Musshoff
 
K
Smithers
 
DW
Tubiana
 
M
Report of the Committee on Hodgkin’s Disease Staging Classification.
Cancer Res
1971
, vol. 
31
 
11
(pg. 
1860
-
1861
)
29
Lister
 
TA
Crowther
 
D
Sutcliffe
 
SB
, et al. 
Report of a committee convened to discuss the evaluation and staging of patients with Hodgkin’s disease: Cotswolds meeting.
J Clin Oncol
1989
, vol. 
7
 
11
(pg. 
1630
-
1636
)
30
Tubiana
 
M
Henry-Amar
 
M
Carde
 
P
, et al. 
Toward comprehensive management tailored to prognostic factors of patients with clinical stages I and II in Hodgkin’s disease. The EORTC Lymphoma Group controlled clinical trials: 1964-1987.
Blood
1989
, vol. 
73
 
1
(pg. 
47
-
56
)
31
Loeffler
 
M
Diehl
 
V
Pfreundschuh
 
M
, et al. 
Dose-response relationship of complementary radiotherapy following four cycles of combination chemotherapy in intermediate-stage Hodgkin’s disease.
J Clin Oncol
1997
, vol. 
15
 
6
(pg. 
2275
-
2287
)
32
Mauch
 
P
Gorshein
 
D
Cunningham
 
J
Hellman
 
S
Influence of mediastinal adenopathy on site and frequency of relapse in patients with Hodgkin’s disease.
Cancer Treat Rep
1982
, vol. 
66
 
4
(pg. 
809
-
817
)
33
Haybittle
 
JL
Hayhoe
 
FG
Easterling
 
MJ
, et al. 
Review of British National Lymphoma Investigation studies of Hodgkin’s disease and development of prognostic index.
Lancet
1985
, vol. 
1
 
8435
(pg. 
967
-
972
)
34
Sutcliffe
 
SB
Gospodarowicz
 
MK
Bergsagel
 
DE
, et al. 
Prognostic groups for management of localized Hodgkin’s disease.
J Clin Oncol
1985
, vol. 
3
 
3
(pg. 
393
-
401
)
35
Carde
 
P
Burgers
 
JM
Henry-Amar
 
M
, et al. 
Clinical stages I and II Hodgkin’s disease: a specifically tailored therapy according to prognostic factors.
J Clin Oncol
1988
, vol. 
6
 
2
(pg. 
239
-
252
)
36
Mauch
 
P
Tarbell
 
N
Weinstein
 
H
, et al. 
Stage IA and IIA supradiaphragmatic Hodgkin’s disease: prognostic factors in surgically staged patients treated with mantle and paraaortic irradiation.
J Clin Oncol
1988
, vol. 
6
 
10
(pg. 
1576
-
1583
)
37
Specht
 
L
Nordentoft
 
AM
Cold
 
S
Clausen
 
NT
Nissen
 
NI
Tumor burden as the most important prognostic factor in early stage Hodgkin’s disease. Relations to other prognostic factors and implications for choice of treatment.
Cancer
1988
, vol. 
61
 
8
(pg. 
1719
-
1727
)
38
Straus
 
DJ
Gaynor
 
JJ
Myers
 
J
, et al. 
Prognostic factors among 185 adults with newly diagnosed advanced Hodgkin’s disease treated with alternating potentially noncross-resistant chemotherapy and intermediate-dose radiation therapy.
J Clin Oncol
1990
, vol. 
8
 
7
(pg. 
1173
-
1186
)
39
Gobbi
 
PG
Comelli
 
M
Grignani
 
GE
Pieresca
 
C
Bertoloni
 
D
Ascari
 
E
Estimate of expected survival at diagnosis in Hodgkin’s disease: a means of weighting prognostic factors and a tool for treatment choice and clinical research. A report from the International Database on Hodgkin’s Disease (IDHD).
Haematologica
1994
, vol. 
79
 
3
(pg. 
241
-
255
)
40
Hagenbeek
 
A
Carde
 
P
Noordjik
 
EM
, et al. 
Prognostic factor tailored treatment of early stage Hodgkin's disease. Results from a prospective randomized phase III clinical trial in 762 patients (H7 study) [abstract].
Blood
1997
, vol. 
90
 
suppl 1
pg. 
585a
 
41
Vlachaki
 
MT
Hagemeister
 
FB
Fuller
 
LM
, et al. 
Long-term outcome of treatment for Ann Arbor Stage I Hodgkin’s disease: prognostic factors for survival and freedom from progression.
Int J Radiat Oncol Biol Phys
1997
, vol. 
38
 
3
(pg. 
593
-
599
)
42
Hasenclever
 
D
Diehl
 
V
A prognostic score for advanced Hodgkin’s disease. International Prognostic Factors Project on Advanced Hodgkin’s Disease.
N Engl J Med
1998
, vol. 
339
 
21
(pg. 
1506
-
1514
)
43
Josting
 
A
Franklin
 
J
May
 
M
, et al. 
New prognostic score based on treatment outcome of patients with relapsed Hodgkin’s lymphoma registered in the database of the German Hodgkin’s lymphoma study group.
J Clin Oncol
2002
, vol. 
20
 
1
(pg. 
221
-
230
)
44
Bonadonna
 
G
Zucali
 
R
Monfardini
 
S
De Lena
 
M
Uslenghi
 
C
Combination chemotherapy of Hodgkin’s disease with adriamycin, bleomycin, vinblastine, and imidazole carboxamide versus MOPP.
Cancer
1975
, vol. 
36
 
1
(pg. 
252
-
259
)
45
Canellos
 
GP
Anderson
 
JR
Propert
 
KJ
, et al. 
Chemotherapy of advanced Hodgkin’s disease with MOPP, ABVD, or MOPP alternating with ABVD.
N Engl J Med
1992
, vol. 
327
 
21
(pg. 
1478
-
1484
)
46
Duggan
 
DB
Petroni
 
GR
Johnson
 
JL
, et al. 
Randomized comparison of ABVD and MOPP/ABV hybrid for the treatment of advanced Hodgkin’s disease: report of an intergroup trial.
J Clin Oncol
2003
, vol. 
21
 
4
(pg. 
607
-
614
)
47
Jagannath
 
S
Dicke
 
KA
Armitage
 
JO
, et al. 
High-dose cyclophosphamide, carmustine, and etoposide and autologous bone marrow transplantation for relapsed Hodgkin’s disease.
Ann Intern Med
1986
, vol. 
104
 
2
(pg. 
163
-
168
)
48
Gribben
 
JG
Linch
 
DC
Singer
 
CR
McMillan
 
AK
Jarrett
 
M
Goldstone
 
AH
Successful treatment of refractory Hodgkin’s disease by high-dose combination chemotherapy and autologous bone marrow transplantation.
Blood
1989
, vol. 
73
 
1
(pg. 
340
-
344
)
49
Phillips
 
GL
Wolff
 
SN
Herzig
 
RH
, et al. 
Treatment of progressive Hodgkin’s disease with intensive chemoradiotherapy and autologous bone marrow transplantation.
Blood
1989
, vol. 
73
 
8
(pg. 
2086
-
2092
)
50
Jones
 
RJ
Piantadosi
 
S
Mann
 
RB
, et al. 
High-dose cytotoxic therapy and bone marrow transplantation for relapsed Hodgkin’s disease.
J Clin Oncol
1990
, vol. 
8
 
3
(pg. 
527
-
537
)
51
Wheeler
 
C
Antin
 
JH
Churchill
 
WH
, et al. 
Cyclophosphamide, carmustine, and etoposide with autologous bone marrow transplantation in refractory Hodgkin’s disease and non-Hodgkin’s lymphoma: a dose-finding study.
J Clin Oncol
1990
, vol. 
8
 
4
(pg. 
648
-
656
)
52
Bierman
 
PJ
Bagin
 
RG
Jagannath
 
S
, et al. 
High dose chemotherapy followed by autologous hematopoietic rescue in Hodgkin’s disease: long-term follow-up in 128 patients.
Ann Oncol
1993
, vol. 
4
 
9
(pg. 
767
-
773
)
53
Goldstone
 
AH
McMillan
 
AK
The place of high-dose therapy with haemopoietic stem cell transplantation in relapsed and refractory Hodgkin’s disease.
Ann Oncol
1993
, vol. 
4
 
Suppl 1
(pg. 
S21
-
S27
)
54
Reece
 
DE
Barnett
 
MJ
Shepherd
 
JD
, et al. 
High-dose cyclophosphamide, carmustine (BCNU), and etoposide (VP16-213) with or without cisplatin (CBV +/- P) and autologous transplantation for patients with Hodgkin’s disease who fail to enter a complete remission after combination chemotherapy.
Blood
1995
, vol. 
86
 
2
(pg. 
451
-
456
)
55
Connors
 
JM
 
The important role of secondary treatment in Hodgkin lymphoma. Oncology (Williston Park). 2012;26 (12):1193-1194, 1199
56
Feinstein
 
AR
Sosin
 
DM
Wells
 
CK
The Will Rogers phenomenon. Stage migration and new diagnostic techniques as a source of misleading statistics for survival in cancer.
N Engl J Med
1985
, vol. 
312
 
25
(pg. 
1604
-
1608
)
57
Skinnider
 
BF
Connors
 
JM
Sutcliffe
 
SB
Gascoyne
 
RD
Anaplastic large cell lymphoma: a clinicopathologic analysis.
Hematol Oncol
1999
, vol. 
17
 
4
(pg. 
137
-
148
)
58
Swerdlow
 
SH
Campo
 
E
Harris
 
NL
, et al. 
WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues
2008
Lyon, France
International Agency for Research on Cancer
59
Rodriguez
 
J
Pugh
 
WC
Cabanillas
 
F
T-cell-rich B-cell lymphoma.
Blood
1993
, vol. 
82
 
5
(pg. 
1586
-
1589
)
60
Greer
 
JP
Macon
 
WR
Lamar
 
RE
, et al. 
T-cell-rich B-cell lymphomas: diagnosis and response to therapy of 44 patients.
J Clin Oncol
1995
, vol. 
13
 
7
(pg. 
1742
-
1750
)
61
Venkataraman
 
G
Song
 
JY
Tzankov
 
A
, et al. 
Aberrant T-cell antigen expression in classical Hodgkin lymphoma is associated with decreased event-free survival and overall survival.
Blood
2013
, vol. 
121
 
10
(pg. 
1795
-
1804
)
62
Scott
 
DW
Chan
 
FC
Hong
 
F
, et al. 
Gene expression-based model using formalin-fixed paraffin-embedded biopsies predicts overall survival in advanced-stage classical Hodgkin lymphoma.
J Clin Oncol
2013
, vol. 
31
 
6
(pg. 
692
-
700
)
63
Sauer
 
M
Plütschow
 
A
Jachimowicz
 
RD
, et al. 
Baseline serum TARC levels predict therapy outcome in patients with Hodgkin lymphoma.
Am J Hematol
2013
, vol. 
88
 
2
(pg. 
113
-
115
)
64
Ouyang
 
J
Plütschow
 
A
Pogge von Strandmann
 
E
, et al. 
Galectin-1 serum levels reflect tumor burden and adverse clinical features in classical Hodgkin lymphoma.
Blood
2013
, vol. 
121
 
17
(pg. 
3431
-
3433
)
65
Jones
 
K
Vari
 
F
Keane
 
C
, et al. 
Serum CD163 and TARC as disease response biomarkers in classical Hodgkin lymphoma.
Clin Cancer Res
2013
, vol. 
19
 
3
(pg. 
731
-
742
)
66
Greaves
 
P
Clear
 
A
Coutinho
 
R
, et al. 
Expression of FOXP3, CD68, and CD20 at diagnosis in the microenvironment of classical Hodgkin lymphoma is predictive of outcome.
J Clin Oncol
2013
, vol. 
31
 
2
(pg. 
256
-
262
)
67
Gharbaran
 
R
Goy
 
A
Tanaka
 
T
, et al. 
Fibroblast growth factor-2 (FGF2) and syndecan-1 (SDC1) are potential biomarkers for putative circulating CD15+/CD30+ cells in poor outcome Hodgkin lymphoma patients.
J Hematol Oncol
2013
, vol. 
6
 pg. 
62
 
68
Tan
 
KL
Scott
 
DW
Hong
 
F
, et al. 
Tumor-associated macrophages predict inferior outcomes in classic Hodgkin lymphoma: a correlative study from the E2496 Intergroup trial.
Blood
2012
, vol. 
120
 
16
(pg. 
3280
-
3287
)
69
Steidl
 
C
Diepstra
 
A
Lee
 
T
, et al. 
Gene expression profiling of microdissected Hodgkin Reed-Sternberg cells correlates with treatment outcome in classical Hodgkin lymphoma.
Blood
2012
, vol. 
120
 
17
(pg. 
3530
-
3540
)
70
Steidl
 
C
Farinha
 
P
Gascoyne
 
RD
Macrophages predict treatment outcome in Hodgkin’s lymphoma.
Haematologica
2011
, vol. 
96
 
2
(pg. 
186
-
189
)
71
Saarinen
 
S
Aavikko
 
M
Aittomäki
 
K
, et al. 
Exome sequencing reveals germline NPAT mutation as a candidate risk factor for Hodgkin lymphoma.
Blood
2011
, vol. 
118
 
3
(pg. 
493
-
498
)
72
Steidl
 
C
Lee
 
T
Shah
 
SP
, et al. 
Tumor-associated macrophages and survival in classic Hodgkin’s lymphoma.
N Engl J Med
2010
, vol. 
362
 
10
(pg. 
875
-
885
)
73
Rautert
 
R
Schinköthe
 
T
Franklin
 
J
, et al. 
Elevated pretreatment interleukin-10 serum level is an International Prognostic Score (IPS)-independent risk factor for early treatment failure in advanced stage Hodgkin lymphoma.
Leuk Lymphoma
2008
, vol. 
49
 
11
(pg. 
2091
-
2098
)
74
Hohaus
 
S
Giachelia
 
M
Di Febo
 
A
, et al. 
Polymorphism in cytokine genes as prognostic markers in Hodgkin’s lymphoma.
Ann Oncol
2007
, vol. 
18
 
8
(pg. 
1376
-
1381
)
75
Diepstra
 
A
van Imhoff
 
GW
Karim-Kos
 
HE
, et al. 
HLA class II expression by Hodgkin Reed-Sternberg cells is an independent prognostic factor in classical Hodgkin’s lymphoma.
J Clin Oncol
2007
, vol. 
25
 
21
(pg. 
3101
-
3108
)
76
Casasnovas
 
RO
Mounier
 
N
Brice
 
P
, et al. 
Groupe d’Etude des Lymphomes de l’Adulte
Plasma cytokine and soluble receptor signature predicts outcome of patients with classical Hodgkin’s lymphoma: a study from the Groupe d’Etude des Lymphomes de l’Adulte.
J Clin Oncol
2007
, vol. 
25
 
13
(pg. 
1732
-
1740
)
77
Montalbán
 
C
García
 
JF
Abraira
 
V
, et al. 
Spanish Hodgkin’s Lymphoma Study Group
Influence of biologic markers on the outcome of Hodgkin’s lymphoma: a study by the Spanish Hodgkin’s Lymphoma Study Group.
J Clin Oncol
2004
, vol. 
22
 
9
(pg. 
1664
-
1673
)
78
Axdorph
 
U
Sjöberg
 
J
Grimfors
 
G
Landgren
 
O
Porwit-MacDonald
 
A
Björkholm
 
M
Biological markers may add to prediction of outcome achieved by the International Prognostic Score in Hodgkin’s disease.
Ann Oncol
2000
, vol. 
11
 
11
(pg. 
1405
-
1411
)
79
Kamper
 
P
Ludvigsen
 
M
Bendix
 
K
, et al. 
Proteomic analysis identifies galectin-1 as a predictive biomarker for relapsed/refractory disease in classical Hodgkin lymphoma.
Blood
2011
, vol. 
117
 
24
(pg. 
6638
-
6649
)
80
Sánchez-Espiridión
 
B
Martín-Moreno
 
AM
Montalbán
 
C
, et al. 
MicroRNA signatures and treatment response in patients with advanced classical Hodgkin lymphoma.
Br J Haematol
2013
, vol. 
162
 
3
(pg. 
336
-
347
)
81
Navarro
 
A
Gaya
 
A
Martinez
 
A
, et al. 
MicroRNA expression profiling in classic Hodgkin lymphoma.
Blood
2008
, vol. 
111
 
5
(pg. 
2825
-
2832
)
82
Kanakry
 
JA
Ambinder
 
RF
EBV-related lymphomas: new approaches to treatment.
Curr Treat Options Oncol
2013
, vol. 
14
 
2
(pg. 
224
-
236
)
83
Diepstra
 
A
van Imhoff
 
GW
Schaapveld
 
M
, et al. 
Latent Epstein-Barr virus infection of tumor cells in classical Hodgkin’s lymphoma predicts adverse outcome in older adult patients.
J Clin Oncol
2009
, vol. 
27
 
23
(pg. 
3815
-
3821
)
84
Keegan
 
TH
Glaser
 
SL
Clarke
 
CA
, et al. 
Epstein-Barr virus as a marker of survival after Hodgkin’s lymphoma: a population-based study.
J Clin Oncol
2005
, vol. 
23
 
30
(pg. 
7604
-
7613
)
85
Jarrett
 
RF
Stark
 
GL
White
 
J
, et al. 
Scotland and Newcastle Epidemiology of Hodgkin Disease Study Group
Impact of tumor Epstein-Barr virus status on presenting features and outcome in age-defined subgroups of patients with classic Hodgkin lymphoma: a population-based study.
Blood
2005
, vol. 
106
 
7
(pg. 
2444
-
2451
)
86
Flavell
 
KJ
Billingham
 
LJ
Biddulph
 
JP
, et al. 
The effect of Epstein-Barr virus status on outcome in age- and sex-defined subgroups of patients with advanced Hodgkin’s disease.
Ann Oncol
2003
, vol. 
14
 
2
(pg. 
282
-
290
)
87
Kayal
 
S
Mathur
 
S
Karak
 
AK
, et al. 
CD68 tumor-associated macrophage marker is not prognostic of clinical outcome in classical Hodgkin lymphoma.
Leuk Lymphoma
2014
, vol. 
55
 
5
(pg. 
1031
-
1037
)
88
Agur
 
A
Amir
 
G
Paltiel
 
O
, et al. 
CD68 staining correlates with the size of residual mass but not with survival in classical Hodgkin lymphoma.
Leuk Lymphoma
 
In press
89
Carella
 
AM
Bellei
 
M
Brice
 
P
, et al. 
High-dose therapy and autologous stem cell transplantation versus conventional therapy for patients with advanced Hodgkin's lymphoma responding to front-line therapy: long term results.
Haematologica
2009
, vol. 
94
 
1
(pg. 
146
-
148
)
90
Federico
 
M
Bellei
 
M
Brice
 
P
, et al. 
EBMT/GISL/ANZLG/SFGM/GELA Intergroup HD01 Trial
High-dose therapy and autologous stem-cell transplantation versus conventional therapy for patients with advanced Hodgkin’s lymphoma responding to front-line therapy.
J Clin Oncol
2003
, vol. 
21
 
12
(pg. 
2320
-
2325
)
91
Forrest
 
DL
Hogge
 
DE
Nevill
 
TJ
, et al. 
High-dose therapy and autologous hematopoietic stem-cell transplantation does not increase the risk of second neoplasms for patients with Hodgkin’s lymphoma: a comparison of conventional therapy alone versus conventional therapy followed by autologous hematopoietic stem-cell transplantation.
J Clin Oncol
2005
, vol. 
23
 
31
(pg. 
7994
-
8002
)
92
Proctor
 
SJ
Mackie
 
M
Dawson
 
A
, et al. 
A population-based study of intensive multi-agent chemotherapy with or without autotransplant for the highest risk Hodgkin’s disease patients identified by the Scotland and Newcastle Lymphoma Group (SNLG) prognostic index. A Scotland and Newcastle Lymphoma Group study (SNLG HD III).
Eur J Cancer
2002
, vol. 
38
 
6
(pg. 
795
-
806
)
93
Johnson
 
PW
Sydes
 
MR
Hancock
 
BW
Cullen
 
M
Radford
 
JA
Stenning
 
SP
Consolidation radiotherapy in patients with advanced Hodgkin’s lymphoma: survival data from the UKLG LY09 randomized controlled trial (ISRCTN97144519).
J Clin Oncol
2010
, vol. 
28
 
20
(pg. 
3352
-
3359
)
94
Laskar
 
S
Gupta
 
T
Vimal
 
S
, et al. 
Consolidation radiation after complete remission in Hodgkin’s disease following six cycles of doxorubicin, bleomycin, vinblastine, and dacarbazine chemotherapy: is there a need?
J Clin Oncol
2004
, vol. 
22
 
1
(pg. 
62
-
68
)
95
Aleman
 
BM
Raemaekers
 
JM
Tirelli
 
U
, et al. 
European Organization for Research and Treatment of Cancer Lymphoma Group
Involved-field radiotherapy for advanced Hodgkin’s lymphoma.
N Engl J Med
2003
, vol. 
348
 
24
(pg. 
2396
-
2406
)
96
Fermé
 
C
Sebban
 
C
Hennequin
 
C
, et al. 
Comparison of chemotherapy to radiotherapy as consolidation of complete or good partial response after six cycles of chemotherapy for patients with advanced Hodgkin’s disease: results of the groupe d’études des lymphomes de l’Adulte H89 trial.
Blood
2000
, vol. 
95
 
7
(pg. 
2246
-
2252
)
97
Meyer
 
RM
Gospodarowicz
 
MK
Connors
 
JM
, et al. 
NCIC Clinical Trials Group; Eastern Cooperative Oncology Group
ABVD alone versus radiation-based therapy in limited-stage Hodgkin’s lymphoma.
N Engl J Med
2012
, vol. 
366
 
5
(pg. 
399
-
408
)
98
Engert
 
A
Plütschow
 
A
Eich
 
HT
, et al. 
Reduced treatment intensity in patients with early-stage Hodgkin’s lymphoma.
N Engl J Med
2010
, vol. 
363
 
7
(pg. 
640
-
652
)
99
Raemaekers
 
JM
André
 
MP
Federico
 
M
, et al. 
Omitting radiotherapy in early positron emission tomography-negative stage I/II Hodgkin lymphoma is associated with an increased risk of early relapse: Clinical results of the preplanned interim analysis of the randomized EORTC/LYSA/FIL H10 trial.
J Clin Oncol
2014
, vol. 
32
 
12
(pg. 
1188
-
1194
)
100
Filippi
 
AR
Botticella
 
A
Bellò
 
M
, et al. 
Interim positron emission tomography and clinical outcome in patients with early stage Hodgkin lymphoma treated with combined modality therapy.
Leuk Lymphoma
2013
, vol. 
54
 
6
(pg. 
1183
-
1187
)
101
Connors
 
JM
Benard
 
F
Gascoyne
 
RD
, et al. 
 
FDG PET/CT scan guided treatment of limited stage Hodgkin lymphoma spares > 80% of patients from radiotherapy while retaining excellent disease control. Haematologica. 2010;95(suppl 4):S15 (Abstract P045)
102
Radford
 
J
Barrington
 
S
Counsell
 
N
, et al. 
 
Involved field radiotherapy versus no further treatment in patients with clinical stages IA and IIA Hodgkin lymphoma and a 'negative' PET scan after 3 cycles ABVD. Results of the UK NCRI RAPID trial [abstract]. ASH Annual Meeting Abstracts. 2012;120(21). Abstract 547
103
Biggi
 
A
Gallamini
 
A
Chauvie
 
S
, et al. 
International validation study for interim PET in ABVD-treated, advanced-stage hodgkin lymphoma: interpretation criteria and concordance rate among reviewers.
J Nucl Med
2013
, vol. 
54
 
5
(pg. 
683
-
690
)
104
Gallamini
 
A
Barrington
 
SF
Biggi
 
A
, et al. 
The predictive role of interim positron emission tomography for Hodgkin lymphoma treatment outcome is confirmed using the interpretation criteria of the Deauville five-point scale.
Haematologica
2014
, vol. 
99
 
6
(pg. 
1107
-
1113
)
105
Gallamini
 
A
Hutchings
 
M
Rigacci
 
L
, et al. 
Early interim 2-[18F]fluoro-2-deoxy-D-glucose positron emission tomography is prognostically superior to international prognostic score in advanced-stage Hodgkin’s lymphoma: a report from a joint Italian-Danish study.
J Clin Oncol
2007
, vol. 
25
 
24
(pg. 
3746
-
3752
)
106
Gallamini
 
A
Patti
 
C
Viviani
 
S
, et al. 
Gruppo Italiano Terapie Innovative nei Linfomi (GITIL)
Early chemotherapy intensification with BEACOPP in advanced-stage Hodgkin lymphoma patients with a interim-PET positive after two ABVD courses.
Br J Haematol
2011
, vol. 
152
 
5
(pg. 
551
-
560
)
107
Hutchings
 
M
Loft
 
A
Hansen
 
M
, et al. 
FDG-PET after two cycles of chemotherapy predicts treatment failure and progression-free survival in Hodgkin lymphoma.
Blood
2006
, vol. 
107
 
1
(pg. 
52
-
59
)
108
Hutchings
 
M
Mikhaeel
 
NG
Fields
 
PA
Nunan
 
T
Timothy
 
AR
Prognostic value of interim FDG-PET after two or three cycles of chemotherapy in Hodgkin lymphoma.
Ann Oncol
2005
, vol. 
16
 
7
(pg. 
1160
-
1168
)
109
Markova
 
J
Kobe
 
C
Skopalova
 
M
, et al. 
FDG-PET for assessment of early treatment response after four cycles of chemotherapy in patients with advanced-stage Hodgkin’s lymphoma has a high negative predictive value.
Ann Oncol
2009
, vol. 
20
 
7
(pg. 
1270
-
1274
)
110
Oki
 
Y
Chuang
 
H
Chasen
 
B
, et al. 
The prognostic value of interim positron emission tomography scan in patients with classical Hodgkin lymphoma.
Br J Haematol
2014
, vol. 
165
 
1
(pg. 
112
-
116
)
111
Evens
 
AM
Hong
 
F
Gordon
 
LI
, et al. 
The efficacy and tolerability of adriamycin, bleomycin, vinblastine, dacarbazine and Stanford V in older Hodgkin lymphoma patients: a comprehensive analysis from the North American intergroup trial E2496.
Br J Haematol
2013
, vol. 
161
 
1
(pg. 
76
-
86
)
112
Böll
 
B
Görgen
 
H
Fuchs
 
M
, et al. 
ABVD in older patients with early-stage Hodgkin lymphoma treated within the German Hodgkin Study Group HD10 and HD11 trials.
J Clin Oncol
2013
, vol. 
31
 
12
(pg. 
1522
-
1529
)
113
Proctor
 
SJ
Wilkinson
 
J
Jones
 
G
, et al. 
Evaluation of treatment outcome in 175 patients with Hodgkin lymphoma aged 60 years or over: the SHIELD study.
Blood
2012
, vol. 
119
 
25
(pg. 
6005
-
6015
)
114
Nabhan
 
C
Smith
 
SM
Helenowski
 
I
, et al. 
Analysis of very elderly (≥80 years) non-hodgkin lymphoma: impact of functional status and co-morbidities on outcome.
Br J Haematol
2012
, vol. 
156
 
2
(pg. 
196
-
204
)
115
Evens
 
AM
Helenowski
 
I
Ramsdale
 
E
, et al. 
A retrospective multicenter analysis of elderly Hodgkin lymphoma: outcomes and prognostic factors in the modern era.
Blood
2012
, vol. 
119
 
3
(pg. 
692
-
695
)
116
Klimm
 
B
Eich
 
HT
Haverkamp
 
H
, et al. 
German Hodgkin Study Group
Poorer outcome of elderly patients treated with extended-field radiotherapy compared with involved-field radiotherapy after chemotherapy for Hodgkin’s lymphoma: an analysis from the German Hodgkin Study Group.
Ann Oncol
2007
, vol. 
18
 
2
(pg. 
357
-
363
)
117
Klimm
 
B
Diehl
 
V
Engert
 
A
 
Hodgkin's lymphoma in the elderly: a different disease in patients over 60. Oncology (Williston Park). 2007;21(8):982-990; discussion 990, 996, 998 passim
118
Diehl
 
V
Franklin
 
J
Pfreundschuh
 
M
, et al. 
German Hodgkin’s Lymphoma Study Group
Standard and increased-dose BEACOPP chemotherapy compared with COPP-ABVD for advanced Hodgkin’s disease [published correction appears in New Engl J Med. 2005;353(7):744].
N Engl J Med
2003
, vol. 
348
 
24
(pg. 
2386
-
2395
)
119
Gianni
 
AM
Rambaldi
 
A
Zinzani
 
PL
, et al. 
Comparable 3-year outcome following ABVD or BEACOPP first-line chemotherapy plus pre-planned high dose salvage in advanced stage Hodgkin lymphoma: a randomized trial of the Michaelangelo, GITIL and IIL cooperative groups [abstract].
J Clin Oncol
2008
, vol. 
26
 
15
 
[Abstract 8506]
120
Martin
 
WG
Ristow
 
KM
Habermann
 
TM
Colgan
 
JP
Witzig
 
TE
Ansell
 
SM
Bleomycin pulmonary toxicity has a negative impact on the outcome of patients with Hodgkin’s lymphoma.
J Clin Oncol
2005
, vol. 
23
 
30
(pg. 
7614
-
7620
)
121
Van Barneveld
 
PW
van der Mark
 
TW
Sleijfer
 
DT
, et al. 
Predictive factors for bleomycin-induced pneumonitis.
Am Rev Respir Dis
1984
, vol. 
130
 
6
(pg. 
1078
-
1081
)
Sign in via your Institution