The majority of patients with Hodgkin lymphoma are cured with frontline therapy; however, 10% to 15% with early-stage disease and 20% to 30% with advanced stage require second-line therapy that includes a potentially curative transplant, of which an additional 50% to 55% are cured. Those with multiply relapsed disease traditionally would receive novel agents on a clinical trial or combination chemotherapy as a potential bridge to an allogeneic stem cell transplant. This treatment paradigm has changed with the availability of brentuximab vedotin, an antibody drug conjugate used pre- and post-ASCT, as well as for palliation. With the availability of the checkpoint inhibitors, nivolumab and pembrolizumab, there will be another shift in treatment, with these agents being used for palliation and potentially replacing allogeneic stem cell transplantation in certain patient populations. Finally, up-front management is also changing and this will have an impact on how patients in the relapsed and refractory setting will be treated.

Learning Objectives
  • Understanding risk factors that predict outcome in relapsed and refractory Hodgkin lymphoma

  • Understanding how to incorporate brentuximab vedotin and the checkpoint inhibitors into the treatment paradigm

This chapter will focus on 5 aspects that are critical in the treatment of patients with relapsed and refractory transplant-eligible Hodgkin lymphoma (HL): the role of pre–allogeneic stem cell transplant (ASCT) positron emission tomography (PET) imaging, incorporation of brentuximab vedotin (BV) into salvage therapy, patients eligible for post-ASCT consolidation, use of checkpoint inhibitors (CPI), and the future role of allogeneic hematopoietic cell transplantation (allo-HCT) in the management of this disease.

The treatment algorithm for the primary management of HL has evolved over the past 3 decades, leading to a change in the patient populations requiring aggressive second-line therapy. The optimal chemotherapy for advanced stage HL (ASHL) can be endlessly debated, but progression-free survival (PFS) has improved by 15% in the past 2 decades.1,2  For early-stage HL (ESHL), PFS is unchanged at close to 90% despite decreasing the number of cycles of doxorubicin (Adriamycin)-Bleomycin-Vinblastine-Dacarbazine administered and reducing the dose and size of radiation therapy fields.3  From 1990 to 2010, relapsed/refractory (rel/ref) HL patients that underwent consultation for autologous stem cell transplantation (ASCT) had received either full-course chemotherapy or combined modality therapy (CMT) as frontline treatment; currently there exists a substantial proportion of patients who received short-course chemotherapy alone or were enrolled in PET-adapted advanced-stage studies, of which the results were not successful. The presumed response that all rel/ref HL patients must receive salvage therapy (ST) followed by high-dose chemo-radiotherapy (HDT)/ASCT, and if chemosensitive disease is achieved, may need to be addressed. In addition, when this second-line approach fails, do all patients require a consultation for a reduced-intensity conditioning (RIC) allo-HCT in the era of excellent new agents, where palliation for prolonged periods of time is now standard and expected? These issues will be discussed in this chapter as well as at the ASH meeting; there are many historical reviews cataloging various salvage regimens and transplant conditioning regimens to which the reader can be referred and these will not be discussed further.4,5 

Currently any patient with adequate end-organ function and performance status, and who are <75 years old, are treated aggressively with second-line therapy with the intent for high-dose therapy (HDT)/ASCT to follow. These patient populations include: early or advanced stage, relapsed and primary refractory HL treated with full chemotherapy; and early-stage HL treated with combined modality therapy with disease either insid,e the radiation field or advanced-stage disease at the time of ST. As therapy for ESHL evolves it is currently unclear whether patients treated with short-course chemotherapy alone (3-4 cycles of doxorubicin [Adriamycin]-Bleomycin-Vinblastine-Dacarbazine), with early stage disease at the time of ST, require HDT/ASCT. For example, in the RAPID study,6  many patients with relapsed disease received radiation therapy alone for salvage. Because so few patients with ESHL relapse, it will be difficult to assess the need for HDT in this specific patient population in a randomized controlled clinical trial.

Pre-ASCT FDG-PET

HDT/ASCT is standard of care for relapsed or primary refractory HL if chemosensitive disease to ST is confirmed, leading to a cure in approximately 50% of the transplanted patients.7  It is very clear that the cure rate ranges from 25% to 75% depending on prognostic factors.8-10  Response to ST, whether complete or partial, is by far the most important predictor of outcome in this setting. In 2010, our group determined that chemosensitive disease should be defined by the pre-transplant 18F-fluorodeoxyglucose (FDG)-PET result.11  These results have been expanded and updated. A negative scan after platinum-based therapy leads to a 10-year overall survival (OS) rate of 75%. For patients with documented improvement on computed tomography (CT), but with a persistent PET-avidity, only 30% are cured (Figure 1).

Figure 1.

All transplanted patients, functional imaging negative vs positive pretransplant treated MSKCC, 1994-2014.

Figure 1.

All transplanted patients, functional imaging negative vs positive pretransplant treated MSKCC, 1994-2014.

Close modal

There are now many clinical trials suggesting that a negative pretransplant FDG-PET has prognostic value,12,13  and this has been confirmed in a recently published meta-analysis review.14,15 

I define the post–ST PET scan as an interim evaluation; remission is defined 90 to 100 days post-ASCT. Based on this premise, nuclear medicine reports need to be interpreted using the 5-point (Deauville) criteria, with scores 4 and 5 defined as a positive scan.16  How many attempts should be made to achieve the goal of a negative scan? There is no correct answer, but it is reasonable to attempt 2 distinct ST programs for advance-stage disease. For stage I/II disease that can be encompassed into a reasonable radiation therapy field, this treatment should also be attempted to achieve a complete response (CR) before ASCT. There is evidence that patients requiring more than one regimen to achieve PET negativity have a nearly similar outcome compared with those who receive only one ST program.17 

Complicating that matter further, not all patients with a post–ST PET negative, complete response are cured and the contrary is also true; as much as 40% of patients transplanted with PET-avid disease achieve long-term PFS rate. Patients with nodal-only disease have a superior outcome to that of patients with stage IV disease. For example, patients with nodal-only disease in remission at the time of ASCT have an 80% to 90% cure rate compared with 55% to 65% for patients with extranodal disease.18-20 

Summary/Recommendation: A negative pre-ASCT PET scan is critical if a favorable outcome with HDT/ASCT is to be achieved and should be required for this treatment to continue to be the standard of care; however, since as much as 40% of patients with a positive scan are cured, ASCT should not be withheld, especially for patients without extranodal involvement.

Primary refractory HL.

Investigators consider primary refractory HL unfavorable. Most series present data only for the transplanted cohort; there is a lack of intent-to-treat second-line data. Compounding the problem, analyses for primary refractory and relapsed patients are often combined, making it somewhat difficult to tease out the data. If one summarizes the information, <50% of patients with primary refractory HL are progression-free.21  We have recently reported on a large intent-to-treat primary refractory HL dataset compiled from patients treated on prospective ST studies at MSKCC. The median 5-year EFS for these 192 patients was 54%. The same 2 risk factors predicted for outcome: stage IV disease before ST and a positive PET scan after ST. A risk stratification model was created assigning 1 point for each factor. The 5-year EFS was 84% for patients with 0 risk factors, 54% with 1 risk factor, and only 28% for patients with both. Another large relapsed/refractory series was recently reported at ASH in 2015 by Broeckelmann et al. Multivariable analysis once again identified stage IV, primary refractory disease, and inadequate response to salvage chemotherapy as significant independent risk factors for PFS.22 

The incorporation of BV into ST

Two to 3 cycles of ST is the standard of care and administered to all patients as part of a curative second-line strategy. Historically, the goal of ST was to achieve chemosensitive disease before curative HDT/ASCT. As stated before, in 2016 the goal of ST is far more significant and a CR or PET-negative response should be the objective. In fact, with the availability of new agents, patients who have a persistently positive scan after >1 ST may be not appropriate candidates for ASCT. Practitioners need to decide whether this patient population should receive novel drugs as part of a chronic or noncurative strategy vs a highly aggressive allo-HCT program.23 

Prospective, randomized trials comparing different salvage regimens in HL are not available. If one summarizes the data for platinum and nonplatinum-containing ST regimens, a reasonable benchmark is a 50% to 60% CR or PET-negative rate with standard ST, but can this number be improved?24  BV is an antibody-drug conjugate (ADC) composed of a CD30-directed monoclonal antibody (recombinant chimeric IgG1) that is covalently linked to the antimicrotubule agent monomethyl auristatin E (MMAE). Binding of the ADC to CD30 on the cell surface initiates internalization of the MMAE-CD30 complex, followed by proteolytic cleavage that releases MMAE, leading to selective apoptosis of the Reed-Sternberg cells with minimal off-target side effects, the major one being neuropathy. BV is approved for the treatment of adult patients with relapsed or refractory HL where ASCT failed as well as part of a maintenance or consolidation strategy post-ASCT for high-risk patients, which will also be discussed here.25,26 

Given that remission status pre-ASCT is highly predictive of outcome, the incorporation of BV with its favorable toxicity profile into an ST regimen is appealing. As a single agent in heavily pretreated patients, the PET-negative rate after 2 to 4 doses of BV is 30% to 40%. Two strategies have evolved to study this agent as part of ST: sequential administration of BV with chemotherapy and combined chemo-immunotherapy.

The MSKCC and the City of Hope (COH) lymphoma groups reported on the sequential approach.27,28  The designs of the studies were similar, with the primary difference being the BV dosing schedule. In the MSKCC trial, patients received 6 weekly doses of BV at 1.2 mg/kg; patients at COH were given 2 to 4 doses of an every-three-week dosing at 1.8 mg/kg. Patients in both studies with a PET-negative response went directly to HDT/ASCT, and additional ST was avoided; if PET-avid disease was persistent, then additional ST was given. The CR rate to BV alone was 30% to 40%, but this rate improved to 80% with the sequential programs. Consistent with previous data, the PET-negative patients had >75% 3-year PFS post-ASCT (Figures 2 and 3). There was no difference in outcome for patients only receiving BV compared with those requiring sequential therapy to achieve a CR. This approach is very appealing because some patients were able to avoid additional cytotoxic ST. However, predicting which of these patients will achieve CR is difficult. Our approach is to offer BV as initial therapy for patients with nodal-only disease.29 

Figure 2.

Deauville 1-3 considered negative, P = .05.

Figure 2.

Deauville 1-3 considered negative, P = .05.

Close modal
Figure 3.

Complete response (CR) vs non-CR.

Figure 3.

Complete response (CR) vs non-CR.

Close modal

At ASH in 2015, 2 very different chemo-immunotherapy ST programs were presented. The first, an outpatient regimen, combined BV with bendamustine. LaCasce et al reported preliminary data from a phase 1/2 study evaluating the safety and efficacy of these 2 agents as a ST program.30  Fifty-five patients were treated; unexpectedly, there were severe infusion reactions seen in >50% of cycles, leading to an amendment and subsequent steroid premedications, which reduced this adverse event to become manageable. A PET-negative response was documented in 76% of patients; stem cell collection was adequate despite the concern of the known side effect of bendamustine on the bone marrow. Although the median follow-up is short at 18 months, 75% of patients are progression-free, making this outpatient ST appealing. In contrast, Garcia-Sanz et al reported data on 36 patients treated with an aggressive inpatient regimen: ESHAP+BV.31  As expected, hematologic toxicity was significant. The CR rate in this ongoing clinical trial was reported at 83% with no stem cell mobilization failures. Both of these phase 2 studies are also administering post-ASCT BV as part of a consolidation strategy based on the results of the Aethera study, which will be described in the next section. Currently, the Seattle and HOVON groups are studying ICE+BV and DHAP+BV, respectively.

Summary/Recommendation: The addition of BV to ST will increase the CR rate by 20%. However, the implication that all patients require this treatment is unclear. It seems reasonable for patients with low disease burden that single-agent BV should be initiated if available or chemotherapy alone if it is not. For patients with stage IV disease, chemo-immunotherapy or ST alone should be the initial treatment.

POST-ASCT BV consolidation

Before 2015, there were only 2 phase 3 studies published for patients with relapsed HL.32,33  Each study confirmed that ASCT was superior to ST alone; nearly all patients had relapsed as opposed to primary refractory disease. Little progress has been made in the past 2 decades except for decreased ASCT treatment–related mortality, which is secondary to improved supportive care. Unfortunately, PFS remains at 50% if data are analyzed by intent-to-treat.

In late 2010, the Aethera study was initiated. It was a phase 3, randomized, placebo-controlled trial that evaluated whether post-ASCT consolidation treatment with BV could prevent disease progression in patients with HL at high risk for relapse.26  A consensus panel determined that primary refractory HL, initial remission duration of <1 year, or extranodal involvement constituted unfavorable risk; patients were required to have one of these factors for study eligibility. Aethera was written before the near-uniform agreement that an abnormal pre–ASCT PET scan is the single most important factor in predicting outcome, and in fact the PET data that are available on study make recommendations for post-ASCT BV consolidation confusing.

Patients were selected to randomly receive BV 1.8 mg/kg every 3 weeks or placebo for 16 cycles (∼12 months), beginning 30 to 45 days after ASCT. Patients with progression of disease after ST were not eligible. Imaging studies were quarterly for the first year and then at 18 and 24 months during long-term follow-up (LTFU). Clinical lymphoma assessments were performed at each cycle of treatment, quarterly during the first year of LTFU, and every 6 months thereafter.

A total of 329 patients were randomized to the BV (n = 165) or placebo (n = 164) treatment arms. Importantly, 60% of patients had primary refractory HL. Median PFS was not reached in the BV arm and was 15.8 months in the placebo arm; 3-year PFS rates were 61% and 43%, respectively (Figure 4). There were only 6 PFS events recorded after the 24-month evaluation, 3 in each cohort, and it is highly likely that patients in either arm at this time point are cured; there is currently no difference in OS.

Figure 4.

3-year progression-free survival (PFS) data in the Aethera study.

Figure 4.

3-year progression-free survival (PFS) data in the Aethera study.

Close modal

Administering 16 doses of BV does come with additional toxicity, namely neuropathy. Among the 112 patients in the BV arm with treatment-related peripheral neuropathy, 88% experienced improvement of symptoms at the time of 3-year analysis. Discontinuation of treatment because of any adverse events (AE) occurred in 54 patients (33%) in the BV arm, primarily as a result of neuropathy. Patients who discontinued treatment as a result of an AE received a median of 9.5 cycles (range, 1-15) in the BV arm. Interestingly, the 2-year PFS rate in these patients was 69% vs 82% for patients who completed all 16 treatment cycles.

Five risk factors predicted for PFS on the Aethera study: initial remission duration <1 year, <CR to most recent salvage therapy, extranodal disease at the time of ST, B symptoms at the time of ST, and >1 ST required to achieve chemosensitive disease. It is clear from the data that patients who have at least 2 of these risk factors achieve benefit from consolidation (Figure 5). If one looks critically at the pre–ASCT PET results, recommendations are fairly straightforward. Because a CR is a favorable risk factor, patients need at least 2 of 3 additional unfavorable risk factors (remission duration <1 year, extranodal disease or requiring >1 ST) for benefit to be achieved with BV consolidation.

Figure 5.

Risk factors associated with poor outcomes in the Aethera study.

Figure 5.

Risk factors associated with poor outcomes in the Aethera study.

Close modal

Summary/recommendations: Consolidation therapy improves PFS and it is reasonable to give a BV-naïve patient 1 year of post-ASCT therapy if he/she has at least 2 adverse risk factors. In patients who have previously received BV pre-ASCT and meet the risk factor requirements to be enrolled in Aethera, a patient should have achieved at least a partial response to BV-based treatment. In general, in this situation, which I suspect will become more frequent in the United States, I reduce the number of doses of BV post-ASCT to 10 or fewer.

In 2009, when the Aethera study was written, the median OS of patients where ASCT failed was 30 months; in 2016, that number has doubled and will likely continue to increase. The reason is the robust drug development program in HL, specifically BV and the CPI. BV has been extensively discussed, but when given for palliation, OS improves by about 1 year. Other agents are also active, and results are summarized in Table 1. Historically, for patients where a good response was documented, a consult for an allo-HCT ensued. These patients were given the option of transplant or palliation. The 1-year mortality from an allo-HCT was 30%, with an equal number of patients being cured. This paradigm is clearly changing with the availability of the CPI. The role of allo-HCT in HL is an area of great disagreement among lymphoma physicians.

Table 1.

New agents in HL

StudyPhaseDrugMechanism of actionRouteNORR, N (%)CRPRSDMedian PFS
Younes et al, 20121  II Panobinsotat Pan HDAC PO 129 35 (27%) 5 (4%) 30 (23%) 71 (55%) 6.1 mo 
Batlevi et al, 2015 (in process) II Entinostat iso-spec HDAC PO 49 6 (12%) 0 (0%) 6 (12%) 6 (12%) 5.5 mo 
Ansell et al, 20152  Nivolumab immune checkpoint IV 23 20 (87%) 4 (17%) 16 (70%) 3 (13%) 86% at 24 wk 
Younes et al, 20123  II Brentuximab vedotin ADC IV 102 76 (75%) 35 (34%) 41 (40%) 22 (22%) 5.6 mo 
Armand et al, 2015 Abstract4  Pembrolizumab Immune checkpoint IV 31 20 (66%) 5 (16%) 15 (48%) 7 (23%) Not reported 
Johnston et al, 2010, Abstract 20125,6  II Everolimus MTOR inhibitor PO 57 24 (42%) 5 (9%) 19 (33%) 20 (35%) 6 mo 
Fehniger et al, 2011 Abstract7  II Lenalidomide Immune modulator PO 38 7 (18%) 1 (3%) 6 (16%) 5 (13%) Not reported 
StudyPhaseDrugMechanism of actionRouteNORR, N (%)CRPRSDMedian PFS
Younes et al, 20121  II Panobinsotat Pan HDAC PO 129 35 (27%) 5 (4%) 30 (23%) 71 (55%) 6.1 mo 
Batlevi et al, 2015 (in process) II Entinostat iso-spec HDAC PO 49 6 (12%) 0 (0%) 6 (12%) 6 (12%) 5.5 mo 
Ansell et al, 20152  Nivolumab immune checkpoint IV 23 20 (87%) 4 (17%) 16 (70%) 3 (13%) 86% at 24 wk 
Younes et al, 20123  II Brentuximab vedotin ADC IV 102 76 (75%) 35 (34%) 41 (40%) 22 (22%) 5.6 mo 
Armand et al, 2015 Abstract4  Pembrolizumab Immune checkpoint IV 31 20 (66%) 5 (16%) 15 (48%) 7 (23%) Not reported 
Johnston et al, 2010, Abstract 20125,6  II Everolimus MTOR inhibitor PO 57 24 (42%) 5 (9%) 19 (33%) 20 (35%) 6 mo 
Fehniger et al, 2011 Abstract7  II Lenalidomide Immune modulator PO 38 7 (18%) 1 (3%) 6 (16%) 5 (13%) Not reported 

The case for chronic administration of CPI

Targeting the PD-1/PDL1 pathway has revolutionized the way some of the common solid tumors are being treated. Studies are behind in the hematologic malignancies but are rapidly catching up. Recently the CPI, nivolumab, been approved for HL palliation and it is likely that pembrolizumab will also be approved. PDL1/2 is nearly uniformly expressed on Reed-Sternberg cells through chromosomal translocation, gene amplification, and EBV-related mechanisms. In HL, as well as primary mediastinal B-cell lymphoma, amplification of chromosome 9p23-4 is the most common genetic abnormality reported and the genes encoding PDL1 and 2 reside there; therefore, these 2 diseases are primed to respond to CPI.34 

Both nivolumab and pembrolizumab are highly active, and phase 1 data were presented at ASH in 2014 and 2015.29,35  Results are quite similar with both agents, and treatment was administered for up to 2 years provided that the HL remained stable and/or the patient continued to have clinical benefit. These are unusual response criteria, and much different compared with traditional agents, when any time progression is documented, the study drug is discontinued. Because of this “rule,” PFS in these phase 1 studies are inflated. However, because clinical benefit included complete resolution of B symptoms, maintenance of palliative treatment was the correct choice, in my opinion.

Beginning with Nivolumab, the dose is 3 mg/kg every 2 weeks and 23 patients with HL were treated. The patients were heavily pretreated with a median of 5 prior regimens received, but some were BV-naïve. The objective response rate was 87%, but only 5 patients achieved a CR based on PET criteria. Amazingly, 13 patients in CR or PR are no longer receiving treatment and the median PFS has not been reached. The 18-month OS was 83% and the majority of deaths were secondary to transplant-related morality in patients referred for an allo-HCT.

Patients enrolled in the study with pembrolizumab were eligible only if BV treatment failed. Patients received pembrolizumab at 10 mg/kg every 2 weeks. Thirty-one patients were enrolled. The CR and PR rates were 16% and 48%, respectively; 70% of the responses lasted longer than 24 weeks, with a median follow-up of 17 months.

These phase 1 studies have led to large phase 2 clinical trials in which response rates were presented at ASCO and EHA this year. Eighty patients were included in the most recent published data for nivolumab; the CR and PR rates were 27% and 45%, respectively. Sixty patients were included in the most recent published data for pembrolizumab; 30 patients failed ASCT and BV, and 30 progressed on BV but were ASCT-ineligible. In the first cohort, the CR and PR rates were 20% and 50%, respectively, and 27% and 53%, respectively, in the second cohort. In both studies, most of the patients with demonstrable response continue to receive treatment.

The side effect profiles of the CPI are different from traditional therapy and are usually immune-related. These include endocrinopathies such as thyroiditis, adrenal and pituitary dysfunction, hepatitis, colitis, and pneumonitis, which is of particular concern in patients with HL. Most of the patients treated with CPI with HL had previously received agents known to cause pulmonary dysfunction such as radiotherapy, bleomycin, gemcitabine, carmustine, and BV. I would caution administering a CPI when a patient has a history of pneumonitis requiring steroid support for symptom control.

Summary/Recommendation: The CPI have been administered to patients with HL for the past 2 to 3 years and the OS of a patient with HL where ASCT failed has increased nearly the same amount. I will continue to use CPI liberally for palliation and keep patients on treatment until obvious disease progression ensues; however, a number of appealing clinical trials are underway combining CPI with other agents including BV and histone deacetylase (HDAC) inhibitors and, if available, I would recommend these options for my patients.

The case for RIC allo-HCT

The early studies of allo-HCT in patients with HL were reported before the RIC era; OS was poor because of treatment-related mortality.36  Therefore, most transplant physicians would agree that an ablative allo-HCT is no longer a viable treatment option in HL. There is clear evidence of the graft-versus-lymphoma effect in HL based on 2 lines of evidence: graft-versus-host disease after allo-HCT is associated with improved PFS and, second, donor lymphocyte infusions (DLI) as part of a T cell–depleted strategy reduces the relapse rate. In 2016, with the availability of umbilical cord and haplo-identical transplants, nearly all patients will have a donor.37  Therefore, if a patient is referred for RIC allo-HCT, this treatment will likely commence provided that the patient meets the standard criteria for an allo-HCT, including some evidence of chemosensitive disease. However, it raises a critical question: which patients, and at what time should a patient be referred?

To answer this question, one must consider the likelihood of cure after RIC. In 2016, registry data from the CIBMTR reported PFS and OS rates of only 20% and 37%, respectively, after a RIC for HL. If one evaluates RIC in the context of chemosensitive disease and performance status before transplant, the EBMT reported that 3-year PFS improved to 42% in favorable patients. We have data from MSKCC on 71 patients who underwent RIC for HL and reported that extranodal disease, less than CR at the time of RIC, and alemtuzumab-containing regimens were all associated with a poor outcome.38  However, patients without any of these risk factors have a 3-year PFS approaching 60%. Confusing the matter further, data from the University of London where all the patients (n = 116) were transplanted with a T cell–depleted, alemtuzumab-based RIC, there was no difference in outcome in patients who had chemosensitive disease pretransplant based on PET status at the time of conditioning, with 3-year PFS rates of 59.4% and 55.7%, respectively, for PET-positive vs PET-negative response (Figure 6).39  These data are helpful in the context of BV treatment as a bridge to RIC. In the pivotal phase 2 clinical trial leading to US Food and Drug Administration approval of BV, 34 of 102 patients achieved a PET-negative response.40  In addition, approximately one third of these CR patients have never relapsed. Therefore, patients achieving a CR to BV after a failed ASCT may benefit from BV treatment of up to 16 cycles and then can be monitored.

Figure 6.

1.4-year PFS according to pre-alloSCT Deauville score.

Figure 6.

1.4-year PFS according to pre-alloSCT Deauville score.

Close modal

Are the results better for RIC in patients who have not received a previous ASCT? There are no modern studies comparing ASCT vs RIC allo-HCT. However, it is clear that there are subgroups of patients that have been described here that have <30% chance of being cured with ASCT.

Summary/recommendation: A reasonable treatment paradigm is depicted in Figure 7.

Figure 7.

Reasonable algorithm for RIC allo-HCT.

Figure 7.

Reasonable algorithm for RIC allo-HCT.

Close modal

A negative pre-ASCT PET scan is critical in the curative outcome of rel/ref HL and the incorporation of BV into ST can improve this metric. Post-ASCT consolidation with BV should be reserved for patients with multiple risk factors because the AEs are significant (namely, neuropathy). The checkpoint inhibitors are now the standard treatment of palliation and the question of how long these agents should be given before allo-HCT demands prospective analysis.

Craig Moskowitz, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY 10065; e-mail: moskowic@mskcc.org.

1.
Press
OW
,
Li
H
,
Schöder
H
, et al
.
US Intergroup Trial of Response-Adapted Therapy for Stage III to IV Hodgkin Lymphoma Using Early Interim Fluorodeoxyglucose-Positron Emission Tomography Imaging: Southwest Oncology Group S0816
.
J Clin Oncol
.
2016
;
34
(
17
):
2020
-
2027
.
2.
Engert
A
.
ABVD or BEACOPP for Advanced Hodgkin Lymphoma
.
J Clin Oncol
.
2016
;
34
(
11
):
1167
-
1169
.
3.
Radford
J
,
Illidge
T
,
Barrington
S
.
PET-Directed Therapy for Hodgkin’s Lymphoma
.
N Engl J Med
.
2015
;
373
(
4
):
392
.
4.
Rancea
M
,
von Tresckow
B
,
Monsef
I
,
Engert
A
,
Skoetz
N
.
High-dose chemotherapy followed by autologous stem cell transplantation for patients with relapsed or refractory Hodgkin lymphoma: a systematic review with meta-analysis
.
Crit Rev Oncol Hematol
.
2014
;
92
(
1
):
1
-
10
.
5.
Hoppe
RT
,
Advani
RH
,
Ai
WZ
, et al
;
National comprehensive cancer network
.
Hodgkin lymphoma, version 2.2015
.
J Natl Compr Canc Netw
.
2015
;
13
(
5
):
554
-
586
.
6.
Radford
J
,
Illidge
T
,
Counsell
N
, et al
.
Results of a trial of PET-directed therapy for early-stage Hodgkin’s lymphoma
.
N Engl J Med
.
2015
;
372
(
17
):
1598
-
1607
.
7.
Perales
MA
,
Ceberio
I
,
Armand
P
, et al
;
American Society for Blood and Marrow Transplantation
.
Role of cytotoxic therapy with hematopoietic cell transplantation in the treatment of Hodgkin lymphoma: guidelines from the American Society for Blood and Marrow Transplantation
.
Biol Blood Marrow Transplant
.
2015
;
21
(
6
):
971
-
983
.
8.
Josting
A
,
Nogová
L
,
Franklin
J
, et al
.
Salvage radiotherapy in patients with relapsed and refractory Hodgkin’s lymphoma: a retrospective analysis from the German Hodgkin Lymphoma Study Group
.
J Clin Oncol
.
2005
;
23
(
7
):
1522
-
1529
.
9.
Josting
A
,
Müller
H
,
Borchmann
P
, et al
.
Dose intensity of chemotherapy in patients with relapsed Hodgkin’s lymphoma
.
J Clin Oncol
.
2010
;
28
(
34
):
5074
-
5080
.
10.
Moskowitz
CH
,
Nimer
SD
,
Zelenetz
AD
, et al
.
A 2-step comprehensive high-dose chemoradiotherapy second-line program for relapsed and refractory Hodgkin disease: analysis by intent to treat and development of a prognostic model
.
Blood
.
2001
;
97
(
3
):
616
-
623
.
11.
Moskowitz
AJ
,
Yahalom
J
,
Kewalramani
T
, et al
.
Pretransplantation functional imaging predicts outcome following autologous stem cell transplantation for relapsed and refractory Hodgkin lymphoma
.
Blood
.
2010
;
116
(
23
):
4934
-
4937
.
12.
Gentzler
RD
,
Patel
JD
.
Optimal first-line and maintenance treatments for advanced-stage nonsquamous non-small cell lung cancer
.
J Natl Compr Canc Netw
.
2014
;
12
(
6
):
889
-
897
.
13.
Smeltzer
JP
,
Cashen
AF
,
Zhang
Q
, et al
.
Prognostic significance of FDG-PET in relapsed or refractory classical Hodgkin lymphoma treated with standard salvage chemotherapy and autologous stem cell transplantation
.
Biol Blood Marrow Transplant
.
2011
;
17
(
11
):
1646
-
1652
.
14.
Adams
HJ
,
Kwee
TC
.
Prognostic value of pretransplant FDG-PET in refractory/relapsed Hodgkin lymphoma treated with autologous stem cell transplantation: systematic review and meta-analysis
.
Ann Hematol
.
2016
;
95
(
5
):
695
-
706
.
15.
Devillier
R
,
Coso
D
,
Castagna
L
, et al
.
Positron emission tomography response at the time of autologous stem cell transplantation predicts outcome of patients with relapsed and/or refractory Hodgkin’s lymphoma responding to prior salvage therapy
.
Haematologica
.
2012
;
97
(
7
):
1073
-
1079
.
16.
Cheson
BD
,
Fisher
RI
,
Barrington
SF
, et al
;
Alliance, Australasian Leukaemia and Lymphoma Group; Eastern Cooperative Oncology Group; European Mantle Cell Lymphoma Consortium; Italian Lymphoma Foundation; European Organisation for Research; Treatment of Cancer/Dutch Hemato-Oncology Group; Grupo Español de Médula Ósea; German High-Grade Lymphoma Study Group; German Hodgkin’s Study Group; Japanese Lymphorra Study Group; Lymphoma Study Association; NCIC Clinical Trials Group; Nordic Lymphoma Study Group; Southwest Oncology Group; United Kingdom National Cancer Research Institute
.
Recommendations for initial evaluation, staging, and response assessment of Hodgkin and non-Hodgkin lymphoma: the Lugano classification
.
J Clin Oncol
.
2014
;
32
(
27
):
3059
-
3068
.
17.
Moskowitz
CH
,
Matasar
MJ
,
Zelenetz
AD
, et al
.
Normalization of pre-ASCT, FDG-PET imaging with second-line, non-cross-resistant, chemotherapy programs improves event-free survival in patients with Hodgkin lymphoma
.
Blood
.
2012
;
119
(
7
):
1665
-
1670
.
18.
Santoro
A
,
Magagnoli
M
,
Spina
M
, et al
.
Ifosfamide, gemcitabine, and vinorelbine: a new induction regimen for refractory and relapsed Hodgkin’s lymphoma
.
Haematologica
.
2007
;
92
(
1
):
35
-
41
.
19.
Sureda
A
,
Arranz
R
,
Iriondo
A
, et al
;
Grupo Español de Linformas/Transplante Autólogo de Médula Osea Spanish Cooperative Group
.
Autologous stem-cell transplantation for Hodgkin’s disease: results and prognostic factors in 494 patients from the Grupo Español de Linfomas/Transplante Autólogo de Médula Osea Spanish Cooperative Group
.
J Clin Oncol
.
2001
;
19
(
5
):
1395
-
1404
.
20.
Brice
P
,
Bouabdallah
R
,
Moreau
P
, et al
.
Prognostic factors for survival after high-dose therapy and autologous stem cell transplantation for patients with relapsing Hodgkin’s disease: analysis of 280 patients from the French registry. Société Française de Greffe de Moëlle
.
Bone Marrow Transplant
.
1997
;
20
(
1
):
21
-
26
.
21.
Collins
GP
,
Parker
AN
,
Pocock
C
, et al
;
British Committee for Standards in Haematology; British Society of Blood and Marrow Transplantation
.
Guideline on the management of primary resistant and relapsed classical Hodgkin lymphoma
.
Br J Haematol
.
2014
;
164
(
1
):
39
-
52
.
22.
Bröckelmann
PJ
,
Müller
H
,
Casasnovas
O
, et al
.
Risk Factors and a Prognostic Score for Progression Free Survival after Treatment with Autologous Stem Cell Transplantation (ASCT) in Patients with Relapsed or Refractory Hodgkin Lymphoma (rrHL)
.
Blood
.
2015
;
126
:
1978
-
1978
.
23.
Shah
GL
,
Bantilan
KS
,
Verwys
SL
, et al
.
Risk Factors Predicting Outcomes for Primary Refractory Hodgkin Lymphoma Patients Treated with Salvage Chemotherapy and Autologous Stem Cell Transplantation
.
Blood
.
2015
;
126
:
520
-
520
.
24.
Castagna
L
,
Bramanti
S
,
Balzarotti
M
, et al
.
Predictive value of early 18F-fluorodeoxyglucose positron emission tomography (FDG-PET) during salvage chemotherapy in relapsing/refractory Hodgkin lymphoma (HL) treated with high-dose chemotherapy
.
Br J Haematol
.
2009
;
145
(
3
):
369
-
372
.
25.
Younes
A
,
Bartlett
NL
,
Leonard
JP
, et al
.
Brentuximab vedotin (SGN-35) for relapsed CD30-positive lymphomas
.
N Engl J Med
.
2010
;
363
(
19
):
1812
-
1821
.
26.
Moskowitz
CH
,
Nademanee
A
,
Masszi
T
, et al
;
AETHERA Study Group
.
Brentuximab vedotin as consolidation therapy after autologous stem-cell transplantation in patients with Hodgkin’s lymphoma at risk of relapse or progression (AETHERA): a randomised, double-blind, placebo-controlled, phase 3 trial
.
Lancet
.
2015
;
385
(
9980
):
1853
-
1862
.
27.
Moskowitz
AJ
,
Schöder
H
,
Yahalom
J
, et al
.
PET-adapted sequential salvage therapy with brentuximab vedotin followed by augmented ifosamide, carboplatin, and etoposide for patients with relapsed and refractory Hodgkin’s lymphoma: a non-randomised, open-label, single-centre, phase 2 study
.
Lancet Oncol
.
2015
;
16
(
3
):
284
-
292
.
28.
Chen
R
,
Palmer
JM
,
Martin
P
, et al
.
Results of a Multicenter Phase II Trial of Brentuximab Vedotin as Second-Line Therapy before Autologous Transplantation in Relapsed/Refractory Hodgkin Lymphoma
.
Biol Blood Marrow Transplant
.
2015
;
21
(
12
):
2136
-
2140
.
29.
Armand
P
,
Shipp
MA
,
Ribrag
V
, et al
.
PD-1 Blockade with Pembrolizumab in Patients with Classical Hodgkin Lymphoma after Brentuximab Vedotin Failure: Safety, Efficacy, and Biomarker Assessment
.
Blood
.
2015
;
126
:
584
-
584
.
30.
LaCasce
AS
,
Bociek
G
,
Sawas
A
, et al
.
Brentuximab Vedotin Plus Bendamustine: A Highly Active Salvage Treatment Regimen for Patients with Relapsed or Refractory Hodgkin Lymphoma
.
Blood
.
2015
;
126
:
3982
-
3982
.
31.
Garcia-Sanz
R
,
Sureda
A
,
Alonso-Alvarez
S
, et al
.
Evaluation of the Regimen Brentuximab Vedotin Plus ESHAP (BRESHAP) in Refractory or Relapsed Hodgkin Lymphoma Patients: Preliminary Results of a Phase I-II Trial from the Spanish Group of Lymphoma and Bone Marrow Transplantation (GELTAMO)
.
Blood
.
2015
;
126
:
582
-
582
.
32.
Linch
DC
,
Winfield
D
,
Goldstone
AH
, et al
.
Dose intensification with autologous bone-marrow transplantation in relapsed and resistant Hodgkin’s disease: results of a BNLI randomised trial
.
Lancet
.
1993
;
341
(
8852
):
1051
-
1054
.
33.
Schmitz
N
,
Pfistner
B
,
Sextro
M
, et al
;
German Hodgkin’s Lymphoma Study Group; Lymphoma Working Party of the European Group for Blood and Marrow Transplantation
.
Aggressive conventional chemotherapy compared with high-dose chemotherapy with autologous haemopoietic stem-cell transplantation for relapsed chemosensitive Hodgkin’s disease: a randomised trial
.
Lancet
.
2002
;
359
(
9323
):
2065
-
2071
.
34.
Roemer
MG
,
Advani
RH
,
Ligon
AH
, et al
.
PD-L1 and PD-L2 Genetic Alterations Define Classical Hodgkin Lymphoma and Predict Outcome
.
J Clin Oncol
.
2016
;
34
(
23
):
2690
-
2697
.
35.
Ansell
SM
,
Lesokhin
AM
,
Borrello
I
, et al
.
PD-1 blockade with nivolumab in relapsed or refractory Hodgkin’s lymphoma
.
N Engl J Med
.
2015
;
372
(
4
):
311
-
319
.
36.
Sureda
A
,
Robinson
S
,
Canals
C
, et al
.
Reduced-intensity conditioning compared with conventional allogeneic stem-cell transplantation in relapsed or refractory Hodgkin’s lymphoma: an analysis from the Lymphoma Working Party of the European Group for Blood and Marrow Transplantation
.
J Clin Oncol
.
2008
;
26
(
3
):
455
-
462
.
37.
Peggs
KS
,
Kayani
I
,
Edwards
N
, et al
.
Donor lymphocyte infusions modulate relapse risk in mixed chimeras and induce durable salvage in relapsed patients after T-cell-depleted allogeneic transplantation for Hodgkin’s lymphoma
.
J Clin Oncol
.
2011
;
29
(
8
):
971
-
978
.
38.
Barba
P
,
Burns
LJ
,
Litzow
MR
, et al
;
American Society for Blood and Marrow Transplantation Committee on Education
.
Success of an International Learning Health Care System in Hematopoietic Cell Transplantation: The American Society of Blood and Marrow Transplantation Clinical Case Forum
.
Biol Blood Marrow Transplant
.
2016
;
22
(
3
):
564
-
570
.
39.
Reyal
Y
,
Kayani
I
,
Bloor
AJ
, et al
.
Impact of Pretransplantation (18)F-Fluorodeoxyglucose-Positron Emission Tomography on Survival Outcomes after T Cell-Depleted Allogeneic Transplantation for Hodgkin Lymphoma
.
Biol Blood Marrow Transplant
.
2016
;
22
(
7
):
1234
-
1241
.
40.
Gopal
AK
,
Chen
R
,
Smith
SE
, et al
.
Durable remissions in a pivotal phase 2 study of brentuximab vedotin in relapsed or refractory Hodgkin lymphoma
.
Blood
.
2015
;
125
(
8
):
1236
-
1243
.

Competing Interests

Conflict-of-interest disclosure: The author has received research funding from Seattle Genetics and Merck, and has consulted for Seattle Genetics, Merck, and BMS.

Author notes

Off-label drug use: None disclosed.