Your activity: 6 p.v.

Treatment of relapsed or refractory classic Hodgkin lymphoma

Treatment of relapsed or refractory classic Hodgkin lymphoma
Author:
Ann S LaCasce, MD
Section Editors:
George P Canellos, MD
Arnold S Freedman, MD
Deputy Editor:
Alan G Rosmarin, MD
Literature review current through: Dec 2022. | This topic last updated: Dec 18, 2020.

INTRODUCTION — Most patients with Hodgkin lymphoma (HL; formerly called Hodgkin's disease) will attain a complete remission after initial treatment and achieve long-term disease control (ie, cure). However, relapse may occur in 10 to 15 percent of patients with favorable prognosis early (stages I to II) HL and in 15 to 30 percent of patients with more advanced HL [1-7]. Approximately 10 to 15 percent of patients have refractory disease that either does not respond to initial therapy or progresses after an initial partial response.

The goal of treatment of relapsed or refractory (r/r) HL should be to achieve long-term disease control while limiting toxicity and complications of therapy. Salvage chemotherapy can achieve a complete response in more than half of patients with first relapse of HL or refractory disease, but long-term disease-free survival generally requires autologous hematopoietic cell transplantation (HCT). (See "Determining eligibility for autologous hematopoietic cell transplantation".)

Treatment of r/r HL with chemotherapy, radiation therapy, and immunotherapy will be reviewed here. The role of HCT in HL is described separately. (See "Hematopoietic cell transplantation in classic Hodgkin lymphoma".)

SPECIAL CONSIDERATIONS DURING THE COVID-19 PANDEMIC — The coronavirus disease 2019 (COVID-19) pandemic has increased the complexity of cancer care. Important issues include balancing the risk from treatment delay versus harm from COVID-19, ways to minimize negative impacts of social distancing during care delivery, and appropriately and fairly allocating limited health care resources. These issues and recommendations for cancer care during the COVID-19 pandemic are discussed separately.

(See "COVID-19: Considerations in patients with cancer".)

DEFINITIONS — In this discussion, the following definitions will apply:

Relapse (or recurrence) is the reappearance of disease at sites of prior disease and/or at new sites after achievement of complete response (CR).

Progression refers to evidence of increasing disease after achieving a stable partial response (PR).

Refractory disease is a failure to achieve an initial CR or PR, and may be described as either primary (failure to respond to initial therapy) or secondary (initial response, but failure to respond after disease relapse).

Progressive disease or relapse usually presents with symptoms related to the enlargement of lymph nodes, liver, or spleen. Progression or relapse are recognized as a 50 percent increase in the longest diameter of a previously identified lesion, appearance of a new site of disease, or new/recurrent involvement of the bone marrow (table 1).

Among patients with relapsed or refractory HL, approximately one-third each are classified as follows [7,8]:

Relapsed less than one year after initial CR

Relapsed more than one year after initial CR

Refractory disease

CONFIRMING REFRACTORY/RELAPSED DISEASE — It is important to confirm the diagnosis of relapsed HL, and to determine if apparent residual or progressive disease indeed represents refractory HL.

Detection of relapsed or refractory disease — The patient treated for HL should be monitored for disease response during treatment, assessed for maintenance of remission following completion of therapy, and evaluated for complications of treatment. The nature and frequency of such evaluations are described separately. (See "Monitoring of the patient with classic Hodgkin lymphoma during and after treatment".)

Most recurrences of HL present with symptoms and/or a mass lesion detected on physical examination. As an example, one study of 210 patients observed that 30 of 37 (81 percent) relapsing patients were symptomatic; in only four cases was relapse detected as a result of routine physical examination of an asymptomatic patient [8].

It is important to distinguish complete response from partial response in patients with residual radiographic abnormalities after initial treatment of HL, in order to distinguish fibrosis from persistent (ie, refractory) HL and thereby avoid unnecessary therapy. This may include positron emission tomography/computed tomographic (PET/CT) scanning to distinguish residual fibrosis/necrosis from persistent disease, and/or biopsy of the suspicious mass.

Biopsy — Diagnosis of relapsed HL generally requires biopsy of a suspicious mass. Clinical judgment often determines whether or not a biopsy is required, but it is sound practice to obtain a tissue diagnosis in most patients. This is especially true in the setting of late relapse, since there is a significant risk for second cancers (non-Hodgkin lymphoma or solid tumors) in these patients. In an instructive report, four patients with HL and PET-avid nodal enlargement at 8 to 48 months post-treatment were found to have benign sarcoid-like lesions [9]. (See "Second malignancies after treatment of classic Hodgkin lymphoma".)

A repeat biopsy may not be necessary in patients with early recurrence in the setting of incomplete remission, especially with persistence of constitutional symptoms. However, if the disease was unusually resistant to therapy, a biopsy may be warranted to confirm the initial diagnosis of HL and exclude other diseases.

Staging — Restaging is recommended at the time of relapse, especially if there appears to be an isolated relapse. Restaging is important for assessing prognosis and for guiding treatment, including selection and duration of chemotherapy, use of radiation therapy, and decisions relating to hematopoietic cell transplantation (HCT) and/or immunotherapy.

Various restaging schemes have been proposed for relapsed HL, but the most straightforward approach employs the criteria of the Ann Arbor staging system with Cotswolds modification to describe the extent of disease at the time of relapse and substitutes the letters RS (relapse stage) for CS (clinical stage) or PS (pathological stage) (table 2). This is discussed in more detail separately. (See "Pretreatment evaluation, staging, and treatment stratification of classic Hodgkin lymphoma" and "Monitoring of the patient with classic Hodgkin lymphoma during and after treatment", section on 'PET/CT scan'.)

Prognosis — The effectiveness of salvage therapy for relapsed HL is influenced by clinical characteristics such as age, stage, symptoms, duration of first remission, anemia, and extent of prior therapy [10-13].

As an example, the German Hodgkin Lymphoma Study Group (GHSG) identified the following adverse risk factors among 422 patients relapsing after primary radiation therapy or chemotherapy [13]:

Time to recurrence ≤12 months

Stage III or IV disease at relapse

Decreased hemoglobin (<10.5 or <12.0 g/dL in females and males, respectively)

These risk factors were incorporated into a relapsed HL prognostic score. Actuarial four-year overall survival for patients relapsing after chemotherapy was 83, 66, 36, and 27 percent for those with zero, one, two, or three unfavorable factors, respectively. Subsequent studies by the GHSG confirmed the adverse prognostic value of these risk factors [14,15].

The likelihood of achieving a complete response and a sustained remission diminishes with second and subsequent relapses.

Outcomes following treatment for refractory HL are even less favorable than following relapse. As an example, an Italian study found that only 8 percent of patients with refractory HL were alive at eight years [10]. However, among patients who are able to undergo autologous HCT, long-term survival can approach 50 percent [16].

PRINCIPLES OF MANAGEMENT

Overview of management — The goal of treatment of relapsed or refractory (r/r) HL should be to achieve long-term disease control (ie, cure) while limiting toxicity and complications of therapy [17,18]. In most cases, long-term disease-free survival requires autologous hematopoietic cell transplantation (HCT) [19,20]. (See "Hematopoietic cell transplantation in classic Hodgkin lymphoma".)

Patients with first relapse of HL or refractory HL are generally treated with intensive combination chemotherapy ("salvage chemotherapy") or targeted chemotherapy (table 3); those who demonstrate a complete response upon restaging by positron emission tomography/computed tomographic (PET/CT) proceed to autologous HCT, if eligible (algorithm 1). (See 'Choice of chemotherapy regimen' below and 'Management of first relapse or refractory HL' below.)

For patients who are not eligible for HCT, treatment decisions must take into consideration the reasons for ineligibility (eg, the type and severity of comorbid illnesses) and the goals of therapy (eg, long-term disease control, palliation of symptoms, prolongation of survival). In this setting, treatment may entail either single agent chemotherapy or combination chemotherapy (as guided by comorbidities), targeted chemotherapy, immunotherapy, and/or radiation therapy (RT). (See 'HCT-ineligible patients' below.)

Patients with a second or subsequent relapse may be candidates for second-line salvage chemotherapy, targeted chemotherapy, immunotherapy, or HCT (if not previously performed). (See 'Second and subsequent relapses' below.)

The therapeutic strategy and sequencing of treatment for r/r HL is influenced by the following factors:

Eligibility for HCT (see "Determining eligibility for autologous hematopoietic cell transplantation" and 'HCT-ineligible patients' below)

Functional response (based on PET/CT examination) to salvage chemotherapy (table 1 and table 2)

Bulky disease (ie, mediastinal disease greater than one-third of thoracic diameter, or mass greater than 10 cm) (see 'Indications for radiation' below)

Duration of remission (see 'Late first relapse' below)

Prior therapy, including chemotherapy, targeted chemotherapy, immunotherapy, HCT, and/or RT

Comorbid illnesses that might limit treatment options

Importance of minimizing disease burden — Long-term survival in the setting of r/r HL is most likely if a complete response (CR) is achieved, as defined by a negative PET/CT (table 1 and table 4). However, some patients with a partial response (PR) to salvage chemotherapy can be cured with autologous HCT.

For most patients, salvage chemotherapy should be followed by autologous HCT. Those who achieve a CR proceed directly with HCT.

For patients who achieve PR to salvage therapy, we favor using further salvage chemotherapy, radiation therapy, or targeted chemotherapy (eg, brentuximab vedotin) to minimize disease burden prior to HCT (algorithm 1). (See 'Management of first relapse or refractory HL' below.)

The importance of achieving a CR prior to transplantation is supported by experience with PET/CT scans obtained after salvage chemotherapy, but prior to autologous HCT. In one series, CR on PET scan had a positive predictive value of 93 percent for progression-free survival (PFS) at two years; in contrast, the majority of PET-positive patients relapsed despite high dose therapy [21].

However, an acceptable option for patients who achieve a PR or lesser response is proceeding directly to autologous HCT rather than treating with a third line of chemotherapy (with or without RT) [22]. It is not clear whether additional therapy prior to autologous HCT improves outcomes. A trial randomly assigned 241 patients with relapsed HL to salvage therapy with DHAP (dexamethasone, cytarabine, and cisplatin) followed by autologous HCT, versus DHAP followed by sequential salvage chemotherapy (cyclophosphamide, methotrexate, and etoposide) prior to autologous HCT [23]. The more intensive treatment regimen caused greater toxicity, but did not improve freedom from treatment failure or overall survival (OS).

Choice of chemotherapy regimen — There is no agreed-upon preferred salvage chemotherapy regimen for r/r HL and clinical practice varies widely (table 3). We generally prefer salvage therapy with ICE (ifosfamide, carboplatin, etoposide) for most patients who are candidates for HCT. Our preference for ICE is largely based on a comparable efficacy to other salvage regimens, its favorable toxicity profile compared with some other treatments, and our familiarity with the protocol.

Most patients are treated with two cycles of salvage chemotherapy, with or without RT, followed by autologous HCT [24,25]. Decisions to incorporate RT, alternative salvage chemotherapy, targeted chemotherapy, and/or immunotherapy are addressed in the sections below, and in the algorithm (algorithm 1).

The most common salvage chemotherapy regimens for r/r HL are:

ICE (ifosfamide, carboplatin, etoposide) [26-28] – Hematologic toxicity is universal with approximately 35 percent of patients requiring transfusion of blood products. Severe (grade 3/4) nonhematologic side effects include infection (23 percent) and nephrotoxicity (1 percent). Stem cell mobilization appears adequate.

Gemcitabine-containing regimens – The most common regimens are:

GVD (gemcitabine, vinorelbine, pegylated liposomal doxorubicin) [29]

GDP (gemcitabine, dexamethasone, cisplatin) [30]

BeGEV (bendamustine, gemcitabine, vinorelbine) [31]

Hematologic toxicity is universal with these gemcitabine-containing regimens. Febrile neutropenia is seen in approximately 15 percent of patients. Stem cell mobilization for autologous HCT is generally adequate after these regimens.

DHAP (dexamethasone, cytarabine, cisplatin) [32] – Hematologic toxicity is universal with more than half of patients requiring transfusion of blood products. Severe (grade 3/4) nonhematologic side effects include infection (24 percent) and nephrotoxicity (6 percent). Stem cell mobilization appears adequate.

Targeted chemotherapy (eg, brentuximab vedotin [BV] plus bendamustine) – Toxicity includes cytopenias and infections, as described below. (See 'Brentuximab vedotin' below.)

There are no randomized trials that directly compare the various salvage regimens for relapsed HL. In phase II studies, response rates for these regimens generally ranged from 60 to 85 percent, but comparison is difficult since most studies involve small numbers of patients with variable clinical status, and many were subsequently treated with HCT.

Toxicity of these regimens is substantial, and patient comorbidities may influence the selection of a particular chemotherapeutic regimen. Hematologic toxicity is common, and many patients will require antibiotics for fever/infection or transfusion with red blood cells and/or platelets.

The ICE regimen is usually administered in the inpatient setting (or may be given in the outpatient setting for select patients) over three days with a cycle length of 14 or 21 days (as permitted by recovery of blood counts) [27]:

Ifosfamide 5 g/m2 with equivalent dose of MESNA given over 24 hours by continuous infusion on day 2

Carboplatin to an area under the curve (AUC) of 5 with a maximum of 800 mg on day 2

Etoposide 100 mg/m2/day on days 1 to 3

Granulocyte colony-stimulating factor (G-CSF) 5 mcg/kg daily on days 5 to 12

Dose adjustments may be necessary for pre-existing renal or liver dysfunction. In addition to monitoring for kidney dysfunction, electrolyte abnormalities, and cytopenias, urine dipstick testing is performed to evaluate for hematuria, a potential sign of hemorrhagic cystitis. ICE is moderately emetogenic; antiemetic prophylaxis is indicated.

For HL that is refractory to salvage therapy with ICE or that relapses after ICE, we generally treat with GVD (algorithm 1). (See 'Second and subsequent relapses' below.)

An acceptable alternative for patients whose disease is refractory to or relapses after ICE is to proceed directly to autologous HCT. (See 'Importance of minimizing disease burden' above.)

For patients with significant comorbid illnesses (eg, those who are considered ineligible for HCT because of comorbidities), we may instead treat with single agent chemotherapy (eg, gemcitabine), targeted chemotherapy, RT (especially if the disease can be encompassed in a radiation port), or immunotherapy with the goal of achieving CR, or to palliate symptoms and extend life. (See 'HCT-ineligible patients' below.)

Hematopoietic cell transplantation (HCT)

Autologous HCT — Autologous HCT should be considered the treatment of choice for most patients with r/r HL (algorithm 1). (See "Hematopoietic cell transplantation in classic Hodgkin lymphoma".)

Sustained remissions after HCT can be achieved in more than half of patients with r/r HL [33,34]. In contrast, long-term disease-free survival is uncommon following salvage chemotherapy (with or without RT) that is not followed by HCT [10-12,35]; the primary exception is patients who experience a late, asymptomatic, isolated relapse of HL. (See 'Late first relapse' below.)

Benefits from HCT compared with chemotherapy alone are especially marked in the following categories of patients:

Early relapse (<12 months after completion of initial therapy)

Generalized systemic relapse, or the presence of other poor prognostic markers

Second (or later) relapse

Refractory HL (ie, induction failure after initial chemotherapy)

Clinical trials that have compared HCT versus salvage chemotherapy (with or without RT) without HCT include the following:

In a randomized study from the German Hodgkin Lymphoma Study Group and the European Group for Blood and Marrow Transplantation, 161 patients with relapsed HL were treated with two cycles of chemotherapy (dexa-BEAM: dexamethasone, carmustine, etoposide, cytarabine, melphalan) [36]. Those with chemosensitive disease were treated with either high dose BEAM followed by autologous HCT or two additional courses of dexa-BEAM without subsequent HCT. At a median follow-up of 39 months:

OS was similar in the two treatment arms

Freedom from treatment failure at three years favored HCT over chemotherapy alone (55 versus 34 percent, respectively)

A prospective, nonrandomized GELA (Groupe d'Etude des Lymphomes de l'Adulte) trial enrolled 157 patients with stage IIIB or IV HL who had refractory disease, PR, or early relapse following initial chemotherapy [37]. Patients were treated with MINE (mesna, ifosfamide, mitoxantrone, etoposide) salvage chemotherapy, with or without subsequent autologous HCT. At a median follow-up of 50 months, the following observations were made:

Five-year estimated OS rates were 71 and 32 percent for patients undergoing HCT or not undergoing autologous HCT, respectively.

Salvage chemotherapy without HCT was an independent predictor of poor OS in multivariate analysis; "B" symptoms at the time of disease progression and refractory disease before HCT were also associated with shorter OS.

Allogeneic HCT — Allogeneic HCT may be offered to patients with HL as a salvage therapy following relapse or progression after autologous HCT. The use of allogeneic HCT in this setting is discussed in more detail separately. (See "Hematopoietic cell transplantation in classic Hodgkin lymphoma" and "Hematopoietic cell transplantation in classic Hodgkin lymphoma", section on 'Allogeneic transplantation'.)

Caution should be exercised in considering allogeneic HCT in patients who previously received a PD-1 inhibitor (nivolumab or pembrolizumab) because of reports of increased toxicity, as discussed below. (See 'GVHD with PD-1 blockade and allogeneic HCT' below.)

Targeted chemotherapy and immunotherapy — The antibody-drug conjugate, brentuximab vedotin, preferentially delivers chemotherapy to CD30-expressing cells, including the Reed-Sternberg cells of HL. Immunotherapy approaches include strategies to overcome evasion of immune surveillance (ie, immune checkpoint blockade) and can achieve high rates of response in r/r HL. It remains unclear if such treatments can achieve long-term remissions and obviate the need for subsequent HCT.

Brentuximab vedotin — Brentuximab vedotin (BV) is an antibody-drug conjugate that comprises an anti-CD30 antibody linked to the anti-tubulin agent, monomethyl auristatin E [38]. In the United States and Europe, BV is approved for the treatment of patients with HL after failure of autologous HCT or after failure of at least two prior multi-agent chemotherapy regimens in patients who are not candidates for HCT. A subset of patients achieved sustained, unmaintained remissions, even in the absence of transplantation [39], while others were able to proceed with reduced intensity allogeneic HCT [40]. Use of BV in patients who have already completed autologous HCT is presented in more detail separately. (See "Hematopoietic cell transplantation in classic Hodgkin lymphoma", section on 'Maintenance therapy'.)

A meta-analysis of six studies (302 patients) reported 61 percent overall response (OR) and 38 percent CR to single agent BV [41]. BV is also effective when combined with other chemotherapy agents (eg, BV plus bendamustine) [42,43].

Informative studies include:

A multicenter phase II study reported 75 percent OR (34 percent CR) among 102 patients with r/r HL following autologous HCT who received BV (1.8 mg/kg every three weeks for up to 16 cycles) [39,44,45]. For the 34 patients who achieved CR, estimated five-year OS and PFS rates were 64 and 52 percent, respectively. Thirteen of these patients (38 percent of patients who achieved CR) remained in remission at the close of study, including nine who remained in sustained remission without any further anticancer therapy or allogeneic HCT.

Two phase I/II studies that treated patients with r/r HL with BV plus bendamustine reported high rates of OR (93 and 78 percent) and CR (74 and 43 percent) [42,43].

Infusion reactions with BV are uncommon, but the rates of reactions are higher when BV is given in combination with bendamustine; this can be mitigated by the use of pretreatment steroids and antihistamines on each day of therapy. Anaphylaxis to BV has been reported, and infections, cytopenias, gastrointestinal complications (eg, hemorrhage, perforation, obstruction), and other adverse effects have been reported [46]. Progressive multifocal leukoencephalopathy and acute pancreatitis are rare complications of brentuximab treatment [47-49]. (See "Neurologic complications of cancer treatment with molecularly targeted and biologic agents" and "Progressive multifocal leukoencephalopathy (PML): Epidemiology, clinical manifestations, and diagnosis".)

PD-1 blockade

Efficacy — Programmed death 1 (PD-1) ligands, PD-L1 and PD-L2, are overexpressed by Hodgkin Reed-Sternberg (HRS) cells in classic HL (cHL), leading to evasion of immune surveillance. Small prospective studies have reported high response rates with PD-1 inhibitors (eg, nivolumab, pembrolizumab), but further study is required to characterize the durability of treatment.

In the United States and Europe, nivolumab is approved for patients with cHL that relapsed or progressed after autologous HCT and post-transplantation brentuximab vedotin (BV). Pembrolizumab is approved in the United States for patients whose disease is refractory or has relapsed after two or more lines of therapy, and in the European Union for patients with progression after autologous HCT and BV, or who are transplant-ineligible and have failed BV. Pembrolizumab is approved for administration as 400 mg every six weeks or 200 mg every three weeks by the US Food and Drug Administration [50].

Adverse events reported with anti-PD-1 monoclonal antibodies include pneumonitis, colitis, hepatitis, thyroid dysfunction, nephritis and renal dysfunction. Rash, pruritus, fatigue, fever, nausea, diarrhea, and metabolic abnormalities (eg, hypophosphatemia, hypercalcemia, and increased lipase levels) are also frequent but generally mild (grade 1/2). Toxicity associated with these agents is discussed separately. (See "Toxicities associated with checkpoint inhibitor immunotherapy".)

Support for anti-PD-1 monoclonal antibody therapy in this setting comes from several prospective trials of heavily pretreated patients:

A multicenter phase II trial of nivolumab reported OR and CR rates of 69 and 16 percent, respectively, among 243 patients with r/r HL, of whom 180 had previously received BV and autologous HCT [51]. Median duration of response and median PFS were 17 and 15 months, respectively.

A phase II study of nivolumab in 80 adults with r/r HL after autologous HCT and/or BV reported a 66 percent OR rate (9 percent CR), with a median duration of response of 7.8 months [52].

The phase II KEYNOTE-087 study reported that pembrolizumab (200 mg every three weeks) was effective for BV-naïve patients and for those treated with BV before or after HCT [53]. The study reported 72 percent OR, including 28 percent CR, in 210 heavily pretreated patients with r/r HL. For two-thirds of patients who achieved CR, at least one year of treatment was required. Responses were durable, with 14 month median PFS; two-year PFS was >60 percent among patients who achieved CR. Grade ≥3 treatment-related toxicity was reported in 11 percent, and none led to death; compared with an earlier report of this study, no unexpected toxicity was noted with more than two years of follow-up [54].

Use of PD-1 blockade as maintenance therapy after autologous HCT for r/r cHL is discussed separately. (See "Hematopoietic cell transplantation in classic Hodgkin lymphoma", section on 'PD-1 blockade maintenance'.)

GVHD with PD-1 blockade and allogeneic HCT — Patients treated with nivolumab or pembrolizumab who proceed to allogeneic HCT have a high rate of transplant-related complications, such as hyperacute graft-versus-host disease (GVHD), severe acute GVHD, steroid-requiring febrile syndrome, hepatic sinusoidal obstruction syndrome, and other immune-related adverse reactions. Conversely, there are reports of acute GVHD developing with nivolumab treatment in patients who had previously undergone allogeneic HCT. (See "Toxicities associated with checkpoint inhibitor immunotherapy", section on 'Combining or sequencing immunotherapy with other therapies'.)

Acute GVHD developed in 6 of 20 (30 percent) who were treated with nivolumab after having previously relapsed following allogeneic HCT; GVHD began within one week after the first infusion of nivolumab [55]. All six patients had a prior history of acute GVHD. Two patients died as a result of GVHD; one died with progressive disease and one died with complications related to a second allogeneic HCT. The OR to nivolumab was 95 percent and median PFS was not reached after more than one year of observation. Another retrospective study reported outcomes in 39 patients with hematologic malignancies who were treated with a PD-1 inhibitor and subsequently underwent allogeneic HCT (all but three with reduced intensity conditioning) [56]. There was a higher than expected rate of severe complications (four treatment-related deaths; one from hepatic sinusoidal obstruction syndrome, three from early acute GVHD), and seven patients developed a noninfectious febrile syndrome that required treatment with steroids. Additional study in larger groups of patients is needed to determine whether prior treatment with a PD-1 inhibitor increases the toxicities of allogeneic HCT.

CAR-T cell therapy — Autologous T cells that bear a chimeric antigen receptor (CAR) targeted against CD30 can achieve a high rate of durable responses in patients treated with lymphodepleting conditioning therapy, with only modest toxicity.

Anti-CD30 CAR-T cells (from 2 x 107 to 2 x 108 cells/m2) were administered to 32 heavily-pretreated, evaluable patients after treatment with one of three different lymphodepleting preconditioning regimens [57]. The overall response rate was 72 percent (including 59 percent CR) and, with median follow-up of nearly 1.5 years, one-year PFS was 36 percent and one-year OS was 94 percent. Neutropenia or thrombocytopenia that did not resolve by day 28 were reported in 10 and 24 percent of patients, respectively. Cytokine release syndrome (CRS) was reported in 24 percent, but all were grade 1; no neurologic toxicity was observed. In an earlier study, administration of CAR-T cells that did not include lymphodepleting preconditioning therapy was associated with only limited activity (33 percent overall response rate) [58].

Radiation therapy (RT)

Indications for radiation — The decision to offer RT must balance the potential improvement of local disease control against the potential for long-term toxicity, including second malignancies and cardiovascular morbidity. Our decision to offer RT depends on the following clinical factors (algorithm 1):

For patients undergoing autologous HCT – We offer RT in order to achieve CR prior to HCT or to consolidate the CR in patients who had bulky disease at the time of relapse, as follows:

Consolidative RT may be administered prior to transplantation for patients who had a PR (by PET/CT) to salvage chemotherapy with residual disease that can be encompassed in a radiation port.

Consolidative RT may be used after autologous HCT for patients who had bulky disease at the time of relapse and for select patients with non-bulky disease that can be encompassed in a radiation field.

There are no randomized trials indicating a survival benefit, but several studies have shown that adjuvant irradiation can control limited residual disease and may contribute to improved prognosis. This is discussed separately. (See "Hematopoietic cell transplantation in classic Hodgkin lymphoma", section on 'Radiation of localized or bulky disease'.)

Late first relapse – Salvage chemotherapy followed by RT may be an option for select patients with a late (generally at least several years after completion of initial treatment), localized, asymptomatic, first recurrence of HL. (See 'Late first relapse' below.)

Palliation without curative intent – RT, with or without systemic chemotherapy or targeted chemotherapy, may provide relief of symptoms in select patients who are not able to undergo chemotherapy and/or HCT with curative intent.

Can radiation be given alone? — In general, long-term control of relapsed or refractory HL requires systemic chemotherapy, with or without RT, followed by HCT (except as described above). (See 'Overview of management' above.)

RT is rarely used as the only salvage treatment for relapsed HL. There are some reports of success in highly selected patients with favorable prognostic criteria (eg, long disease-free intervals, absence of extranodal sites, and no B symptoms) whose disease can be encompassed in a reasonable radiation port [59-61].

A retrospective analysis of 100 patients who received salvage RT reported a 77 percent CR rate for the 80 patients who received either mantle or involved field RT, but the five-year freedom from progression (FFP) rate was only 28 percent [62]. This low FFP rate indicates that RT salvage alone is not generally sufficient for long-term disease control.

SPECIFIC CLINICAL SCENARIOS

Management of first relapse or refractory HL — A guiding principle for our approach to managing first relapse of HL or refractory disease is to attempt to achieve a complete response (CR) and then proceed to autologous hematopoietic cell transplantation (HCT). In some cases, achieving CR may entail the sequential use of systemic chemotherapy, radiation therapy (RT), targeted chemotherapy, or immunotherapy to achieve CR prior to HCT (algorithm 1).

Management of HCT-ineligible patients is described below. (See 'HCT-ineligible patients' below.)

Salvage chemotherapy (often with RT) is an acceptable alternative to HCT in select patients with a late (generally at least several years after completion of initial treatment), localized (ie, single site of relapse), asymptomatic, first relapse of HL. (See 'Late first relapse' below.)

We initially treat relapsed or refractory HL with two cycles of salvage therapy with ICE (ifosfamide, carboplatin, etoposide). Alternative regimens for initial salvage chemotherapy may be selected based on the presence of comorbid illnesses, toxicities, and other clinical features. (See 'Choice of chemotherapy regimen' above.)

Three weeks after completion of salvage therapy with ICE, we perform a positron emission tomography/computed tomographic (PET/CT) examination to determine response (algorithm 1 and table 1).

Subsequent therapy is guided by PET/CT results, as follows:

CR (Deauville 1 to 3):

HCT-eligible patients should proceed to autologous HCT. (See "Determining eligibility for autologous hematopoietic cell transplantation" and "Hematopoietic cell transplantation in classic Hodgkin lymphoma".)

Those who had bulky disease at the time of relapse, or localized disease that was not previously irradiated, may receive involved-site RT consolidation after HCT. (See 'Indications for radiation' above.)

These patients should be evaluated for brentuximab vedotin maintenance therapy after HCT. (See "Hematopoietic cell transplantation in classic Hodgkin lymphoma", section on 'Maintenance therapy'.)

Partial response (PR, Deauville 4 or 5):

For patients with residual disease after two cycles of ICE, we generally treat with two cycles of second-line salvage chemotherapy (eg, GVD [gemcitabine, vinorelbine, pegylated liposomal doxorubicin] or other alternative salvage regimens) (see 'Choice of chemotherapy regimen' above). Other experts offer HCT to select patients at this time rather than proceeding with further chemotherapy [22].

PET/CT is performed three weeks later, and management is guided by the results, as follows:

CR (Deauville 1 to 3): HCT-eligible patients should proceed to autologous HCT.

PR (Deauville 4 or 5) or progressive disease:

Disease that can be encompassed in a radiation port is treated with involved-site RT. PET/CT is repeated three weeks after completion of RT. We proceed to HCT if there is no evidence of progression outside of the radiation field; note that PET/CT may demonstrate persistent inflammation-related activity within the radiation field at this point in time. (See 'Radiation therapy (RT)' above.)

For disease that cannot be encompassed in a radiation port, we treat with alternative salvage chemotherapy (eg, DHAP [dexamethasone, cytarabine, cisplatin]), targeted chemotherapy (eg, brentuximab vedotin), or immunotherapy (eg, PD-1 blockade). (See 'Choice of chemotherapy regimen' above and 'Targeted chemotherapy and immunotherapy' above.)

-For patients who achieve CR by PET/CT after completing this therapy, we proceed to HCT.

-For patients who achieve PR after completing this therapy, we treat with involved-site RT (if disease can be encompassed in a radiation field) prior to HCT.

-For patients with PD, we treat with RT (if appropriate) and may offer targeted chemotherapy, immunotherapy, or HCT, if the patient is eligible for these treatments; if not, we focus on palliation of symptoms.

Progressive disease (PD): We treat with serial systemic therapies aimed at achieving CR. Acceptable options include alternative salvage chemotherapy (single agent or combination), targeted chemotherapy, immunotherapy, proceeding to autologous HCT, or participation in a clinical trial.

For some patients, the goal of relieving symptoms or palliative care may be the proper course of action. (See 'Clinical trials' below.)

HCT-ineligible patients — Patients who are ineligible for HCT because of comorbid illnesses or advanced age (the age of eligibility for HCT is usually determined by treating institution) are treated with the goal of achieving CR and long-term disease control; in some cases the goals may be relief of symptoms and prolongation of survival. Selection of systemic treatment may be influenced by likelihood of tolerance of salvage therapy. As examples, we usually offer brentuximab vedotin or single agent gemcitabine, rather than intensive combination chemotherapy, such as ICE. Serial administration of single agent chemotherapy regimens may offer benefit with tolerable toxicity. (See 'Second and subsequent relapses' below.)

This may be followed by RT, if appropriate, with the goal of achieving CR.

Following completion of therapy, we observe patients with follow-up visits every three months for the first two years, and every six months thereafter. Follow-up visits include history and physical examination to monitor for recurrence of disease or side effects of therapy, and laboratory studies (eg, complete blood count with differential, erythrocyte sedimentation rate [ESR], and chemistry studies, including electrolytes, kidney function, and liver function tests). We do not perform routine surveillance PET/CT following CR.

Late first relapse — Salvage chemotherapy (often with RT) is an acceptable alternative to HCT in select patients with a late (generally at least several years after completion of initial treatment), localized (ie, single site of relapse), asymptomatic, first relapse of HL. In this setting, treatment with conventional dose chemotherapy, with or without RT, can achieve long-term survival in 50 to 80 percent of patients, even without subsequent HCT [10,63-65]. However, a majority of such patients may achieve long-term lymphoma-free survival after autologous HCT in this setting [66].

In this setting, we offer two to three cycles of salvage chemotherapy (eg, ICE) followed by consolidative RT to the site of relapse. (See 'Choice of chemotherapy regimen' above.)

Risks and benefits of consolidation with autologous HCT should be discussed with the young fit patient who achieves CR in this setting. (See 'Autologous HCT' above.)

Rarely, such a patient may have received initial induction therapy with a regimen that did not include an anthracycline (eg, MOPP). If so, that patient may be considered for treatment with four to six cycles of ABVD (doxorubicin, bleomycin, vinblastine, and dacarbazine) or another first-line regimen that was not used initially, with or without RT (table 5) [10,35,67,68].

A decision to forego HCT in this setting must weigh the toxicity of cumulative doses of chemotherapy (both prior and planned therapy) and should specifically consider possible cardiac toxicity from anthracycline therapy and radiation, pulmonary toxicity from bleomycin, and neurotoxicity from vinca alkaloids.

Second and subsequent relapses — Special consideration must be given to patients in the setting of second or subsequent relapse of HL. Such individuals will have already received at least one form of salvage chemotherapy, targeted chemotherapy, radiation therapy, immunotherapy, and/or HCT. The likelihood of achieving a sustained complete remission diminishes with additional relapses. In this setting, toxicities associated with prior therapy, comorbidities, and other considerations will inform the therapeutic strategy.

Options for treatment are influenced by the number of prior relapses, the nature of prior treatments (eg, initial and salvage chemotherapy regimens, prior autologous and/or allogeneic HCT, immunotherapy), and the individual's general medical condition and goals of treatment (ie, treatment with curative intent versus relief of symptoms).

Therapeutic options include:

Further salvage chemotherapy, which must consider residual and/or likely toxicities from prior treatment (eg, cardiotoxicity from anthracyclines, nephrotoxicity and/or neuropathy from platinum-containing regimens)

Targeted chemotherapy (eg, brentuximab vedotin)

Immunotherapy approaches (eg, PD-1 blockade)

Autologous HCT, if not previously performed (or reduced intensity allogeneic HCT, if the patient previously underwent autologous HCT) (see "Hematopoietic cell transplantation in classic Hodgkin lymphoma")

Sequential administration of targeted chemotherapy, immunotherapy, and single agent chemotherapy (eg, gemcitabine, etoposide, vinorelbine, vinblastine, liposomal doxorubicin, bendamustine) may offer palliation of symptoms with relatively low toxicity

Participation in clinical trials

CLINICAL TRIALS — Often there is no better therapy to offer a patient than enrollment onto a well-designed, scientifically valid, peer-reviewed clinical trial. Additional information and instructions for referring a patient to an appropriate research center can be obtained from the United States National Institutes of Health (www.clinicaltrials.gov).

Many agents are under active investigation or in development for relapsed or refractory HL. Examples include combinations of agents already used in HL, agents studied in other diseases (eg, rituximab [69,70], ibrutinib [71], everolimus [72], lenalidomide [73], bendamustine [74,75], histone deacetylase inhibitors [76]), novel monoclonal antibodies [77], and other novel agents.

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Management of Hodgkin lymphoma".)

INFORMATION FOR PATIENTS — UpToDate offers two types of patient education materials, "The Basics" and "Beyond the Basics." The Basics patient education pieces are written in plain language, at the 5th to 6th grade reading level, and they answer the four or five key questions a patient might have about a given condition. These articles are best for patients who want a general overview and who prefer short, easy-to-read materials. Beyond the Basics patient education pieces are longer, more sophisticated, and more detailed. These articles are written at the 10th to 12th grade reading level and are best for patients who want in-depth information and are comfortable with some medical jargon.

Here are the patient education articles that are relevant to this topic. We encourage you to print or e-mail these topics to your patients. (You can also locate patient education articles on a variety of subjects by searching on "patient education" and the keyword(s) of interest.)

Basics topics (see "Patient education: Autologous bone marrow transplant (The Basics)")

Beyond the Basics topics (see "Patient education: Hodgkin lymphoma in adults (Beyond the Basics)" and "Patient education: Hematopoietic cell transplantation (bone marrow transplantation) (Beyond the Basics)")

SUMMARY AND RECOMMENDATIONS

Evaluation of suspected relapsed or refractory (r/r) Hodgkin lymphoma (HL) should include biopsy confirmation of relapse and restaging to assess the extent of disease. (See 'Definitions' above and 'Confirming refractory/relapsed disease' above.)

Management depends on the patient's eligibility for autologous hematopoietic cell transplantation (HCT), prior treatment (including chemotherapy, radiation, and/or immunotherapy), the response to salvage chemotherapy, the presence of bulky or localized disease at recurrence, and comorbid medical conditions. (See 'Overview of management' above.)

For most patients with r/r HL, we recommend treatment with salvage combination chemotherapy followed by autologous HCT, rather than chemotherapy without HCT (Grade 1B). (See 'Hematopoietic cell transplantation (HCT)' above.)

No randomized trials have identified a particular salvage regimen as having a survival advantage or superior response rates in this setting. The most commonly used regimens are ICE (ifosfamide, carboplatin, etoposide) and GVD (gemcitabine, vinorelbine, pegylated liposomal doxorubicin) (table 5).

For most patients with r/r HL, we suggest ICE chemotherapy (Grade 2B). This preference is largely based on comparable efficacy, a more favorable toxicity profile, and our greater familiarity with that regimen. (See 'Choice of chemotherapy regimen' above.)

Management after initial salvage chemotherapy is guided by the results of positron emission tomography/computerized tomography (PET/CT). Long-term survival is most likely if a complete response (CR) is achieved; some patients with a partial response (PR) to salvage chemotherapy can be cured with autologous HCT (algorithm 1). (See 'Importance of minimizing disease burden' above.)

Subsequent management is guided by findings from PET/CT after salvage chemotherapy:

Patients who achieve CR proceed to HCT, which may be followed by radiation therapy (RT) and/or maintenance brentuximab vedotin.

For those who have a PR, we suggest further therapy to minimize disease burden prior to HCT rather than proceeding directly with HCT (Grade 2C). Our approach depends on the nature and location of disease:

-For residual disease that can be encompassed in a single radiation port, we treat with RT prior to HCT. (See 'Indications for radiation' above.)

-For residual disease that cannot be encompassed in a radiation port, we offer alternative salvage chemotherapy, targeted chemotherapy, or immunotherapy. We prefer GVD chemotherapy in this setting. (See 'Choice of chemotherapy regimen' above.)

For those with progressive disease, we offer sequential therapies (as for those with PR) with an aim to achieve CR prior to HCT, or for palliation of symptoms and prolongation of life.

Patients who are ineligible for HCT because of comorbid illnesses are treated with the goal of achieving long-term disease control; in some cases the goal may simply be to palliate symptoms. For HCT-ineligible patients, we suggest less intensive therapy (eg, brentuximab vedotin, single agent gemcitabine, or immunotherapy), with or without RT, rather than intensive combination chemotherapy (eg, ICE) (Grade 2C).

For select patients with a late (generally at least several years after completion of initial treatment), localized (ie, single site of relapse), asymptomatic, first relapse of HL, salvage chemotherapy (often with RT) is an acceptable alternative to HCT. (See 'Late first relapse' above.)

Patients with second or later relapse and those who relapse following autologous HCT may be treated with brentuximab vedotin, immune checkpoint inhibitors (eg, nivolumab, pembrolizumab), allogeneic HCT, and experimental approaches in clinical trials. This choice should consider disease tempo, prior therapy, comorbidities, treatment toxicities, and patient/clinician preferences (table 5). (See "Hematopoietic cell transplantation in classic Hodgkin lymphoma".)

ACKNOWLEDGMENTS — The editorial staff at UpToDate would like to acknowledge Peter M Mauch, MD, who contributed to an earlier version of this topic review.

  1. Eich HT, Diehl V, Görgen H, et al. Intensified chemotherapy and dose-reduced involved-field radiotherapy in patients with early unfavorable Hodgkin's lymphoma: final analysis of the German Hodgkin Study Group HD11 trial. J Clin Oncol 2010; 28:4199.
  2. Meyer RM, Gospodarowicz MK, Connors JM, et al. Randomized comparison of ABVD chemotherapy with a strategy that includes radiation therapy in patients with limited-stage Hodgkin's lymphoma: National Cancer Institute of Canada Clinical Trials Group and the Eastern Cooperative Oncology Group. J Clin Oncol 2005; 23:4634.
  3. Engert A, Plütschow A, Eich HT, et al. Reduced treatment intensity in patients with early-stage Hodgkin's lymphoma. N Engl J Med 2010; 363:640.
  4. 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; 372:1598.
  5. 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; 31:684.
  6. Borchmann P, Haverkamp H, Diehl V, et al. Eight cycles of escalated-dose BEACOPP compared with four cycles of escalated-dose BEACOPP followed by four cycles of baseline-dose BEACOPP with or without radiotherapy in patients with advanced-stage hodgkin's lymphoma: final analysis of the HD12 trial of the German Hodgkin Study Group. J Clin Oncol 2011; 29:4234.
  7. Engert A, Diehl V, Franklin J, et al. Escalated-dose BEACOPP in the treatment of patients with advanced-stage Hodgkin's lymphoma: 10 years of follow-up of the GHSG HD9 study. J Clin Oncol 2009; 27:4548.
  8. Radford JA, Eardley A, Woodman C, Crowther D. Follow up policy after treatment for Hodgkin's disease: too many clinic visits and routine tests? A review of hospital records. BMJ 1997; 314:343.
  9. London J, Grados A, Fermé C, et al. Sarcoidosis occurring after lymphoma: report of 14 patients and review of the literature. Medicine (Baltimore) 2014; 93:e121.
  10. Bonfante V, Santoro A, Viviani S, et al. Outcome of patients with Hodgkin's disease failing after primary MOPP-ABVD. J Clin Oncol 1997; 15:528.
  11. Lohri A, Barnett M, Fairey RN, et al. Outcome of treatment of first relapse of Hodgkin's disease after primary chemotherapy: identification of risk factors from the British Columbia experience 1970 to 1988. Blood 1991; 77:2292.
  12. Brice P, Bastion Y, Divine M, et al. Analysis of prognostic factors after the first relapse of Hodgkin's disease in 187 patients. Cancer 1996; 78:1293.
  13. 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; 20:221.
  14. Sieniawski M, Franklin J, Nogova L, et al. Outcome of patients experiencing progression or relapse after primary treatment with two cycles of chemotherapy and radiotherapy for early-stage favorable Hodgkin's lymphoma. J Clin Oncol 2007; 25:2000.
  15. Böll B, Goergen H, Arndt N, et al. Relapsed hodgkin lymphoma in older patients: a comprehensive analysis from the German hodgkin study group. J Clin Oncol 2013; 31:4431.
  16. Moskowitz CH, Kewalramani T, Nimer SD, et al. Effectiveness of high dose chemoradiotherapy and autologous stem cell transplantation for patients with biopsy-proven primary refractory Hodgkin's disease. Br J Haematol 2004; 124:645.
  17. Brice P. Managing relapsed and refractory Hodgkin lymphoma. Br J Haematol 2008; 141:3.
  18. Alinari L, Blum KA. How I treat relapsed classical Hodgkin lymphoma after autologous stem cell transplant. Blood 2016; 127:287.
  19. Collins GP, Parker AN, Pocock C, et al. Guideline on the management of primary resistant and relapsed classical Hodgkin lymphoma. Br J Haematol 2014; 164:39.
  20. http://www.nccn.org/professionals/physician_gls/pdf/hodgkins.pdf (Accessed on February 13, 2014).
  21. 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; 145:369.
  22. Gerrie AS, Power MM, Shepherd JD, et al. Chemoresistance can be overcome with high-dose chemotherapy and autologous stem-cell transplantation for relapsed and refractory Hodgkin lymphoma. Ann Oncol 2014; 25:2218.
  23. Josting A, Müller H, Borchmann P, et al. Dose intensity of chemotherapy in patients with relapsed Hodgkin's lymphoma. J Clin Oncol 2010; 28:5074.
  24. Byrne BJ, Gockerman JP. Salvage therapy in Hodgkin's lymphoma. Oncologist 2007; 12:156.
  25. Mendler JH, Friedberg JW. Salvage therapy in Hodgkin's lymphoma. Oncologist 2009; 14:425.
  26. 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; 97:616.
  27. Moskowitz CH, Bertino JR, Glassman JR, et al. Ifosfamide, carboplatin, and etoposide: a highly effective cytoreduction and peripheral-blood progenitor-cell mobilization regimen for transplant-eligible patients with non-Hodgkin's lymphoma. J Clin Oncol 1999; 17:3776.
  28. Moskowitz C. Risk-adapted therapy for relapsed and refractory lymphoma using ICE chemotherapy. Cancer Chemother Pharmacol 2002; 49 Suppl 1:S9.
  29. Bartlett NL, Niedzwiecki D, Johnson JL, et al. Gemcitabine, vinorelbine, and pegylated liposomal doxorubicin (GVD), a salvage regimen in relapsed Hodgkin's lymphoma: CALGB 59804. Ann Oncol 2007; 18:1071.
  30. Kuruvilla J, Nagy T, Pintilie M, et al. Similar response rates and superior early progression-free survival with gemcitabine, dexamethasone, and cisplatin salvage therapy compared with carmustine, etoposide, cytarabine, and melphalan salvage therapy prior to autologous stem cell transplantation for recurrent or refractory Hodgkin lymphoma. Cancer 2006; 106:353.
  31. Santoro A, Mazza R, Pulsoni A, et al. Bendamustine in Combination With Gemcitabine and Vinorelbine Is an Effective Regimen As Induction Chemotherapy Before Autologous Stem-Cell Transplantation for Relapsed or Refractory Hodgkin Lymphoma: Final Results of a Multicenter Phase II Study. J Clin Oncol 2016; 34:3293.
  32. Brandwein JM, Callum J, Sutcliffe SB, et al. Evaluation of cytoreductive therapy prior to high dose treatment with autologous bone marrow transplantation in relapsed and refractory Hodgkin's disease. Bone Marrow Transplant 1990; 5:99.
  33. Josting A, Reiser M, Rueffer U, et al. Treatment of primary progressive Hodgkin's and aggressive non-Hodgkin's lymphoma: is there a chance for cure? J Clin Oncol 2000; 18:332.
  34. Rancea M, Monsef I, von Tresckow B, et al. High-dose chemotherapy followed by autologous stem cell transplantation for patients with relapsed/refractory Hodgkin lymphoma. Cochrane Database Syst Rev 2013; :CD009411.
  35. Harker WG, Kushlan P, Rosenberg SA. Combination chemotherapy for advanced Hodgkin's disease after failure of MOPP: ABVD and B-CAVe. Ann Intern Med 1984; 101:440.
  36. Schmitz N, Pfistner B, Sextro M, et al. 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; 359:2065.
  37. Fermé C, Mounier N, Diviné M, et al. Intensive salvage therapy with high-dose chemotherapy for patients with advanced Hodgkin's disease in relapse or failure after initial chemotherapy: results of the Groupe d'Etudes des Lymphomes de l'Adulte H89 Trial. J Clin Oncol 2002; 20:467.
  38. Okeley NM, Miyamoto JB, Zhang X, et al. Intracellular activation of SGN-35, a potent anti-CD30 antibody-drug conjugate. Clin Cancer Res 2010; 16:888.
  39. Chen R, Gopal AK, Smith SE, et al. Five-year survival and durability results of brentuximab vedotin in patients with relapsed or refractory Hodgkin lymphoma. Blood 2016; 128:1562.
  40. Chen R, Palmer JM, Thomas SH, et al. Brentuximab vedotin enables successful reduced-intensity allogeneic hematopoietic cell transplantation in patients with relapsed or refractory Hodgkin lymphoma. Blood 2012; 119:6379.
  41. Chen R, Wang F, Zhang H, Chen B. Brentuximab vedotin for treatment of relapsed or refractory malignant lymphoma: results of a systematic review and meta-analysis of prospective studies. Drug Des Devel Ther 2015; 9:2277.
  42. LaCasce AS, Bociek RG, Sawas A, et al. Brentuximab vedotin plus bendamustine: a highly active first salvage regimen for relapsed or refractory Hodgkin lymphoma. Blood 2018; 132:40.
  43. O'Connor OA, Lue JK, Sawas A, et al. Brentuximab vedotin plus bendamustine in relapsed or refractory Hodgkin's lymphoma: an international, multicentre, single-arm, phase 1-2 trial. Lancet Oncol 2018; 19:257.
  44. Younes A, Gopal AK, Smith SE, et al. Results of a pivotal phase II study of brentuximab vedotin for patients with relapsed or refractory Hodgkin's lymphoma. J Clin Oncol 2012; 30:2183.
  45. 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; 125:1236.
  46. https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/125388s094lbl.pdf (Accessed on November 15, 2017).
  47. Wagner-Johnston ND, Bartlett NL, Cashen A, Berger JR. Progressive multifocal leukoencephalopathy in a patient with Hodgkin lymphoma treated with brentuximab vedotin. Leuk Lymphoma 2012; 53:2283.
  48. Jalan P, Mahajan A, Pandav V, et al. Brentuximab associated progressive multifocal leukoencephalopathy. Clin Neurol Neurosurg 2012; 114:1335.
  49. Urru SA, Mariotti E, Carta P, et al. Acute pancreatitis following brentuximab vedotin therapy for refractory Hodgkin lymphoma: a case report. Drugs R D 2014; 14:9.
  50. Pembrolizumab injection. United States Prescribing Information. US National Library of Medicine. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/125514s059s064s076s083lbl.pdf (Accessed on April 29, 2020).
  51. Armand P, Engert A, Younes A, et al. Nivolumab for Relapsed/Refractory Classic Hodgkin Lymphoma After Failure of Autologous Hematopoietic Cell Transplantation: Extended Follow-Up of the Multicohort Single-Arm Phase II CheckMate 205 Trial. J Clin Oncol 2018; 36:1428.
  52. Younes A, Santoro A, Shipp M, et al. Nivolumab for classical Hodgkin's lymphoma after failure of both autologous stem-cell transplantation and brentuximab vedotin: a multicentre, multicohort, single-arm phase 2 trial. Lancet Oncol 2016; 17:1283.
  53. Chen R, Zinzani PL, Lee HJ, et al. Pembrolizumab in relapsed or refractory Hodgkin lymphoma: 2-year follow-up of KEYNOTE-087. Blood 2019; 134:1144.
  54. Chen R, Zinzani PL, Fanale MA, et al. Phase II Study of the Efficacy and Safety of Pembrolizumab for Relapsed/Refractory Classic Hodgkin Lymphoma. J Clin Oncol 2017; 35:2125.
  55. Herbaux C, Gauthier J, Brice P, et al. Efficacy and tolerability of nivolumab after allogeneic transplantation for relapsed Hodgkin lymphoma. Blood 2017; 129:2471.
  56. Merryman RW, Kim HT, Zinzani PL, et al. Safety and efficacy of allogeneic hematopoietic stem cell transplant after PD-1 blockade in relapsed/refractory lymphoma. Blood 2017; 129:1380.
  57. Ramos CA, Grover NS, Beaven AW, et al. Anti-CD30 CAR-T Cell Therapy in Relapsed and Refractory Hodgkin Lymphoma. J Clin Oncol 2020; 38:3794.
  58. Ramos CA, Ballard B, Zhang H, et al. Clinical and immunological responses after CD30-specific chimeric antigen receptor-redirected lymphocytes. J Clin Invest 2017; 127:3462.
  59. Wirth A, Corry J, Laidlaw C, et al. Salvage radiotherapy for Hodgkin's disease following chemotherapy failure. Int J Radiat Oncol Biol Phys 1997; 39:599.
  60. Campbell B, Wirth A, Milner A, et al. Long-term follow-up of salvage radiotherapy in Hodgkin's lymphoma after chemotherapy failure. Int J Radiat Oncol Biol Phys 2005; 63:1538.
  61. Goda JS, Massey C, Kuruvilla J, et al. Role of salvage radiation therapy for patients with relapsed or refractory hodgkin lymphoma who failed autologous stem cell transplant. Int J Radiat Oncol Biol Phys 2012; 84:e329.
  62. 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; 23:1522.
  63. Viviani S, Santoro A, Negretti E, et al. Salvage chemotherapy in Hodgkin's disease. Results in patients relapsing more than twelve months after first complete remission. Ann Oncol 1990; 1:123.
  64. Radman I, Basić N, Labar B, et al. Long-term results of conventional-dose salvage chemotherapy in patients with refractory and relapsed Hodgkin's disease (Croatian experience). Ann Oncol 2002; 13:1650.
  65. Yuen AR, Rosenberg SA, Hoppe RT, et al. Comparison between conventional salvage therapy and high-dose therapy with autografting for recurrent or refractory Hodgkin's disease. Blood 1997; 89:814.
  66. Bröckelmann PJ, Goergen H, Kohnhorst C, et al. Late Relapse of Classical Hodgkin Lymphoma: An Analysis of the German Hodgkin Study Group HD7 to HD12 Trials. J Clin Oncol 2017; 35:1444.
  67. 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; 327:1478.
  68. Bonadonna G, Santoro A, Gianni AM, et al. Primary and salvage chemotherapy in advanced Hodgkin's disease: the Milan Cancer Institute experience. Ann Oncol 1991; 2 Suppl 1:9.
  69. Younes A, Oki Y, McLaughlin P, et al. Phase 2 study of rituximab plus ABVD in patients with newly diagnosed classical Hodgkin lymphoma. Blood 2012; 119:4123.
  70. Kasamon YL, Jacene HA, Gocke CD, et al. Phase 2 study of rituximab-ABVD in classical Hodgkin lymphoma. Blood 2012; 119:4129.
  71. Hamadani M, Balasubramanian S, Hari PN. Ibrutinib in Refractory Classic Hodgkin's Lymphoma. N Engl J Med 2015; 373:1381.
  72. Johnston PB, Inwards DJ, Colgan JP, et al. A Phase II trial of the oral mTOR inhibitor everolimus in relapsed Hodgkin lymphoma. Am J Hematol 2010; 85:320.
  73. Fehniger TA, Larson S, Trinkaus K, et al. A phase 2 multicenter study of lenalidomide in relapsed or refractory classical Hodgkin lymphoma. Blood 2011; 118:5119.
  74. Moskowitz AJ, Hamlin PA Jr, Perales MA, et al. Phase II study of bendamustine in relapsed and refractory Hodgkin lymphoma. J Clin Oncol 2013; 31:456.
  75. Corazzelli G, Angrilli F, D'Arco A, et al. Efficacy and safety of bendamustine for the treatment of patients with recurring Hodgkin lymphoma. Br J Haematol 2013; 160:207.
  76. Younes A, Sureda A, Ben-Yehuda D, et al. Panobinostat in patients with relapsed/refractory Hodgkin's lymphoma after autologous stem-cell transplantation: results of a phase II study. J Clin Oncol 2012; 30:2197.
  77. Rothe A, Sasse S, Topp MS, et al. A phase 1 study of the bispecific anti-CD30/CD16A antibody construct AFM13 in patients with relapsed or refractory Hodgkin lymphoma. Blood 2015; 125:4024.
Topic 4765 Version 53.0

References