Mediastinal Lymphoma

Back

Practice Essentials

Primary mediastinal B-cell lymphoma (PMBCL) is relatively rare B-cell non-Hodgkin lymphoma that comprises 6–12% of all diffuse large B-cell lymphomas (DLBCLs) and 2–4% of all non-Hodgkin lymphomas. PMBCL affects young adults in their third to fourth decade of life and has a slight female predominance.[1]  

PMBCL was initially recognized in the Revised European-American Classification of Lymphoid Neoplasms (REAL) as a subtype of DLBCL that involves the mediastinum. However, PMBCL became a fully recognized separate entity in 2001 when the World Health organization (WHO) identified that PMBCL represents a distinct clinicopathologic disease and should be classified as such.

Gene expression studies have confirmed that PMBCL is molecularly different from DLBCL and that it may resemble Hodgkin lymphoma.[2, 3, 4] Because of its skewed age distribution, PMBCL accounts for a much higher proportion of lymphomas in patients who have undergone autologous hematopoietic stem cell transplantation (auto-HSCT). Importantly, the majority of patients are cured with modern intensive combination chemoimmunotherapy that is often followed by either involved-field radiation or auto-HSCT. In fact, in some retrospective studies, PMBCL was shown to even have better prognosis than DLBCL when all other variables were equal.[5, 6] Important to note, however, is that the role of radiotherapy (RT) and/or auto-HSCT is being challenged in the current modern era of highly sophisticated imaging modalities such as positron emission tomography (PET) scanning. See the image below.



View Image

Example of mediastinal lymphoma at diagnosis. A large tumor mass is present in the anterior mediastinum, and an associated pleural effusion can also b....

While PMBCL is generally limited to the mediastinum, hematogenous and extranodal involvements are common in relapsed patients, who have generally a poor prognosis.[7, 8]  For more information, see B-Cell Lymphoma.

For patient education information, see Non-Hodgkin Lymphoma.

 

Pathology and Etiology

The tumor cells resemble immunoblasts or centroblasts, and, in many cases, sclerosis and fibrosis can be identified. Flow cytometry studies identify a unique immunophenotype profile for primary mediastinal B-cell lymphoma (PMBCL). Malignant cells express B-cell antigens (CD20, CD19, CD22, and CD79a), and, in some patients, weak CD30 expression is noted. The weak CD30 expression can aid in differentiating PMBCL from anaplastic large cell lymphoma and classic Hodgkin lymphoma. This is critical, as Reed-Sternberg–like cells can be present in patients with PMBCL. Additionally, the malignant cells also lack CD15 expression but have frequent positivity of the transcriptional regulators BCL6, PAX5, and BOB1.

PMBCL is hypothesized to originate from the germinal or postgerminal centers; however, surface immunoglobulin expression is frequently absent.

Several studies have explored the underlying genetic and molecular features that lead to the evolution of primary mediastinal B-cell lymphoma (PMBCL). Gene expression profiling has clearly demonstrated that this disease entity is different from diffuse large B-cell lymphoma (DLBCL) and has identified several deregulated pathways involved in the pathogenesis of PMBCL. Steidl and Gascoyne eloquently summarized the recurrent gene alterations involved in the pathogenesis of PMBCL.[9]

Gene expression profiling has demonstrated overexpression of genes encoding the NF-kappa-B pathway, suggesting its involvement in the pathogenesis of PMBCL. Further, inhibiting I-kappa-B, which activates NF-kappa-B signaling, was shown to induce cell kill in vitro in PMBCL cell-lines.

Chromosomal gains and amplifications of the REL gene locus on band 2p16.1 have been found in 50% of cases.[9, 10, 11]  Other chromosomal aberrations that affect NF-kappa-B pathways include BCL-10 (1p22) and MALT-1 (18q21).

Song et al identified a tumor suppressor gene that encodes the A-20 protein, which acts as an inhibitor to the NF-kappa-B pathway downstream from the tumor necrosis factor (TNF) receptor.[12]  Mutations in that gene have been found in over 30% of PMBCL cases, but it has also been described in other lymphoid malignancies.[13, 14]  Another mutated tumor suppressor gene is SOCS1 (suppressor of cytokine signaling). SOCS1 usually acts as an inhibitor to the JAK-STAT6 pathway, preventing continued activation and proliferation.[15]  In fact, Mottok et al suggested that SOCS1 deletion mutations were present in 45% of 20 studied patients.[16]

More recent studies have also shown a role of immune deregulation in PMBCL pathogenesis. Specifically, the reduced expression of major histocompatibility II complex genes led to decreased infiltrating cytotoxic T-cells, and some investigators have suggested this can lead to inferior outcomes, arguing that more studies exploring the role of immune escape in PMBCL are warranted. Another mechanism by which PMBCL cells escape immune surveillance has been by overexpressing certain surface molecules such as PD-1 ligands and receptors, which, in turn, leads to inactivating infiltrating effector T cells.[17]

Clinical Presentation

History

Primary mediastinal B-cell lymphoma (PMBCL) patients usually present with a bulky anterior mediastinal mass in their third or fourth decade in life. Superior vena cava (SVC) syndrome is common, and, in some reports, 50-80% of patients can have some form of SVC compromise. Phrenic nerve palsy, dysphagia, hoarseness, and breast swelling (in women) can occur. Shortness of breath can be due to pleural effusion, massive mediastinal mass, pericardial effusion, or airway compression. Systemic symptoms (fever, weight loss, night sweats) occur in 30-47% of patients.

Most patients have localized symptoms that depend on the bulk and extent of the disease; however, if recurrence develops, a hematogenous pattern of spread to parenchymal organs, such as the liver, kidneys, or brain, is common. Symptoms in recurrent disease accordingly vary and depend on the organ involved.

Physical examination

Physical examination may reveal the following:

Performance status should be noted because this is an important prognostic indicator.[18]

Additional findings

Laboratory studies are not diagnostic of PMBL, but over 70% of patients can have elevated lactate dehydrogenase (LDH) levels. By definition, patients do not have bone marrow involvement, and 75-80% of patients are usually staged as having stage I or II disease. Most patients present with bulky disease defined as a mass greater than 10 cm in largest diameter.

Differential Diagnosis

The differential diagnosis of primary mediastinal B-cell lymphoma (PMBCL) includes the following:

Diagnostic considerations

Differentiating PMBCL from other malignancies that involve the mediastinum is extremely important because the diagnosis affects management and outcome.[19] Similarly important is differentiating PMBCL from systemic diffuse large B-cell lymphoma (DLBCL) with mediastinal involvement. This latter entity often affects older patients and usually has involvement of distant lymph nodes (away from the mediastinum) and for some, bone marrow is affected with disease.

Distinguishing Hodgkin lymphoma from PMBCL can be challenging. Both diseases can affect younger adults and both entities can be similar histologically. Flow cytometry studies can be helpful, as many patients with classic Hodgkin lymphoma have neoplastic cells that express CD15 and CD30, while they lack expression of B-cell markers. As mentioned above, PMBCL patients express B-cell markers and have weak CD30 expression.

Despite significant advances, cases continue to be difficult to diagnose. Some patients have features similar to classic Hodgkin lymphoma (CHL) and PMBCL at the same time. These patients are diagnosed with mediastinal gray zone lymphoma (MGZL), which is described as B-cell lymphoma with features intermediate between DLBCL and CHL. Prognosis is worse than that of both CHL and PMBCL. Management of these patients is not well defined, but National Comprehensive Cancer Network (NCCN) guidelines recommend treatment with aggressive DLBCL regimens.[20]  Data suggest that adding rituximab to chemotherapy regimens improves outcomes.[20, 21]  

Workup

Laboratory studies

Perform a CBC count with differential and platelets. Perform an electrolyte panel and liver function tests.

Elevation in the serum lactic dehydrogenase (LDH) or beta-2 microglobulin level value is an adverse prognostic feature and usually is seen in the majority of patients.

The markers alpha-fetoprotein and beta-human chorionic gonadotropin (beta-hCG) are often highly elevated in patients with mediastinal germ cell tumors, constituting an important differential diagnosis in males.

Imaging studies

Obtain a chest radiograph (posteroanterior, lateral). A mass larger than one third the diameter of the thorax is considered bulky and indicates a poor prognosis. This might have therapeutic implications later in treatment if radiotherapy is being considered.

Obtain CT scans (chest, abdomen, pelvis). Extension to the pleura, pericardium, and even the chest wall is common. Invasion of the liver, kidneys, and peripheral lymph nodes is more common at the time of recurrence. On occasion, obtaining a CT scan of the neck and soft tissues might be warranted.

Positron emission tomography (PET) scans are considered standard of care procedure before initiating therapy. PET scans are expected to become negative upon completion of successful treatment. Additional diagnostic or treatment considerations (as discussed below) are suggested if the PET scans remain positive after completion of therapy. Treatment decisions based solely on PET scan results are not encouraged, and patients might need to undergo additional diagnostic procedures to confirm whether the PET scan findings are truly positive.

Consider other imaging studies if they are clinically indicated (eg, head CT scan or MRI, if the patient has neurologic problems; see image below). Consider performing a multiple-gated acquisition (MUGA) scan to assess cardiac function before anthracycline-based chemotherapy.



View Image

Isolated CNS relapse of primary mediastinal B-cell lymphoma.

Diagnostic procedures

Adequate diagnostic biopsy is needed and may require surgery to obtain a sufficient sample for accurate diagnosis. Not uncommon, biopsies can initially be nondiagnostic owing to extensive fibrosis and necrosis, and additional studies might be needed. Most commonly, patients either undergo mediastinoscopy or thoracoscopy depending on the location and feasibility.[22]

Ancillary studies, which include immunohistochemistry, immunophenotyping (flow cytometry), and gene rearrangement studies, are often necessary to establish the diagnosis.

Bone marrow aspirate and biopsy are necessary for staging. A unilateral sample is sufficient if the biopsy specimen is larger than 2 cm. By definition, the bone marrow is not usually involved. Patients with marrow involvement might have systemic diffuse large B-cell lymphoma (DLBCL) with secondary mediastinal disease, as opposed to primary mediastinal B-cell lymphoma (PMBCL).

Other tests should be performed if clinically indicated (eg, thoracentesis for pleural effusion, lumbar puncture for neurologic symptoms).

Treatment & Management

For patients who present with superior vena cava (SVC) syndrome, establishing the diagnosis in a timely and efficient manner is critical. Radiation therapy (RT) can alter the pathologic findings and would impair accurate diagnosis; thus, it should be avoided except in extreme circumstances. Patients who are relatively stable should undergo emergent diagnostic evaluation (as summarized above) followed by treatment initiation.[23]

Chemoimmunotherapy

Combination anthracycline-based chemotherapy is the mainstay of treatment for primary mediastinal B-cell lymphoma (PMBCL). The standard front-line regimen in the United States is cyclophosphamide, doxorubicin (Adriamycin), vincristine, and prednisone combined with rituximab (CHOP-R). Rituximab is a chimeric monoclonal anti-CD20 antibody that has transformed how B-cell lymphomas are treated and has become a standard component of treating all B-cell lymphoma histologies that express CD20. 

A few studies, mainly from Europe, have advocated themethotrexate, doxorubicin (Adriamycin), cyclophosphamide, vincristine (Oncovin), prednisone, and bleomycin (MACOP-B) regimen, combined with rituximab.[24, 25, 26] The standard CHOP-R regimen is now being challenged by a combination program that contains etoposide (dose-adjusted EPOCH) plus rituximab (DA-EPOCH-R), although prospective randomized trials to accurately compare these 2 programs (CHOP-R vs DA-EPOCH-R) have not been completed.[27]  Nevertheless, the National Comprehensive Cancer Network (NCCN) guidelines recommend DA-EPOCH-R as the preferred first-line treatment for PMBCL.[20]

As PMBCL was recognized only recently as being a distinct entity, original studies that have established CHOP-R as a standard therapy in diffuse large B-cell lymphoma (DLBCL) did not include PMBCL patients. Accordingly, the German Lymphoma Study Group sought to confirm the impact of chemoimmunotherapy specifically on the PMBCL subset of patients who were enrolled on their MiNT trial (MabThera (Rituximab) International Trial).[28]

In this trial, patients were younger than 60 years with DLBCL and had 0-1 risk factors according to the age-adjusted International Prognostic Index (aaIPI). Patients were randomly assigned to 6 cycles of CHOP-like regimens with or without rituximab. Consolidating XRT was given to sites of primary bulky disease. Of 824 patients enrolled, 87 had PMBCL. Rituximab increased the rates of complete remission (unconfirmed) in PMBCL (from 54% to 80%; P =.015). In PMBCL, rituximab virtually eliminated progressive disease (2.5% vs 24%; P =.006).With a median observation time of 62 months for PMBCL, the 5-year event-free survival was improved (79.1% vs 47.3%; P =.011). Furthermore, 5-year progression-free survival was improved by rituximab (89.8% vs 60.1%; P =.006). These data further confirmed that the addition of rituximab to 6 cycles of CHOP-likechemotherapy improved long-term outcome for young patients with PMBCL.

Multiple analyses have indicated an improvement in PMBCL outcomes when rituximab is added to a polychemotherapy backbone. This, coupled with concern regarding long‐term sequelae of RT, have led to questions about the utility of radiotherapy when rituximab is added to standard chemotherapy. A National Cancer Database (NCDB) study of 465 patients receiving multiagent chemotherapy in 2006-2011 were with a median follow‐up of 36 months demonstrated that the use of radiation therapy as a component of combined modality therapy was associated with a significant improvement in overall survival (56% reduction in hazard for death on multivariate analysis) when compared with systemic therapy alone in the years following rituximab approval. 5‐year overall survival (OS) for the entire cohort was 87%.  Patients who received radiation therapy (RT) had an OS of 93% compared to an OS of  83% among those who did not receive RT.  Although the lack of specific chemotherapy data is a limitation of the study, the data does supports the use of RT in all PMBCL stages.[29]

Patients should be evaluated clinically and radiographically to assure continued response. Interim positron emission tomography (PET) scan evaluation is discussed separately below. Patients usually undergo 6 cycles of CHOP-R administered every 3-weeks. The regimen has expected adverse effects and toxicities as discussed below, and the use of growth factors (filgrastim or peg-filgrastim) is dependent on the patient's age and comorbidities. Given the relatively younger age at PMBCL presentation, the authors advocate against the routine use of growth factors as primary prophylaxis. Secondary prophylaxis, however, is recommended to ensure adequate dose density and intensity.

Consolidation therapy and interim PET scans

Prior to the wide use and adaptability to PET scans, most patients underwent consolidative RT or high-dose chemotherapy and autologous hematopoietic stem cell transplantation (auto-HSCT) after completion of systemic therapy. While this approach continues to be commonly used, it has become increasingly controversial, especially in patients who attain complete PET scan negativity upon completion of systemic chemoimmunotherapy.

Several reports have suggested inferior survival in patients with DLBCL who have residual PET scan positivity at the end of chemotherapy and other studies have shown that patients who have a positive interim PET scan (after 2-4 cycles of therapy) predict higher risk of subsequent relapse.[30, 31, 32] However, treatment decisions in patients who remain PET positive after completion of systemic therapy should never be based solely on the PET scan interpretation. Moskowitz et al showed in a large phase II study that the majority of DLBCL patients who remained PET positive had no residual disease when diagnostic biopsies were performed.[33] In that report, 30% of the 98 enrolled patients had PMBCL.

Whether RT should be delivered to all patients with PMBCL regardless of PET scan results or whether this approach should be individualized based on PET and/or other clinical or prognostic features remain unknown.

Savage et al reported on the British Columbia experience.[34] CHOP-R followed by consolidative RT was the adapted approach to all PMBCL patients diagnosed and treated from 2001–2005. After 2005, PET scanning was used to guide RT following 6 cycles of CHOP-R. To that end, if the PET scan was negative, patients were observed and if the PET scan was positive, consolidative RT was given. In total, 176 patients were identified: 96 received CHOP-R and 80 received CHOP. For the CHOP-R treated patients, 46 were treated in the “RT era” with 80% receiving RT; 50 were treated in the “PET era”; 38% received RT. Comparing between eras, no overall survival was suggested by adding RT. Further, when PET-positive patients received RT consolidation, no significant difference in outcome was observed between PET-positive and PET-negative patients, suggesting that some patients can avoid undergoing RT when PET is used to guide therapy.

In an attempt to eliminate RT, Dunleavy et al recently reported a phase II trial on 51 patients who were treated with DA-EPOCH-R and showed excellent results. With a median follow-up of 5 years, event-free survival was 93% and overall survival was 97%. For patients who had a PET on this study, the negative predictive value was 100%, while the positive predictive value was 17%. This finding is in line with the data from Moskowitz et al (see above), for which patients have false-positive PET scans after completion of therapy, and underscores the importance of not making therapeutic decisions based on PET findings only. In all, only 2 patients (4%) underwent RT when DA-EPOCH-R was used.

The use of consolidative auto-HSCT in patients with PMBCL stems from the effectiveness of this approach in patients with relapsed DLBCL.[35] Residual disease radiographically was hypothesized to represent persistent lymphoma and patients underwent the aggressive salvage therapy as PET scans were not available then. With the advent of PET and continued standardization of its interpretation, The European Society for Medical Oncology (ESMO) guidelines recommend against routine use of auto-HSCT as a primary consolidative approach.[36] Whether patients who have residual disease that is established histologically should undergo RT or auto-HSCT is unknown. Enrolling these patients in clinical trials is an option, if available. Outside of clinical trials, the decision needs to be individualized and would factor prognostic features, morbid conditions, and patients' wishes. 

Relapsed/refractory disease

While PMBCL is often cured with standard first-line therapies, approximately 200 patients per year in the United States are diagnosed with relapsed/refractory PMBCL, which has a poor prognosis with a two-year survival of 15%.[37] Due to its rarity, no standard of care has been identified and relapsed/refractory disease is generally treated following protocols for other DLBCL subtypes.[20]

Patients with relapsed disease often have systemic involvement and many also have extranodal disease. These patients are recommended to undergo salvage systemic chemotherapy followed by stem cell collection and subsequent transplantation. Rituximab, ifosfamide, carboplatin, and etoposide (RICE) is a common salvage approach, although other regimens are acceptable. Patients who demonstrate chemosensitive disease (improvement radiographically and on PET) are taken to transplantation. Patients who have refractory disease should be offered clinical trials, although some can be considered for allogeneic bone marrow transplantation.

PMBCL frequently involves PD-1 ligand overexpression, potentially making PMBCL susceptible to PD-1 blockade. NCCN has included pembrolizumab, a humanized anti–PD-1 monoclonal antibody blocking interaction of PD-1 with its ligands, PD-L1 and PD-L2, among the recommended treatments for relapsed/refractory PMBCL.[20]  

Treatment in pregnancy

Some patients are young women who may be pregnant at the time of diagnosis. The management of malignancy during pregnancy raises specific and complex issues. Concern for the patient's health needs to be balanced with the potential teratogenicity of the chemotherapy and the radiation administered for diagnostic examinations or as part of treatment.

Termination of pregnancy is often recommended if the diagnosis is made in the first trimester. However, this is not acceptable to all patients. In cases in which pregnancy is continued, the administration of chemotherapy drugs without undue teratogenicity is often possible. Staging and restaging examinations are minimized. Radiography is avoided, and MRI or ultrasonography procedures are used instead.

The administration of corticosteroids may exacerbate problems such as preeclampsia or glucose intolerance. Close collaboration with an obstetrician is required.

Consultations

Patients should be referred to a medical hematologist or oncologist for treatment.

Follow-up

The vast majority of patients can be successfully treated in an outpatient setting for front-line care.

After completion of treatment, patients are usually seen in the outpatient clinic at regular intervals of 2-3 months for the first year. Patients are seen every 3-4 months until 5 years. The authors recommend seeing patients annually after that indefinitely. Routine surveillance CT scans and/or PET scans are not recommended after establishing complete remission. The authors consider performing CT scans on some patients upon their request, especially if they have high likelihood of relapse.

Complications

The chemotherapeutic drugs used for the management of lymphoma have numerous adverse effects. Nausea and vomiting are common but can be avoided with the use of appropriate antiemetics. Hair loss occurs in most patients but is completely reversible after the completion of treatment.

Mild peripheral neuropathy due to chemotherapy is common. Patients experience numbness in fingertips and toes. Motor neuropathy is unusual.

Myelosuppression (bone marrow suppression) and moderate pancytopenia occur after every treatment cycle. Blood counts typically reach their nadir approximately 10 days after the completion of a treatment cycle. Fatigue is common.

Neutropenic fever and infection are common complications of chemotherapy and require immediate treatment. Approximately 10-20% of patients develop excessive neutropenia or an infectious complication. Primary prophylaxis with antibiotics is not recommended, although it is used for some patients. The use of growth factors is discussed above.

Cardiac toxicity due to chemotherapy is unusual but can occur. Cardiac toxicity from anthracyclines is dose dependent and rare in the typical young patient with PMBCL. Serial monitoring with echocardiograms or multiple-gated acquisition (MUGA) scans may be necessary in individual cases. Typically, patients undergo a MUGA scan to evaluate the left ventricular ejection fraction prior to the initiation of chemotherapy. A MUGA scan is performed in most centers only if clinical concerns arise about cardiomyopathy. Patients should not receive more than 400 mg/m2 of doxorubicin in their lifetime. The incidence of cardiomyopathy if this dose is exceeded is 7-8%. The use of cardioprotectant agents may allow the administration of higher doses of anthracyclines, but these cardioprotectant agents might affect the efficacy of chemotherapy. Therefore, cardioprotectant agents are not routinely recommended.

Rituximab is generally safe. It can cause fever and chills, particularly during the first administration. Rare cases of anaphylactic reactions have been reported. Cases of hepatitis B virus (HBV) reactivation that have resulted in fulminant hepatitis and death have been reported. Persons at high risk of HBV infection should be screened before the initiation of rituximab. Carriers of HBV should be closely monitored for clinical and laboratory signs of active HBV infection and hepatitis during and up to several months after rituximab therapy. All patients should have their hepatitis titers checked before rituximab initiation.

Acute adverse effects of radiation are usually limited and include erythema of the skin and, sometimes, radiation pneumonitis.

Late adverse effects related to treatment include decreased fertility, a slightly increased incidence of secondary cancers in radiation fields (especially breast cancer among women treated during adolescence), and a slightly increased risk for secondary leukemia, especially among patients treated with combined-modality therapy (ie, chemotherapy and radiation).

In addition, coronary artery disease may be more common and may have an earlier onset if substantial areas of the heart are exposed to radiation. Smoking and alcohol abuse should be avoided because of their association with cancer and heart disease.

Guidelines Summary

In 2016, the European Society for Medical Oncology (ESMO) released guidelines for the management of primary mediastinal B-cell lymphoma (PMBCL).[36]  The National Comprehensive Cancer Network (NCCN) guidelines for the treatment of diffuse large B-cell lymphona include recommendations for diagnosis and managament of PMBCL.[20]

ESMO guidelines require FDG-PET/CT scan to assess disease extent and to obtain better definition of the residual mediastinal masses at the completion of treatment.[36]  NCCN guidelines require clinical pathologic correlation to establish a diagnosis of PMBCL.[20]

Treatment

NCCN guidelines recommend the following first line treatment regimens (in order of preference)[20] :

ESMO guidelines recommend rituximab in combination with any of the following regimens[36] :

ESMO also recommends consolidative mediastinal RT (doses in the range 30–36 Gy, in 1.5–2.0 Gy fractions) in patients treated with standard-dose chemoimmunotherapy but could be omitted in patients with a complete metabolic response only after DA-EPOCH-R. 

Autologous stem cell transplantation (HDCT/ASCT) is not recommended in patients who achieved complete remission even in initially poor-risk patients who attain an adequate response to initial therapy.[36]

Post-treatment evaluation

Due to the high frequency of false-positive scans, ESMO recommends further investigaton of a positive PET scan before modifying planned therapy.  A post-treatment PET/CT response evaluation should not be carried out until at least 5–6 weeks from the last infusion of chemotherapy to minimise the incidence of false-positive scans. Serial scanning may be required to fully evaluate areas of residual post-treatment PET tracer uptake, with many patients manifesting gradual resolution.[36]

The NCCN notes that PET/CT scan is essential posttreatment. Biopsy of a PET/CT scan–positive mass is recommended if additional systemic treatment is being considered [20]

Treatment of relapsed/refractory PMBCL

For treatment of relapse/refractory disease, NCCN guidelines recommends either pembrolizumab or following the guidelines for DLBCL.[20]

ESMO recommends salvage treatment strategies similar to nodal DLBCLs. If radiation has not been a component of initial therapy, RT should be incorporated into the salvage treatment for patients with relapsed disease, ideally post-transplant if significant mediastinal or lung volumes are involved.[36]

What is primary mediastinal B-cell lymphoma (PMBCL)?What is the pathophysiology of primary mediastinal B-cell lymphoma (PMBCL)?Which clinical history findings are characteristic of primary mediastinal B-cell lymphoma (PMBCL)?Which physical findings are characteristic of primary mediastinal B-cell lymphoma (PMBCL)?Which lab test findings suggest primary mediastinal B-cell lymphoma (PMBCL)?Which conditions are included in the differential diagnoses of primary mediastinal B-cell lymphoma (PMBCL)?How is primary mediastinal B-cell lymphoma (PMBCL) differentiated from diffuse large B-cell lymphoma (DLBCL)?What is the role of lab testing in the workup of primary mediastinal B-cell lymphoma (PMBCL)?What is the role of imaging in the workup of primary mediastinal B-cell lymphoma (PMBCL)?What is the role of biopsy in the workup of primary mediastinal B-cell lymphoma (PMBCL)?How is primary mediastinal B-cell lymphoma (PMBCL) treated?What is the role of chemoimmunotherapy in the treatment of primary mediastinal B-cell lymphoma (PMBCL)?What is the role of consolidation therapy in the treatment of primary mediastinal B-cell lymphoma (PMBCL)?How is relapsed or refractory primary mediastinal B-cell lymphoma (PMBCL) treated?How is primary mediastinal B-cell lymphoma (PMBCL) treated during pregnancy?Which specialist consultations are beneficial to patients with primary mediastinal B-cell lymphoma (PMBCL)?What is included in long-term monitoring of primary mediastinal B-cell lymphoma (PMBCL)?What are the possible complications of primary mediastinal B-cell lymphoma (PMBCL) treatment?Which organizations have issued guidelines on the treatment of primary mediastinal B-cell lymphoma (PMBCL)?What are the guidelines on the diagnosis of primary mediastinal B-cell lymphoma (PMBCL)?What are the NCCN guidelines on first-line treatment regimens for primary mediastinal B-cell lymphoma (PMBCL)?What are the ESMO guidelines on first-line treatment for primary mediastinal B-cell lymphoma (PMBCL)?What are the ESMO guidelines on post-treatment evaluation of primary mediastinal B-cell lymphoma (PMBCL)?What are the NCCN guidelines on post-treatment evaluation of primary mediastinal B-cell lymphoma (PMBCL)?What are the NCCN treatment guidelines for relapsed/refractory primary mediastinal B-cell lymphoma (PMBCL)?What are the ESMO treatment guidelines for relapsed/refractory primary mediastinal B-cell lymphoma (PMBCL)?

Author

Sonali M Smith, MD, Associate Professor, Section of Hematology/Oncology, Director, Lymphoma Program, The University of Chicago Pritzker School of Medicine

Disclosure: Received consulting fee from Genentech/BiogenIdec for speaking and teaching; Received consulting fee from Lilly for speaking and teaching; Received consulting fee from Cephalon for consulting; Received consulting fee from Allos for speaking and teaching; Received consulting fee from Spectrum Pharmaceuticals for consulting; Received consulting fee from GlaxoSmithKline for consulting; Received consulting fee from Novartis for consulting; Received consulting fee from Celgene for consulting.

Coauthor(s)

Andrew S Artz, MD, MS, Associate Professor, Department of Medicine, Section of Hematology/Oncology, University of Chicago Division of the Biological Sciences, The Pritzker School of Medicine

Disclosure: Nothing to disclose.

Chadi Nabhan, MD, FACP, Associate Professor of Medicine, Department of Medicine, Section of Hematology and Oncology, Medical Director, Ambulatory Clinical Cancer Care Services, The University of Chicago Comprehensive Cancer Center

Disclosure: Received grant/research funds from Genentech for other; Received honoraria from Genentech for speaking and teaching.

Koen W Van Besien, MD, Director of Stem Cell Transplantation and Lymphoma, Associate Professor, Department of Internal Medicine, Section of Hematology/Oncology, The University of Chicago Pritzker School of Medicine

Disclosure: Nothing to disclose.

Specialty Editors

Francisco Talavera, PharmD, PhD, Adjunct Assistant Professor, University of Nebraska Medical Center College of Pharmacy; Editor-in-Chief, Medscape Drug Reference

Disclosure: Received salary from Medscape for employment. for: Medscape.

Ronald A Sacher, MBBCh, FRCPC, DTM&H, Professor of Internal Medicine and Pathology, Director, Hoxworth Blood Center, University of Cincinnati Academic Health Center

Disclosure: Nothing to disclose.

Chief Editor

Emmanuel C Besa, MD, Professor Emeritus, Department of Medicine, Division of Hematologic Malignancies and Hematopoietic Stem Cell Transplantation, Kimmel Cancer Center, Jefferson Medical College of Thomas Jefferson University

Disclosure: Nothing to disclose.

Additional Contributors

Koyamangalath Krishnan, MD, FRCP, FACP, Dishner Endowed Chair of Excellence in Medicine, Professor of Medicine, James H Quillen College of Medicine at East Tennessee State University

Disclosure: Nothing to disclose.

References

  1. Lisenko K, Dingeldein G, Cremer M, Kriegsmann M, Ho AD, Rieger M, et al. Addition of rituximab to CHOP-like chemotherapy in first line treatment of primary mediastinal B-cell lymphoma. BMC Cancer. 2017 May 22. 17 (1):359. [View Abstract]
  2. Miles RR, Mankey CC, Seiler CE 3rd, Smith LB, Teruya-Feldstein J, Hsi ED, et al. Expression of Grb2 distinguishes classical Hodgkin lymphomas from primary mediastinal B-cell lymphomas and other diffuse large B-cell lymphomas. Hum Pathol. 2009 Dec. 40(12):1731-7. [View Abstract]
  3. Feuerhake F, Kutok JL, Monti S, Chen W, LaCasce AS, Cattoretti G, et al. NFkappaB activity, function, and target-gene signatures in primary mediastinal large B-cell lymphoma and diffuse large B-cell lymphoma subtypes. Blood. 2005 Aug 15. 106(4):1392-9. [View Abstract]
  4. Savage KJ, Monti S, Kutok JL, Cattoretti G, Neuberg D, De Leval L, et al. The molecular signature of mediastinal large B-cell lymphoma differs from that of other diffuse large B-cell lymphomas and shares features with classical Hodgkin lymphoma. Blood. 2003 Dec 1. 102(12):3871-9. [View Abstract]
  5. Savage KJ, Al-Rajhi N, Voss N, Paltiel C, Klasa R, Gascoyne RD. Favorable outcome of primary mediastinal large B-cell lymphoma in a single institution: the British Columbia experience. Ann Oncol. 2006 Jan. 17(1):123-30. [View Abstract]
  6. Rieger M, Osterborg A, Pettengell R, White D, Gill D, Walewski J. Primary mediastinal B-cell lymphoma treated with CHOP-like chemotherapy with or without rituximab: results of the Mabthera International Trial Group study. Ann Oncol. 2011 Mar. 22(3):664-70. [View Abstract]
  7. Dubashi B, Cyriac S, Tenali SG. Clinicopathological analysis and outcome of primary mediastinal malignancies - A report of 91 cases from a single institute. Ann Thorac Med. 2009 Jul. 4(3):140-2. [View Abstract]
  8. van Besien K, Kelta M, Bahaguna P. Primary mediastinal B-cell lymphoma: a review of pathology and management. J Clin Oncol. 2001 Mar 15. 19(6):1855-64. [View Abstract]
  9. Steidl C, Gascoyne RD. The molecular pathogenesis of primary mediastinal large B-cell lymphoma. Blood. 2011 Sep 8. 118(10):2659-69. [View Abstract]
  10. Bea S, Zettl A, Wright G, Salaverria I, Jehn P, Moreno V. Diffuse large B-cell lymphoma subgroups have distinct genetic profiles that influence tumor biology and improve gene-expression-based survival prediction. Blood. 2005 Nov 1. 106(9):3183-90. [View Abstract]
  11. Weniger MA, Gesk S, Ehrlich S, Martin-Subero JI, Dyer MJ, Siebert R. Gains of REL in primary mediastinal B-cell lymphoma coincide with nuclear accumulation of REL protein. Genes Chromosomes Cancer. 2007 Apr. 46(4):406-15. [View Abstract]
  12. Song HY, Rothe M, Goeddel DV. The tumor necrosis factor-inducible zinc finger protein A20 interacts with TRAF1/TRAF2 and inhibits NF-kappaB activation. Proc Natl Acad Sci U S A. 1996 Jun 25. 93(13):6721-5. [View Abstract]
  13. Honma K, Tsuzuki S, Nakagawa M, Tagawa H, Nakamura S, Morishima Y, et al. TNFAIP3/A20 functions as a novel tumor suppressor gene in several subtypes of non-Hodgkin lymphomas. Blood. 2009 Sep 17. 114(12):2467-75. [View Abstract]
  14. Kato M, Sanada M, Kato I, Sato Y, Takita J, Takeuchi K. Frequent inactivation of A20 in B-cell lymphomas. Nature. 2009 Jun 4. 459(7247):712-6. [View Abstract]
  15. Davey GM, Heath WR, Starr R. SOCS1: a potent and multifaceted regulator of cytokines and cell-mediated inflammation. Tissue Antigens. 2006 Jan. 67(1):1-9. [View Abstract]
  16. Mottok A, Renné C, Seifert M, Oppermann E, Bechstein W, Hansmann ML. Inactivating SOCS1 mutations are caused by aberrant somatic hypermutation and restricted to a subset of B-cell lymphoma entities. Blood. 2009 Nov 12. 114(20):4503-6. [View Abstract]
  17. Mottok A, Woolcock B, Chan FC, Tong KM, Chong L, Farinha P, et al. Genomic Alterations in CIITA Are Frequent in Primary Mediastinal Large B Cell Lymphoma and Are Associated with Diminished MHC Class II Expression. Cell Rep. 2015 Nov 17. 13 (7):1418-31. [View Abstract]
  18. Lazzarino M, Orlandi E, Paulli M, Sträter J, Klersy C, Gianelli U, et al. Treatment outcome and prognostic factors for primary mediastinal (thymic) B-cell lymphoma: a multicenter study of 106 patients. J Clin Oncol. 1997 Apr. 15(4):1646-53. [View Abstract]
  19. Traverse-Glehen A, Pittaluga S, Gaulard P, Sorbara L, Alonso MA, Raffeld M, et al. Mediastinal gray zone lymphoma: the missing link between classic Hodgkin's lymphoma and mediastinal large B-cell lymphoma. Am J Surg Pathol. 2005 Nov. 29(11):1411-21. [View Abstract]
  20. [Guideline] National Comprehensive Cancer Network. NCCN Clinical Practice Guidelines in Oncology: B-Cell Lymphomas. NCCN.org. Available at https://www.nccn.org/professionals/physician_gls/pdf/b-cell.pdf. Version 4.2019 — June 18, 2019; Accessed: September 14, 2019.
  21. Song HN, Kim SJ, Ko YH, Kim WS. Mediastinal Gray Zone Lymphoma with Features Intermediate between Classical Hodgkin Lymphoma and Primary Mediastinal B-Cell Lymphoma. Acta Haematol. 2016. 136 (3):186-90. [View Abstract]
  22. Sun W, Song K, Zervos M, Pass H, Cangiarella J, Bizekis C, et al. The diagnostic value of endobronchial ultrasound-guided needle biopsy in lung cancer and mediastinal adenopathy. Diagn Cytopathol. 2010 May. 38(5):337-42. [View Abstract]
  23. Jackson MW, Rusthoven CG, Jones BL, Kamdar M, Rabinovitch R. Improved Survival With Radiation Therapy in Stage I-II Primary Mediastinal B Cell Lymphoma: A Surveillance, Epidemiology, and End Results Database Analysis. Int J Radiat Oncol Biol Phys. 2015 Sep 25. [View Abstract]
  24. Zinzani PL, Stefoni V, Finolezzi E, Brusamolino E, Cabras MG, Chiappella A, et al. Rituximab combined with MACOP-B or VACOP-B and radiation therapy in primary mediastinal large B-cell lymphoma: a retrospective study. Clin Lymphoma Myeloma. 2009 Oct. 9(5):381-5. [View Abstract]
  25. Savage KJ, Al-Rajhi N, Voss N, Paltiel C, Klasa R, Gascoyne RD, et al. Favorable outcome of primary mediastinal large B-cell lymphoma in a single institution: the British Columbia experience. Ann Oncol. 2006 Jan. 17(1):123-30. [View Abstract]
  26. Zinzani PL, Martelli M, Magagnoli M, Pescarmona E, Scaramucci L, Palombi F, et al. Treatment and clinical management of primary mediastinal large B-cell lymphoma with sclerosis: MACOP-B regimen and mediastinal radiotherapy monitored by (67)Gallium scan in 50 patients. Blood. 1999 Nov 15. 94(10):3289-93. [View Abstract]
  27. Dunleavy K, Pittaluga S, Maeda LS, Advani R, Chen CC, Hessler J. Dose-adjusted EPOCH-rituximab therapy in primary mediastinal B-cell lymphoma. N Engl J Med. 2013 Apr 11. 368(15):1408-16. [View Abstract]
  28. Witzens-Harig M, Ho AD, Kuhnt E, Trneny M, Rieger M, Österborg A, et al. Primary Mediastinal B Cell Lymphoma Treated with CHOP-Like Chemotherapy with or without Rituximab: 5-Year Results of the Mabthera International Trial Group (MInT) Study. Blood (ASH Annual Meeting Abstracts). 2012. 120:Abstract 1612.
  29. Jackson MW, Rusthoven CG, Jones BL, Kamdar M, Rabinovitch R. Improved survival with combined modality therapy in the modern era for primary mediastinal B-cell lymphoma. Am J Hematol. 2016 May. 91 (5):476-80. [View Abstract]
  30. Spaepen K, Stroobants S, Dupont P, Van Steenweghen S, Thomas J, Vandenberghe P. Prognostic value of positron emission tomography (PET) with fluorine-18 fluorodeoxyglucose ([18F]FDG) after first-line chemotherapy in non-Hodgkin's lymphoma: is [18F]FDG-PET a valid alternative to conventional diagnostic methods?. J Clin Oncol. 2001 Jan 15. 19(2):414-9. [View Abstract]
  31. Jerusalem G, Beguin Y, Fassotte MF, Najjar F, Paulus P, Rigo P. Persistent tumor 18F-FDG uptake after a few cycles of polychemotherapy is predictive of treatment failure in non-Hodgkin's lymphoma. Haematologica. 2000 Jun. 85(6):613-8. [View Abstract]
  32. Haioun C, Itti E, Rahmouni A, Brice P, Rain JD, Belhadj K. [18F]fluoro-2-deoxy-D-glucose positron emission tomography (FDG-PET) in aggressive lymphoma: an early prognostic tool for predicting patient outcome. Blood. 2005 Aug 15. 106(4):1376-81. [View Abstract]
  33. Moskowitz CH, Schöder H, Teruya-Feldstein J, Sima C, Iasonos A, Portlock CS. Risk-adapted dose-dense immunochemotherapy determined by interim FDG-PET in Advanced-stage diffuse large B-Cell lymphoma. J Clin Oncol. 2010 Apr 10. 28(11):1896-903. [View Abstract]
  34. Savage KJ, Yenson PR, Shenkier T, Klasa R, Villa D, Goktepe O, et al. The Outcome of Primary Mediastinal Large B-Cell Lymphoma (PMBCL) in the R-CHOP Treatment Era. Blood (ASH Annual Meeting Abstracts). 2012. 120:Abstract 303.
  35. Popat U, Przepiork D, Champlin R, Pugh W, Amin K, Mehra R, et al. High-dose chemotherapy for relapsed and refractory diffuse large B-cell lymphoma: mediastinal localization predicts for a favorable outcome. J Clin Oncol. 1998 Jan. 16(1):63-9. [View Abstract]
  36. [Guideline] Vitolo U, Seymour JF, Martelli M, Illerhaus G, Illidge T, Zucca E, et al. Extranodal diffuse large B-cell lymphoma (DLBCL) and primary mediastinal B-cell lymphoma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2016 Sep. 27 (suppl 5):v91-v102. [View Abstract]
  37. Zinzani PL, Ribrag V, Moskowitz CH, Michot JM, Kuruvilla J, Balakumaran A, et al. Safety and tolerability of pembrolizumab in patients with relapsed/refractory primary mediastinal large B-cell lymphoma. Blood. 2017 Jul 20. 130 (3):267-270. [View Abstract]

Example of mediastinal lymphoma at diagnosis. A large tumor mass is present in the anterior mediastinum, and an associated pleural effusion can also be seen.

Isolated CNS relapse of primary mediastinal B-cell lymphoma.

Example of mediastinal lymphoma at diagnosis. A large tumor mass is present in the anterior mediastinum, and an associated pleural effusion can also be seen.

Isolated CNS relapse of primary mediastinal B-cell lymphoma.