Aggressive B-cell non-Hodgkin lymphoma (NHL) includes the following subtypes:
- Diffuse large B-cell lymphoma.
- Primary mediastinal large B-cell lymphoma.
- Intravascular large B-cell lymphoma (intravascular lymphomatosis).
- Follicular lymphoma (grade 3b).
- Mantle cell lymphoma.
- Burkitt lymphoma/diffuse small noncleaved-cell lymphoma.
- B-cell lymphoblastic lymphoma.
- Primary effusion lymphoma.
- Plasmablastic lymphoma.
- Polymorphic posttransplant lymphoproliferative disorder.
- Lymphomatoid granulomatosis.
Diffuse Large B-Cell Lymphoma
Diffuse large B-cell lymphoma (DLBCL) is the most common type of NHL and makes up 30% of newly diagnosed cases. Most patients present with rapidly enlarging masses, often with both local and systemic symptoms (designated B symptoms with fever, recurrent night sweats, or weight loss). For more information about weight loss, see Nutrition in Cancer Care.
Some cases of large B-cell lymphoma have a prominent background of reactive T cells and often of histiocytes, so-called T-cell/histiocyte-rich large B-cell lymphoma. This subtype of large cell lymphoma has frequent liver, spleen, and bone marrow involvement; however, the outcome is equivalent to that of similarly staged patients with DLBCL. At diagnosis, some patients with DLBCL have a concomitant indolent small B-cell component. While overall survival (OS) appears similar to de novo DLBCL after multidrug chemotherapy, there is a higher risk of indolent relapse.
Prognosis
Most patients with localized disease are curable with combined-modality therapy or combination chemotherapy alone. Among patients with advanced-stage disease, 50% are cured with doxorubicin-based combination chemotherapy and rituximab, typically R-CHOP (rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone).
The National Comprehensive Cancer Network International Prognostic Index (IPI) for aggressive NHL (diffuse large cell lymphoma) identifies the following five significant risk factors prognostic of overall survival (OS) and their associated risk scores:
- Age.
- 40 years: 0.
- 41–60 years: 1.
- 61–75 years: 2.
- >75 years: 3.
- Stage III/IV: 1.
- Performance status (PS) 2/3/4: 1.
- Serum lactate dehydrogenase (LDH).
- Normalized: 0.
- >1x–3x: 1.
- >3x: 2.
- Number of extranodal sites ≥2: 1.
Risk scores:
- Low (0 or 1): 5-year OS rate, 96%; progression-free survival (PFS) rate, 91%.
- Low intermediate (2 or 3): 5-year OS rate, 82%; PFS rate, 74%.
- High intermediate (4 or 5): 5-year OS rate, 64%; PFS rate, 51%.
- High (>6): 5-year OS rate, 33%; PFS rate, 30%.
Age-adjusted and stage-adjusted modifications of this IPI are used for younger patients with localized disease. Shorter intervals of time between diagnosis and treatment appear to be a surrogate for poor prognostic biological factors.
The BCL2 gene and rearrangement of the MYC gene or dual overexpression of the MYC gene, or both, confer a particularly poor prognosis. Dose-intensive therapies, infusional therapies, and stem cell transplant (SCT) consolidation are being explored in this high-risk group. A retrospective review evaluated 159 patients with previously untreated DLBCL who underwent double-hit genetic testing by fluorescence in situ hybridization (FISH) and achieved complete response. The induction therapy did not alter 3-year relapse-free survival or OS when autologous SCT was used.
In a retrospective review of 117 patients with relapsed or refractory DLBCL who underwent autologous SCT, the 4-year OS rate was 25% for patients with double-hit lymphomas (rearrangement of BCL2 and MYC), 61% for patients with double-expressor lymphomas (no rearrangement, but increased expression of BCL2 and MYC), and 70% for patients without these features. Patients at high risk of relapse consider for clinical trials.
Molecular profiles of gene expression using DNA microarrays may help to stratify patients in the future for therapies directed at specific targets and to better predict survival after standard chemotherapy. For example, true anaplastic lymphoma kinase (ALK)-positive large B-cell lymphomas are extremely rare, and they do not respond well to conventional R-CHOP therapy. Anecdotal responses to ALK inhibitors like lorlatinib or alectinib have been reported. Coexpression of CD20 and CD30 may define a subgroup of patients with DLBCL with a unique molecular signature and a more favorable prognosis. Patients in this subgroup may be treated with an anti-CD30–specific therapy, such as brentuximab vedotin. Patients with DLBCL who are event-free after 2 years have a subsequent OS equivalent to that of the age- and sex-matched general population.
Central nervous system (CNS) prophylaxis
The CNS-IPI tool predicts which patients have a CNS relapse risk exceeding 10%. It was developed by the German Lymphoma Study Group and validated by the British Columbia Cancer Agency database. The presence of four to six of the CNS-IPI risk factors (age >60 years, performance status ≥2, elevated LDH, stage III or IV disease, >1 extranodal site, or involvement of the kidneys or adrenal glands) was used to define a high-risk group for CNS recurrence (a 12% risk of CNS involvement by 2 years).
CNS prophylaxis (usually with four to six doses of intrathecal methotrexate) is often recommended for patients with testicular involvement. A retrospective analysis of the German RICOVER studies compared intrathecal methotrexate with no prophylaxis in patients with DLBCL. This study was completed during the R-CHOP treatment era. With the possible exception of patients with testicular involvement, the analysis showed that intrathecal methotrexate did not reduce the risk of CNS disease. Some clinicians use high-dose intravenous (IV) methotrexate (usually four doses) as an alternative to intrathecal therapy because drug delivery is improved and patient morbidity is decreased. A retrospective study evaluated 1,162 patients from 21 U.S. academic centers where 77% received intrathecal methotrexate, 20% received high-dose IV methotrexate, and 3% received both sequentially (because of toxicity). There was no difference in CNS relapse rates between patients who received intrathecal methotrexate or high-dose IV methotrexate (5.4% vs. 6.8%, P = .40). Testicular involvement, nongerminal center subtype, and high extranodal involvement predicted increased CNS relapse regardless of the route of prophylaxis. Two retrospective studies evaluating high-dose methotrexate in patients with high-risk DLBCL also showed no improvement in CNS relapse rate. Patients deemed at high risk for CNS relapse (e.g., patients with testicular, renal, or adrenal disease and three or more extranodal sites) often receive intrathecal methotrexate or high-dose IV methotrexate, but the lack of confirmatory randomized studies calls this standard into question and shows an urgent need for better therapeutics verified in clinical trials. While there is insufficient evidence to support a significant benefit for CNS prophylaxis in most high-risk patients, the perceived risk of not treating for CNS relapse has often outweighed the lack of evidence for its efficacy. Patients with testicular involvement are an exception, as they show benefit from intrathecal or high-dose IV methotrexate.
The addition of rituximab to cyclophosphamide, doxorubicin, vincristine, prednisone (CHOP)-based regimens has significantly reduced the risk of CNS relapse in retrospective analyses. Patients with CNS dissemination at diagnosis or at relapse usually receive rituximab and high doses of methotrexate and/or cytarabine followed by autologous SCT, but this approach has not been assessed in randomized trials.
Primary Mediastinal Large B-Cell Lymphoma
Primary mediastinal (thymic) large B-cell lymphoma (PMBCL) is a subset of DLBCL with molecular characteristics that are most similar to nodular-sclerosing Hodgkin lymphoma (HL). Mediastinal lymphomas with features intermediate between primary mediastinal B-cell lymphoma and nodular-sclerosing HL are called mediastinal gray-zone lymphomas. Patients are usually female and young (median age, 30–40 years). Patients present with a locally invasive anterior mediastinal mass that may cause respiratory symptoms or superior vena cava syndrome.
Prognosis and therapy are the same as for other comparably staged patients with DLBCL. Uncontrolled phase II studies using dose-adjusted R-EPOCH (etoposide, prednisone, vincristine, cyclophosphamide, and doxorubicin plus rituximab) or R-CHOP showed high cure rates while avoiding any mediastinal radiation. These results suggest that patients who receive R-CHOP–based regimens may avoid the serious long-term complications of radiation therapy when given with chemotherapy. Posttreatment fluorine F 18-fludeoxyglucose (18F-FDG) positron emission tomography–computed tomography (PET-CT) scans are controversial; it remains unclear if PET scans can reliably identify patients who can take or omit radiation therapy consolidation.
A retrospective review of 109 patients with PMBCL showed that 63% had a negative end-of-treatment PET-CT (EOT-PET-CT) (Deauville score 1–3). No radiation therapy was offered and the 5-year time-to-progression rate (similar to disease-free survival, but restricted to lymphoma relapse) was 90%, and the 5-year OS rate was 97%. Patients with a positive EOT-PET-CT received radiation therapy consolidation. It is unclear from this study whether those patients might have done just as well without radiation therapy. Clinicians may monitor patients with Deauville 4 scores on EOT-PET-CT scans for improvement over time, as an alternative to giving radiation therapy. However, this approach has not been studied in a clinical trial.
In situations where mediastinal radiation therapy would encompass the left side of the heart or would increase breast cancer risk in young female patients, proton therapy may be considered to reduce radiation dose to organs at risk. For more information, see the Superior Vena Cava Syndrome section in Cardiopulmonary Syndromes.
Because PMBCL is characterized by high expression of programmed death-ligand 1 (PD-L1) and variable expression of CD30, a phase II study evaluated nivolumab plus brentuximab vedotin in 30 patients with relapsed disease. With a median follow-up of 11.1 months, the objective response rate was 73% (95% CI, 54%−88%). Similarly, a phase II trial of pembrolizumab in 53 patients with relapsed or refractory disease showed an objective response rate of 41.5%. With a median follow-up of 48.7 months, the 4-year PFS rate was 33.0% and the 4-year OS rate was 45.3%. Among the 11 patients who achieved a complete response, all remained in complete response at the time of this final analysis.
Among those who had received two prior lines of therapy, more than one-half of patients who received CAR T-cell therapy with lisocabtagene maraleucel had disease response.
Intravascular Large B-Cell Lymphoma (Intravascular Lymphomatosis)
Intravascular lymphomatosis is characterized by large cell lymphoma confined to the intravascular lumen. The brain, kidneys, lungs, and skin are the organs most likely affected by intravascular lymphomatosis.
With the use of aggressive R-CHOP–based combination chemotherapy, as is used in DLBCL, the prognosis is similar to that of conventional stage IV DLBCL.
Follicular Lymphoma (Grade 3b)
Prognosis
The natural history of follicular large cell lymphoma remains controversial. While there is agreement about the significant number of long-term disease-free survivors with early-stage disease, the potential for cure in patients with advanced disease (stage III or stage IV) remains uncertain. Some groups report a continuous relapse rate similar to the other follicular lymphomas (a pattern of indolent lymphoma). Other investigators report a plateau in freedom from progression at levels expected for an aggressive lymphoma (40% at 10 years). This discrepancy may be caused by variations in histological classification between institutions and the rarity of patients with follicular large cell lymphoma. A retrospective review of 252 patients, all treated with anthracycline-containing combination chemotherapy, showed that patients with more than 50% diffuse components on biopsy had a worse OS than other patients with follicular large cell lymphoma.
Therapeutic approaches
Treatment of follicular large cell lymphoma is more similar to treatment of aggressive NHL than it is to the treatment of indolent NHL. In support of this approach, treatment with high-dose chemotherapy and autologous hematopoietic peripheral SCT shows the same curative potential in patients with follicular large cell lymphoma who relapse as it does in patients with diffuse large cell lymphoma who relapse.
Among those who had received two prior lines of therapy, more than one-half of patients who received CAR T-cell therapy with lisocabtagene maraleucel had disease response.
Mantle Cell Lymphoma
Mantle cell lymphoma (MCL) is found in lymph nodes, the spleen, bone marrow, blood, and sometimes the gastrointestinal system (lymphomatous polyposis). MCL is characterized by CD5-positive follicular mantle B cells, a translocation of chromosomes 11 and 14, and an overexpression of the cyclin D1 protein. MCL may be divided into two clinical subtypes. The first is a classical version with lymphadenopathy with high SOX-11 expression that manifests with an aggressive clinical course and a worse prognosis. The second is a leukemic, non-nodal version with low SOX-11 expression and a more indolent course and a better prognosis. A complex karyotype predicts poor response to induction therapy and inferior survival. There is frequent overlap on presentation with these subtypes, and the therapeutic implication remains unclear. However, both of these versions can converge later in their course into a blastoid phenotype or treatment-resistant phenotype due to genomic instability and selection. TP53 mutations are particularly associated with early disease progression and death for patients receiving conventional chemoimmunotherapy and second-line Bruton tyrosine kinase (BTK) inhibitors.
Like the low-grade lymphomas, MCL appears incurable with anthracycline-based chemotherapy and occurs in older patients with generally asymptomatic advanced-stage disease. The median survival, however, is significantly shorter (5–7 years) than that of other lymphomas. This histology is now considered to be an aggressive lymphoma. A diffuse pattern and the blastoid variant have an aggressive course with shorter survival, while the mantle zone type may have a more indolent course. A high cell-proliferation rate (increased Ki-67, mitotic index, beta-2-microglobulin) may be associated with a poorer prognosis.
Therapeutic approaches
Asymptomatic patients with low-risk scores on the IPI may do well when initial therapy is deferred. There is no standard approach to MCL. Several induction chemotherapy regimens may be used for symptomatic progressing disease. These regimens range in intensity from rituximab alone to rituximab plus ibrutinib, rituximab plus bendamustine, R-CHOP, or high-dose intensive regimens such as R-hyper C-VAD (hyperfractionated cyclophosphamide, vincristine, doxorubicin, and dexamethasone alternating with methotrexate and cytarabine). Some physicians use autologous SCT or allogeneic SCT consolidation next, while others prefer rituximab maintenance, reserving high-dose consolidation for a later time. Ibrutinib, lenalidomide, and bortezomib have shown activity in relapsing patients, and these drugs are being incorporated up front.
It is unclear which therapeutic approach offers the best long-term survival in this clinicopathological entity.
In a phase II trial of previously untreated patients older than 64 years with MCL, 50 patients received the B-cell receptor-inhibitor ibrutinib plus rituximab. With a median follow-up of 45 months, the overall response rate was 96%, the complete response rate was 71%, the 3-year PFS rate was 87%, and the 3-year OS rate was 94%. In a phase II trial of 131 previously untreated patients with MCL aged 65 years or younger, 1 year of ibrutinib plus 4 weeks of rituximab resulted in a complete response rate of 89% prior to any chemotherapy consolidation. Another phase II trial using ibrutinib plus rituximab included asymptomatic patients with previously untreated MCL; the complete response rate was 87%. Previously treated patients who received ibrutinib had a response rate of 86% (21% complete response rate) and a median PFS of 14 months. In a prospective randomized trial, 280 patients with relapsed or refractory MCL received either ibrutinib or temsirolimus. With a median follow-up of 15 months, the median PFS favored ibrutinib (14.6 months vs. 6.2 months; hazard ratio , 0.43; 95% CI, 0.32–0.58, P .0001). Ibrutinib was combined with another active agent, venetoclax, in a phase II study of 23 patients with relapsed or refractory MCL. An unprecedented 71% of patients had a complete response and 78% of responding patients maintained response at 15 months.
A prospective randomized trial included 523 patients aged 65 years and older with MCL. Patients were randomly assigned to receive either ibrutinib, bendamustine, and rituximab or bendamustine and rituximab alone. With a median follow-up of 84.7 months, the median PFS was 80.6 months for patients who received ibrutinib, and 52.9 months for patients who received bendamustine and rituximab alone (HR, 0.75; 95% CI, 0.59–0.96; P = .01). There was no difference in the 7-year OS rate (55.0% vs. 56.8%; HR, 1.07; 95% CI, 0.81–1.40). It is unclear if patients who received ibrutinib alone could have achieved these same results without receiving conventional chemotherapy. The magnitude of benefit demonstrated by the PFS results contrasted with the insufficient OS benefit after 7 years may cast doubt on the long-term safety of this combination.
In a prospective randomized trial, 560 patients older than 60 years and not eligible for SCT were given either R-CHOP or R-FC (rituximab, fludarabine, cyclophosphamide) for six to eight cycles, followed by maintenance therapy in responders randomly assigned to rituximab or interferon-alpha maintenance therapy. With a median follow-up of 7.6 years, the median OS was significantly shorter after R-FC than after R-CHOP (3.9 years vs. 6.4 years; P = .0054). In the same trial, with a median follow-up of 8 years for the 316 responding patients, rituximab maintenance resulted in improved OS over interferon maintenance (median OS, 9.8 years vs. 7.1 years; P = .009). Patients responsive to R-CHOP benefitted most from rituximab in OS (median, 9.8 years vs. 6.4 years; P = .0026). A randomized trial compared bendamustine plus rituximab (BR) with R-CHOP and showed improved PFS (35 vs. 22 months; HR, 0.49; 95% CI, 0.28–0.79; P = .004) but no difference in OS. However, this trial failed to show any benefit for rituximab maintenance after BR. A prospective randomized trial of 487 patients compared VR-CAP (bortezomib, rituximab, cyclophosphamide, doxorubicin, prednisone) with R-CHOP. With a median follow-up of 82 months, the median OS was longer for VR-CAP (90.7 months) compared with R-CHOP (55.7 months) (HR, 0.66; 95% CI, 0.51−0.85; P = .001).
A prospective randomized trial of 497 patients younger than 65 years compared six cycles of R-CHOP with six cycles of alternating R-CHOP and R-DHAP (rituximab, dexamethasone, cytarabine, and cisplatin), with both groups then receiving autologous SCT. With a median follow-up of 10.6 years, the 10-year PFS rate was 73% for patients who received R-DHAP and 57% for patients who received R-CHOP (HR, 0.56; P = .038), but there was no difference in the 10-year OS rates (60% vs. 55% ; HR, 0.80; 95% CI, 0.61–1.06; P = .12). This is the randomized trial referenced by all subsequent articles establishing a role for cytarabine in induction therapy; the ultimate lack of survival advantage casts doubt on this assertion.
Randomized trials have not confirmed an OS benefit in patients who receive consolidation therapy with autologous SCT since the introduction of rituximab. A retrospective analysis of 1,265 patients younger than 65 years who were transplant-eligible showed no benefit for autologous SCT in time-to-next treatment (HR, 0.84; 95% CI, 0.68–1.03) or OS (HR, 0.86; 95% CI, 0.63–1.18). This same retrospective analysis of real-world cohorts found benefit for maintenance rituximab (after BR induction) in time-to-next treatment (HR, 1.96; 95% CI, 1.61–2.38; P .001) and OS (HR, 1.51; 95% CI, 1.19–1.92; P .001).
In a prospective trial (NCT00921414) of 299 patients with previously untreated MCL, 257 responders received four courses of R-DHAP and autologous SCT. The patients were randomly assigned to receive rituximab maintenance therapy for 3 years versus no maintenance therapy. After randomization and a median follow-up of 50.2 months, the 4-year PFS rate was 83% in the rituximab-maintenance arm (95% CI, 73%–88%) and 64% in the no-maintenance arm (95% CI, 55%–73%; P .001). The 4-year OS rate also favored the rituximab-maintenance arm at 89% (95% CI, 81%–94%) versus the no-maintenance arm at 80% (95% CI, 72%–88%; P = .04).
Lenalidomide with or without rituximab also shows response rates of around 50% in relapsed patients, with even higher response rates for previously untreated patients.
Acalabrutinib (another B-cell receptor inhibitor via the BTK pathway) was studied in 124 patients with relapsed or refractory MCL. In a phase II study, there was an 81% overall response rate, 40% complete response rate, and 67% 1-year PFS rate. The BTK inhibitor zanubrutinib was evaluated in a phase II study of 86 patients with relapsed or refractory MCL. After a median follow-up of 35.3 months, the overall response rate was 84%, the complete response rate was 78%, and the median PFS was 33.0 months.
Patients with relapsed or refractory MCL whose disease did not respond to ibrutinib or acalabrutinib were enrolled in a phase II trial (ZUMA-2 ) of brexucabtagene autoleucel, an anti-CD19 CAR T-cell therapy. With a median follow-up of 36 months, 68 patients had an objective response rate of 91% (95% CI, 82%−97%) and a complete response rate of 68% (95% CI, 55%−78%). Median PFS and OS were 25.8 months (95% CI, 10–48) and 46.6 months (95% CI, 24.9–not estimable), respectively. Grade 3 or higher cytokine release syndrome occurred in 15% of patients, and neurological events occurred in 31% of patients. A subsequent retrospective evaluation at 16 institutions included 168 patients who received brexucabtagene autoleucel as part of the U.S. Lymphoma CAR-T Consortium. The study showed similar results to the ZUMA-2 trial.
The reversible, noncovalent, BTK inhibitor pirtobrutinib was evaluated in a phase I/II study of 164 patients with MCL. Seventy-nine patients (87.8%) received at least one dose at the recommended phase II dose of 200 mg once daily. Among 90 patients previously treated with covalent BTK inhibitors included in the primary efficacy cohort, the overall response rate was 57.8% (95% CI, 46.9%–68.1%), including a complete response rate of 20.0%. With a median follow-up of 12 months, the median duration of response was 21.6 months (95% CI, 7.5–not reached). In the safety cohort of 164 patients with MCL, the most common treatment-emergent adverse events were fatigue (29.9%), diarrhea (21.3%), and dyspnea (16.5%). Grade 3 or higher hemorrhage occurred in 3.7% of patients, and atrial fibrillation/flutter occurred in 1.2%. Only 3% of patients discontinued pirtobrutinib because of a treatment-related adverse event. The U.S. Food and Drug Administration approved pirtobrutinib for patients who have received at least two prior lines of therapy, including another BTK inhibitor.
In summary, the optimal sequencing of these various therapies is unclear and is the subject of an ongoing Intergroup clinical trial. Rituximab, lenalidomide, ibrutinib, acalabrutinib, zanubrutinib, pirtobrutinib, and venetoclax represent directed biological agents that may lead to chemotherapy-free treatment strategies for patients with MCL. Most studies support 2 to 3 years of rituximab maintenance therapy after induction therapy (with or without consolidation).
Routine administration of CNS prophylaxis in high-risk MCL has never been studied in a prospective randomized trial. The use of intrathecal or high-dose methotrexate or the use of systemic therapies with CNS penetration like ibrutinib, high-dose cytarabine, or venetoclax, have not been studied and proven efficacious in this situation.
Burkitt Lymphoma/Diffuse Small Noncleaved-Cell Lymphoma
Burkitt lymphoma/diffuse small noncleaved-cell lymphoma typically involves younger patients and represents the most common type of pediatric NHL. These types of aggressive extranodal B-cell lymphomas are characterized by translocation and deregulation of the MYC gene on chromosome 8. A subgroup of patients with dual translocation of MYC and BCL2 appear to have an extremely poor outcome despite aggressive therapy (median OS, 5 months).
In some patients with larger B cells, there is morphological overlap with DLBCL. These Burkitt-like large cell lymphomas show MYC deregulation, extremely high proliferation rates, and a gene-expression profile as expected for classic Burkitt lymphoma. Endemic cases, usually from Africa, involve the facial bones or jaws of children, mostly containing Epstein-Barr virus (EBV) genomes. Sporadic cases usually involve the gastrointestinal system, ovaries, or kidneys. Patients present with rapidly growing masses and a very high LDH but are potentially curable with intensive doxorubicin-based combination chemotherapy.
Therapeutic approaches
Treatment of Burkitt lymphoma/diffuse small noncleaved-cell lymphoma involves aggressive multidrug regimens in combination with rituximab, similar to those used for the advanced-stage aggressive lymphomas (diffuse large cell). Aggressive combination chemotherapy, which is modeled after that used in childhood Burkitt lymphoma, has been very successful for adult patients with more than 60% of advanced-stage patients free of disease at 5 years. Adverse prognostic factors include bulky abdominal disease and high serum LDH. Patients with Burkitt lymphoma have a 20% to 30% lifetime risk of CNS involvement. Prophylaxis with intrathecal chemotherapy is required as part of induction therapy. Patients with HIV-associated Burkitt lymphoma also benefit from less-toxic modification of the aggressive multidrug regimens in combination with rituximab. For more information, see Primary CNS Lymphoma Treatment and AIDS-Related Lymphoma Treatment.
B-Cell Lymphoblastic Lymphoma
B-cell lymphoblastic lymphoma (precursor T-cell) is a very aggressive form of NHL. It is less common than T-cell lymphoblastic lymphoma.
Treatment is usually modeled after that for acute lymphoblastic leukemia. Intensive combination chemotherapy with or without bone marrow transplant is the standard treatment for this aggressive histological type of NHL. Radiation therapy is sometimes given to areas of bulky tumor masses. Because these forms of NHL tend to progress quickly, combination chemotherapy is instituted rapidly once the diagnosis is confirmed. Careful review of the pathological specimens, bone marrow aspirate, biopsy specimen, cerebrospinal fluid cytology, and lymphocyte marker constitute the most important aspects of the pretreatment staging workup. For more information, see Acute Lymphoblastic Leukemia Treatment.
Primary Effusion Lymphoma
Primary effusion lymphoma presents exclusively or mainly in the pleural, pericardial, or abdominal cavities in the absence of an identifiable tumor mass. Patients are usually HIV seropositive, and the tumor usually contains Kaposi sarcoma–associated herpes virus/human herpes virus 8.
Prognosis
The prognosis of primary effusion lymphoma is extremely poor.
Therapeutic approaches
Therapy is usually modeled after the treatment of comparably staged diffuse large cell lymphomas.
Plasmablastic Lymphoma
Plasmablastic lymphoma is most often seen in patients with HIV infection and is characterized by CD20-negative large B cells with plasmacytic features. This type of lymphoma has a very aggressive clinical course, including poor responses and short remissions with standard chemotherapy. Anecdotal reports suggest using aggressive chemotherapy for Burkitt or lymphoblastic lymphoma, followed by SCT consolidation in responding patients, when feasible.
Polymorphic Posttransplant Lymphoproliferative Disorder
Patients who undergo transplant of the heart, lung, liver, kidney, or pancreas usually require lifelong immunosuppression. This may result in posttransplant lymphoproliferative disorder (PTLD) in 1% to 3% of recipients, which appears as an aggressive lymphoma. Pathologists can distinguish a polyclonal B-cell hyperplasia from a monoclonal B-cell lymphoma; both are almost always associated with EBV.
Prognosis
Poor performance status, grafted organ involvement, high IPI, elevated LDH, and multiple sites of disease are poor prognostic factors for PTLD.
Therapeutic options
In some cases, withdrawal of immunosuppression results in eradication of the lymphoma. When this is unsuccessful or not feasible, a course of rituximab may be considered, because it has shown durable remissions in approximately 60% of patients and a favorable toxicity profile. If these measures fail, doxorubicin-based combination chemotherapy (R-CHOP) is recommended, although some patients can avoid cytotoxic therapy. Localized presentations can be controlled with surgery or radiation therapy alone. These localized mass lesions, which may grow over a period of months, are often phenotypically polyclonal and tend to occur within weeks or a few months after transplant. Multifocal, rapidly progressive disease occurs late after transplant (>1 year) and is usually phenotypically monoclonal and associated with EBV. These patients may have durable remissions using standard chemotherapy regimens for aggressive lymphoma. Instances of EBV-negative PTLD occur even later (median, 5 years posttransplant) and have a worse prognosis; R-CHOP chemotherapy can be applied directly in this circumstance. A sustained clinical response after failure from chemotherapy was attained using an immunotoxin (anti-CD22 B-cell surface antigen antibody linked with ricin, a plant toxin). An anti-interleukin-6 monoclonal antibody is also under clinical evaluation.
Lymphomatoid Granulomatosis
Lymphomatoid granulomatosis is an EBV-positive large B-cell lymphoma with a predominant T-cell background. The histology shows association with angioinvasion and vasculitis, usually manifesting as pulmonary lesions or paranasal sinus involvement.
Patients are managed like others with diffuse large cell lymphoma and require doxorubicin-based combination chemotherapy.