National Cancer Institute National Cancer Institute
U.S. National Institutes of Health National Cancer Institute
Send to Printer
Adult Non-Hodgkin Lymphoma Treatment (PDQ®)     
Last Modified: 01/09/2009
Health Professional Version
Cellular Classification of Adult Non-Hodgkin Lymphoma

Updated REAL/WHO Classification
PDQ Modification of REAL Classification of Lymphoproliferative Diseases
Indolent NHL
Aggressive NHL

Note: Some citations in the text of this section are followed by a level of evidence. The PDQ editorial boards use a formal ranking system to help the reader judge the strength of evidence linked to the reported results of a therapeutic strategy. (Refer to the PDQ summary on Levels of Evidence 1 for more information.)

A pathologist should be consulted prior to a biopsy because some studies require special preparation of tissue (e.g., frozen tissue). Knowledge of cell surface markers and immunoglobulin and T-cell receptor gene rearrangements may help with diagnostic and therapeutic decisions. The clonal excess of light chain immunoglobulin may differentiate malignant from reactive cells. Since the prognosis and the approach to treatment are influenced by histopathology, outside biopsy specimens should be carefully reviewed by a hematopathologist who is experienced in diagnosing lymphomas. Although lymph node biopsies are recommended whenever possible, sometimes immunophenotypic data are sufficient to allow diagnosis of lymphoma when fine-needle aspiration cytology is preferred.[1,2]

Historically, uniform treatment of patients with non-Hodgkin lymphoma (NHL) has been hampered by the lack of a uniform classification system. In 1982, results of a consensus study were published as the Working Formulation.[3] The Working Formulation combined results from six major classification systems into one classification. This allowed comparison of studies from different institutions and countries. The Rappaport Classification, which also follows, is no longer in common use.

Historical Classification Systems for Non-Hodgkin Lymphoma
Working Formulation [3]  Rappaport Classification 
Low grade
A. Small lymphocytic, consistent with chronic lymphocytic leukemia Diffuse lymphocytic, well-differentiated
B. Follicular, predominantly small-cleaved cell Nodular lymphocytic, poorly differentiated
C. Follicular, mixed small-cleaved, and large cell Nodular mixed, lymphocytic, and histiocytic
Intermediate grade
D. Follicular, predominantly large cell Nodular histiocytic
E. Diffuse, small-cleaved cell Diffuse lymphocytic, poorly differentiated
F. Diffuse mixed, small and large cell Diffuse mixed, lymphocytic, and histiocytic
G. Diffuse, large cell, cleaved, or noncleaved cell Diffuse histiocytic
High grade
H. Immunoblastic, large cell Diffuse histiocytic
I. Lymphoblastic, convoluted, or nonconvoluted cell Diffuse lymphoblastic
J. Small noncleaved-cell, Burkitt, or non-Burkitt Diffuse undifferentiated Burkitt or non-Burkitt

As the understanding of NHL has improved and as the histopathologic diagnosis of NHL has become more sophisticated with the use of immunologic and genetic techniques, a number of new pathologic entities have been described.[4] In addition, the understanding and treatment of many of the previously described pathologic subtypes have changed. As a result, the Working Formulation has become outdated and less useful to clinicians and pathologists. Thus, European and American pathologists have proposed a new classification, the Revised European American Lymphoma (REAL) Classification.[5-8] Since 1995, members of the European and American Hematopathology societies have been collaborating on a new World Health Organization (WHO) classification, which represents an updated version of the REAL system.[9-11]

The WHO modification of the REAL classification recognizes three major categories of lymphoid malignancies based on morphology and cell lineage: B-cell neoplasms, T-cell/natural killer (NK)–cell neoplasms, and Hodgkin lymphoma. Both lymphomas and lymphoid leukemias are included in this classification because both solid and circulating phases are present in many lymphoid neoplasms and distinction between them is artificial. For example, B-cell chronic lymphocytic leukemia and B-cell small lymphocytic lymphoma are simply different manifestations of the same neoplasm, as are lymphoblastic lymphomas and acute lymphocytic leukemias. Within the B-cell and T-cell categories, two subdivisions are recognized: precursor neoplasms, which correspond to the earliest stages of differentiation, and more mature differentiated neoplasms.[9-11]

Updated REAL/WHO Classification

B-cell neoplasms

  1. Precursor B-cell neoplasm: precursor B-acute lymphoblastic leukemia/lymphoblastic lymphoma (LBL).
  2. Peripheral B-cell neoplasms.
    1. B-cell chronic lymphocytic leukemia/small lymphocytic lymphoma.
    2. B-cell prolymphocytic leukemia.
    3. Lymphoplasmacytic lymphoma/immunocytoma.
    4. Mantle cell lymphoma.
    5. Follicular lymphoma.
    6. Extranodal marginal zone B-cell lymphoma of mucosa-associated lymphatic tissue (MALT) type.
    7. Nodal marginal zone B-cell lymphoma (± monocytoid B-cells).
    8. Splenic marginal zone lymphoma (± villous lymphocytes).
    9. Hairy cell leukemia.
    10. Plasmacytoma/plasma cell myeloma.
    11. Diffuse large B-cell lymphoma.
    12. Burkitt lymphoma.

T-cell and putative NK-cell neoplasms

  1. Precursor T-cell neoplasm: precursor T-acute lymphoblastic leukemia/LBL.
  2. Peripheral T-cell and NK-cell neoplasms.
    1. T-cell chronic lymphocytic leukemia/prolymphocytic leukemia.
    2. T-cell granular lymphocytic leukemia.
    3. Mycosis fungoides/Sézary syndrome.
    4. Peripheral T-cell lymphoma, not otherwise characterized.
    5. Hepatosplenic gamma/delta T-cell lymphoma.
    6. Subcutaneous panniculitis-like T-cell lymphoma.
    7. Angioimmunoblastic T-cell lymphoma.
    8. Extranodal T-/NK-cell lymphoma, nasal type.
    9. Enteropathy-type intestinal T-cell lymphoma.
    10. Adult T-cell lymphoma/leukemia (human T-lymphotrophic virus [HTLV] 1+).
    11. Anaplastic large cell lymphoma, primary systemic type.
    12. Anaplastic large cell lymphoma, primary cutaneous type.
    13. Aggressive NK-cell leukemia.

Hodgkin lymphoma

  1. Nodular lymphocyte–predominant Hodgkin lymphoma.
  2. Classical Hodgkin lymphoma.
    1. Nodular sclerosis Hodgkin lymphoma.
    2. Lymphocyte-rich classical Hodgkin lymphoma.
    3. Mixed-cellularity Hodgkin lymphoma.
    4. Lymphocyte-depleted Hodgkin lymphoma.

The REAL classification encompasses all the lymphoproliferative neoplasms. Refer to the following PDQ summaries for more information:

The more than 20 clinicopathologic entities described here can be divided into the more clinically useful indolent or aggressive lymphomas as follows:

PDQ Modification of REAL Classification of Lymphoproliferative Diseases
  1. Plasma cell disorders. (Refer to the PDQ summary on Multiple Myeloma and Other Plasma Cell Neoplasms Treatment 8 for more information.)
    1. Bone.
    2. Extramedullary.
      1. Monoclonal gammopathy of undetermined significance.
      2. Plasmacytoma.
      3. Multiple myeloma.
      4. Amyloidosis.
  2. Hodgkin lymphoma. (Refer to the PDQ summary on Adult Hodgkin Lymphoma Treatment 4 for more information.)
    1. Nodular sclerosis Hodgkin lymphoma.
    2. Lymphocyte-rich classical Hodgkin lymphoma.
    3. Mixed-cellularity Hodgkin lymphoma.
    4. Lymphocyte-depleted Hodgkin lymphoma.
  3. Indolent lymphoma/leukemia.
    1. Follicular lymphoma (follicular small-cleaved cell [grade 1], follicular mixed small-cleaved, and large cell [grade 2], and diffuse small-cleaved cell).
    2. Chronic lymphocytic leukemia/small lymphocytic lymphoma. (Refer to the PDQ summary on Chronic Lymphocytic Leukemia Treatment 6 for more information.)
    3. Lymphoplasmacytic lymphoma (Waldenström macroglobulinemia).
    4. Extranodal marginal zone B-cell lymphoma (MALT lymphoma).
    5. Nodal marginal zone B-cell lymphoma (monocytoid B-cell lymphoma).
    6. Splenic marginal zone lymphoma (splenic lymphoma with villous lymphocytes).
    7. Hairy cell leukemia. (Refer to the PDQ summary on Hairy Cell Leukemia Treatment 7 for more information.)
    8. Mycosis fungoides/Sézary syndrome. (Refer to the PDQ summary on Mycosis Fungoides/Sézary Syndrome Treatment 9 for more information.)
    9. T-cell granular lymphocytic leukemia. (Refer to the PDQ summary on Chronic Lymphocytic Leukemia Treatment 6 for more information.)
    10. Primary cutaneous anaplastic large cell lymphoma/lymphomatoid papulosis (CD30+).
    11. Nodular lymphocyte–predominant Hodgkin lymphoma. (Refer to the PDQ summary on Adult Hodgkin Lymphoma Treatment 4 for more information.)
  4. Aggressive lymphoma/leukemia.
    1. Diffuse large cell lymphoma (includes diffuse mixed-cell, diffuse large cell, immunoblastic, and T-cell rich large B-cell lymphoma).

      Distinguish:

      1. Mediastinal large B-cell lymphoma.
      2. Follicular large cell lymphoma (grade 3).
      3. Anaplastic large cell lymphoma (CD30+).
      4. Extranodal NK-/T-cell lymphoma, nasal type/aggressive NK-cell leukemia/blastic NK-cell lymphoma.
      5. Lymphomatoid granulomatosis (angiocentric pulmonary B-cell lymphoma).
      6. Angioimmunoblastic T-cell lymphoma.
      7. Peripheral T-cell lymphoma, unspecified.
        1. Subcutaneous panniculitis-like T-cell lymphoma.
        2. Hepatosplenic T-cell lymphoma.
      8. Enteropathy-type T-cell lymphoma.
      9. Intravascular large B-cell lymphoma.
    2. Burkitt lymphoma/Burkitt cell leukemia/Burkitt-like lymphoma.
    3. Precursor B-cell or T-cell lymphoblastic lymphoma/leukemia. (Refer the PDQ summary on Adult Acute Lymphoblastic Leukemia Treatment 3 for more information.)
    4. Primary central nervous system (CNS) lymphoma. (Refer to the PDQ summary on Primary Central Nervous System Lymphoma Treatment 10 for more information.)
    5. Adult T-cell leukemia/lymphoma (HTLV 1+).
    6. Mantle cell lymphoma.
    7. Polymorphic posttransplantation lymphoproliferative disorder (PTLD).
    8. AIDS-related lymphoma. (Refer to the PDQ summary on AIDS-Related Lymphoma Treatment 5 for more information.)
    9. True histiocytic lymphoma.
    10. Primary effusion lymphoma.
    11. B-cell or T-cell prolymphocytic leukemia. (Refer to the PDQ summary on Chronic Lymphocytic Leukemia Treatment 6 for more information.)
Indolent NHL

Follicular lymphoma

Follicular lymphoma comprises 20% of all non-Hodgkin lymphomas and as many as 70% of the indolent lymphomas reported in American and European clinical trials.[7,8,11] Most patients with follicular lymphoma are 50 years and older and present with widespread disease at diagnosis. Nodal involvement is most common and is often accompanied by splenic and bone marrow disease. Rearrangement of the bcl-2 gene is present in more than 90% of patients with follicular lymphoma; overexpression of the bcl-2 protein is associated with the inability to eradicate the lymphoma by inhibiting apoptosis.[12]

Despite the advanced stage, the median survival ranges from 8 to 15 years, leading to the designation of being indolent.[13-15] Patients with advanced-stage follicular lymphoma are not cured with current therapeutic options.[16] The rate of relapse is fairly consistent over time, even in patients who have achieved complete responses to treatment.[17] Watchful waiting, i.e., the deferring of treatment until the patient becomes symptomatic, is an option for patients with advanced-stage follicular lymphoma.[18] An international index for follicular lymphoma (i.e., the Follicular Lymphoma International Prognostic Index [FLIPI])[19-21] identified five significant risk factors prognostic of overall survival (OS):

  1. Age (≤60 years vs. >60 years).
  2. Serum lactate dehydrogenase (normal vs. elevated).
  3. Stage (stage I or stage II vs. stage III or stage IV).
  4. Hemoglobin level (≥120 g/L vs. <120 g/L).
  5. Number of nodal areas (≤4 vs. >4).

Patients with 0 to 1 risk factors have an 85% 10-year survival rate, while three or more risk factors confer a 40% 10-year survival rate.[19] Gene expression profiles of tumor biopsy specimens suggest that follicular lymphoma that is surrounded by infiltrating T-lymphocytes has a much longer median survival (13.6 years) than follicular lymphoma that is surrounded by dendritic and monocytic cells (3.9 years) (P < .001).[22] These infiltrating nonmalignant cells may be valuable therapeutic targets.[23]

Follicular small-cleaved cell lymphoma and follicular mixed small-cleaved and large cell lymphoma do not have reproducibly different disease-free survival or OS.[10] Therapeutic options include watchful waiting; rituximab, an anti-CD20 monoclonal antibody, alone or with purine nucleoside analogs; oral alkylating agents; and combination chemotherapy.[24] Radiolabeled monoclonal antibodies, vaccines, and autologous or allogeneic bone marrow or peripheral stem cell transplantation are also under clinical evaluation.[24] Currently, no randomized trials guide clinicians about the initial choice of rituximab, nucleoside analogs, alkylating agents, combination chemotherapy, radiolabeled monoclonal antibodies, or combinations of these options. On a comparative basis, it is difficult to prove benefit when relapsing disease is followed with watchful waiting, or when the median survival is more than 10 years.

Patients with indolent lymphoma may experience a relapse with a more aggressive histology. If the clinical pattern of relapse suggests that the disease is behaving in a more aggressive manner, a biopsy should be performed. Documentation of conversion to a more aggressive histology requires an appropriate change to a therapy applicable to that histologic type.[25] Rapid growth or discordant growth between various disease sites may indicate a histologic conversion. The risk of histologic transformation was 30% by 10 years in a retrospective review of 325 patients from diagnosis between 1972 and 1999.[26] In this series, high-risk factors for subsequent histologic transformation were advanced stage, high-risk Follicular Lymphoma International Prognostic Index (FLIPI), and expectant management. The median survival after transformation was 1 to 2 years, with 25% of patients alive at 5 years and with approximately 10% to 20% of patients alive 10 years after retreatment.[27] Histologic conversions should be treated with the regimens described in the Aggressive, Recurrent Adult Non-Hodgkin's Lymphoma 11 section of this summary. The durability of the second remission may be short, and clinical trials should be considered.[27-29]

Lymphoplasmacytic lymphoma (Waldenström macroglobulinemia)

Lymphoplasmacytic lymphoma is usually associated with a monoclonal serum paraprotein of immunoglobulin M (IgM) type (Waldenström macroglobulinemia).[30-32] Most patients have bone marrow, lymph node, and splenic involvement, and some patients may develop hyperviscosity syndrome. Other lymphomas may also be associated with serum paraproteins.

The management of lymphoplasmacytic lymphoma is similar to that of other low-grade lymphomas, especially diffuse small lymphocytic lymphoma/chronic lymphocytic leukemia.[31-35] If the viscosity relative to water is greater than four, the patient may have manifestations of hyperviscosity. Plasmapheresis is useful for temporary, acute symptoms (such as retinopathy, congestive heart failure, and CNS dysfunction) but should be combined with chemotherapy for prolonged control of the disease. Symptomatic patients with a serum viscosity of not more than four are usually started directly on chemotherapy. Therapy may be required to correct hemolytic anemia in patients with chronic cold agglutinin disease; chlorambucil, with or without prednisone, is the mainstay. Occasionally, a heated room is required for patients whose cold agglutinins become activated by even minor chilling.

Asymptomatic patients can be monitored for evidence of disease progression without immediate need for chemotherapy.[18] First-line regimens include rituximab, the nucleoside analogs, and alkylating agents, either as single agents or as part of combination chemotherapy.[36,37] Rituximab shows 60% to 80% response rates in previously untreated patients, but close monitoring of the serum IgM is required because of a sudden rise in this paraprotein at the start of therapy.[36,38,39][Level of evidence: 3iiiDiv] The nucleoside analogues 2-chlorodeoxyadenosine and fludarabine have shown similar response rates for previously untreated patients with lymphoplasmacytic lymphoma.[40-42][Level of evidence: 3iiiDiv] Single-agent alkylators and combination chemotherapy also show similar response rates.[43][Level of evidence: 3iiiDiv] Currently, no randomized trials guide clinicians about the initial choice of rituximab, nucleoside analogs, alkylating agents, combination chemotherapy, or combinations of these options.[31,32,36]

Interferon-alpha also shows activity in this disease, in contrast to poor responses in patients with multiple myeloma.[44] Myeloablative therapy with autologous hematopoietic stem cell support is under clinical evaluation.[45,46] Candidates for this approach should avoid long-term use of alkylating agents or purine nucleoside analogs, which can deplete hematopoietic stem cells.[36] After relapse from alkylating-agent therapy, 92 patients with lymphoplasmacytic lymphoma were randomized to fludarabine versus cyclophosphamide, doxorubicin, and prednisone. Although relapse-free survival favored fludarabine (median duration 19 months vs. 3 months, P < .01), no difference was observed in OS.[47][Level of evidence: 1iiDii] Among patients with concomitant hepatitis C virus (HCV) infection, some will attain a complete or partial remission after loss of detectable HCV RNA with treatment using interferon-alpha with or without ribavirin.[48][Level of evidence: 3iiiDiv]

Marginal zone lymphoma

Marginal zone lymphomas were previously included among the diffuse small lymphocytic lymphomas. When marginal zone lymphomas involve the nodes, they are called monocytoid B-cell lymphomas or nodal marginal zone B-cell lymphomas, and when they involve extranodal sites (e.g., gastrointestinal tract, thyroid, lung, breast, orbit, and skin), they are called MALT lymphomas.[4,49-57]

Many patients have a history of autoimmune disease, such as Hashimoto thyroiditis or Sjögren syndrome, or of Helicobacter gastritis. Most patients present with stage I or stage II extranodal disease, which is most often in the stomach. Treatment of Helicobacter pylori infection may resolve many cases of localized gastric involvement.[56,58-62] After standard antibiotic regimens, 50% of patients show resolution of gastric MALT by endoscopy after 3 months. Other patients may show resolution after 12 to 18 months of observation. Of the patients who attain complete remission, 30% demonstrate monoclonality by immunoglobulin heavy chain rearrangement on stomach biopsies with a 5-year median follow-up.[63] The clinical implication of this finding is unknown. Translocation t(11;18) in patients with gastric MALT predicts for poor response to antibiotic therapy, for Helicobacter pylori–negative testing, and for poor response to oral alkylator chemotherapy.[64-66] Stable asymptomatic patients with persistently positive biopsies have been successfully followed on a watchful waiting approach until disease progression.[61,62] Patients who progress are treated with radiation therapy,[67-70] rituximab,[71] surgery (total gastrectomy or partial gastrectomy plus radiation therapy),[72] chemotherapy,[54] or combined modality therapy.[73] The use of endoscopic ultrasonography may help clinicians to follow responses in these patients.[74] Three small case series (two retrospective and one prospective) reported durable complete remissions after treatment of Helicobacter pylori in patients with aggressive lymphoma (complete remission rate of 35%–88% and a median duration of 21–60 months).[75-77]

Localized involvement of other sites can be treated with radiation or surgery.[68-70,78] Patients with extragastric MALT lymphoma have a higher relapse rate than patients with gastric MALT lymphoma in some series, with relapses many years and even decades later.[79] Many of these recurrences involve different MALT sites than the original location. When disseminated to lymph nodes, bone marrow, or blood, this entity behaves like other low-grade lymphomas.[55,80] For patients with ocular adnexal MALT, antibiotic therapy using doxycycline targeting Chlamydia psittaci resulted in durable remissions for half of the patients in a small series of 27 patients.[81][Level of evidence: 3iiiDiv] Large B-cell lymphomas of MALT sites are classified and treated as diffuse large cell lymphomas.[82]

Patients with nodal marginal zone lymphoma (monocytoid B-cell lymphoma) are treated with the same paradigm of watchful waiting or therapies as described for follicular lymphoma. Among patients with concomitant HCV infection, the majority attain a complete or partial remission after loss of detectable HCV RNA with treatment using interferon-alpha with or without ribavirin.[48][Level of evidence: 3iiiDiv]

The disease variously known as Mediterranean abdominal lymphoma, heavy chain disease, or immunoproliferative small intestinal disease (IPSID), which occurs in young adults in eastern Mediterranean countries, is another version of MALT lymphoma, which responds to antibiotics in its early stages.[83] Campylobacter jejuni has been identified as one of the bacterial species associated with IPSID, and antibiotic therapy may result in remission of the disease.[84]

Splenic marginal zone lymphoma

Splenic marginal zone lymphoma is an indolent lymphoma that is marked by massive splenomegaly and peripheral blood and bone marrow involvement, usually without adenopathy.[85-87] This type of lymphoma is otherwise known as splenic lymphoma with villous lymphocytes. Splenectomy may result in prolonged remission.[57,88] Management is similar to that of other low-grade lymphomas and usually involves rituximab alone or rituximab in combination with purine analogs or alkylating agent chemotherapy.[89] Splenic marginal zone lymphoma responds less well to chemotherapy, which would ordinarily be effective for chronic lymphocytic leukemia.[86,87,89] Among small numbers of patients with splenic marginal zone lymphoma (splenic lymphoma with villous lymphocytes) and infection with HCV, the majority attained a complete or partial remission after loss of detectable HCV RNA with treatment using interferon-alpha with or without ribavirin.[48,90,91][Level of evidence: 3iiiDiv] In contrast, no responses to interferon were seen in six HCV-negative patients.

Primary cutaneous anaplastic large cell lymphoma

Primary cutaneous anaplastic large cell lymphoma presents in the skin only with no pre-existing lymphoproliferative disease and no extracutaneous sites of involvement.[92,93] Patients with this type of lymphoma encompass a spectrum ranging from clinically benign lymphomatoid papulosis, marked by localized nodules that may regress spontaneously, to a progressive and systemic disease requiring aggressive doxorubicin-based combination chemotherapy. This spectrum has been called the primary cutaneous CD30-positive T-cell lymphoproliferative disorder. Patients with localized disease usually undergo radiation therapy. With more disseminated involvement, watchful waiting or doxorubicin-based combination chemotherapy is applied.[92,93]

(Refer to the PDQ summaries on Chronic Lymphocytic Leukemia Treatment 6; Mycosis Fungoides/Sézary Syndrome Treatment 9; Hairy Cell Leukemia Treatment 7; and Adult Hodgkin Lymphoma Treatment 4 for more information.)

Aggressive NHL

Diffuse large cell lymphoma

Diffuse large B-cell lymphoma is the most common of the non-Hodgkin lymphomas and comprises 30% of newly diagnosed cases.[7] Most patients present with rapidly enlarging masses, often with symptoms both locally and systemically (designated B symptoms with fever, recurrent night sweats, or weight loss). (Refer to the PDQ summary on Fever, Sweats, and Hot Flashes 12and for more information on weight loss, refer to the Nutrition in Cancer Care 13 summary.) The vast majority of patients with localized disease are curable with combined modality therapy or combination chemotherapy alone.[94] For patients with advanced-stage disease, 50% of presenting patients are cured with doxorubicin-based combination chemotherapy and rituximab.[95-97]

An International Prognostic Index (IPI) for aggressive NHL (diffuse large cell lymphoma) identifies five significant risk factors prognostic of OS:[98]

  1. Age (≤60 years of age vs. >60 years of age).
  2. Serum lactate dehydrogenase (LDH) (normal vs. elevated).
  3. Performance status (0 or 1 vs. 2–4).
  4. Stage (stage I or stage II vs. stage III or stage IV).
  5. Extranodal site involvement (0 or 1 vs. 2–4).

Patients with two or more risk factors have a less than 50% chance of relapse-free survival and OS at 5 years. This study also identifies patients at high risk of relapse based on specific sites of involvement, including bone marrow, CNS, liver, lung, and spleen. Age-adjusted and stage-adjusted modifications of this IPI are used for younger patients with localized disease.[99] Patients at high risk of relapse may be considered for clinical trials.[100] 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.[101-104]

CNS prophylaxis (usually with four to six injections of methotrexate intrathecally) is recommended for patients with paranasal sinus or testicular involvement. Some clinicians are employing high-dose intravenous methotrexate (usually four doses) as an alternative to intrathecal therapy because drug delivery is improved, and patient morbidity is decreased.[105] CNS prophylaxis for bone marrow involvement is controversial; some investigators recommend it, others do not.[106] A retrospective analysis of 605 patients with diffuse large cell lymphoma who did not receive prophylactic intrathecal therapy identified an elevated serum LDH and more than one extranodal site as independent risk factors for CNS recurrence. Patients with both risk factors have a 17% probability of CNS recurrence at 1 year after diagnosis (95% confidence interval [CI], 7%–28%) versus 2.8% (95% CI, 2.7%–2.9%) for the remaining patients.[107][Level of evidence: 3iiiDiii] Some cases of large B-cell lymphoma have a prominent background of reactive T-cells and often of histiocytes, so-called T-cell/histocyte-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 diffuse large B-cell lymphoma.[108-110] Some patients with diffuse large B-cell lymphoma at diagnosis have a concomitant indolent small B-cell component; while OS appears similar after multidrug chemotherapy, there is a higher risk of indolent relapses.[111]

Mediastinal large B-cell lymphoma (primary mediastinal large B-cell lymphoma)

Primary mediastinal (thymic) large B-cell lymphoma is a subset of diffuse large cell lymphoma characterized by significant fibrosis on histology.[112-118] 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. Therapy and prognosis are the same as for other comparably staged patients with diffuse large cell lymphoma, except for advanced-stage patients with a pleural effusion, who have an extremely poor prognoses (progression-free survival is less than 20%) whether the effusion is cytologically positive or negative. (For information on superior vena cava syndrome and pleural effusion, refer to the Cardiopulmonary Syndromes 14 summary.) High-dose chemotherapy with hematopoietic stem cell rescue has been applied to these poor prognosis patients. Evidence for this approach is anecdotal.[118]

Follicular large cell lymphoma

The natural history of follicular large cell lymphoma remains controversial.[119] While there is agreement about the significant number of long-term disease-free survivors with early stage disease, the curability of 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).[120] Other investigators report a plateau in freedom-from-progression at levels expected for an aggressive lymphoma (40% at 10 years).[121,122] This discrepancy may be caused by variations in histologic 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.[123] Treatment of these patients 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 stem cell transplantation 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.[124][Level of evidence: 3iiiA]

Anaplastic large cell lymphoma

Anaplastic large cell lymphomas (ALCL) may be confused with carcinomas and are associated with the Ki-1 (CD30) antigen. These lymphomas are usually of T-cell origin, often present with extranodal disease, and are found especially in the skin. The translocation of chromosomes 2 and 5 creates a unique fusion protein with a nucleophosmin-ALK.[125] Patients whose lymphomas express ALK (immunohistochemistry) are usually younger and may have systemic symptoms, extranodal disease, and advanced stage disease; however, they have a more favorable survival rate than that of ALK-negative patients.[126] Patients with these types of lymphomas are generally treated the same as patients with diffuse large cell lymphomas and have as good a prognosis as comparably staged patients, as evidenced in the NHL-BFM-90 15 trial. Anaplastic large cell lymphoma in children is usually characterized by systemic and cutaneous disease and has high response rates and good OS with doxorubicin-based combination chemotherapy.[127]

Extranodal NK-/T-cell lymphoma

Extranodal NK-/T-cell lymphoma (nasal type) is an aggressive lymphoma marked by extensive necrosis and angioinvasion, most often presenting in extranodal sites, in particular the nasal or paranasal sinus region.[128-133] Other extranodal sites include the palate, trachea, skin, and gastrointestinal tract. Hemophagocytic syndrome may occur; historically these tumors were considered part of lethal midline granuloma.[134] In most cases, Epstein-Barr virus (EBV) genomes are detectable in the tumor cells and immunophenotyping shows CD56 positivity. Cases with blood and marrow involvement are considered NK-cell leukemia. In addition to doxorubicin-based combination chemotherapy, the increased risk of CNS involvement and of local recurrence has led to recommendations for radiation therapy locally, often prior to the start of chemotherapy, and for intrathecal prophylaxis and/or prophylactic cranial radiation therapy.[129,133,135-138] The highly aggressive course, with poor response and short survival with standard therapies, especially for patients with advanced stage disease, has led some investigators to recommend bone marrow or peripheral stem cell transplantation consolidation.[130-132] NK-/T-cell lymphoma that presents only in the skin has a more favorable prognosis, especially in patients with coexpression of CD30 with CD56.[139]

Lymphomatoid granulomatosis

Lymphomatoid granulomatosis is an EBV-positive large B-cell lymphoma with a predominant T-cell background.[140,141] 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.

Angioimmunoblastic T-cell lymphoma

Angioimmunoblastic T-cell lymphoma was formerly called angioimmunoblastic lymphadenopathy with dysproteinemia. Characterized by clonal T-cell receptor gene rearrangement, this entity is managed like diffuse large cell lymphoma.[142-144] Patients present with profound lymphadenopathy, fever, night sweats, weight loss, skin rash, a positive Coomb test, and polyclonal hypergammaglobulinemia.[134] (For information on night sweats, weight loss, and skin rash, refer to the PDQ summaries on Fever, Sweats, and Hot Flashes 12, Nutrition in Cancer Care 13, and Pruritus 16, respectively.) Opportunistic infections are frequent because of an underlying immune deficiency. Doxorubicin-based combination chemotherapy is recommended as it is for other aggressive lymphomas.[142] Myeloablative chemotherapy and radiation therapy with autologous peripheral stem cell support has been described in anecdotal reports.[145] Occasional spontaneous remissions and protracted responses to steroids only have been reported. B-cell EBV genomes are detected in most affected patients.[146]

Peripheral T-cell lymphoma

Patients with peripheral T-cell lymphoma have diffuse large cell or diffuse mixed lymphoma that expresses a cell surface phenotype of a postthymic (or peripheral) T-cell expressing CD4 or CD8 but not both together.[147] Peripheral T-cell lymphoma encompasses a group of heterogeneous nodal T-cell lymphomas that will require future delineation.[134] This includes the so-called Lennert lymphoma, a T-cell lymphoma admixed with a preponderance of lymphoepithelioid cells. Most investigators report worse response and survival rates for patients with peripheral T-cell lymphomas than for patients with comparably staged B-cell aggressive lymphomas.[148-150] Therapy involves doxorubicin-based combination chemotherapy, which is also used for B-cell diffuse large cell lymphoma. Most patients present with multiple adverse prognostic factors (i.e., older age, stage IV, multiple extranodal sites, and elevated LDH), and these patients have a low (<20%) failure-free survival and OS at 5 years.[150] High-dose chemotherapy with hematopoietic stem cell support has been applied to patients with advanced-stage peripheral T-cell lymphoma. Evidence for this approach is anecdotal.[145,151] Anecdotal responses have also been seen with alemtuzumab, an anti-CD52 monoclonal antibody, or denileukin difitox, a toxin-antibody ligand, after relapse from previous chemotherapy.[152,153] An unusual type of peripheral T-cell lymphoma occurring mostly in young men, hepatosplenic T-cell lymphoma, appears to be localized to the hepatic and splenic sinusoids, with cell surface expression of the T-cell receptor gamma/delta.[154-158] Another variant, subcutaneous panniculitis-like T-cell lymphoma, is localized to subcutaneous tissue associated with hemophagocytic syndrome.[159-162] These patients have cells that express alpha/beta phenotype. Those with gamma-delta phenotype have a more aggressive clinical course and are classified as cutaneous gamma-delta T-cell lymphoma.[163-165] These patients may manifest involvement of the epidermis, dermis, subcutaneous region, or mucosa. These entities have extremely poor prognoses with an extremely aggressive clinical course and are treated with the same paradigm as for the highest-risk groups with diffuse large B-cell lymphoma.

Enteropathy-type intestinal T-cell lymphoma

Enteropathy-type intestinal T-cell lymphoma involves the small bowel of patients with gluten-sensitive enteropathy (celiac sprue).[134,166,167] Since a gluten-free diet prevents the development of lymphoma, patients diagnosed with celiac sprue in childhood rarely develop lymphoma. The diagnosis of celiac disease is usually made by finding villous atrophy in the resected intestine. Surgery is often required for diagnosis and to avoid perforation during therapy. Therapy is with doxorubicin-based combination chemotherapy, but relapse rates appear higher than for comparably staged diffuse large cell lymphoma.[167,168] Complications of treatment include gastrointestinal bleeding, small bowel perforation, and enterocolic fistulae; patients often require parenteral nutrition. (For information on parenteral nutrition, refer to the Gastrointestinal Complications 17 summary and the Nutrition in Cancer Care 13 summary.) Multifocal intestinal perforations and visceral abdominal involvement are seen at the time of relapse. High-dose therapy with hematopoietic stem cell rescue has been applied in first remission or at relapse.[167] Evidence for this approach is anecdotal.

Intravascular large B-cell lymphoma (intravascular lymphomatosis)

Intravascular lymphomatosis is characterized by large cell lymphoma confined to the intravascular lumen; with the use of aggressive combination chemotherapy, the prognosis is similar to more conventional presentations.[169,170] The brain, kidneys, lungs, and skin are the organs most likely affected by intravascular lymphomatosis.

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 non-Hodgkin lymphoma.[171] These types of aggressive extranodal B-cell lymphomas are characterized by translocation and deregulation of the C-myc gene on chromosome 8.[172] A subgroup of patients with dual translocation of C-myc and bcl-2 appear to have an extremely poor outcome despite aggressive therapy (5-month OS).[173][Level of evidence: 3iiiA] In some patients with larger B cells, there is morphologic overlap with diffuse large B-cell lymphoma. These Burkitt-like large cell lymphomas show C-myc deregulation, extremely high proliferation rates, and a gene-expression profile as expected for classic Burkitt lymphoma.[10,174,175] Endemic cases, usually from Africa, involve the facial bones or jaws of children, mostly containing EBV genomes. Sporadic cases usually involve the gastrointestinal system, ovaries, or kidneys. Patients present with rapidly growing masses and a very high lactate dehydrogenase but are potentially curable with intensive doxorubicin-based combination chemotherapy. Treatment of Burkitt lymphoma/diffuse small noncleaved-cell lymphoma involves aggressive multidrug regimens similar to those used for the advanced-stage aggressive lymphomas (diffuse large cell).[176-178] Aggressive combination chemotherapy, which is patterned after that used in childhood Burkitt lymphoma, has been described in CALGB-9251 18, for example, and has been very successful for adult patients with more than 60% of advanced-stage patients free of disease at 5 years.[179-184] Adverse prognostic factors include bulky abdominal disease and high serum LDH. In some institutions, treatment includes the use of consolidative bone marrow transplantation (BMT).[185,186] 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.[187] (Refer to the PDQ summaries on Primary Central Nervous System Lymphoma Treatment 10 and AIDS-Related Lymphoma Treatment 5 for more information.)

Lymphoblastic lymphoma

Lymphoblastic lymphoma (precursor T-cell) is a very aggressive form of NHL. It often occurs in young patients but not exclusively.[188] It is commonly associated with large mediastinal masses and has a high predilection for disseminating to bone marrow and to the CNS. Treatment is usually patterned after that for acute lymphoblastic leukemia. Intensive combination chemotherapy with or without BMT is the standard treatment of this aggressive histologic type of NHL.[189-191] Radiation therapy is sometimes given to areas of bulky tumor masses. Since these forms of NHL tend to progress so quickly, combination chemotherapy is instituted rapidly once the diagnosis has been confirmed. Careful review of the pathologic specimens, bone marrow aspirate, biopsy specimen, cerebrospinal fluid cytology, and lymphocyte marker constitute the most important aspects of the pretreatment staging workup. (Refer to the PDQ summary on Adult Acute Lymphoblastic Leukemia Treatment 3 for more information.)

Adult T-cell leukemia/lymphoma

Adult T-cell leukemia/lymphoma is caused by infection with the retrovirus human T-cell lymphotropic virus type I and is frequently associated with lymphadenopathy, hypercalcemia, circulating leukemic cells, bone and skin involvement, hepatosplenomegaly, a rapidly progressive course, and poor response to chemotherapy.[192,193] (Refer to the PDQ summary on Hypercalcemia 19 for more information.) Using combination chemotherapy, only 10% to 20% of patients survived even 3 years in a trial of 118 patients.[194] The combination of zidovudine and interferon-alpha has activity against adult T-cell leukemia/lymphoma, even for patients who failed previous cytotoxic therapy. Durable remissions are seen in 66% of presenting patients with this combination, but long-term disease-free survival rates are not yet available.[195-197]

Mantle cell lymphoma

Mantle cell lymphoma is found in lymph nodes, the spleen, bone marrow, blood, and sometimes the gastrointestinal system (lymphomatous polyposis).[4,198,199] Mantle cell lymphoma is characterized by CD5-positive follicular mantle B cells, a translocation of chromosomes 11 and 14, and an overexpression of the cyclin D1 protein.[200] Like the low-grade lymphomas, mantle cell lymphoma appears incurable with anthracycline-based chemotherapy and occurs in older patients with generally asymptomatic advanced-stage disease.[201] The median survival, however, is significantly shorter (3–5 years) than that of other lymphomas, and this histology is now considered to be an aggressive lymphoma.[202] 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.[49,203] A high cell proliferation rate (increased Ki-67, mitotic index, beta-2-microglobulin) may be associated with a poorer prognosis.[200,204] It is unclear which chemotherapeutic approach offers the best long-term survival in this clinicopathologic entity; refractoriness to chemotherapy is a usual feature.[202,205-210] Many investigators are exploring high-dose therapy with stem cell/marrow support or the use of interferon or anti-CD20 antibodies after CHOP (cyclophosphamide, doxorubicin, vincristine, prednisone) chemotherapy.[207-209,211-218] Thus far, randomized trials have not shown OS benefits from these newer approaches.[218] Bortezomib shows response rates close to 50% in relapsed patients, prompting clinical trials combining this proteasome inhibitor with rituximab and cytotoxic agents in first-line therapy.[219-221][Level of evidence: 3iiiDiv]

Polymorphic posttransplantation lymphoproliferative disorder (PTLD)

Patients who undergo transplantation of the heart, lung, liver, kidney, or pancreas usually require life-long immunosuppression. This may result in PTLD in 1% to 3% of recipients, which appears as an aggressive lymphoma.[222] Pathologists can distinguish a polyclonal B-cell hyperplasia from a monoclonal B-cell lymphoma; both are almost always associated with EBV.[223] Poor performance status, grafted organ involvement, high IPI, elevated LDH, and multiple sites of disease are poor prognostic factors for PTLD.[224,225] In some cases, withdrawal of immunosuppression results in eradication of the lymphoma.[226] When this is unsuccessful or not feasible, a trial of rituximab may be considered, because it has shown durable remissions in approximately 60% of patients and a favorable toxicity profile.[227] Sometimes, a combination of acyclovir and interferon-alpha has been used.[222,228] If these measures fail, doxorubicin-based combination chemotherapy is recommended, though most patients can avoid cytotoxic therapy.[229] 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 transplantation.[223] Multifocal, rapidly progressive disease occurs late after transplantation (>1 year) and is usually phenotypically monoclonal and associated with EBV.[230] These patients may have durable remissions using standard chemotherapy regimens for aggressive lymphoma.[230-232] Instances of EBV-negative PTLD occur late (median, 5 years posttransplant) and have particularly poor prognoses.[233] 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).[234] An anti-interleukin-6 monoclonal antibody is also under clinical evaluation.[235]

True histiocytic lymphoma

True histiocytic lymphomas are very rare tumors that show histiocytic differentiation and express histiocytic markers in the absence of B-cell or T-cell lineage-specific immunologic markers.[236,237] Care must be taken with immunophenotypic tests to exclude anaplastic large cell lymphoma or hemophagocytic syndromes caused by viral infections, especially EBV. Therapy is modeled after the treatment of comparably staged diffuse large cell lymphomas, but the optimal approach remains to be defined.

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.[238] Patients are usually HIV-seropositive, and the tumor usually contains Kaposi sarcoma-associated herpes virus/human herpes virus 8. Therapy is usually modeled after the treatment of comparably staged diffuse large cell lymphomas, but the prognosis is extremely poor.

References

  1. Zeppa P, Marino G, Troncone G, et al.: Fine-needle cytology and flow cytometry immunophenotyping and subclassification of non-Hodgkin lymphoma: a critical review of 307 cases with technical suggestions. Cancer 102 (1): 55-65, 2004.  [PUBMED Abstract]

  2. Young NA, Al-Saleem T: Diagnosis of lymphoma by fine-needle aspiration cytology using the revised European-American classification of lymphoid neoplasms. Cancer 87 (6): 325-45, 1999.  [PUBMED Abstract]

  3. National Cancer Institute sponsored study of classifications of non-Hodgkin's lymphomas: summary and description of a working formulation for clinical usage. The Non-Hodgkin's Lymphoma Pathologic Classification Project. Cancer 49 (10): 2112-35, 1982.  [PUBMED Abstract]

  4. Pugh WC: Is the working formulation adequate for the classification of the low grade lymphomas? Leuk Lymphoma 10(Suppl): 1-8, 1993. 

  5. Harris NL, Jaffe ES, Stein H, et al.: A revised European-American classification of lymphoid neoplasms: a proposal from the International Lymphoma Study Group. Blood 84 (5): 1361-92, 1994.  [PUBMED Abstract]

  6. Pittaluga S, Bijnens L, Teodorovic I, et al.: Clinical analysis of 670 cases in two trials of the European Organization for the Research and Treatment of Cancer Lymphoma Cooperative Group subtyped according to the Revised European-American Classification of Lymphoid Neoplasms: a comparison with the Working Formulation. Blood 87 (10): 4358-67, 1996.  [PUBMED Abstract]

  7. Armitage JO, Weisenburger DD: New approach to classifying non-Hodgkin's lymphomas: clinical features of the major histologic subtypes. Non-Hodgkin's Lymphoma Classification Project. J Clin Oncol 16 (8): 2780-95, 1998.  [PUBMED Abstract]

  8. A clinical evaluation of the International Lymphoma Study Group classification of non-Hodgkin's lymphoma. The Non-Hodgkin's Lymphoma Classification Project. Blood 89 (11): 3909-18, 1997.  [PUBMED Abstract]

  9. Pileri SA, Milani M, Fraternali-Orcioni G, et al.: From the R.E.A.L. Classification to the upcoming WHO scheme: a step toward universal categorization of lymphoma entities? Ann Oncol 9 (6): 607-12, 1998.  [PUBMED Abstract]

  10. Harris NL, Jaffe ES, Armitage JO, et al.: Lymphoma classification: from R.E.A.L. to W.H.O. and beyond. Cancer: Principles and Practice of Oncology Updates 13(3): 1-14, 1999. 

  11. Society for Hematopathology Program.: Society for Hematopathology Program. Am J Surg Pathol 21(1): 114-121, 1997. 

  12. López-Guillermo A, Cabanillas F, McDonnell TI, et al.: Correlation of bcl-2 rearrangement with clinical characteristics and outcome in indolent follicular lymphoma. Blood 93 (9): 3081-7, 1999.  [PUBMED Abstract]

  13. Peterson BA, Petroni GR, Frizzera G, et al.: Prolonged single-agent versus combination chemotherapy in indolent follicular lymphomas: a study of the cancer and leukemia group B. J Clin Oncol 21 (1): 5-15, 2003.  [PUBMED Abstract]

  14. Swenson WT, Wooldridge JE, Lynch CF, et al.: Improved survival of follicular lymphoma patients in the United States. J Clin Oncol 23 (22): 5019-26, 2005.  [PUBMED Abstract]

  15. Liu Q, Fayad L, Cabanillas F, et al.: Improvement of overall and failure-free survival in stage IV follicular lymphoma: 25 years of treatment experience at The University of Texas M.D. Anderson Cancer Center. J Clin Oncol 24 (10): 1582-9, 2006.  [PUBMED Abstract]

  16. Hiddemann W, Buske C, Dreyling M, et al.: Treatment strategies in follicular lymphomas: current status and future perspectives. J Clin Oncol 23 (26): 6394-9, 2005.  [PUBMED Abstract]

  17. Fisher RI, LeBlanc M, Press OW, et al.: New treatment options have changed the survival of patients with follicular lymphoma. J Clin Oncol 23 (33): 8447-52, 2005.  [PUBMED Abstract]

  18. Ardeshna KM, Smith P, Norton A, et al.: Long-term effect of a watch and wait policy versus immediate systemic treatment for asymptomatic advanced-stage non-Hodgkin lymphoma: a randomised controlled trial. Lancet 362 (9383): 516-22, 2003.  [PUBMED Abstract]

  19. Solal-Céligny P, Roy P, Colombat P, et al.: Follicular lymphoma international prognostic index. Blood 104 (5): 1258-65, 2004.  [PUBMED Abstract]

  20. Perea G, Altés A, Montoto S, et al.: Prognostic indexes in follicular lymphoma: a comparison of different prognostic systems. Ann Oncol 16 (9): 1508-13, 2005.  [PUBMED Abstract]

  21. Buske C, Hoster E, Dreyling M, et al.: The Follicular Lymphoma International Prognostic Index (FLIPI) separates high-risk from intermediate- or low-risk patients with advanced-stage follicular lymphoma treated front-line with rituximab and the combination of cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP) with respect to treatment outcome. Blood 108 (5): 1504-8, 2006.  [PUBMED Abstract]

  22. Dave SS, Wright G, Tan B, et al.: Prediction of survival in follicular lymphoma based on molecular features of tumor-infiltrating immune cells. N Engl J Med 351 (21): 2159-69, 2004.  [PUBMED Abstract]

  23. Küppers R: Prognosis in follicular lymphoma--it's in the microenvironment. N Engl J Med 351 (21): 2152-3, 2004.  [PUBMED Abstract]

  24. Peterson BA: Current treatment of follicular low-grade lymphomas. Semin Oncol 26 (5 Suppl 14): 2-11, 1999.  [PUBMED Abstract]

  25. Tsimberidou AM, O'Brien S, Khouri I, et al.: Clinical outcomes and prognostic factors in patients with Richter's syndrome treated with chemotherapy or chemoimmunotherapy with or without stem-cell transplantation. J Clin Oncol 24 (15): 2343-51, 2006.  [PUBMED Abstract]

  26. Montoto S, Davies AJ, Matthews J, et al.: Risk and clinical implications of transformation of follicular lymphoma to diffuse large B-cell lymphoma. J Clin Oncol 25 (17): 2426-33, 2007.  [PUBMED Abstract]

  27. Yuen AR, Kamel OW, Halpern J, et al.: Long-term survival after histologic transformation of low-grade follicular lymphoma. J Clin Oncol 13 (7): 1726-33, 1995.  [PUBMED Abstract]

  28. Bastion Y, Sebban C, Berger F, et al.: Incidence, predictive factors, and outcome of lymphoma transformation in follicular lymphoma patients. J Clin Oncol 15 (4): 1587-94, 1997.  [PUBMED Abstract]

  29. Williams CD, Harrison CN, Lister TA, et al.: High-dose therapy and autologous stem-cell support for chemosensitive transformed low-grade follicular non-Hodgkin's lymphoma: a case-matched study from the European Bone Marrow Transplant Registry. J Clin Oncol 19 (3): 727-35, 2001.  [PUBMED Abstract]

  30. Facon T, Brouillard M, Duhamel A, et al.: Prognostic factors in Waldenström's macroglobulinemia: a report of 167 cases. J Clin Oncol 11 (8): 1553-8, 1993.  [PUBMED Abstract]

  31. Vijay A, Gertz MA: Waldenström macroglobulinemia. Blood 109 (12): 5096-103, 2007.  [PUBMED Abstract]

  32. Dimopoulos MA, Kyle RA, Anagnostopoulos A, et al.: Diagnosis and management of Waldenstrom's macroglobulinemia. J Clin Oncol 23 (7): 1564-77, 2005.  [PUBMED Abstract]

  33. Leblond V, Ben-Othman T, Deconinck E, et al.: Activity of fludarabine in previously treated Waldenström's macroglobulinemia: a report of 71 cases. Groupe Coopératif Macroglobulinémie. J Clin Oncol 16 (6): 2060-4, 1998.  [PUBMED Abstract]

  34. Foran JM, Rohatiner AZ, Coiffier B, et al.: Multicenter phase II study of fludarabine phosphate for patients with newly diagnosed lymphoplasmacytoid lymphoma, Waldenström's macroglobulinemia, and mantle-cell lymphoma. J Clin Oncol 17 (2): 546-53, 1999.  [PUBMED Abstract]

  35. Baldini L, Goldaniga M, Guffanti A, et al.: Immunoglobulin M monoclonal gammopathies of undetermined significance and indolent Waldenstrom's macroglobulinemia recognize the same determinants of evolution into symptomatic lymphoid disorders: proposal for a common prognostic scoring system. J Clin Oncol 23 (21): 4662-8, 2005.  [PUBMED Abstract]

  36. Gertz MA, Anagnostopoulos A, Anderson K, et al.: Treatment recommendations in Waldenstrom's macroglobulinemia: consensus panel recommendations from the Second International Workshop on Waldenstrom's Macroglobulinemia. Semin Oncol 30 (2): 121-6, 2003.  [PUBMED Abstract]

  37. Dimopoulos MA, Anagnostopoulos A, Kyrtsonis MC, et al.: Primary treatment of Waldenström macroglobulinemia with dexamethasone, rituximab, and cyclophosphamide. J Clin Oncol 25 (22): 3344-9, 2007.  [PUBMED Abstract]

  38. Dimopoulos MA, Zervas C, Zomas A, et al.: Treatment of Waldenström's macroglobulinemia with rituximab. J Clin Oncol 20 (9): 2327-33, 2002.  [PUBMED Abstract]

  39. Treon SP, Branagan AR, Hunter Z, et al.: Paradoxical increases in serum IgM and viscosity levels following rituximab in Waldenstrom's macroglobulinemia. Ann Oncol 15 (10): 1481-3, 2004.  [PUBMED Abstract]

  40. Dimopoulos MA, Kantarjian H, Weber D, et al.: Primary therapy of Waldenström's macroglobulinemia with 2-chlorodeoxyadenosine. J Clin Oncol 12 (12): 2694-8, 1994.  [PUBMED Abstract]

  41. Dimopoulos MA, Alexanian R: Waldenstrom's macroglobulinemia. Blood 83 (6): 1452-9, 1994.  [PUBMED Abstract]

  42. Hellmann A, Lewandowski K, Zaucha JM, et al.: Effect of a 2-hour infusion of 2-chlorodeoxyadenosine in the treatment of refractory or previously untreated Waldenström's macroglobulinemia. Eur J Haematol 63 (1): 35-41, 1999.  [PUBMED Abstract]

  43. García-Sanz R, Montoto S, Torrequebrada A, et al.: Waldenström macroglobulinaemia: presenting features and outcome in a series with 217 cases. Br J Haematol 115 (3): 575-82, 2001.  [PUBMED Abstract]

  44. Rotoli B, De Renzo A, Frigeri F, et al.: A phase II trial on alpha-interferon (alpha IFN) effect in patients with monoclonal IgM gammopathy. Leuk Lymphoma 13 (5-6): 463-9, 1994.  [PUBMED Abstract]

  45. Dreger P, Glass B, Kuse R, et al.: Myeloablative radiochemotherapy followed by reinfusion of purged autologous stem cells for Waldenström's macroglobulinaemia. Br J Haematol 106 (1): 115-8, 1999.  [PUBMED Abstract]

  46. Desikan R, Dhodapkar M, Siegel D, et al.: High-dose therapy with autologous haemopoietic stem cell support for Waldenström's macroglobulinaemia. Br J Haematol 105 (4): 993-6, 1999.  [PUBMED Abstract]

  47. Leblond V, Lévy V, Maloisel F, et al.: Multicenter, randomized comparative trial of fludarabine and the combination of cyclophosphamide-doxorubicin-prednisone in 92 patients with Waldenström macroglobulinemia in first relapse or with primary refractory disease. Blood 98 (9): 2640-4, 2001.  [PUBMED Abstract]

  48. Vallisa D, Bernuzzi P, Arcaini L, et al.: Role of anti-hepatitis C virus (HCV) treatment in HCV-related, low-grade, B-cell, non-Hodgkin's lymphoma: a multicenter Italian experience. J Clin Oncol 23 (3): 468-73, 2005.  [PUBMED Abstract]

  49. Fisher RI, Dahlberg S, Nathwani BN, et al.: A clinical analysis of two indolent lymphoma entities: mantle cell lymphoma and marginal zone lymphoma (including the mucosa-associated lymphoid tissue and monocytoid B-cell subcategories): a Southwest Oncology Group study. Blood 85 (4): 1075-82, 1995.  [PUBMED Abstract]

  50. Isaacson PG: Lymphomas of mucosa-associated lymphoid tissue (MALT). Histopathology 16: 617-619, 1990. 

  51. Nizze H, Cogliatti SB, von Schilling C, et al.: Monocytoid B-cell lymphoma: morphological variants and relationship to low-grade B-cell lymphoma of the mucosa-associated lymphoid tissue. Histopathology 18 (5): 403-14, 1991.  [PUBMED Abstract]

  52. Pimpinelli N, Santucci M, Mori M, et al.: Primary cutaneous B-cell lymphoma: a clinically homogeneous entity? J Am Acad Dermatol 37 (6): 1012-6, 1997.  [PUBMED Abstract]

  53. Li G, Hansmann ML, Zwingers T, et al.: Primary lymphomas of the lung: morphological, immunohistochemical and clinical features. Histopathology 16 (6): 519-31, 1990.  [PUBMED Abstract]

  54. Zinzani PL, Magagnoli M, Galieni P, et al.: Nongastrointestinal low-grade mucosa-associated lymphoid tissue lymphoma: analysis of 75 patients. J Clin Oncol 17 (4): 1254, 1999.  [PUBMED Abstract]

  55. Nathwani BN, Drachenberg MR, Hernandez AM, et al.: Nodal monocytoid B-cell lymphoma (nodal marginal-zone B-cell lymphoma). Semin Hematol 36 (2): 128-38, 1999.  [PUBMED Abstract]

  56. Isaacson PG: Mucosa-associated lymphoid tissue lymphoma. Semin Hematol 36 (2): 139-47, 1999.  [PUBMED Abstract]

  57. Bertoni F, Zucca E: State-of-the-art therapeutics: marginal-zone lymphoma. J Clin Oncol 23 (26): 6415-20, 2005.  [PUBMED Abstract]

  58. Wotherspoon AC, Doglioni C, Diss TC, et al.: Regression of primary low-grade B-cell gastric lymphoma of mucosa-associated lymphoid tissue type after eradication of Helicobacter pylori. Lancet 342 (8871): 575-7, 1993.  [PUBMED Abstract]

  59. Neubauer A, Thiede C, Morgner A, et al.: Cure of Helicobacter pylori infection and duration of remission of low-grade gastric mucosa-associated lymphoid tissue lymphoma. J Natl Cancer Inst 89 (18): 1350-5, 1997.  [PUBMED Abstract]

  60. Roggero E, Zucca E, Pinotti G, et al.: Eradication of Helicobacter pylori infection in primary low-grade gastric lymphoma of mucosa-associated lymphoid tissue. Ann Intern Med 122 (10): 767-9, 1995.  [PUBMED Abstract]

  61. Zucca E, Roggero E, Pileri S: B-cell lymphoma of MALT type: a review with special emphasis on diagnostic and management problems of low-grade gastric tumours. Br J Haematol 100 (1): 3-14, 1998.  [PUBMED Abstract]

  62. Steinbach G, Ford R, Glober G, et al.: Antibiotic treatment of gastric lymphoma of mucosa-associated lymphoid tissue. An uncontrolled trial. Ann Intern Med 131 (2): 88-95, 1999.  [PUBMED Abstract]

  63. Wündisch T, Thiede C, Morgner A, et al.: Long-term follow-up of gastric MALT lymphoma after Helicobacter pylori eradication. J Clin Oncol 23 (31): 8018-24, 2005.  [PUBMED Abstract]

  64. Ye H, Liu H, Raderer M, et al.: High incidence of t(11;18)(q21;q21) in Helicobacter pylori-negative gastric MALT lymphoma. Blood 101 (7): 2547-50, 2003.  [PUBMED Abstract]

  65. Lévy M, Copie-Bergman C, Gameiro C, et al.: Prognostic value of translocation t(11;18) in tumoral response of low-grade gastric lymphoma of mucosa-associated lymphoid tissue type to oral chemotherapy. J Clin Oncol 23 (22): 5061-6, 2005.  [PUBMED Abstract]

  66. Nakamura S, Ye H, Bacon CM, et al.: Clinical impact of genetic aberrations in gastric MALT lymphoma: a comprehensive analysis using interphase fluorescence in situ hybridisation. Gut 56 (10): 1358-63, 2007.  [PUBMED Abstract]

  67. Schechter NR, Yahalom J: Low-grade MALT lymphoma of the stomach: a review of treatment options. Int J Radiat Oncol Biol Phys 46 (5): 1093-103, 2000.  [PUBMED Abstract]

  68. Tsang RW, Gospodarowicz MK, Pintilie M, et al.: Stage I and II MALT lymphoma: results of treatment with radiotherapy. Int J Radiat Oncol Biol Phys 50 (5): 1258-64, 2001.  [PUBMED Abstract]

  69. Tsang RW, Gospodarowicz MK, Pintilie M, et al.: Localized mucosa-associated lymphoid tissue lymphoma treated with radiation therapy has excellent clinical outcome. J Clin Oncol 21 (22): 4157-64, 2003.  [PUBMED Abstract]

  70. Tsai HK, Li S, Ng AK, et al.: Role of radiation therapy in the treatment of stage I/II mucosa-associated lymphoid tissue lymphoma. Ann Oncol 18 (4): 672-8, 2007.  [PUBMED Abstract]

  71. Martinelli G, Laszlo D, Ferreri AJ, et al.: Clinical activity of rituximab in gastric marginal zone non-Hodgkin's lymphoma resistant to or not eligible for anti-Helicobacter pylori therapy. J Clin Oncol 23 (9): 1979-83, 2005.  [PUBMED Abstract]

  72. Cogliatti SB, Schmid U, Schumacher U, et al.: Primary B-cell gastric lymphoma: a clinicopathological study of 145 patients. Gastroenterology 101 (5): 1159-70, 1991.  [PUBMED Abstract]

  73. Thieblemont C, Bastion Y, Berger F, et al.: Mucosa-associated lymphoid tissue gastrointestinal and nongastrointestinal lymphoma behavior: analysis of 108 patients. J Clin Oncol 15 (4): 1624-30, 1997.  [PUBMED Abstract]

  74. Pavlick AC, Gerdes H, Portlock CS: Endoscopic ultrasound in the evaluation of gastric small lymphocytic mucosa-associated lymphoid tumors. J Clin Oncol 15 (5): 1761-6, 1997.  [PUBMED Abstract]

  75. Morgner A, Miehlke S, Fischbach W, et al.: Complete remission of primary high-grade B-cell gastric lymphoma after cure of Helicobacter pylori infection. J Clin Oncol 19 (7): 2041-8, 2001.  [PUBMED Abstract]

  76. Chen LT, Lin JT, Shyu RY, et al.: Prospective study of Helicobacter pylori eradication therapy in stage I(E) high-grade mucosa-associated lymphoid tissue lymphoma of the stomach. J Clin Oncol 19 (22): 4245-51, 2001.  [PUBMED Abstract]

  77. Chen LT, Lin JT, Tai JJ, et al.: Long-term results of anti-Helicobacter pylori therapy in early-stage gastric high-grade transformed MALT lymphoma. J Natl Cancer Inst 97 (18): 1345-53, 2005.  [PUBMED Abstract]

  78. Uno T, Isobe K, Shikama N, et al.: Radiotherapy for extranodal, marginal zone, B-cell lymphoma of mucosa-associated lymphoid tissue originating in the ocular adnexa: a multiinstitutional, retrospective review of 50 patients. Cancer 98 (4): 865-71, 2003.  [PUBMED Abstract]

  79. Raderer M, Streubel B, Woehrer S, et al.: High relapse rate in patients with MALT lymphoma warrants lifelong follow-up. Clin Cancer Res 11 (9): 3349-52, 2005.  [PUBMED Abstract]

  80. Raderer M, Wöhrer S, Streubel B, et al.: Assessment of disease dissemination in gastric compared with extragastric mucosa-associated lymphoid tissue lymphoma using extensive staging: a single-center experience. J Clin Oncol 24 (19): 3136-41, 2006.  [PUBMED Abstract]

  81. Ferreri AJ, Ponzoni M, Guidoboni M, et al.: Bacteria-eradicating therapy with doxycycline in ocular adnexal MALT lymphoma: a multicenter prospective trial. J Natl Cancer Inst 98 (19): 1375-82, 2006.  [PUBMED Abstract]

  82. Kuo SH, Chen LT, Yeh KH, et al.: Nuclear expression of BCL10 or nuclear factor kappa B predicts Helicobacter pylori-independent status of early-stage, high-grade gastric mucosa-associated lymphoid tissue lymphomas. J Clin Oncol 22 (17): 3491-7, 2004.  [PUBMED Abstract]

  83. Isaacson PG: Gastrointestinal lymphoma. Hum Pathol 25 (10): 1020-9, 1994.  [PUBMED Abstract]

  84. Lecuit M, Abachin E, Martin A, et al.: Immunoproliferative small intestinal disease associated with Campylobacter jejuni. N Engl J Med 350 (3): 239-48, 2004.  [PUBMED Abstract]

  85. Franco V, Florena AM, Iannitto E: Splenic marginal zone lymphoma. Blood 101 (7): 2464-72, 2003.  [PUBMED Abstract]

  86. Iannitto E, Ambrosetti A, Ammatuna E, et al.: Splenic marginal zone lymphoma with or without villous lymphocytes. Hematologic findings and outcomes in a series of 57 patients. Cancer 101 (9): 2050-7, 2004.  [PUBMED Abstract]

  87. Arcaini L, Paulli M, Boveri E, et al.: Splenic and nodal marginal zone lymphomas are indolent disorders at high hepatitis C virus seroprevalence with distinct presenting features but similar morphologic and phenotypic profiles. Cancer 100 (1): 107-15, 2004.  [PUBMED Abstract]

  88. Parry-Jones N, Matutes E, Gruszka-Westwood AM, et al.: Prognostic features of splenic lymphoma with villous lymphocytes: a report on 129 patients. Br J Haematol 120 (5): 759-64, 2003.  [PUBMED Abstract]

  89. Arcaini L, Lazzarino M, Colombo N, et al.: Splenic marginal zone lymphoma: a prognostic model for clinical use. Blood 107 (12): 4643-9, 2006.  [PUBMED Abstract]

  90. Hermine O, Lefrère F, Bronowicki JP, et al.: Regression of splenic lymphoma with villous lymphocytes after treatment of hepatitis C virus infection. N Engl J Med 347 (2): 89-94, 2002.  [PUBMED Abstract]

  91. Kelaidi C, Rollot F, Park S, et al.: Response to antiviral treatment in hepatitis C virus-associated marginal zone lymphomas. Leukemia 18 (10): 1711-6, 2004.  [PUBMED Abstract]

  92. de Bruin PC, Beljaards RC, van Heerde P, et al.: Differences in clinical behaviour and immunophenotype between primary cutaneous and primary nodal anaplastic large cell lymphoma of T-cell or null cell phenotype. Histopathology 23 (2): 127-35, 1993.  [PUBMED Abstract]

  93. Willemze R, Beljaards RC: Spectrum of primary cutaneous CD30 (Ki-1)-positive lymphoproliferative disorders. A proposal for classification and guidelines for management and treatment. J Am Acad Dermatol 28 (6): 973-80, 1993.  [PUBMED Abstract]

  94. Miller TP, Dahlberg S, Cassady JR, et al.: Chemotherapy alone compared with chemotherapy plus radiotherapy for localized intermediate- and high-grade non-Hodgkin's lymphoma. N Engl J Med 339 (1): 21-6, 1998.  [PUBMED Abstract]

  95. Coiffier B, Lepage E, Briere J, et al.: CHOP chemotherapy plus rituximab compared with CHOP alone in elderly patients with diffuse large-B-cell lymphoma. N Engl J Med 346 (4): 235-42, 2002.  [PUBMED Abstract]

  96. Coiffier B: State-of-the-art therapeutics: diffuse large B-cell lymphoma. J Clin Oncol 23 (26): 6387-93, 2005.  [PUBMED Abstract]

  97. Habermann TM, Weller EA, Morrison VA, et al.: Rituximab-CHOP versus CHOP alone or with maintenance rituximab in older patients with diffuse large B-cell lymphoma. J Clin Oncol 24 (19): 3121-7, 2006.  [PUBMED Abstract]

  98. A predictive model for aggressive non-Hodgkin's lymphoma. The International Non-Hodgkin's Lymphoma Prognostic Factors Project. N Engl J Med 329 (14): 987-94, 1993.  [PUBMED Abstract]

  99. Møller MB, Christensen BE, Pedersen NT: Prognosis of localized diffuse large B-cell lymphoma in younger patients. Cancer 98 (3): 516-21, 2003.  [PUBMED Abstract]

  100. Canellos GP: CHOP may have been part of the beginning but certainly not the end: issues in risk-related therapy of large-cell lymphoma. J Clin Oncol 15 (5): 1713-6, 1997.  [PUBMED Abstract]

  101. Lossos IS, Czerwinski DK, Alizadeh AA, et al.: Prediction of survival in diffuse large-B-cell lymphoma based on the expression of six genes. N Engl J Med 350 (18): 1828-37, 2004.  [PUBMED Abstract]

  102. Rosenwald A, Wright G, Chan WC, et al.: The use of molecular profiling to predict survival after chemotherapy for diffuse large-B-cell lymphoma. N Engl J Med 346 (25): 1937-47, 2002.  [PUBMED Abstract]

  103. Abramson JS, Shipp MA: Advances in the biology and therapy of diffuse large B-cell lymphoma: moving toward a molecularly targeted approach. Blood 106 (4): 1164-74, 2005.  [PUBMED Abstract]

  104. de Jong D, Rosenwald A, Chhanabhai M, et al.: Immunohistochemical prognostic markers in diffuse large B-cell lymphoma: validation of tissue microarray as a prerequisite for broad clinical applications--a study from the Lunenburg Lymphoma Biomarker Consortium. J Clin Oncol 25 (7): 805-12, 2007.  [PUBMED Abstract]

  105. Glantz MJ, Cole BF, Recht L, et al.: High-dose intravenous methotrexate for patients with nonleukemic leptomeningeal cancer: is intrathecal chemotherapy necessary? J Clin Oncol 16 (4): 1561-7, 1998.  [PUBMED Abstract]

  106. Fisher RI, Gaynor ER, Dahlberg S, et al.: Comparison of a standard regimen (CHOP) with three intensive chemotherapy regimens for advanced non-Hodgkin's lymphoma. N Engl J Med 328 (14): 1002-6, 1993.  [PUBMED Abstract]

  107. van Besien K, Ha CS, Murphy S, et al.: Risk factors, treatment, and outcome of central nervous system recurrence in adults with intermediate-grade and immunoblastic lymphoma. Blood 91 (4): 1178-84, 1998.  [PUBMED Abstract]

  108. Delabie J, Vandenberghe E, Kennes C, et al.: Histiocyte-rich B-cell lymphoma. A distinct clinicopathologic entity possibly related to lymphocyte predominant Hodgkin's disease, paragranuloma subtype. Am J Surg Pathol 16 (1): 37-48, 1992.  [PUBMED Abstract]

  109. Achten R, Verhoef G, Vanuytsel L, et al.: T-cell/histiocyte-rich large B-cell lymphoma: a distinct clinicopathologic entity. J Clin Oncol 20 (5): 1269-77, 2002.  [PUBMED Abstract]

  110. Bouabdallah R, Mounier N, Guettier C, et al.: T-cell/histiocyte-rich large B-cell lymphomas and classical diffuse large B-cell lymphomas have similar outcome after chemotherapy: a matched-control analysis. J Clin Oncol 21 (7): 1271-7, 2003.  [PUBMED Abstract]

  111. Ghesquières H, Berger F, Felman P, et al.: Clinicopathologic characteristics and outcome of diffuse large B-cell lymphomas presenting with an associated low-grade component at diagnosis. J Clin Oncol 24 (33): 5234-41, 2006.  [PUBMED Abstract]

  112. Lazzarino M, Orlandi E, Paulli M, et al.: Primary mediastinal B-cell lymphoma with sclerosis: an aggressive tumor with distinctive clinical and pathologic features. J Clin Oncol 11 (12): 2306-13, 1993.  [PUBMED Abstract]

  113. Kirn D, Mauch P, Shaffer K, et al.: Large-cell and immunoblastic lymphoma of the mediastinum: prognostic features and treatment outcome in 57 patients. J Clin Oncol 11 (7): 1336-43, 1993.  [PUBMED Abstract]

  114. Aisenberg AC: Primary large-cell lymphoma of the mediastinum. J Clin Oncol 11 (12): 2291-4, 1993.  [PUBMED Abstract]

  115. Abou-Elella AA, Weisenburger DD, Vose JM, et al.: Primary mediastinal large B-cell lymphoma: a clinicopathologic study of 43 patients from the Nebraska Lymphoma Study Group. J Clin Oncol 17 (3): 784-90, 1999.  [PUBMED Abstract]

  116. Cazals-Hatem D, Lepage E, Brice P, et al.: Primary mediastinal large B-cell lymphoma. A clinicopathologic study of 141 cases compared with 916 nonmediastinal large B-cell lymphomas, a GELA ("Groupe d'Etude des Lymphomes de l'Adulte") study. Am J Surg Pathol 20 (7): 877-88, 1996.  [PUBMED Abstract]

  117. Popat U, Przepiork D, Champlin 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 16 (1): 63-9, 1998.  [PUBMED Abstract]

  118. van Besien K, Kelta M, Bahaguna P: Primary mediastinal B-cell lymphoma: a review of pathology and management. J Clin Oncol 19 (6): 1855-64, 2001.  [PUBMED Abstract]

  119. Longo DL: What's the deal with follicular lymphomas? J Clin Oncol 11 (2): 202-8, 1993.  [PUBMED Abstract]

  120. Anderson JR, Vose JM, Bierman PJ, et al.: Clinical features and prognosis of follicular large-cell lymphoma: a report from the Nebraska Lymphoma Study Group. J Clin Oncol 11 (2): 218-24, 1993.  [PUBMED Abstract]

  121. Bartlett NL, Rizeq M, Dorfman RF, et al.: Follicular large-cell lymphoma: intermediate or low grade? J Clin Oncol 12 (7): 1349-57, 1994.  [PUBMED Abstract]

  122. Wendum D, Sebban C, Gaulard P, et al.: Follicular large-cell lymphoma treated with intensive chemotherapy: an analysis of 89 cases included in the LNH87 trial and comparison with the outcome of diffuse large B-cell lymphoma. Groupe d'Etude des Lymphomes de l'Adulte. J Clin Oncol 15 (4): 1654-63, 1997.  [PUBMED Abstract]

  123. Hans CP, Weisenburger DD, Vose JM, et al.: A significant diffuse component predicts for inferior survival in grade 3 follicular lymphoma, but cytologic subtypes do not predict survival. Blood 101 (6): 2363-7, 2003.  [PUBMED Abstract]

  124. Vose JM, Bierman PJ, Lynch JC, et al.: Effect of follicularity on autologous transplantation for large-cell non-Hodgkin's lymphoma. J Clin Oncol 16 (3): 844-9, 1998.  [PUBMED Abstract]

  125. Bai RY, Ouyang T, Miething C, et al.: Nucleophosmin-anaplastic lymphoma kinase associated with anaplastic large-cell lymphoma activates the phosphatidylinositol 3-kinase/Akt antiapoptotic signaling pathway. Blood 96 (13): 4319-27, 2000.  [PUBMED Abstract]

  126. Gascoyne RD, Aoun P, Wu D, et al.: Prognostic significance of anaplastic lymphoma kinase (ALK) protein expression in adults with anaplastic large cell lymphoma. Blood 93 (11): 3913-21, 1999.  [PUBMED Abstract]

  127. Seidemann K, Tiemann M, Schrappe M, et al.: Short-pulse B-non-Hodgkin lymphoma-type chemotherapy is efficacious treatment for pediatric anaplastic large cell lymphoma: a report of the Berlin-Frankfurt-Münster Group Trial NHL-BFM 90. Blood 97 (12): 3699-706, 2001.  [PUBMED Abstract]

  128. Lipford EH Jr, Margolick JB, Longo DL, et al.: Angiocentric immunoproliferative lesions: a clinicopathologic spectrum of post-thymic T-cell proliferations. Blood 72 (5): 1674-81, 1988.  [PUBMED Abstract]

  129. Logsdon MD, Ha CS, Kavadi VS, et al.: Lymphoma of the nasal cavity and paranasal sinuses: improved outcome and altered prognostic factors with combined modality therapy. Cancer 80 (3): 477-88, 1997.  [PUBMED Abstract]

  130. Liang R, Todd D, Chan TK, et al.: Treatment outcome and prognostic factors for primary nasal lymphoma. J Clin Oncol 13 (3): 666-70, 1995.  [PUBMED Abstract]

  131. Cheung MM, Chan JK, Lau WH, et al.: Primary non-Hodgkin's lymphoma of the nose and nasopharynx: clinical features, tumor immunophenotype, and treatment outcome in 113 patients. J Clin Oncol 16 (1): 70-7, 1998.  [PUBMED Abstract]

  132. Hausdorff J, Davis E, Long G, et al.: Non-Hodgkin's lymphoma of the paranasal sinuses: clinical and pathological features, and response to combined-modality therapy. Cancer J Sci Am 3 (5): 303-11, 1997 Sep-Oct.  [PUBMED Abstract]

  133. Liang R: Diagnosis and management of primary nasal lymphoma of T-cell or NK-cell origin. Clin Lymphoma 1 (1): 33-7; discussion 38, 2000.  [PUBMED Abstract]

  134. Rizvi MA, Evens AM, Tallman MS, et al.: T-cell non-Hodgkin lymphoma. Blood 107 (4): 1255-64, 2006.  [PUBMED Abstract]

  135. Kim GE, Cho JH, Yang WI, et al.: Angiocentric lymphoma of the head and neck: patterns of systemic failure after radiation treatment. J Clin Oncol 18 (1): 54-63, 2000.  [PUBMED Abstract]

  136. Li YX, Yao B, Jin J, et al.: Radiotherapy as primary treatment for stage IE and IIE nasal natural killer/T-cell lymphoma. J Clin Oncol 24 (1): 181-9, 2006.  [PUBMED Abstract]

  137. Lee J, Suh C, Park YH, et al.: Extranodal natural killer T-cell lymphoma, nasal-type: a prognostic model from a retrospective multicenter study. J Clin Oncol 24 (4): 612-8, 2006.  [PUBMED Abstract]

  138. Li CC, Tien HF, Tang JL, et al.: Treatment outcome and pattern of failure in 77 patients with sinonasal natural killer/T-cell or T-cell lymphoma. Cancer 100 (2): 366-75, 2004.  [PUBMED Abstract]

  139. Mraz-Gernhard S, Natkunam Y, Hoppe RT, et al.: Natural killer/natural killer-like T-cell lymphoma, CD56+, presenting in the skin: an increasingly recognized entity with an aggressive course. J Clin Oncol 19 (8): 2179-88, 2001.  [PUBMED Abstract]

  140. Guinee D Jr, Jaffe E, Kingma D, et al.: Pulmonary lymphomatoid granulomatosis. Evidence for a proliferation of Epstein-Barr virus infected B-lymphocytes with a prominent T-cell component and vasculitis. Am J Surg Pathol 18 (8): 753-64, 1994.  [PUBMED Abstract]

  141. Myers JL, Kurtin PJ, Katzenstein AL, et al.: Lymphomatoid granulomatosis. Evidence of immunophenotypic diversity and relationship to Epstein-Barr virus infection. Am J Surg Pathol 19 (11): 1300-12, 1995.  [PUBMED Abstract]

  142. Siegert W, Agthe A, Griesser H, et al.: Treatment of angioimmunoblastic lymphadenopathy (AILD)-type T-cell lymphoma using prednisone with or without the COPBLAM/IMVP-16 regimen. A multicenter study. Kiel Lymphoma Study Group. Ann Intern Med 117 (5): 364-70, 1992.  [PUBMED Abstract]

  143. Jaffe ES: Angioimmunoblastic T-cell lymphoma: new insights, but the clinical challenge remains. Ann Oncol 6 (7): 631-2, 1995.  [PUBMED Abstract]

  144. Siegert W, Nerl C, Agthe A, et al.: Angioimmunoblastic lymphadenopathy (AILD)-type T-cell lymphoma: prognostic impact of clinical observations and laboratory findings at presentation. The Kiel Lymphoma Study Group. Ann Oncol 6 (7): 659-64, 1995.  [PUBMED Abstract]

  145. Reimer P, Schertlin T, Rüdiger T, et al.: Myeloablative radiochemotherapy followed by autologous peripheral blood stem cell transplantation as first-line therapy in peripheral T-cell lymphomas: first results of a prospective multicenter study. Hematol J 5 (4): 304-11, 2004.  [PUBMED Abstract]

  146. Bräuninger A, Spieker T, Willenbrock K, et al.: Survival and clonal expansion of mutating "forbidden" (immunoglobulin receptor-deficient) epstein-barr virus-infected b cells in angioimmunoblastic t cell lymphoma. J Exp Med 194 (7): 927-40, 2001.  [PUBMED Abstract]

  147. Rüdiger T, Weisenburger DD, Anderson JR, et al.: Peripheral T-cell lymphoma (excluding anaplastic large-cell lymphoma): results from the Non-Hodgkin's Lymphoma Classification Project. Ann Oncol 13 (1): 140-9, 2002.  [PUBMED Abstract]

  148. López-Guillermo A, Cid J, Salar A, et al.: Peripheral T-cell lymphomas: initial features, natural history, and prognostic factors in a series of 174 patients diagnosed according to the R.E.A.L. Classification. Ann Oncol 9 (8): 849-55, 1998.  [PUBMED Abstract]

  149. Gisselbrecht C, Gaulard P, Lepage E, et al.: Prognostic significance of T-cell phenotype in aggressive non-Hodgkin's lymphomas. Groupe d'Etudes des Lymphomes de l'Adulte (GELA). Blood 92 (1): 76-82, 1998.  [PUBMED Abstract]

  150. Sonnen R, Schmidt WP, Müller-Hermelink HK, et al.: The International Prognostic Index determines the outcome of patients with nodal mature T-cell lymphomas. Br J Haematol 129 (3): 366-72, 2005.  [PUBMED Abstract]

  151. Rodriguez J, Munsell M, Yazji S, et al.: Impact of high-dose chemotherapy on peripheral T-cell lymphomas. J Clin Oncol 19 (17): 3766-70, 2001.  [PUBMED Abstract]

  152. Enblad G, Hagberg H, Erlanson M, et al.: A pilot study of alemtuzumab (anti-CD52 monoclonal antibody) therapy for patients with relapsed or chemotherapy-refractory peripheral T-cell lymphomas. Blood 103 (8): 2920-4, 2004.  [PUBMED Abstract]

  153. Talpur R, Apisarnthanarax N, Ward S, et al.: Treatment of refractory peripheral T-cell lymphoma with denileukin diftitox (ONTAK). Leuk Lymphoma 43 (1): 121-6, 2002.  [PUBMED Abstract]

  154. Farcet JP, Gaulard P, Marolleau JP, et al.: Hepatosplenic T-cell lymphoma: sinusal/sinusoidal localization of malignant cells expressing the T-cell receptor gamma delta. Blood 75 (11): 2213-9, 1990.  [PUBMED Abstract]

  155. Wong KF, Chan JK, Matutes E, et al.: Hepatosplenic gamma delta T-cell lymphoma. A distinctive aggressive lymphoma type. Am J Surg Pathol 19 (6): 718-26, 1995.  [PUBMED Abstract]

  156. François A, Lesesve JF, Stamatoullas A, et al.: Hepatosplenic gamma/delta T-cell lymphoma: a report of two cases in immunocompromised patients, associated with isochromosome 7q. Am J Surg Pathol 21 (7): 781-90, 1997.  [PUBMED Abstract]

  157. Belhadj K, Reyes F, Farcet JP, et al.: Hepatosplenic gammadelta T-cell lymphoma is a rare clinicopathologic entity with poor outcome: report on a series of 21 patients. Blood 102 (13): 4261-9, 2003.  [PUBMED Abstract]

  158. Chanan-Khan A, Islam T, Alam A, et al.: Long-term survival with allogeneic stem cell transplant and donor lymphocyte infusion following salvage therapy with anti-CD52 monoclonal antibody (Campath) in a patient with alpha/beta hepatosplenic T-cell non-Hodgkin's lymphoma. Leuk Lymphoma 45 (8): 1673-5, 2004.  [PUBMED Abstract]

  159. Go RS, Wester SM: Immunophenotypic and molecular features, clinical outcomes, treatments, and prognostic factors associated with subcutaneous panniculitis-like T-cell lymphoma: a systematic analysis of 156 patients reported in the literature. Cancer 101 (6): 1404-13, 2004.  [PUBMED Abstract]

  160. Marzano AV, Berti E, Paulli M, et al.: Cytophagic histiocytic panniculitis and subcutaneous panniculitis-like T-cell lymphoma: report of 7 cases. Arch Dermatol 136 (7): 889-96, 2000.  [PUBMED Abstract]

  161. Hoque SR, Child FJ, Whittaker SJ, et al.: Subcutaneous panniculitis-like T-cell lymphoma: a clinicopathological, immunophenotypic and molecular analysis of six patients. Br J Dermatol 148 (3): 516-25, 2003.  [PUBMED Abstract]

  162. Salhany KE, Macon WR, Choi JK, et al.: Subcutaneous panniculitis-like T-cell lymphoma: clinicopathologic, immunophenotypic, and genotypic analysis of alpha/beta and gamma/delta subtypes. Am J Surg Pathol 22 (7): 881-93, 1998.  [PUBMED Abstract]

  163. Massone C, Chott A, Metze D, et al.: Subcutaneous, blastic natural killer (NK), NK/T-cell, and other cytotoxic lymphomas of the skin: a morphologic, immunophenotypic, and molecular study of 50 patients. Am J Surg Pathol 28 (6): 719-35, 2004.  [PUBMED Abstract]

  164. Arnulf B, Copie-Bergman C, Delfau-Larue MH, et al.: Nonhepatosplenic gammadelta T-cell lymphoma: a subset of cytotoxic lymphomas with mucosal or skin localization. Blood 91 (5): 1723-31, 1998.  [PUBMED Abstract]

  165. Toro JR, Liewehr DJ, Pabby N, et al.: Gamma-delta T-cell phenotype is associated with significantly decreased survival in cutaneous T-cell lymphoma. Blood 101 (9): 3407-12, 2003.  [PUBMED Abstract]

  166. Egan LJ, Walsh SV, Stevens FM, et al.: Celiac-associated lymphoma. A single institution experience of 30 cases in the combination chemotherapy era. J Clin Gastroenterol 21 (2): 123-9, 1995.  [PUBMED Abstract]

  167. Gale J, Simmonds PD, Mead GM, et al.: Enteropathy-type intestinal T-cell lymphoma: clinical features and treatment of 31 patients in a single center. J Clin Oncol 18 (4): 795-803, 2000.  [PUBMED Abstract]

  168. Daum S, Ullrich R, Heise W, et al.: Intestinal non-Hodgkin's lymphoma: a multicenter prospective clinical study from the German Study Group on Intestinal non-Hodgkin's Lymphoma. J Clin Oncol 21 (14): 2740-6, 2003.  [PUBMED Abstract]

  169. Murase T, Yamaguchi M, Suzuki R, et al.: Intravascular large B-cell lymphoma (IVLBCL): a clinicopathologic study of 96 cases with special reference to the immunophenotypic heterogeneity of CD5. Blood 109 (2): 478-85, 2007.  [PUBMED Abstract]

  170. Ponzoni M, Ferreri AJ, Campo E, et al.: Definition, diagnosis, and management of intravascular large B-cell lymphoma: proposals and perspectives from an international consensus meeting. J Clin Oncol 25 (21): 3168-73, 2007.  [PUBMED Abstract]

  171. Blum KA, Lozanski G, Byrd JC: Adult Burkitt leukemia and lymphoma. Blood 104 (10): 3009-20, 2004.  [PUBMED Abstract]

  172. Onciu M, Schlette E, Zhou Y, et al.: Secondary chromosomal abnormalities predict outcome in pediatric and adult high-stage Burkitt lymphoma. Cancer 107 (5): 1084-92, 2006.  [PUBMED Abstract]

  173. Macpherson N, Lesack D, Klasa R, et al.: Small noncleaved, non-Burkitt's (Burkit-Like) lymphoma: cytogenetics predict outcome and reflect clinical presentation. J Clin Oncol 17 (5): 1558-67, 1999.  [PUBMED Abstract]

  174. Dave SS, Fu K, Wright GW, et al.: Molecular diagnosis of Burkitt's lymphoma. N Engl J Med 354 (23): 2431-42, 2006.  [PUBMED Abstract]

  175. Hummel M, Bentink S, Berger H, et al.: A biologic definition of Burkitt's lymphoma from transcriptional and genomic profiling. N Engl J Med 354 (23): 2419-30, 2006.  [PUBMED Abstract]

  176. Longo DL, Duffey PL, Jaffe ES, et al.: Diffuse small noncleaved-cell, non-Burkitt's lymphoma in adults: a high-grade lymphoma responsive to ProMACE-based combination chemotherapy. J Clin Oncol 12 (10): 2153-9, 1994.  [PUBMED Abstract]

  177. McMaster ML, Greer JP, Greco FA, et al.: Effective treatment of small-noncleaved-cell lymphoma with high-intensity, brief-duration chemotherapy. J Clin Oncol 9 (6): 941-6, 1991.  [PUBMED Abstract]

  178. Thomas DA, Faderl S, O'Brien S, et al.: Chemoimmunotherapy with hyper-CVAD plus rituximab for the treatment of adult Burkitt and Burkitt-type lymphoma or acute lymphoblastic leukemia. Cancer 106 (7): 1569-80, 2006.  [PUBMED Abstract]

  179. Soussain C, Patte C, Ostronoff M, et al.: Small noncleaved cell lymphoma and leukemia in adults. A retrospective study of 65 adults treated with the LMB pediatric protocols. Blood 85 (3): 664-74, 1995.  [PUBMED Abstract]

  180. Magrath I, Adde M, Shad A, et al.: Adults and children with small non-cleaved-cell lymphoma have a similar excellent outcome when treated with the same chemotherapy regimen. J Clin Oncol 14 (3): 925-34, 1996.  [PUBMED Abstract]

  181. Adde M, Shad A, Venzon D, et al.: Additional chemotherapy agents improve treatment outcome for children and adults with advanced B-cell lymphomas. Semin Oncol 25 (2 Suppl 4): 33-9; discussion 45-8, 1998.  [PUBMED Abstract]

  182. Hoelzer D, Ludwig WD, Thiel E, et al.: Improved outcome in adult B-cell acute lymphoblastic leukemia. Blood 87 (2): 495-508, 1996.  [PUBMED Abstract]

  183. Lee EJ, Petroni GR, Schiffer CA, et al.: Brief-duration high-intensity chemotherapy for patients with small noncleaved-cell lymphoma or FAB L3 acute lymphocytic leukemia: results of cancer and leukemia group B study 9251. J Clin Oncol 19 (20): 4014-22, 2001.  [PUBMED Abstract]

  184. Mead GM, Sydes MR, Walewski J, et al.: An international evaluation of CODOX-M and CODOX-M alternating with IVAC in adult Burkitt's lymphoma: results of United Kingdom Lymphoma Group LY06 study. Ann Oncol 13 (8): 1264-74, 2002.  [PUBMED Abstract]

  185. Freedman AS, Takvorian T, Anderson KC, et al.: Autologous bone marrow transplantation in B-cell non-Hodgkin's lymphoma: very low treatment-related mortality in 100 patients in sensitive relapse. J Clin Oncol 8 (5): 784-91, 1990.  [PUBMED Abstract]

  186. Sweetenham JW, Pearce R, Philip T, et al.: High-dose therapy and autologous bone marrow transplantation for intermediate and high grade non-Hodgkin's lymphoma in patients aged 55 years and over: results from the European Group for Bone Marrow Transplantation. The EBMT Lymphoma Working Party. Bone Marrow Transplant 14 (6): 981-7, 1994.  [PUBMED Abstract]

  187. Rizzieri DA, Johnson JL, Niedzwiecki D, et al.: Intensive chemotherapy with and without cranial radiation for Burkitt leukemia and lymphoma: final results of Cancer and Leukemia Group B Study 9251. Cancer 100 (7): 1438-48, 2004.  [PUBMED Abstract]

  188. Morel P, Lepage E, Brice P, et al.: Prognosis and treatment of lymphoblastic lymphoma in adults: a report on 80 patients. J Clin Oncol 10 (7): 1078-85, 1992.  [PUBMED Abstract]

  189. Verdonck LF, Dekker AW, de Gast GC, et al.: Autologous bone marrow transplantation for adult poor-risk lymphoblastic lymphoma in first remission. J Clin Oncol 10 (4): 644-6, 1992.  [PUBMED Abstract]

  190. Thomas DA, O'Brien S, Cortes J, et al.: Outcome with the hyper-CVAD regimens in lymphoblastic lymphoma. Blood 104 (6): 1624-30, 2004.  [PUBMED Abstract]

  191. Sweetenham JW, Santini G, Qian W, et al.: High-dose therapy and autologous stem-cell transplantation versus conventional-dose consolidation/maintenance therapy as postremission therapy for adult patients with lymphoblastic lymphoma: results of a randomized trial of the European Group for Blood and Marrow Transplantation and the United Kingdom Lymphoma Group. J Clin Oncol 19 (11): 2927-36, 2001.  [PUBMED Abstract]

  192. Höllsberg P, Hafler DA: Seminars in medicine of the Beth Israel Hospital, Boston. Pathogenesis of diseases induced by human lymphotropic virus type I infection. N Engl J Med 328 (16): 1173-82, 1993.  [PUBMED Abstract]

  193. Foss FM, Aquino SL, Ferry JA: Case records of the Massachusetts General Hospital. Weekly clinicopathological exercises. Case 10-2003. A 72-year-old man with rapidly progressive leukemia, rash, and multiorgan failure. N Engl J Med 348 (13): 1267-75, 2003.  [PUBMED Abstract]

  194. Tsukasaki K, Utsunomiya A, Fukuda H, et al.: VCAP-AMP-VECP compared with biweekly CHOP for adult T-cell leukemia-lymphoma: Japan Clinical Oncology Group Study JCOG9801. J Clin Oncol 25 (34): 5458-64, 2007.  [PUBMED Abstract]

  195. Gill PS, Harrington W Jr, Kaplan MH, et al.: Treatment of adult T-cell leukemia-lymphoma with a combination of interferon alfa and zidovudine. N Engl J Med 332 (26): 1744-8, 1995.  [PUBMED Abstract]

  196. Matutes E, Taylor GP, Cavenagh J, et al.: Interferon alpha and zidovudine therapy in adult T-cell leukaemia lymphoma: response and outcome in 15 patients. Br J Haematol 113 (3): 779-84, 2001.  [PUBMED Abstract]

  197. Hermine O, Allard I, Lévy V, et al.: A prospective phase II clinical trial with the use of zidovudine and interferon-alpha in the acute and lymphoma forms of adult T-cell leukemia/lymphoma. Hematol J 3 (6): 276-82, 2002.  [PUBMED Abstract]

  198. Norton AJ, Matthews J, Pappa V, et al.: Mantle cell lymphoma: natural history defined in a serially biopsied population over a 20-year period. Ann Oncol 6 (3): 249-56, 1995.  [PUBMED Abstract]

  199. Zucca E, Roggero E, Pinotti G, et al.: Patterns of survival in mantle cell lymphoma. Ann Oncol 6 (3): 257-62, 1995.  [PUBMED Abstract]

  200. Campo E, Raffeld M, Jaffe ES: Mantle-cell lymphoma. Semin Hematol 36 (2): 115-27, 1999.  [PUBMED Abstract]

  201. Weisenburger DD, Armitage JO: Mantle cell lymphoma-- an entity comes of age. Blood 87 (11): 4483-94, 1996.  [PUBMED Abstract]

  202. Hiddemann W, Unterhalt M, Herrmann R, et al.: Mantle-cell lymphomas have more widespread disease and a slower response to chemotherapy compared with follicle-center lymphomas: results of a prospective comparative analysis of the German Low-Grade Lymphoma Study Group. J Clin Oncol 16 (5): 1922-30, 1998.  [PUBMED Abstract]

  203. Majlis A, Pugh WC, Rodriguez MA, et al.: Mantle cell lymphoma: correlation of clinical outcome and biologic features with three histologic variants. J Clin Oncol 15 (4): 1664-71, 1997.  [PUBMED Abstract]

  204. Tiemann M, Schrader C, Klapper W, et al.: Histopathology, cell proliferation indices and clinical outcome in 304 patients with mantle cell lymphoma (MCL): a clinicopathological study from the European MCL Network. Br J Haematol 131 (1): 29-38, 2005.  [PUBMED Abstract]

  205. Velders GA, Kluin-Nelemans JC, De Boer CJ, et al.: Mantle-cell lymphoma: a population-based clinical study. J Clin Oncol 14 (4): 1269-74, 1996.  [PUBMED Abstract]

  206. Teodorovic I, Pittaluga S, Kluin-Nelemans JC, et al.: Efficacy of four different regimens in 64 mantle-cell lymphoma cases: clinicopathologic comparison with 498 other non-Hodgkin's lymphoma subtypes. European Organization for the Research and Treatment of Cancer Lymphoma Cooperative Group. J Clin Oncol 13 (11): 2819-26, 1995.  [PUBMED Abstract]

  207. Vandenberghe E, Ruiz de Elvira C, Loberiza FR, et al.: Outcome of autologous transplantation for mantle cell lymphoma: a study by the European Blood and Bone Marrow Transplant and Autologous Blood and Marrow Transplant Registries. Br J Haematol 120 (5): 793-800, 2003.  [PUBMED Abstract]

  208. Lenz G, Dreyling M, Hoster E, et al.: Immunochemotherapy with rituximab and cyclophosphamide, doxorubicin, vincristine, and prednisone significantly improves response and time to treatment failure, but not long-term outcome in patients with previously untreated mantle cell lymphoma: results of a prospective randomized trial of the German Low Grade Lymphoma Study Group (GLSG). J Clin Oncol 23 (9): 1984-92, 2005.  [PUBMED Abstract]

  209. Romaguera JE, Fayad L, Rodriguez MA, et al.: High rate of durable remissions after treatment of newly diagnosed aggressive mantle-cell lymphoma with rituximab plus hyper-CVAD alternating with rituximab plus high-dose methotrexate and cytarabine. J Clin Oncol 23 (28): 7013-23, 2005.  [PUBMED Abstract]

  210. Witzig TE: Current treatment approaches for mantle-cell lymphoma. J Clin Oncol 23 (26): 6409-14, 2005.  [PUBMED Abstract]

  211. Khouri IF, Lee MS, Saliba RM, et al.: Nonablative allogeneic stem-cell transplantation for advanced/recurrent mantle-cell lymphoma. J Clin Oncol 21 (23): 4407-12, 2003.  [PUBMED Abstract]

  212. Khouri IF, Romaguera J, Kantarjian H, et al.: Hyper-CVAD and high-dose methotrexate/cytarabine followed by stem-cell transplantation: an active regimen for aggressive mantle-cell lymphoma. J Clin Oncol 16 (12): 3803-9, 1998.  [PUBMED Abstract]

  213. Howard OM, Gribben JG, Neuberg DS, et al.: Rituximab and CHOP induction therapy for newly diagnosed mantle-cell lymphoma: molecular complete responses are not predictive of progression-free survival. J Clin Oncol 20 (5): 1288-94, 2002.  [PUBMED Abstract]

  214. Lefrère F, Delmer A, Suzan F, et al.: Sequential chemotherapy by CHOP and DHAP regimens followed by high-dose therapy with stem cell transplantation induces a high rate of complete response and improves event-free survival in mantle cell lymphoma: a prospective study. Leukemia 16 (4): 587-93, 2002.  [PUBMED Abstract]

  215. Gopal AK, Rajendran JG, Petersdorf SH, et al.: High-dose chemo-radioimmunotherapy with autologous stem cell support for relapsed mantle cell lymphoma. Blood 99 (9): 3158-62, 2002.  [PUBMED Abstract]

  216. Gianni AM, Magni M, Martelli M, et al.: Long-term remission in mantle cell lymphoma following high-dose sequential chemotherapy and in vivo rituximab-purged stem cell autografting (R-HDS regimen). Blood 102 (2): 749-55, 2003.  [PUBMED Abstract]

  217. Forstpointner R, Dreyling M, Repp R, et al.: The addition of rituximab to a combination of fludarabine, cyclophosphamide, mitoxantrone (FCM) significantly increases the response rate and prolongs survival as compared with FCM alone in patients with relapsed and refractory follicular and mantle cell lymphomas: results of a prospective randomized study of the German Low-Grade Lymphoma Study Group. Blood 104 (10): 3064-71, 2004.  [PUBMED Abstract]

  218. Dreyling M, Lenz G, Hoster E, et al.: Early consolidation by myeloablative radiochemotherapy followed by autologous stem cell transplantation in first remission significantly prolongs progression-free survival in mantle-cell lymphoma: results of a prospective randomized trial of the European MCL Network. Blood 105 (7): 2677-84, 2005.  [PUBMED Abstract]

  219. Goy A, Younes A, McLaughlin P, et al.: Phase II study of proteasome inhibitor bortezomib in relapsed or refractory B-cell non-Hodgkin's lymphoma. J Clin Oncol 23 (4): 667-75, 2005.  [PUBMED Abstract]

  220. O'Connor OA, Wright J, Moskowitz C, et al.: Phase II clinical experience with the novel proteasome inhibitor bortezomib in patients with indolent non-Hodgkin's lymphoma and mantle cell lymphoma. J Clin Oncol 23 (4): 676-84, 2005.  [PUBMED Abstract]

  221. Fisher RI, Bernstein SH, Kahl BS, et al.: Multicenter phase II study of bortezomib in patients with relapsed or refractory mantle cell lymphoma. J Clin Oncol 24 (30): 4867-74, 2006.  [PUBMED Abstract]

  222. Morrison VA, Dunn DL, Manivel JC, et al.: Clinical characteristics of post-transplant lymphoproliferative disorders. Am J Med 97 (1): 14-24, 1994.  [PUBMED Abstract]

  223. Knowles DM, Cesarman E, Chadburn A, et al.: Correlative morphologic and molecular genetic analysis demonstrates three distinct categories of posttransplantation lymphoproliferative disorders. Blood 85 (2): 552-65, 1995.  [PUBMED Abstract]

  224. Leblond V, Dhedin N, Mamzer Bruneel MF, et al.: Identification of prognostic factors in 61 patients with posttransplantation lymphoproliferative disorders. J Clin Oncol 19 (3): 772-8, 2001.  [PUBMED Abstract]

  225. Ghobrial IM, Habermann TM, Maurer MJ, et al.: Prognostic analysis for survival in adult solid organ transplant recipients with post-transplantation lymphoproliferative disorders. J Clin Oncol 23 (30): 7574-82, 2005.  [PUBMED Abstract]

  226. Armitage JM, Kormos RL, Stuart RS, et al.: Posttransplant lymphoproliferative disease in thoracic organ transplant patients: ten years of cyclosporine-based immunosuppression. J Heart Lung Transplant 10 (6): 877-86; discussion 886-7, 1991 Nov-Dec.  [PUBMED Abstract]

  227. Kuehnle I, Huls MH, Liu Z, et al.: CD20 monoclonal antibody (rituximab) for therapy of Epstein-Barr virus lymphoma after hemopoietic stem-cell transplantation. Blood 95 (4): 1502-5, 2000.  [PUBMED Abstract]

  228. Shapiro RS, Chauvenet A, McGuire W, et al.: Treatment of B-cell lymphoproliferative disorders with interferon alfa and intravenous gamma globulin. N Engl J Med 318 (20): 1334, 1988.  [PUBMED Abstract]

  229. Leblond V, Sutton L, Dorent R, et al.: Lymphoproliferative disorders after organ transplantation: a report of 24 cases observed in a single center. J Clin Oncol 13 (4): 961-8, 1995.  [PUBMED Abstract]

  230. Mamzer-Bruneel MF, Lomé C, Morelon E, et al.: Durable remission after aggressive chemotherapy for very late post-kidney transplant lymphoproliferation: A report of 16 cases observed in a single center. J Clin Oncol 18 (21): 3622-32, 2000.  [PUBMED Abstract]

  231. Swinnen LJ: Durable remission after aggressive chemotherapy for post-cardiac transplant lymphoproliferation. Leuk Lymphoma 28 (1-2): 89-101, 1997.  [PUBMED Abstract]

  232. McCarthy M, Ramage J, McNair A, et al.: The clinical diversity and role of chemotherapy in lymphoproliferative disorder in liver transplant recipients. J Hepatol 27 (6): 1015-21, 1997.  [PUBMED Abstract]

  233. Leblond V, Davi F, Charlotte F, et al.: Posttransplant lymphoproliferative disorders not associated with Epstein-Barr virus: a distinct entity? J Clin Oncol 16 (6): 2052-9, 1998.  [PUBMED Abstract]

  234. Senderowicz AM, Vitetta E, Headlee D, et al.: Complete sustained response of a refractory, post-transplantation, large B-cell lymphoma to an anti-CD22 immunotoxin. Ann Intern Med 126 (11): 882-5, 1997.  [PUBMED Abstract]

  235. Haddad E, Paczesny S, Leblond V, et al.: Treatment of B-lymphoproliferative disorder with a monoclonal anti-interleukin-6 antibody in 12 patients: a multicenter phase 1-2 clinical trial. Blood 97 (6): 1590-7, 2001.  [PUBMED Abstract]

  236. Soslow RA, Davis RE, Warnke RA, et al.: True histiocytic lymphoma following therapy for lymphoblastic neoplasms. Blood 87 (12): 5207-12, 1996.  [PUBMED Abstract]

  237. Kamel OW, Gocke CD, Kell DL, et al.: True histiocytic lymphoma: a study of 12 cases based on current definition. Leuk Lymphoma 18 (1-2): 81-6, 1995.  [PUBMED Abstract]

  238. Nador RG, Cesarman E, Chadburn A, et al.: Primary effusion lymphoma: a distinct clinicopathologic entity associated with the Kaposi's sarcoma-associated herpes virus. Blood 88 (2): 645-56, 1996.  [PUBMED Abstract]



Glossary Terms

Level of evidence 1iiDii
Randomized, controlled, nonblinded clinical trial with disease-free survival as an endpoint. See Levels of Evidence for Adult and Pediatric Cancer Treatment Studies (PDQ®) for more information.
Level of evidence 3iiiA
Nonconsecutive case series with total mortality as an endpoint. See Levels of Evidence for Adult and Pediatric Cancer Treatment Studies (PDQ®) for more information.
Level of evidence 3iiiDiii
Nonconsecutive case series with progression-free survival as an endpoint. See Levels of Evidence for Adult and Pediatric Cancer Treatment Studies (PDQ®) for more information.
Level of evidence 3iiiDiv
Nonconsecutive case series with tumor response rate as an endpoint. See Levels of Evidence for Adult and Pediatric Cancer Treatment Studies (PDQ®) for more information.


Table of Links

1http://www.cancer.gov/cancertopics/pdq/levels-evidence-adult-treatment/HealthPr
ofessional
2http://www.cancer.gov/cancertopics/pdq/treatment/adult-non-hodgkins/HealthProfe
ssional/Table1
3http://www.cancer.gov/cancertopics/pdq/treatment/adultALL/HealthProfessional
4http://www.cancer.gov/cancertopics/pdq/treatment/adulthodgkins/HealthProfession
al
5http://www.cancer.gov/cancertopics/pdq/treatment/AIDS-related-lymphoma/HealthPr
ofessional
6http://www.cancer.gov/cancertopics/pdq/treatment/CLL/healthprofessional
7http://www.cancer.gov/cancertopics/pdq/treatment/hairy-cell-leukemia/HealthProf
essional
8http://www.cancer.gov/cancertopics/pdq/treatment/myeloma/HealthProfessional
9http://www.cancer.gov/cancertopics/pdq/treatment/mycosisfungoides/HealthProfess
ional
10http://www.cancer.gov/cancertopics/pdq/treatment/primary-CNS-lymphoma/HealthPro
fessional
11http://www.cancer.gov/cancertopics/pdq/treatment/adult-non-hodgkins/HealthProfe
ssional/174.cdr#Section_174
12http://www.cancer.gov/cancertopics/pdq/supportivecare/fever/healthprofessional/
allpages
13http://www.cancer.gov/cancertopics/pdq/supportivecare/nutrition/healthprofessio
nal/allpages
14http://www.cancer.gov/cancertopics/pdq/supportivecare/cardiopulmonary/HealthPro
fessional
15http://www.cancer.gov/search/viewclinicaltrials.aspx?version= heal
thprofessional &cdrid=77645
16http://www.cancer.gov/cancertopics/pdq/supportivecare/pruritus/healthprofession
al/allpages
17http://www.cancer.gov/cancertopics/pdq/supportivecare/gastrointestinalcomplicat
ions/HealthProfessional/183.cdr#Section_183
18http://www.cancer.gov/search/viewclinicaltrials.aspx?version= heal
thprofessional &cdrid=77643
19http://www.cancer.gov/cancertopics/pdq/supportivecare/hypercalcemia/healthprofe
ssional/allpages