NCCN Clinical Practice Guidelines in Oncology for Non-Hodgkin's Lymphomas
NCCN Categories of Evidence and Consensus
Category 1: The recommendation is based on high-level evidence (e.g., randomized controlled trials) and there is uniform NCCN consensus.
Category 2A: The recommendation is based on lower-level evidence and there is uniform NCCN consensus.
Category 2B: The recommendation is based on lower-level evidence and there is nonuniform NCCN consensus (but no major disagreement).
Category 3: The recommendation is based on any level of evidence but reflects major disagreement.
All recommendations are category 2A unless otherwise noted.
The full NCCN Clinical Practice Guidelines in Oncology for Non-Hodgkin's Lymphomas are not printed in this issue of JNCCN, but can be accessed online at www.NCCN.org.
Clinical trials: NCCN believes that the best management for any cancer patient is in a clinical trial. Participation in clinical trials is especially encouraged.
Overview
Non-Hodgkin's lymphomas (NHLs) are a heterogeneous group of lymphoproliferative disorders originating in B-, T-, or natural killer (NK) lymphocytes. In the United States, B-cell lymphomas represent 80% to 85% of the cases, with 15% to 20% being T-cell lymphomas; NK lymphomas are rare. In 2010, an estimated 65,540 new cases of NHL were diagnosed and 20,210 patients died of the disease.1 NHL is the sixth leading site of new cancer diagnoses among men and women, accounting for 4% of new cancer cases and 4% of cancer-related deaths.1
The incidence of NHL increased dramatically between 1970 and 1995, but has moderated since the mid-90s. This increase is attributed partly to the HIV epidemic and the development of AIDS-related NHL, but much of it has been observed in patients in their sixth and seventh decades. A large part of this increased incidence has paralleled a major decrease in mortality from other causes. The median age of individuals with NHL has risen in the past 2 decades.2 As a result, patients with NHL may also have significant comorbid conditions, which complicate treatment options.
NOTE: This manuscript highlights only a portion of the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines) for Non-Hodgkin's Lymphomas. Please refer to www.NCCN.org for the complete guidelines.
Classification
In 1956, Rappaport et al.3 proposed a lymphoma classification based on the pattern of cell growth (nodular or diffuse), and size and shape of the tumor cells.4 This classification, though widely used in the Unites States, quickly became outdated with the discovery and existence of distinct types of lymphocytes (B, T, and NK). The Kiel classification, which divided the lymphomas into low- and high-grade based on histologic features, became the first and most significant classification to apply this new information to lymphomas.5–7 This classification was widely used in Europe. However, the use of different classification systems in clinical studies made results difficult to compare. Hence, the International Working Formulation (IWF) for NHLs was developed to standardize the classification of lymphomas.

Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046

Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046
Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046

Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046

Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046
Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046

Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046

Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046
Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046

Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046

Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046
Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046

Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046

Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046
Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046

Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046

Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046
Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046

Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046

Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046
Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046

Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046

Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046
Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046

Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046

Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046
Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046

Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046

Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046
Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046

Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046

Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046
Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046

Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046

Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046
Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046

Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046

Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046
Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046

Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046

Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046
Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046

Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046

Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046
Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046

Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046

Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046
Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046

Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046

Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046
Version 2.2011, 03-01-11 ©2011 National Comprehensive Cancer Network, Inc. All rights reserved. These guidelines and this illustration may not be reproduced in any form without the express written permission of NCCN.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 9, 5; 10.6004/jnccn.2011.0046
IWF Classification
The IWF classified NHL as either low-, intermediate-, or high-grade based on the morphology and natural history.8 This classification divided diffuse large B-cell lymphoma (DLBCL) into intermediate- and high-grade groups. However, these distinctions were not reproducible. Because this classification did not include immunophenotyping, the categories were not reproducible.9 In addition, after this classification was published, many new diseases were described that were not included in the IWF classification.
Revised European-American Lymphoma Classification
In 1994, the International Lymphoma Study Group developed the Revised European-American Lymphoma (REAL) classification, which classified lymphomas based on the cell of origin (B, T, or NK) and included morphology, immunophenotype, genetic, and clinical features to define diseases.10 In 1997, the International Lymphoma Classification Project performed a clinical evaluation of the REAL classification in a cohort of 1403 cases of NHL,11,12 confirming the diagnosis of NHL in 1378 (98.2%). This study identified the 13 most common histologic types, constituting approximately 90% of the cases of NHL in the United States. The findings were as follows: DLBCL, 31%; follicular lymphoma (FL), 22%; small lymphocytic lymphoma/chronic lymphocytic leukemia (SLL/CLL), 6%; mantle cell lymphoma (MCL), 6%; peripheral T-cell lymphoma (PTCL), 6%; and mucosa-associated lymphoid tissue (MALT) lymphoma, 5%. The remaining subtypes each occurred in fewer than 2% of cases. Importantly, in the United States more than 50% of lymphoma cases are either DLBCL or FL. The study investigators concluded that the REAL classification can be readily applied and identifies clinically distinctive types of NHL.
WHO Classification
In 2001, the WHO updated the classification of hematopoietic and lymphoid neoplasms13,14 to apply the principles of the REAL classification, representing the first international consensus on classification of hematologic malignancies. The REAL/WHO classification of NHL includes many entities not recognized by the IWF.13,14 After consideration of cell of origin (B, T, or NK), the classification subdivides lymphomas into those from precursor lymphocytes versus those derived from mature lymphocytes. The classification is further refined based on immunophenotype and genetic and clinical features. These considerations have helped define active treatment for specific subtypes of lymphoma.
In 2008, the International T-Cell Lymphoma Project evaluated the WHO classification of T-cell lymphoma in a cohort of 1314 cases of PTCL and NK/T-cell lymphomas (NKTCL). The diagnosis of PTCL or NKTCL was confirmed in 1153 cases (88%). The most common subtypes were PTCL-not otherwise specified (PTCL-NOS; 25.9%); angioimmunoblastic lymphoma (18.5%); NKTCL (10.4%); adult T-cell leukemia/lymphoma (9.6%); anaplastic large cell lymphoma (ALCL); anaplastic lymphoma kinase (ALK)-positive (6.6%); and ALCL, ALK-negative (5.5%).15 The findings of this study validated the efficacy of the WHO classification for defining subtypes of T-cell lymphomas. The WHO classification was updated again in September 2008 to add new diseases and subtypes that have been recognized in the past decade, and to better define some of the heterogeneous and ambiguous categories based on recent advances16,17 (available online, in these guidelines, at www.NCCN.org [ST-1]).
Response Criteria
In 1999, the IWG published guidelines for response criteria for lymphoma based on the reduction in size of the enlarged lymph node as measured by CT scan and the extent of bone marrow involvement determined through bone marrow aspirate and biopsy.18 These guidelines were revised in 2007 by the International Harmonization Project to incorporate immunohistochemistry, flow cytometry, and 18-flourodeoxyglucose (FDG)–PET scans in the definition of response.19 In the revised guidelines, the complete response, unconfirmed category was essentially eliminated because residual masses were defined as a partial or complete response based on the result of a PET scan. Using the revised system, response is categorized as complete response, partial response, stable disease, and relapsed disease or progressive disease. However, the application of PET to responses is limited to histologies that show reliable FDG uptake in active tumor. However, the revised response criteria have thus far only been validated for DLBCL and Hodgkin lymphoma. The application of the revised response criteria to other histologies requires validation, and the original IWG guidelines should be used.
Diagnosis
Genetic features, detected using cytogenetics or fluorescence in situ hybridization (FISH), are increasingly important in defining specific NHL subtypes. In addition, detection of viruses, particularly Epstein-Barr virus (EBV), human herpesvirus 8, and human T-cell leukemia virus type 1, is often necessary to establish a specific diagnosis. In all cases, the most important first step is to make an accurate pathologic diagnosis. The basic pathologic evaluation is the same in each guideline, although some further evaluation may be useful in certain circumstances to clarify a particular diagnosis; these are outlined in the pathologic evaluation of the individual guideline.
An incisional or excisional lymph node biopsy is recommended to establish the diagnosis of NHL. Core needle biopsy is discouraged unless the clinical situation dictates that this is the only safe means of obtaining diagnostic tissue. Fine needle aspiration (FNA) biopsy is widely used in the diagnosis of malignant neoplasms, but its role in diagnosing lymphoma is still controversial.20,21 Because the revised REAL/WHO classification is based on both morphology and immunophenotyping, FNA alone is not acceptable as a reliable diagnostic tool for NHL. However, its use in combination with ancillary techniques may provide precise diagnosis, thereby obviating the need for a more invasive biopsy in highly selected circumstances. Recent studies have shown that the diagnostic accuracy of FNA improves significantly when it is used in combination with immunohistochemistry and flow cytometry.22–24
In the NCCN Guidelines, FNA results alone are not suitable for making an initial diagnosis of NHL, although it may be sufficient to establish relapse. However, in certain circumstances, when a lymph node is not easily accessible, a combination of core biopsy and FNA in conjunction with appropriate ancillary techniques (polymerase chain reaction [PCR] for IGHV and/or T-cell receptor [TCR] gene rearrangements; FISH for major translocations; immunophenotypic analysis) may be sufficient for diagnosis. This is particularly true for the diagnosis of CLL. In other entities presenting in leukemic phase, such as FL or MCL, a biopsy is still preferred to clarify histologic subtype.
Immunophenotypic analysis is essential for differentiating the various subtypes of NHL to establish the proper diagnosis. It can be performed using flow cytometry and/or immunohistochemistry; the choice depends on the antigens and the expertise and resources available to the hematopathologist. In some cases, flow cytometry and immunohistochemistry are complementary diagnostic tools.25 Cytogenetic or molecular genetic analysis may be necessary under certain circumstances to identify the specific chromosomal translocations that are characteristic of some NHL subtypes or to establish clonality.
After publication of the 2008 WHO classification, the panel developed a series of algorithms for using immunophenotyping to diagnose mature lymphoid neoplasms. These algorithms should be used in conjunction with clinical and pathologic correlation. They were developed as a guide for surgical pathologists and to help clinicians interpret pathology reports.
Initial assessment begins with morphologic, clinical, and immunophenotypic analysis. Morphologic assessment involves determining the cell size (small, medium-sized, or large cells), with or without anaplastic morphology. Clinical features include patient's age and the location (nodal, extranodal, and, among extranodal site, skin vs. other specific sites). The initial immunophenotyping panel should include Pan-B and Pan-T-cell antigens. Based on the morphologic and clinical features, some of the B- and T-cell subset antigens may also be added in the initial panel (see Immunophenotyping and Genetic Testing in the Differential Diagnosis of Mature B-cell and T/NK-cell Neoplasms, available online, in these guidelines, at www.NCCN.org [NHODG-A]).
Workup
Essential workup procedures include a complete physical examination, with particular attention to node-bearing areas; determination of the size of liver and spleen, symptoms present, and performance status; and laboratory studies, including CBC, serum lactate dehydrogenase (LDH), hepatitis B testing (see later discussion), comprehensive metabolic panel, and CT chest/abdominal/pelvic with oral and intravenous contrast (unless coexistent renal insufficiency). Multiple-gated acquisition (MUGA) scan or echocardiograms are recommended when anthracyclines and anthracenedione-containing regimens are used. Bone marrow biopsy with or without aspirate is essential whenever treatment is considered; however, it may be deferred in certain circumstances (see later discussion).
Because of the risk of hepatitis B reactivation, the panel included hepatitis B testing (hepatitis B surface antigen [HBsAg] and hepatitis B core antibody [HBcAb]) as part of essential workup in all patients before initiating anti-CD20 monoclonal antibody-based regimens. Furthermore, hepatitis B reactivation has been reported with chemotherapy alone, and therefore testing should be considered in anyone with a risk factor (e.g., blood transfusion, intravenous drug abuse) or from a region with a nonnegligible prevalence of hepatitis B infection. Further discussion is provided in Hepatitis B Virus Reactivation on the facing page. Hepatitis C testing is needed in high-risk patients and patients with splenic marginal zone lymphoma (MZL).
Optional procedures (depending on specific lymphoma type) include β2-microglobulin, CT or PET-CT scans, endoscopic ultrasound (gastric MALT lymphoma), head CT or brain MRI, and lumbar puncture to analyze cerebrospinal fluid (MCL and DLBCL). Discussion of fertility issues and sperm banking should be addressed when appropriate.26
Bone marrow biopsy is usually included in the workup for all patients with NHL, except those with SLL/CLL in whom a clonal lymphocytosis is identified with flow cytometry. Bone marrow involvement occurs in 39% of low-grade, 36% of intermediate-grade, and 18% of high-grade lymphomas. Bone marrow involvement was associated with significantly shorter survivals in patients with intermediate- or high-grade lymphomas.27 A recent retrospective analysis analyzing the incidence of involvement and parameters predicting bone marrow involvement in 192 patients with stage I and II DLBCL found the overall incidence of bone marrow involvement to be 3.6%. The authors concluded that bone marrow biopsy may be safely omitted in selected patients with early-stage DLBCL.28 In cutaneous B-cell lymphomas, bone marrow biopsy is essential for primary cutaneous DLBCL, leg-type (PCDLBCL-LT) because it is an aggressive lymphoma that will probably require systemic treatment, whereas the role of bone marrow biopsy in primary cutaneous follicle center lymphoma (PCFCL) and primary cutaneous MZL is less clear. Recent studies indicate that bone marrow biopsy is an essential component of staging in patients with PCFCL first presenting in the skin, whereas it seems to have limited value in patients with MZL presenting in the skin, and may be considered only in selected cases.29,30
In these guidelines, bone marrow biopsy with or without aspirate is included as part of the essential workup for all lymphomas. However, in patients with low-bulk indolent disease with radiographic clinical stage III disease, an initial staging bone marrow evaluation can be deferred if observation is recommended because it will not change the clinical recommendations. However, in the evaluation of potentially early-stage indolent lymphoma (stage I or II), bone marrow biopsy is essential; some panel members advocate bilateral core biopsies in this situation.31 Bilateral cores are recommended if radioimmunotherapy is considered.
FDG-PET scan has been used for initial staging, restaging, and follow-up of patients with NHL.32 In a recent meta-analysis, PET showed a high positivity and specificity when used to stage and restage patients with lymphoma.33 FDG-PET is nearly universally positive at diagnosis in Hodgkin lymphoma, DLBCL, and FL34; approximately 90% in T-cell lymphoma35 and nodal MZL; but less sensitive for extranodal MZL.36 However, several benign conditions, including sarcoid, infection, and inflammation, can result in false-positive PET scans complicating the interpretation. Lesions smaller than 1 cm are not reliably visualized with PET scans. PET scan is now part of the pretreatment evaluation in Hodgkin lymphoma and DLBCL, and may be useful in selected cases with other histologies. The pretreatment PET scan is particularly important in helping to interpret posttreatment response evaluation according to new response criteria (see previous discussion). At diagnosis, PET scans may detect additional disease sites, although the clinical stage is only modified in 15% to 20% of patients and the additional information provided by the scans results in a change in treatment in only 8% of patients. PET scan has generally been used in conjunction with diagnostic CT scans.
Integrated PET-CT has largely replaced dedicated CT scans in the United States. This diagnostic study has distinct advantages in both staging and restaging compared with full-dose diagnostic CT or PET alone.37,38 In a retrospective study, PET-CT performed with low-dose nonenhanced CT was found to be more sensitive and specific than routine contrast-enhanced CT in evaluating lymph node and organ involvement in patients with Hodgkin disease or high-grade NHL.38 Preliminary results of another recent prospective study (47 patients; patients who had undergone prior diagnostic CT were excluded) showed a good correlation between low-dose unenhanced PET-CT and full-dose enhanced PET-CT in the evaluation of lymph nodes and extranodal disease in lymphomas.37 However, the lack of intravenous contrast and the diminished resolution can make the anatomic localization and significance of FDG-avid sites difficult to interpret in some cases. Further studies are needed to determine if PET-CT scans can replace diagnostic CT scans in the initial staging and response evaluation of lymphomas. The panel has included PET-CT scan as an optional workup procedure for selected patients.
Supportive Care
Hepatitis B Virus Reactivation
Hepatitis B virus (HBV) reactivation has been reported to occur in patients treated with chemotherapy with or without anti-CD20 monoclonal antibody. Treatment with rituximab alone is also a risk for hepatitis B reactivation.39 HBV-reactivation may result in a fulminant hepatitis, hepatic failure, and death. The median time to hepatitis diagnosis was approximately 4 months after the initiation, according to the package insert (www.fda.gov).
Testing of patients at risk for hepatitis B reactivation should include HBsAg and HBcAb. In a prospective study of all patients undergoing immunosuppressive therapy (chemotherapy, antibody therapy, high-dose dexamethasone) at Memorial Sloan-Kettering Cancer Center, 1% of patients were HBsAg-positive and 9% were HBcAb-positive.40 A retrospective study conducted by MD Anderson Cancer Center reported similar findings (HBsAg and HBcAb were positive in 2% and 8% of patients, respectively).41 Patients positive for HBsAg are at a greater risk for HBV reactivation than those positive for HBcAb.39 In a prospective study of 100 Chinese patients undergoing chemotherapy for lymphoma, hepatitis developed in 67% of HBsAg-positive patients and 14% HBsAg-negative patients during cytotoxic therapy.42 Other risk factors for reactivation include young age, male gender, elevated pretreatment viral load, and prolonged immunosuppression.43,44 The use of rituximab in HBcAb-positive patients has been reported to cause fatal HBV-related liver disease. A retrospective study of Italian HBcAb-positive patients with lymphoma found that 2.7% of patients treated with rituximab and chemotherapy developed HBV-related liver disease compared with 0.8% of patients treated with chemotherapy alone. HBV-related liver disease was not seen in patients who were observed or underwent other therapy (radiation, antibiotics, interferon).45
Antiviral prophylaxis has been effective in preventing hepatitis B reactivation during chemoimmunotherapy in HBsAg-positive patients.46–48 The results of a systematic review of 14 studies involving HBsAg-positive patients undergoing chemotherapy showed that lamivudine prophylaxis reduced the risk for HBV reactivation by 79% or greater; HBV-associated hepatic failure and death may also be reduced.46 None of the patients in the preventive lamivudine group developed HBV-related hepatic failure, compared with 21 of 162 patients in the control group, and only 4 deaths were attributable to HBV in the preventive lamivudine group compared with 27 deaths in the control group. Lamivudine was well tolerated with no adverse effects. In a small randomized study, Lau et al.49 showed that preemptive antiviral treatment with lamivudine was superior to deferred treatment. This study randomized 30 patients with HBsSAg-positive lymphoma to receive lamivudine either before chemotherapy or for the treatment of increased viral load based on HBV DNA PCR levels. HBV reactivation was observed in 53% of monitored patients and none in the prophylaxis group. Interestingly, clinical cancer-related outcomes were also significantly better in the prophylaxis group than the treatment group.
The guidelines recommend HBsAg and HBcAb testing for all patients receiving rituximab. For patients in whom one or both of these tests are positive, a baseline hepatitis B viral load should be determined using quantitative PCR. However, a negative baseline PCR does not preclude the possibility of activation. In patients from areas with high prevalence (Asia, Africa, Eastern Europe, and portions of South America) or regions where the prevalence of HBV is unknown, all of those undergoing immunotherapy, chemotherapy, or chemoimmunotherapy should be tested for HBsAg and HBcAb. Patients receiving intravenous immunoglobulin (IVIG) may be HBcAb-positive as a consequence of IVIG therapy. Empiric antiviral therapy with oncologic treatment is recommended for any patient who is either HBsAg- or HBcAb-positive. During the treatment period, viral load should be monitored monthly with PCR and 3 months thereafter. Patients treated with chemotherapy alone should receive prophylaxis if they have a measurable viral load independent of the viral serology. If viral load is consistently undetectable, prophylaxis should be given to HBsAg-positive patients and may be considered in patients HBcAb-positive. If viral load fails to drop, consultation with a hepatologist is recommended. However, because of the potential emergence of resistance to lamivudine, it is not the optimal drug for prophylaxis. Several appropriate agents for viral prophylaxis are available; good choice will be driven by institutional standard or recommendation from the consultant. The optimal duration of prophylaxis remains undefined, but the panel recommends it be maintained for at least 6 months after the completion of oncologic treatment.
Progressive Multifocal Leucoencephalopathy
Progressive multifocal leucoencephalopathy (PML) is a serious and usually fatal central nervous system infection caused by JC polyoma virus. In a recent report of 57 cases from the Research on Adverse Drug Events and Reports project, 52 patients with lymphoproliferative disorders developed PML after treatment with rituximab and other treatments, including hematopoietic stem cell transplantation (HSCT) or chemotherapy with purine analogs or alkylating agents.50 Median time from last rituximab dose to PML diagnosis was 5.5 months, and median time to death after diagnosis was 2.0 months. The case-fatality rate was 90%.
PML is usually diagnosed based on PCR of cerebrospinal fluid, or sometimes brain biopsy. No effective treatment exists for PML. Patients must be carefully monitored for the development of any neurologic symptoms. Currently, no pretreatment evaluation can predict for the subsequent development of PML.
Tumor Lysis Syndrome
Tumor lysis syndrome (TLS) is characterized by metabolic abnormalities caused by the abrupt release of intracellular contents into the blood from cellular disintegration induced by chemotherapy. It is usually observed within 12 to 72 hours after chemotherapy initiation.51 Untreated TLS can induce profound metabolic changes, resulting in cardiac arrhythmias, seizures, loss of muscle control, acute renal failure, and death.
Cairo and Bishop52 recently classified TLS into laboratory or clinical types. Laboratory TLS is defined as a 25% increase in the levels of serum uric acid, potassium, or phosphorus or a 25% decrease in calcium levels. Clinical TLS refers to laboratory TLS with clinical toxicity that requires intervention. Clinical complications may include renal insufficiency, cardiac arrhythmia, or seizures. The 4 primary electrolyte abnormalities of TLS are hyperkalemia, hyperuricemia, hyperphosphatemia, and hypocalcemia. Symptoms associated with TLS may include nausea and vomiting, diarrhea, seizures, shortness of breath, or cardiac arrhythmias.
TLS is best managed if it is anticipated and treatment is initiated before chemotherapy. The cornerstone of management is hydration and the control of hyperuricemia. Allopurinol should be administered before the initiation of chemotherapy. When the uric acid level remains elevated despite treatment with allopurinol, or when renal insufficiency is present, treatment with rasburicase is indicated. Electrolytes and renal function should be monitored every 6 to 8 hours with appropriate interventions for hyperphosphatemia and hyperkalemia. Careful clinical monitoring will help to preempt complications, and in many cases admission to the intensive care unit is appropriate. Cardiac monitoring or serial electrocardiogram may be beneficial to identify early electrolyte-related cardiac abnormalities. Dialysis may be necessary in cases of anuric acute renal failure.
Allopurinol is a xanthine analog and a competitive inhibitor of xanthine oxidase, thereby blocking conversion of purine metabolites to uric acid. Allopurinol will decrease uric acid production and has been shown to reduce the incidence of uric acid uropathy.53 Because the drug inhibits new uric acid formation rather than reduces existing uric acid, elevated levels of uric acid can take several days to normalize after initiation of treatment, thereby delaying the start of chemotherapy. Furthermore, allopurinol may lead to the accumulation of xanthine crystals in renal tubules, leading to acute obstructive uropathy. Allopurinol will also reduce clearance of 6-mercaptopurine and high-dose methotrexate.
Rasburicase is a recombinant urate oxidase that catalyzes the oxidation of uric acid to a highly soluble nontoxic metabolite that is readily excreted. It has been shown to be safe and highly effective in the prevention and treatment of chemotherapy-induced hyperuricemia in both children and adults.54 The GRAAL1 (Groupe d'Etude des Lymphomes de l'Adulte Trial on Rasburicase Activity in Adult Lymphoma) study evaluated the efficacy and safety of rasburicase for the prevention and treatment of hyperuricemia in patients with NHL during induction chemotherapy.55 Uric acid levels decreased within 4 hours after the first injection of the drug. Creatinine levels and other metabolites were also controlled with the administration of rasburicase.
Cortes et al.56 recently reported the results of a prospective, randomized controlled trial that compared the efficacy of rasburicase and allopurinol in adult patients with hematologic malignancies at high or potential risk for TLS. The plasma uric acid response rate was 87% for rasburicase, 78% for rasburicase plus allopurinol, and 66% for allopurinol. Rasburicase was superior to allopurinol in the overall study population, in patients at high risk for TLS (89% vs. 68%), and in patients with baseline hyperuricemia (90% vs. 53%). The time to control serum uric acid in hyperuricemic patients was 4 hours for rasburicase and 27 hours for allopurinol. However, rasburicase can induce anaphylactic reactions. Other adverse reactions include methemoglobinemia and severe hemolysis in patients with glucose-6-phosphate dehydrogenase (G6PD) deficiency.
The risk factors for TLS include bone marrow involvement, bulky tumors that are chemosensitive, rapidly proliferative or aggressive hematologic malignancies, an elevated leukocyte count or pretreatment LDH level, preexisting elevated uric acid, renal disease, or renal involvement of tumor. Patients diagnosed with lymphoblastic lymphoma or Burkitt lymphoma are at a higher risk for developing TLS. Occasionally, patients with bulky presentation of DLBCL and those with CLL and high white blood cell count may experience TLS at a moderately high frequency.
These guidelines recommend that allopurinol be started 2 to 3 days before chemotherapy and continued for 10 to 14 days. Rasburicase is recommended for patients with any of the following risk factors: presence of any high-risk feature; bulky disease requiring immediate therapy; inability to achieve adequate hydration; allopurinol is ineffective; or acute renal failure. One dose is adequate in most cases; repeat dosing should be individualized.
CLL/SLL
CLL and SLL are different manifestations of the same disease and are managed in much the same way.57 The major difference is that in CLL a significant number of the abnormal lymphocytes are also found in the bone marrow and blood, whereas in SLL the abnormal lymphocytes are predominantly found in the lymph nodes.
Cytogenetic abnormalities that can be detected with FISH are present in approximately 80% of patients with CLL. The most common abnormality is del(13q) (55%), followed by del(11q) (18%), trisomy 12 (16%), del(17p) (7%), and del(6q) (7%).58 Del(13q) is associated with a favorable prognosis and the longest median survival (133 months). Del(11q) is associated with extensive lymphadenopathy, disease progression, and shorter median survival (79 months).58,59 Among patients with del(11q), those with a complete loss of ATM function might have impaired response to irradiation or cytotoxic drugs, resulting in poor clinical outcome.60 Alkylating agent–based chemoimmunotherapy regimens have significantly improved clinical outcomes by overcoming the adverse prognostic significance of del(11q) in previously untreated patients with CLL.61 Del(17p) affecting TP53 is associated with shorter treatment-free intervals, shorter median survival (32 months), and poor response to chemotherapy.58 Recent studies have identified TP53 mutations as an independent predictor of short survival and resistance to chemotherapy.62 Resistance to chemotherapy has been attributed to the presence of mutation in the remaining TP53 allele.63 However, the natural history of patients with del(17p) can be heterogeneous, with some patients having an indolent disease course.64,65 TP53 mutation also carries a poor prognosis regardless of the presence of del(17p) when treated with fludarabine-based chemotherapy.66,67
The impact of these prognostic factors on the clinical outcome of patients has been examined in large prospective randomized studies. In the CALGB 9712 study,68 unmutated IGHV (≥ 98%), del(11q), and del(17p) were identified as independent prognostic factors for overall and progression-free survival; two other studies (E299769 and LRF CLL470) identified these features as prognostic indicators for progression-free survival. In the LRF CLL4 trial, patients with 5% to 20% of cells with del(17p) had similar response rates and survival as those without del(17p).70 In contrast, those with 20% or more cells with del(17p) had a poor outcome, with a 13% response rate and median overall survival of only 11 months. The finding that del(17p) is more frequently observed in treated patients than in untreated ones (20% vs. 5%–10%) suggests that treatment-driven clonal selection occurs during therapy.
New prognostic markers such as β2-microglobulin, IGHV mutational status, CD38, and zeta-associated protein 70 (ZAP-70) have been identified.71 Higher β2-microglobulin was an independent adverse prognostic factor in patients treated with front-line fludarabine-based chemotherapy.72 Unmutated IGHV is associated with a poor prognosis irrespective of disease stage.73 The extent of mutation is also important, with the longest survivals observed in patients with more than 3% mutations, and slightly shorter survivals seen in those with 1% to 2% mutations.74 A subset of patients in whom the IGHV3-21 variable region is rearranged have more aggressive disease and shorter survival regardless of the mutation status.75
Overexpression of CD38 (≥ 30%)76–78 and ZAP-70 (≥ 20%)79–82 is also associated with a poor prognosis. However, the fact that these cutoffs are based on flow cytometry and reproducibility across laboratories remains a problem. Recent studies have shown that the combined analysis of ZAP-70 and CD38 expression provides a more discriminatory prediction of treatment-free interval than each factor alone.83–86 Wierda et al.87 developed a nomogram using age, β2-microglobulin, absolute lymphocyte count, sex, Rai stage, and number of involved lymph nodes, which may help stratify untreated patients with CLL into 3 different risk groups (low, intermediate, and high).87 The estimated median survival times were not reached for low-risk groups. The median survival times for intermediate- and high-risk groups were 10 and 5 years, respectively. The 5-and 10-year overall survival rates were 97% and 80%, 80% and 52%, and 55% and 26%, respectively. This prognostic model must be validated in prospective studies.
Staging
The nearly universal involvement of the bone marrow and peripheral blood in CLL/SLL limits the utility of the Ann Arbor staging system. Two different staging systems, Rai and Binet, are currently used worldwide.88,89 The modified Rai classification is most useful clinically and provides important prognostic information.88 The survival rate of patients with low-risk disease (Rai stage 0) is essentially the same as that of age-matched controls. Patients with intermediate-risk disease (Rai stage I–II) have a shorter survival, particularly when other adverse factors coexist, such as a lymphocyte doubling time of less than 1 year. Patients with high-risk disease (Rai stage III–IV) have a poor prognosis. The Binet staging system is based on the number of involved areas and the level of hemoglobin and platelets and, like the Rai system, has a good correlation with clinical outcome.89
Diagnosis
The diagnosis of CLL requires the presence of at least 5000 malignant B cells/mm3. The presence of fewer B cells in the absence of lymphadenopathy or other clinical features characteristic of a lymphoproliferative disorder is defined as monoclonal B lymphocytosis (MBL). MBL is a relatively recent diagnostic category consisting of individuals with an abnormal B-cell population but do not meet the diagnostic criteria for CLL.90 Most cases of MBL have the immunophenotype of CLL (discussed later). The “favorable” molecular lesions, mutated IGHV and del(13q), are commonly seen in patients with MBL.91 The estimated rate of progression of MBL to CLL requiring treatment is 1.1% per year. A CT scan is essential to distinguish MBL from SLL. The CLL/SLL guideline now includes an initial stratification between CLL/SLL and MBL (absolute lymphocyte count < 5000 B cells/mm3, lymph nodes < 1.5 cm, no anemia or thrombocytopenia). Observation is recommended for all patients diagnosed with MBL.
Adequate immunophenotyping using flow cytometry of peripheral blood or paraffin-section immunohistochemistry is required to confirm the diagnosis of CLL/SLL. The recommended panel for immunohistochemistry includes CD3, CD5, CD10, CD20, CD23, and cyclin D1. These can be useful, particularly for diagnosing CLL/SLL type without circulating cells. Cell surface markers for flow cytometric studies include kappa/lambda, CD19, CD20, CD5, CD23, and CD10. Additional paraffin-embedded material may be used for immunophenotyping to determine lineage and clonality.
The typical immunophenotype includes CD5+, CD10–, CD19+, CD20, dim expression of surface immunoglobulin, CD23+, CD43+/–, and cyclin D1–. Distinguishing CLL/SLL from MCL is essential because they are both CD5+ B-cell tumors. Although CD23 is often helpful, cyclin D1– is critical in this differentiation of tumor types. FISH for detecting t(11;14) can help distinguish MCL from CLL. Cytogenetics and/or FISH for detecting del(11q), del(13q), trisomy 12, and del(17p), and molecular genetic analysis to detect IGHV mutation status, can provide prognostic information and guide selection of therapy. Although FISH is optional for patients with Rai low-risk disease for whom observation would be recommended, it should be evaluated any time therapy is considered. Cytogenetic abnormalities can evolve over time, and therefore reevaluation of FISH is necessary to direct treatment options in patients with indications for treatment. CD38 and/or ZAP-70 expression can be determined using immunohistochemistry or flow cytometry. Evaluation of ZAP-70 expression using flow cytometry can be challenging and is not recommended outside a clinical trial.
Conventional metaphase cytogenetics is difficult in CLL because of the very low proliferative activity of leukemic cells in vitro. Therefore, interphase cytogenetic analysis with FISH has been the standard method to detect chromosomal abnormalities that have important prognostic significance. However, FISH can only detect abnormalities specific to the probes used. Cytokine or CpG oligonucleotide stimulation has been used to promote efficient metaphase analysis.92 Recent studies have shown that stimulation with CpG oligonucleotide and interleukin-2 is more effective than that with 12-O-tetradecanoylphorbol-13-acetate (TPA) for detecting chromosomal abnormalities in CLL.93,94 A prospective study conducted by the CLL Research Consortium confirmed that abnormal clones in CLL are more readily detected with CpG oligonucleotide stimulation than with traditional B-cell mitogens, and the clonal abnormalities revealed by CpG-stimulated metaphase cytogenetics are consistent with those detected with interphase FISH and are reproducible among different cytogenetic laboratories.95 However, the use of CpG stimulation for CLL cytogenetics is not yet universally available.
Workup
The workup for CLL/SLL is similar to that for other lymphoid neoplasms. Quantitative immunoglobulins may be particularly informative in patients with recurrent infections. Measurement of β2-microglobulin may provide useful prognostic information.72,87 Although classically the pattern of bone marrow involvement (diffuse vs. nodular) had prognostic significance, this is no longer a factor when more reliable prognostic markers are used, such as IGHV mutation (or its surrogate ZAP-70) and cytogenetic abnormalities determined with FISH, all of which can be obtained through analysis of circulating lymphocytes. Thus, bone marrow biopsy is no longer considered a required part of evaluation for patients with CLL, although it remains useful in evaluating the cause of cytopenias.
CT scans are useful for following and monitoring disease progression when peripheral adenopathy is present. For anemic patients, reticulocyte counts and a direct Coombs test should be performed to evaluate for the possibility of hemolysis. PET scan is generally not useful in CLL but can assist in directing nodal biopsy if Richter's transformation is suspected.
Response Criteria
The NCI–sponsored working group first published the guidelines for the diagnosis and treatment of CLL in 1996.96 Recent developments in the use of prognostic markers and treatment options for CLL have led to revision of these guidelines, particularly the response criteria.97 Complete and partial responses are considered clinically beneficial. Relapsed disease progresses after 12 months or more of complete or partial response, whereas refractory disease does not respond to purine analog-based therapy or progresses within 12 months after this therapy is given.
Treatment Options
First-Line Therapy: In earlier clinical trials, chlorambucil plus prednisone had comparable efficacy to CVP (cyclophosphamide, vincristine, and prednisone) and CHOP (cyclophosphamide, doxorubicin, vincristine, and prednisone) regimens in patients with advanced CLL.98 In the CALGB 9011 study, 509 patients were randomized to first-line therapy with either fludarabine, chlorambucil, or the combination.99 The combination arm was stopped because of excessive toxicity. Complete remission (20% vs. 4% for chlorambucil), partial remission (43% vs. 33% for chlorambucil), median duration of remission (25 vs. 14 months for chlorambucil), and median progression-free survival (20 vs. 14 months for chlorambucil) were significantly better in patients treated with fludarabine. The study found no significant differences in overall survival between the fludarabine and chlorambucil arms (66 vs. 56 months for chlorambucil).
In a phase III randomized trial conducted by the German CLL Study Group (GCLLSG), fludarabine was significantly better than chlorambucil in terms of overall response rate (72% vs. 51%, respectively) and time to treatment failure (18 vs. 11 months, respectively) in patients older than 65 years (median age, 70 years), but fludarabine showed no survival benefit over chlorambucil (overall and progression-free survivals were 46 and 19 months, respectively, for fludarabine, and 64 and 19 months, respectively, for chlorambucil).100
A European randomized study compared fludarabine with 2 alkylating agent–based combination regimens, CAP (cyclophosphamide, doxorubicin, and prednisone) and CHOP, as first-line treatment in patients with advanced CLL.101 Fludarabine and CHOP produced similar overall remission rates (71%) to CAP (58%), but fludarabine was better tolerated than CHOP. In large randomized trials, the combination of fludarabine and cyclophosphamide (FC) was associated with an increase in overall response, complete response, and progression-free survival compared with fludarabine alone.70,102,103
CALGB study 9712 compared the efficacy of fludarabine with concurrent or sequential administration of rituximab in untreated patients with CLL.104,105 The concurrent regimen was associated with a higher overall response rate (90% vs. 77% for the sequential regimen) at the expense of higher grade 3 or 4 toxicity. However, comparison of the outcomes of patients treated with fludarabine alone in the CALGB 9011 trial and the pooled results from the CALGB 9712 study suggest that the addition of rituximab to fludarabine prolongs progression-free and overall survival.106
The combination of fludarabine, cyclophosphamide, and rituximab (FCR) evaluated at MD Anderson Cancer Center as initial therapy produced high overall and complete response rates.107–109 At a median follow-up of 6 years, the overall response rate was 95% (72% complete response) for the 300 study patients.108 Recently, a large international randomized phase III clinical trial (CLL8) showed that the addition of rituximab to fludarabine-based chemotherapy improved response rates and progression-free and overall survival in patients with CLL compared with fludarabine-based chemotherapy alone.110 In this trial, 817 patients with previously untreated CD20+ CLL were randomized to 6 courses of either FCR or FC regimen. At 3 years after randomization, the progression-free and overall survival rates were 65% and 87%, respectively, for patients randomized to FCR compared with 45% and 82.5%, respectively, for those who received FC. FCR also induced a higher overall response rate (95% vs. 88%) and more complete responses (44% vs. 22%) than FC. Median progression-free survival was 52 and 33 months for FCR and FC, respectively. Based on the results of this trial, the FDA approved rituximab in combination with FC for patients with previously untreated CD20+ CLL.
In a trial initiated by the CLL Research Consortium, pentostatin, cyclophosphamide, and rituximab showed significant clinical activity despite poor risk–based prognoses in previously untreated patients.111 Responses were observed in 91% of patients (41% complete response, 21% nodular partial response, and 28% partial response). In a subsequent study, the combination of higher-dose pentostatin and rituximab resulted in an overall response rate of 76%, with a complete response rate of 27%.112 However, in historical comparison, the response rates were higher and median treatment-free survival (16 vs. 30 months for pentostatin, cyclophosphamide, and rituximab) was notably longer in all accrued patients treated with pentostatin, cyclophosphamide, and rituximab compared with pentostatin and rituximab.
Bendamustine is an alkylating agent with a low cross-resistance with other alkylating agents (chlorambucil, cyclophosphamide, ifosfamide) and fludarabine. A pivotal phase III randomized study (n = 319) comparing bendamustine with chlorambucil in patients with untreated CLL113,114 showed that the overall response (68% vs. 31%, respectively) and complete response rates (31% vs. 2%, respectively) were significantly higher for bendamustine. Median progression-free survival (22 vs. 8.3 months for chlorambucil) and median duration of remission (22 vs. 8 months with chlorambucil) were also better for bendamustine. However, no differences were seen in overall survival between the groups, and the efficacy of bendamustine compared with first-line therapies other than chlorambucil has not yet been established. In a multicenter phase II trial (CLL2M) from the GCLLSG, bendamustine in combination with rituximab (BR) resulted in an overall response rate of 91% (33% complete response and 55% partial response) for the entire study population (n = 117) with untreated CLL.115 The GCLLSG is currently comparing FCR and BR (CLL10).
Alemtuzumab, a monoclonal antibody targeting CD52, has been effective as a first-line treatment for patients with CLL.116,117 An international multicenter study (CAM307) that randomized 297 patients with CLL to receive either alemtuzumab or chlorambucil as first-line treatment117 showed that alemtuzumab had superior progression-free survival, with a 42% reduction in risk of progression or death. The overall response rate for alemtuzumab was significantly better than for chlorambucil (83% with 24% complete response vs. 55% with 2% complete response). In patients with del(17p), the overall response rate and median progression-free survival for alemtuzumab were 64% and 11 months, respectively, which were higher than those observed with chlorambucil (20% and 2 months, respectively).
Relapsed or Refractory Disease: The FCR regimen also induced higher response rates in previously treated patients (n = 177).118 The overall response rate was 73% (complete remission, nodular partial remission, and partial remissions were achieved in 25%, 16%, and 32% of patients, respectively). Recently, the REACH (Rituximab in the Study of Relapsed Chronic Lymphocytic Leukemia) trial compared 6 cycles of FCR with 6 cycles of FC in 552 patients with previously treated CLL.119 After a median follow-up of 25 months, patients in the FCR group had significantly improved progression-free survival compared with those in the FC group (31 vs. 21 months, respectively). The overall response rate was also significantly higher for the FCR regimen (70%, including 24% complete response and 46% partial response vs. 58%, including 13% complete response and 45% partial response for FC regimen). At median follow-up of 25 months, overall survival was not significantly improved for FCR. Based on the results of this trial, the FDA approved rituximab in combination with FC for patients with previously treated CD20+ CLL.
The combination of pentostatin and cyclophosphamide (PC) with or without rituximab (R) has shown significant activity in previously treated patients with relapsed or refractory disease.120,121 In a small number of previously treated patients (n = 46), the response rates were similar for PC and PCR. However, based on a historical retrospective comparison with PC regimen, PCR has a longer median duration of response (25 vs. 7 months) and median survival (44 vs. 16 months).120
In a phase I/II trial, the combination of oxaliplatin, fludarabine, cytarabine, and rituximab (OFAR) was highly active in fludarabine-refractory patients with CLL and those with Richter's syndrome.122,123 The overall response rates were 50% in patients with Richter's syndrome and 33% in those with fludarabine-refractory CLL.122 Responses were achieved in 7 of 20 patients (35%) with del(17p), 2 of 7 patients (29%) with del(11q), all 4 patients with trisomy 12, and 2 of 5 patients (40%) with del(13q). The median response duration was 10 months. The overall response rate in patients aged 70 years or older (n = 14) was 50%.
The GCLLSG conducted a trial on BR for patients with relapsed CLL, which resulted in an overall response rate of 77% in 62 evaluable patients with relapsed or refractory disease.124 Complete and partial responses were seen in 14.5% and 63% of patients, respectively. Stable disease was achieved in 18% of patients, and 5% had progressive disease.
High-dose methylprednisolone (HDMP) with rituximab is well tolerated and an effective therapy for patients with refractory CLL, including those with unfavorable prognostic features.125–127 In a study of 28 patients with fludarabine-refractory CLL, the overall response rate was 96% (32% complete response).126 In a follow-up of 37 patients with CLL treated with HDMP and rituximab at Mayo Clinic, 29 (78%) had an objective response, including 5 of 9 patients with del(17p),125 and 8 (22%) had a complete clinical response. The 3-year survival rate was 41%. One study showed that although the combination of HDMP and rituximab induced superior overall (93%) and complete (14%) response rates compared with HDMP alone (43% and 0%, respectively) in heavily pretreated patients with advanced disease, it was also associated with a high rate of opportunistic infections.127
Alemtuzumab also induced significant responses in patients for whom fludarabine-based therapy failed.128 Overall objective response in the intent-to-treat population (n = 93) was 33% (2% complete response and 31% partial response). Median time to progression was 4.7 months overall (9.5 months for responders) and median overall survival was 16 months (32 months for responders). Other studies have also shown that alemtuzumab is effective in patients with fludarabine-refractory CLL and del(17p) or p53 gene mutations.129–131 Subcutaneous alemtuzumab seems to be as effective and safe as intravenous alemtuzumab in patients with advanced-stage relapsed or refractory CLL.132–135 In a recent retrospective analysis, favorable overall response rate and progression-free and overall survivals (49%, and 7 and 19 months, respectively) were observed with alemtuzumab in pretreated patients with del(17p).136 However, nodal sites of disease have generally not responded well with single-agent alemtuzumab. Combinations of alemtuzumab and fludarabine137,138; alemtuzumab and rituximab139,140; and cyclophosphamide, fludarabine, alemtuzumab, and rituximab (CFAR)141 have shown promising results in patients with relapsed or refractory disease.
Ofatumumab, a human CD20 monoclonal antibody, was found to be well tolerated in patients with relapsed or refractory CLL in a phase I/II study.142 In October 2009, ofatumumab was approved by the FDA for treatment of patients with CLL refractory to fludarabine and alemtuzumab based on the interim analysis of the pivotal international clinical trial, which included data from 138 patients with fludarabine- and alemtuzumab-refractory (FA-ref) CLL or patients with fludarabine-refractory CLL with bulky lymphadenopathy (BF-ref).143 In the interim analysis, the overall response rate was 58% and 47% in the FA-ref and BF-ref groups, respectively. The final results from the pivotal trial showed the safety and efficacy of ofatumumab in this heavily pretreated patient population. Median progression-free survival was 5.5 months for both groups.144 The median overall survival was 14 and 17 months for the FA-ref and BF-ref groups, respectively.
Allogeneic HSCT has been shown to improve the prognosis in patients with advanced disease and those with poor-risk features.145–151 In a retrospective analysis of the European Group for Blood and Marrow Transplantation (EBMT), allogeneic HSCT induced long-term remission in patients with del(17p).150 At median follow-up of 39 months, 3-year overall and progression-free survival rates were 44% and 37%, respectively. Final results of the prospective multicenter trial (GCLLSG CLL3X study) also showed that nonmyeloablative allogeneic stem cell transplant can induce sustained minimal residual disease–negative event-free survival in a significant proportion of patients with del(17p).151 Patients with and without del(17p) had 3-year event-free survivals rates of 43% and 41%, respectively. Of 13 patients with del(17p), 5 were minimal residual disease–negative at 12 months. This study also identified refractory disease at the time of transplant as a negative prognostic factor for overall and event-free survivals. It is understood that studies involving allogeneic HSCTs are subject to strong selection biases. Nonetheless, available evidence from nonrandomized clinical studies suggests that allogeneic HSCT is an effective treatment option for patients refractory to chemoimmunotherapy or who develop recurrence within 12 months after purine analog treatment.152
NCCN Recommendations
Localized SLL (Ann Arbor Stage I): Locoregional radiation therapy (RT) is an appropriate induction therapy for this group of patients. In rare patients, RT may be contraindicated or may be a suboptimal therapy because of the presence of comorbidities or potential for long-term toxicity. Patients with localized SLL that has progressed after initial RT are treated as described for patients with SLL (Ann Arbor stage II–IV).
SLL (Ann Arbor Stage II–IV) or CLL (Rai Stages 0–IV): In some patients, early-stage disease may have an indolent course, and in others it may progress rapidly to advanced disease requiring immediate treatment. Absolute lymphocyte count alone is not an indication for treatment unless it is greater than 200 to 300 x 109/L or symptoms related to leukostasis are present. Therefore, in patients with SLL (Ann Arbor stage II–IV) or CLL (Rai stages 0–II), treatment options depend on the presence or absence of the following indications: significant disease-related symptoms, including severe fatigue, weight loss, night sweats, and fever without infection; threatened end-organ function; progressive bulky disease (enlarged spleen or lymph nodes); lymphocyte doubling time of 6 months or less; or progression to more advanced-stage CLL with progressive anemia or thrombocytopenia. Patients with no indications for treatment can be observed until disease progression or the appearance of any indications for treatment. Patients with advanced-stage CLL (Rai stage III–IV) require immediate treatment.
Given the incurability of the disease, these guidelines recommend enrollment in clinical trials, when locally available, as the preferred first-line therapy for all patients. For patients presenting with indications for treatment and not eligible or do not have access to clinical trials, the treatment recommendations included in the guidelines are based on the presence or absence of del(17p) or del(11q), age, and performance status of the patient. Reevaluation of FISH is necessary to direct treatment options in patients with indications for treatment.
CLL/SLL Without del(17p) or del(11q): First-Line Therapy: Patients are stratified according to their age and associated comorbid conditions. Comorbidities can be assessed using the cumulative illness rating scale.153
For frail patients with significant comorbidities and unable to tolerate purine analogs, options include monotherapy with chlorambucil (with or without prednisone), rituximab, or pulse corticosteroids.
For patients aged 70 years or older, or those younger with significant comorbidities, these NCCN Guidelines include alkylating agent–based chemotherapy or chemoimmunotherapy; monotherapy with alemtuzumab or rituximab; fludarabine with or without rituximab; or cladribine as options. Based on the results of the CLL8 trial,110 the guidelines include rituximab in combination with purine analog–based chemotherapy or bendamustine as options for patients aged 70 years or younger or older patients without significant comorbidities. See Suggested Treatment Regimens for CLL Without del(11q) or del(17p) on page 493 for a list of specific regimens.
Based on several trials, chemoimmunotherapy has emerged as the standard of care in patients younger than 70 without significant comorbidities.105,110 A randomized comparison of FCR versus PCR showed a higher complete response rate for FCR, but the overall response and overall survival rates were no different between the regimens.154 Both FCR and FR are highly active regimens, but category 1 evidence is not available to help choose between these 2 regimens. In the absence of del(11q), whether these regimens have different long-term outcomes is uncertain.
Second-Line Therapy: In patients for whom first-line therapy fails, treatment options depend on the duration of response after first-line therapy. Among patients in whom initial therapy with FCR chemoimmunotherapy failed, those with a time to treatment failure of 3 years or more had better median survival (44 months) than those with a time to treatment failure of less than 3 years.155 If the response duration is more than 3 years (long response), these NCCN Guidelines recommend additional cycles of the same regimen that was used as first-line therapy for all patients.
If the response duration is less than 2 years (short response), treatment options depend on patient age. For patients 70 years or older, or younger patients with comorbidities, options include reduced-dose FCR or PCR; bendamustine with or without rituximab; HDMP with rituximab; monotherapy with ofatumumab; alemtuzumab with or without rituximab; or dose-dense rituximab. For patients younger than 70 or older patients without significant comorbidities, the guidelines have included chemoimmunotherapy, monotherapy with ofatumumab, or alemtuzumab with or without rituximab as options. See Suggested Treatment Regimens for CLL Without del(11q) or del(17p) on page 493 for a list of specific regimens.
Allogeneic HSCT can be considered for patients with short response but would generally be used after reinduction of remission.
CLL/SLL With del(17p): Del(17p) is associated with low response rates with all treatments. Because no standard treatment exists, a clinical trial is recommended. First-line therapy options include FCR or FR, HDMP plus rituximab, alemtuzumab with or without rituximab, or BR.
Patients who have experienced a complete or partial response to first-line therapy should be treated with allogeneic HSCT if they are eligible. Patients experiencing a complete or partial response after transplant can either be observed or enrolled in a clinical trial. Alternatively, patients with a partial response could also be treated with chemoimmunotherapy. See Suggested Treatment Regimens for CLL With del(17p) on page 494 for a list of specific regimens.
Patients who experience no response to first-line therapy, experience response to first-line therapy but are not eligible for allogenic HSCT, or experience no response to transplant should be enrolled in a clinical trial or can be treated with second-line therapy for relapsed or refractory disease. The guidelines have included chemoimmunotherapy, monotherapy with ofatumumab, alemtuzumab with or without rituximab, HDMP, or bendamustine with or without rituximab as options. See Suggested Treatment Regimens for CLL With del(17p) on page 494 for a list of specific regimens.
CLL/SLL With del(11q): First-line therapy options are based on the age and associated comorbid conditions. If patients have a del(11q), an alkylator must be included in the regimen. Response rates and duration improve when an alkylator is added to a purine analog. In patients older than 70 or with significant comorbidities, single-agent alemtuzumab or rituximab should be used only if an alkylator is considered unsafe or was intolerable. For a list of specific regimens, see Suggested Treatment Regimens for CLL With del(11q), page 495.
Patients who experience complete response to first-line therapy can either be observed until disease progression or enrolled in a clinical trial. For those experiencing disease progression after complete response, treatment options depend on the duration of response to first-line therapy. (See Suggested Treatment Regimens for CLL With del(11q), page 495, for a list of specific regimens.) Patients with a partial response to first-line therapy should be treated with allogeneic HSCT, if they are eligible. After transplant, treatment options are similar to those described for patients with del(17p).
Patients with no response to first-line therapy, or those with a partial response to first-line therapy but not eligible for allogenic HSCT should be enrolled in clinical trials or treated with second-line therapy for relapsed or refractory disease. (See Suggested Treatment Regimens for CLL With del(11q), page 495, for a list of specific regimens.)
Histologic Transformation to DLBCL or Hodgkin Lymphoma: Approximately 2% to 5% of patients with CLL will develop Richter's syndrome (transformation into DLBCL or Hodgkin lymphoma) during the course of treatment.156–158 The incidence of transformation increases with the number of prior regimens. Patients with Richter's syndrome should be treated with a combination of chemoimmunotherapy regimens initially developed for DLBCL.159 In addition to these regimens, the guidelines have also included R-hyperCVAD (cyclophosphamide, vincristine, doxorubicin, and dexamethasone alternating with high-dose methotrexate and cytarabine, followed by rituximab) as an option for patients with histologic transformation and for relapsed or refractory CLL.160,161
Allogeneic HSCT has also shown promising results in patients with Richter's syndrome who experience response to initial therapy. In a nonrandomized comparison, the estimated cumulative 3-year survival rate was significantly higher (75%) in patients who underwent allogeneic HSCT after experiencing complete or partial response to initial therapy than in those who experienced response to initial therapy and underwent no allogeneic HSCT or who underwent allogeneic HSCT for relapsed or refractory Richter's syndrome (27% and 21%, respectively).159 Allogenic HSCT can be considered after initial therapy.
Supportive Care for Patients With CLL
Infections: Infectious complications in patients with CLL are influenced by the reduction in immunoglobulin levels and are more common in previously treated patients.162 Hypoglobulinemia is present in 38% of patients up to 3 years before CLL diagnosis.163 In a retrospective analysis, 89% of patients with fludarabine-refractory CLL developed infectious complications (78% bacterial and 12% viral).164 IVIG, antibacterial or antiviral prophylaxis, and vaccinations are the 3 options available for reducing infectious complications.
In randomized studies, IVIG has been associated with a significant decrease in the occurrence of infections but no improvement in overall survival.165–169 Antibacterial prophylaxis may be a useful alternative option. Protein and conjugate vaccines have been shown to induce better responses than plain polysaccharide vaccines.170,171 Some studies have reported that H2-receptor blockers can enhance vaccine response.172,173
In selected patients (serum IgG < 500 mg/dL) with recurrent sinopulmonary infections requiring intravenous antibiotics or hospitalization, the NCCN Guidelines recommend monitoring IVIG levels and administering 0.3 to 0.5 g/kg of IVIG monthly to maintain nadir levels of approximately 500 mg /dL. Antibacterial prophylaxis is also appropriate for managing infections. Antiviral and pneumocystis prophylaxis is recommended for patients receiving purine-analog and/or alemtuzumab during treatment and thereafter. Acyclovir or its equivalent is recommended for herpes virus, and sulfamethoxazole trimethoprim or equivalent is recommended for pneumocystis carinii pneumonia. Annual influenza vaccine and pneumococcal vaccine (every 5 years) is recommended for all patients. All live vaccines should be avoided. Patients with CLL have a poor immune response to influenza vaccine and therefore should be cautious during influenza season even after vaccination. Patients who have undergone rituximab therapy do not respond to influenza vaccination until B-cell recovery.
Cytomegalovirus Reactivation: Cytomegalovirus reactivation is a well-documented side effect in patients receiving alemtuzumab.116,128,174 Cytomegalovirus reactivation is associated with relatively mild or no symptoms when prophylactic measures are used during treatment with alemtuzumab. The current appropriate management is controversial; some clinicians use ganciclovir (oral or intravenous) prophylactically if viremia is present, others use it only if the viral load is increasing.175,176
Clinicians must be aware of the high risk of cytomegalovirus reactivation. Regularly monitoring patients for cytomegalovirus reactivation using PCR is an effective approach.177 These NCCN Guidelines recommend cytomegalovirus viremia monitoring (every 2–3 weeks). Consultation with an infectious disease expert may be necessary.
Autoimmune Cytopenias: Autoimmune hemolytic anemia (AIHA); immune-mediated thrombocytopenia, also known as immune thrombocytopenic purpura (ITP); and pure red blood cell aplasia (PRCA) are the most frequent autoimmune cytopenias in patients with CLL.178,179
AIHA is the most common form of autoimmune cytopenia. The direct antiglobulin test (DAT) has been used to diagnose AIHA. However, most patients with AIHA have negative DAT, and therefore additional markers, such as low haptoglobin and elevated reticulocyte and LDH, are required to confirm the diagnosis.180 Patients with advanced disease, unmutated IGHV, increased serum β2-microglobulin level, and high expression of ZAP-70 are also at a higher risk of developing AIHA.181–183 ITP in patients with CLL is associated with poorer survival independent of common clinical prognostic variables. In a recent Italian study, high WBC count, unmutated IGHV, and DAT and ZAP-70 positivity were associated with the development of ITP in patients with CLL.184 PRCA is less common in these patients.
Bone marrow evaluation is recommended to confirm the diagnosis of autoimmune cytopenia. Evaluation of parvovirus B19 is also recommended to exclude parvovirus-induced PRCA. AIHA and ITP can be managed with corticosteroids in most cases. IVIG, cyclosporin,185 and splenectomy should be used in steroid-refractory cases. Rituximab has also been effective for the treatment of autoimmune cytopenias.186–192 Corticosteroids tend to be less effective in PRCA than in ITP or AIHA. If corticosteroids are not effective, cyclosporin can be used. In very refractory cases, allogeneic HSCT maybe necessary. More recently, synthetic thrombopoietin-like agents, such as romiplostim and eltrombopag, have shown promising results in the treatment of thrombocytopenia associated with ITP.193–195 In 2008, both romiplostim and eltrombopag received FDA approval for the treatment of thrombocytopenia in patients with ITP refractory to steroids, IVIG, and splenectomy.
Purine analog–based therapy has been associated with AIHA. Recent studies have reported a higher incidence of AIHA in patients treated with fludarabine or chlorambucil than in those treated with fludarabine-based combination regimens (FC or FCR).180,196 AIHA should not preclude the use of combination therapy containing fludarabine, and patients should be observed carefully. In the case of severe AIHA, fludarabine therapy should be discontinued and subsequent use avoided.
Tumor Lysis Syndrome: Patients with CLL and high WBC count may occasionally experience TLS and should be managed as outlined in Tumor Lysis Syndrome, page 524.
Follicular Lymphoma
Diagnosis
FL is the most common indolent subtype of NHL, accounting for approximately 22% of all newly diagnosed cases. Approximately 90% of the cases have a t(14;18) translocation, which juxtaposes BCL2 with the IGHV locus and results in the deregulated expression of BCL2.
FL has a characteristic immunophenotype that includes CD20+, CD10+, BCL2+, CD23+/–, CD43–, CD5–, CCDN1–, and BCL6+. Rare cases of FL may be CD10– or BCL2–. In young patients with localized BCL2– disease, pediatric FL may be considered. This diagnosis is easily established based on histology, but immunophenotyping is encouraged to rule out a nodular MCL or SLL. Low-grade FL with a high proliferation index determined through Ki-67 immunostaining has been shown to be associated with aggressive clinical behavior, but no evidence shows that it should guide selection of therapy.197,198 Molecular genetic analysis to detect BCL2 rearrangement; cytogenetics or FISH to identify t(14;18); and paraffin section immunohistochemistry for Ki-67 will be useful under certain circumstances.
In FL, pathologic grading according to the number of centroblasts is considered to be a clinical predictor of outcome. In the 2001 WHO classification, 3 grades were recommended: FL1, FL2, and FL3; FL3 could be optionally stratified into 3A (centrocytes still present) or 3B (sheets of centroblasts). However, clinical outcomes for patients with FL1 and FL2 do not differ and classification is unreliable. Therefore, the updated 2008 WHO classifies these as one grade (FL1–2).16,17 Hans et al.199 reported no difference in survival between grade 3A and 3B, whereas patients with FL3 with more than 50% diffuse components had an inferior survival similar to that of those with DLBCL. FL3B with cytogenetic abnormalities of BCL6 (at 3q27) is believed to be genetically more akin to germinal center–type DLBCL than FL1–3A and has a more aggressive clinical course. Patients with FL3B with BCL2 translocation seem to have similar clinical behavior to patients with FL1–3A.200 Because FL3B is rare, in most studies clinical behavior of FL3 is based mainly on FL3A cases. The 2008 WHO classification mandates stratifying FL3 into either 3A or 3B. FL is thus still divided into 3 grades (FL1–2, FL3A, and FL3B) based on the number of centroblasts. Any diffuse areas in FL should be given a separate diagnosis of DLBCL if they meet the criteria for FL3A or 3B.
The Follicular Lymphoma International Prognostic Index (FLIPI) is based on patient age, Ann Arbor stage, number of involved nodal sites involved, hemoglobin levels, and serum LDH levels.201 In the National LymphoCare study, which analyzed the treatment options and outcomes of 2728 patients with newly diagnosed FL, FLIPI was used to classify patients into 3 distinct groups, with survival ranging from 52% to 90% at 5 years.202 In a recent study conducted by the International Follicular Lymphoma Prognostic Factor Project, a prognostic model was developed from prospective accumulated data that includes age, hemoglobin, dimension of the longest lymph node, β2-microglobulin, and bone marrow involvement. FLIPI2 was highly predictive of treatment outcome in patients with newly diagnosed FL treated with chemoimmunotherapy.203 With follow-up to date, FLIPI2 does not predict for overall survival; furthermore, it is only applicable to patients requiring therapy. Both the FLIPI1 and -2 predict for prognosis, but they have not yet been established as a means of selecting treatment options.
Workup
The diagnostic workup for FL is similar to that for other indolent lymphomas. Most patients present with disseminated disease. The approach to therapy differs dramatically for localized and disseminated disease. Bone marrow biopsy with aspirate is essential to document clinical stage I and II disease. This can be deferred if observation is the initial treatment option. Most NCCN investigators routinely use chest, abdominal, and pelvic CT as part of the diagnostic evaluation. Neck CT may also help define the extent of local disease. In patients presenting with what appears to be localized disease, a PET scan may help identify occult sites of disease or be useful if concern exists about histologic transformation.204 PET does not replace histologic confirmation of the diagnosis; however, areas with discordant high FDG-avidity represent the most likely sites of transformation.
Treatment Options Based on Clinical Stage
The guidelines for FL apply to FL1 and FL2. FL3A and FL3B are commonly treated according to guidelines for DLBCL. In most centers, the proportion of patients diagnosed with FL3 is greater than that previously diagnosed as follicular large cell lymphoma in the IWF.
Stages I and II: Involved-field RT (IFRT; 24–30 Gy, with an additional 6 Gy in selected patients with bulky disease) is the preferred treatment for patients with stage I or contiguous stage II disease. In a retrospective analysis of 43 patients with stage I and II disease, carefully selected patients (meeting criteria of large abdominal radiation field, advanced age, concern for xerostomia or patient refusal) who did not undergo immediate treatment had comparable outcomes to those treated with RT.205 In selected cases in which toxicity of IFRT outweighs the potential clinical benefit, observation may be appropriate. Alternate treatment options include immunotherapy with or without chemotherapy with or without RT. The addition of cyclophosphamide, vincristine, prednisone, and bleomycin (COP-bleomycin) or CHOP-bleomycin improved failure-free survival but did not impact overall survival in patients with early-stage disease.206 The addition of adjuvant CHOP to RT did not improve relapse-free survival in patients with early-stage, low-grade lymphoma.207 Therefore, chemotherapy plus RT is included as a category 2B recommendation.
For patients with a partial or complete response, clinical follow-up with examination and laboratory assessment is initially performed every 3 months, with repeat imaging every 6 months or as clinically indicated. Patients with no response to initial therapy should be managed in the same manner as those with advanced disease.
Stage II (Bulky Disease) and Stages III and IV: Rituximab has shown single-agent activity in previously untreated patients and in those with relapsed or refractory disease.208–210 The addition of rituximab to combination chemotherapy regimens has consistently increased the overall response rate, response duration, and progression-free survival. In addition, some studies have shown a benefit in overall survival, with a recent meta-analysis confirming this despite what is still limited follow-up for FL.211
A small study evaluating the safety and efficacy of R-CHOP showed excellent long-term results.212,213 A prospective randomized phase III study in 428 patients by the German Low-Grade Lymphoma Study Group (GLSG) established the superiority of R-CHOP over CHOP in treatment-naïve patients. R-CHOP was associated with a 60% reduction in the relative risk for treatment failure, significantly prolonged time to treatment failure, higher overall response rate, and prolonged duration of remission.214 Overall survival analysis is complicated by a second randomization that included high-dose therapy followed by autologous stem cell rescue (HDT/ASCR). In this study, overall survival was the same with and without rituximab, if there was consolidation with HDT/ASCR. However, overall survival was significantly improved for patients treated with R-CHOP followed by interferon compared with those treated with CHOP followed by interferon. R-CHOP also improved outcome of elderly patients with previously untreated FL.215
The addition of rituximab to CVP (cyclophosphamide, vincristine, and prednisone) chemotherapy (R-CVP) significantly improved outcome in patients with previously untreated FL, with no significant increase in toxicity.216 At a median follow-up of 53 months, R-CVP was associated with an improved overall response rate (81% vs. 57%), median time to progression (34 vs. 15 months), and 4-year overall survival (83% vs. 77%).217
The addition of rituximab to fludarabine or fludarabine-based combination has improved outcomes in various clinical studies.218–221 In a prospective randomized trial, the FCMR regimen (fludarabine, cyclophosphamide, mitoxantrone, and rituximab) was associated with superior outcomes in patients with relapsed or refractory FL and MCL.219 In another randomized trial, concurrent administration of rituximab with the FND regimen (fludarabine, mitoxantrone, and dexamethasone) resulted in a significantly higher 3-year failure-free survival rate (84% vs. 59% for sequential arm) in a subset of patients with FL.220
Bendamustine, as a single agent or in combination with rituximab (BR), has shown promising results with acceptable toxicity in both newly diagnosed and heavily pretreated patients with relapsed or refractory indolent or mantle cell histologies and transformed NHL.222–226 A randomized phase III study conducted by the Study Group Indolent Lymphomas (StiL) compared BR with R-CHOP in patients with advanced follicular, indolent, and mantle cell lymphomas. The overall response rate was similar in both arms, although the complete response rate was significantly higher in the BR arm (40% vs. 31%).222 However, patients treated with BR had significantly longer median progression-free (55 vs. 35 months) and event-free survivals (54 vs. 31 months). BR also showed a better toxicity profile. Overall survival is similar.
Radioimmunotherapy with 131I-tositumumab227–230 and 90Y-ibritumomab tiuxetan231–233 was also evaluated in patients with newly diagnosed and those with relapsed, refractory, or histologically transformed FL. Initial treatment with a single 1-week course of 131I-tositumomab induced prolonged clinical and molecular remissions in patients with advanced FL.227 After a median follow-up of 10 years, the median duration of response was 6 years. For the 57 complete responders, median progression-free survival was 11 years234; 10-year progression-free and overall survival rates were approximately 40% and 82%, respectively. In an international phase II trial, 90Y-ibritumomab used as first-line therapy resulted in an overall response rate of 72% (52% showing a complete response, and 20% showing a partial response) at 12 months after therapy. At a median follow-up of 23 months, the progression-free survival was 18 months.235
A single course of 131I-tositumumab was significantly more efficacious than the last qualifying chemotherapy in extensively pretreated patients with refractory, low-grade, or transformed NHL.229 The final results of the study showed that 131I-tositumumab resulted in long-term, durable complete response in a subset of patients who had received no prior rituximab.236 In a randomized phase III study, 90Y-ibritumomab tiuxetan also produced statistically and clinically significant higher overall response and complete response rates than rituximab alone in patients with relapsed or refractory low-grade, follicular, or transformed lymphoma.232 At a median follow-up of 44 months, median time to progression (15 vs. 10.2 months) and duration of response (16.7 vs. 11.2 months) were longer in patients treated with 90Y-ibritumomab than those treated with rituximab.233
NCCN Recommendations for Stage II (Bulky Disease) and Stage III and IV: Despite therapeutic advances that have improved the survival of patients with FL, it remains an incurable disease with conventional therapy. Four prospective randomized trials have failed to show a survival advantage for immediate treatment.237–240 Modified Groupe d'Etude des Lymphomes Folliculaire (GELF) criteria are used to decide when to initiate therapy in patients with advanced-stage disease, including symptoms attributable to FL (not limited to B symptoms); threatened end-organ function; cytopenia secondary to lymphoma; bulky disease (a singe mass > 7 cm, or 3 or more masses > 3 cm); splenomegaly; and steady progression over at least 6 months. Patient preference should be considered; however, patients wanting treatment without a clinical indication should be referred for an appropriate clinical trial. Treatment selection should be highly individualized according to age, extent of disease, comorbid conditions, and goals of therapy. When choosing an initial therapy, care should be given to avoid excessively myelotoxic regimens in patients who may be candidates for HDT/ASCR. In patients with hepatitis B, treatment with an antiviral should be given if rituximab is used (see Hepatitis B Virus Reactivation, page 523).
First-Line Therapy: In the absence of an appropriate clinical trial, patients with indications for treatment should undergo systemic therapy. In selected cases, such as elderly frail patients unable to tolerate chemotherapy, IFRT (4 Gy) may be used for local palliation. Asymptomatic patients, especially those older than 70 years, can be observed.239 At 36 months after randomization, the results of an interim analysis of the Intergroup trial of rituximab versus a watch-and-wait strategy showed that the estimated progression-free survival was significantly better for asymptomatic patients with stage II and IV nonbulky disease receiving rituximab alone or rituximab followed by rituximab maintenance than those undergoing observation, but no difference was seen in overall survival between the treatment arms.240 Further follow-up is needed to determine if immediate treatment has an impact on time to second therapy. The panel thought that these data were not sufficiently compelling to indicate a change of practice. The ECOG RESORT trial is examining rituximab maintenance versus rituximab delayed until progression in a similar patient population, and will provide some additional insight.
Based on the reported data, BR, CHOP, or CVP chemotherapy for first-line therapy in patients with advanced FL are all category 1 recommendations. BR has been shown to have less toxicity and a superior progression-free survival compared with R-CHOP; however, overall survival is not different. Furthermore, limited data are available on the risk of secondary myelodysplastic syndromes/acute myeloid leukemia (MDS/AML) after bendamustine. Data from a limited subset of patients suggest that peripheral blood stem cells can be collected after both BR and R-CHOP; more data are needed to confirm this finding. No randomized trials have compared R-CHOP and R-CVP. Therefore, choice of first-line therapy in advanced-stage FL remains a challenge for the clinician. Other suggested regimens include rituximab either as a single agent or in combination with fludarabine-based chemotherapy. Radioimmunotherapy is included as category 2B option for first-line treatment. IFRT (4–30 Gy) with or without systemic therapy can be considered for palliation in patients with locally bulky or symptomatic disease if they are unable to tolerate systemic therapy.
Single-agent rituximab is the preferred first-line therapy for elderly or infirm patients. Single-agent cyclophosphamide had equivalent overall survival and complete response rates as cyclophosphamide-based combination chemotherapy.241 The NCCN Guidelines also include radioimmunotherapy and alkylating agent–based chemotherapy (cyclophosphamide or chlorambucil) with or without rituximab as alternative options for elderly or infirm patients.
First-Line Consolidation or Extended Dosing: Chemotherapy Followed by Radioimmunotherapy: First-line chemotherapy followed by radioimmunotherapy with either 131I-tositumumab242–245 or 90Y-ibritumomab246–249 has been evaluated in several phase II studies. In the SWOG S9911 trial, CHOP followed by 131I-tositumomab resulted in an overall response rate of 91%, including a 69% complete remission rate in patients with previously untreated FL.244 After a median follow-up of 5 years, the estimated 5-year overall survival and progression-free survival rates were 87% and 67%, respectively.243 In historical comparison, these statistics were better than those reported for CHOP alone. In a multicenter phase II study in untreated patients with FL, CVP chemotherapy followed by 131I-tositumomab resulted in overall and complete response rates of 100% and 93%, respectively. The 5-year progression-free and overall survival rates were 56% and 83%, respectively.245
In the international phase III FIT trial (First-Line Indolent Trial), 414 patients with advanced-stage FL responding to first-line induction therapy were randomized to receive either 90Y-ibritumomab or no further treatment.248 After a median follow-up of 5.5 years, the 5-year progression-free survival rates were 47% and 29%, respectively. Median progression-free survival was 49 and 14 months, respectively.250 No significant difference was seen in overall survival. The rate of secondary malignancies (MDS/AML) were higher among patients in the consolidation group (3%) than those in the control group (1%). This trial included only a limited number of patients (14%) who received rituximab in combination with chemotherapy as induction therapy. Among these patients, the 5-year progression-free survival rates were 64% and 48%, respectively, for the 90Y-ibritumomab and control groups.
Maintenance Therapy With Rituximab: Prolonged administration of rituximab significantly improved event-free survival in chemotherapy-naïve patients experiencing response to rituximab induction, but did not extend overall survival.251–253 In another study, maintenance rituximab improved progression-free survival (31 vs. 7 months). However, retreatment with rituximab at progression provided the same duration of benefit as maintenance rituximab (31 vs. 27 months).254 The randomized phase III study (ECOG 1496) showed a progression-free survival benefit for rituximab maintenance in patients with advanced indolent lymphoma responding to first-line chemotherapy.255 The 3-year progression-free survival rate was 68% for maintenance rituximab compared with 33% for observation for all patients with advanced indolent lymphoma experiencing response or stable disease after CVP chemotherapy. The corresponding progression-free survival rates were 64% and 33%, respectively, for patients with FL.255
The PRIMA trial prospectively evaluated the role of rituximab maintenance in patients experiencing response to first-line chemotherapy in combination with rituximab (R-CVP, R-CHOP, or FCMR). This study randomized 1018 eligible patients to either observation or rituximab maintenance for 2 years.256 The interm analysis with a median follow-up of 24 months showed that rituximab maintenance significantly improved progression-free survival (primary end point) compared with observation. After a median follow-up of 36 months, the 3-year progression-free survival rate was 75% in the rituximab maintenance arm and 58% in the observation arm.257 At 2 years after randomization, 72% of patients in the rituximab maintenance group were experiencing a complete response or complete response unconfirmed257; however, no significant differences were seen in overall survival. Follow-up is ongoing to evaluate the effect of rituximab maintenance on overall survival.
NCCN Recommendations: Patients experiencing complete or partial response to first-line therapy can be observed or treated with consolidation therapy. Based on the results of the PRIMA study,256 maintenance rituximab up to 2 years is recommended (category 1) for patients experiencing response to first-line chemoimmunotherapy. Based on the results of the FIT trial, radioimmunotherapy is recommended (category 1) only for patients who received first-line chemotherapy.248 The recommendation to limit radioimmunotherapy to patients receiving induction chemotherapy rather than chemoimmunotherapy is based on the small proportion of patients who received chemoimmunotherapy induction in the FIT trial.
Second-Line Therapy for Relapsed or Progressive Disease: Frequently, patients experiencing progression after first-line therapy will benefit from a second period of observation. Thus, treatment for relapsed or progressive disease is based on the modified GELF criteria as used in first-line therapy. Progressive disease should be histologically documented to exclude transformations, especially in the presence of rising LDH levels, disproportional growth in one area, development of extranodal disease, or new B symptoms. Nonuniform uptake on a FDG-PET scan can be an indication of transformation; areas of high standardized uptake value, especially exceeding 13.1, are suspicious for transformation. However, a PET scan does not replace a biopsy; it should be used to direct a biopsy to enhance the diagnostic yield. Options include chemoimmunotherapy regimens used for first-line treatment, the FCMR regimen (category 1), radioimmunotherapy (category 1), or any of the second-line regimens used for patients with DLBCL.
Second-Line Consolidation or Extended Dosing: Two large randomized trials have established that rituximab maintenance after second-line therapy for relapsed/refractory disease provides a progression-free survival advantage over observation for patients treated with chemoimmunotherapy.258–260
In a prospective randomized study by the GLSG, rituximab maintenance after second-line treatment with FCMR significantly prolonged duration of response in patients with recurring or refractory FL and, to a lesser degree, those with MCL.258 In a phase III Intergroup trial (EORTC 20981), maintenance rituximab significantly improved median progression-free and overall survivals in patients with relapsed or resistant FL responding to CHOP or R-CHOP.259 With a median follow-up of 6 years, the 5-year overall survival rate was 74% and 64% in the rituximab maintenance and observation arms, respectively.260
HDT/ASCR has been shown to prolong overall and progression-free survivals in patients with relapsed or refractory disease.261–263 The GELA recently conducted a retrospective analysis of patients with relapsed or refractory FL after first-line treatment with chemotherapy alone and found that event-free survival and survival after relapse were superior in those treated with rituximab-containing regimens compared with chemotherapy only–based HDT/ASCR.264 The combination of rituximab-based second-line therapy followed by HDT/ASCR had the best results, with a 90% survival rate after relapse at 5 years. Allogeneic HSCT is associated with high treatment-related mortality rates (30%–38% for myeloablative and 25% for nonmyeloablative).265,266 In a recent report from the Center for International Blood and Marrow Transplant Research, both myeloablative and nonmyeloablative transplant had similar TRM rates, but nonmyeloablative allogeneic HSCT was associated with an increased risk of disease progression.267
NCCN Recommendations: Rituximab maintenance is recommended (category 1) for patients in second-line remission. However, the panel recognized that the efficacy of maintenance rituximab in second-line remission would likely be impacted by first-line maintenance. If a patient experienced progression during or within 6 months of first-line maintenance rituximab, the value of maintenance in the second-line is likely minimal. HDT/ASCR is an appropriate consolidative therapy for patients experiencing second or third remission. Allogeneic HSCT may be considered for highly selected patients. For patients receiving consolidation therapy, clinical follow-up is initially recommended every 3 months with repeat imaging every 6 months and/or as clinically indicated.
Histologic Transformation to DLBCL: Transformation to DLBCL in patients with FL occurs at a rate of approximately 2% to 3% per year for at least 15 years, with the risk of transformation decreasing thereafter for reasons that remain unclear.268 Transformation to DLBCL is generally associated with a poor clinical outcome. However, patients with limited disease and no previous exposure to chemotherapy can have favorable outcomes similar to those with de novo DLBCL.269 In patients who have had multiple prior therapies, the prognosis is much poorer and enrollment in a clinical trial is the preferred option. In the absence of a clinical trial, treatment options include radioimmunotherapy, chemotherapy with or without rituximab, IFRT, or best supportive care. HDT/ASCR or allogeneic HSCT can be considered for patients with responsive disease after initial treatment.
For patients who have had minimal (IFRT alone or one course of single-agent therapy, including rituximab) or no prior chemotherapy, anthracycline-based chemotherapy with rituximab, with or without RT, is included as a treatment option. Enrollment in a clinical trial is recommended for all patients after initial therapy. Patients experiencing response to initial treatment could be considered for HDT/ASCR or allogeneic HSCT. Alternatively, patients experiencing a complete response to initial therapy can be observed, whereas radioimmunotherapy may be considered for those experiencing a partial response. Patients experiencing no response or progressive disease after initial therapy should be treated with radioimmunotherapy or best supportive care.
DLBCL
Diagnosis
DLBCLs are the most common lymphoid neoplasms in adults. DLBCL, NOS; FL (grade 3); DLBCL coexistent with FL of any grade; DLBCL coexistent with gastric MALT or nongastric MALT lymphoma; intravascular large B-cell lymphoma; DLBCL associated with chronic inflammation; ALK-positive DLBCL; EBV-positive DLBCL of the elderly; and T-cell/histiocyte–rich large B-cell lymphoma are also managed according to the DLBCL guidelines.
Recent studies with gene expression microarray analysis of DLBCL have revealed significant heterogeneity within this diagnosis.270 Gene expression profiling has been used to identify 3 different subtypes of DLBCL: germinal center B-cell, activated B-cell, and type 3, which includes primary mediastinal B-cell lymphoma (PMBL) and cases that cannot be classified as germinal center B-cell or activated B-cell subtypes.270 Gene expression profiling is not yet recommended for routine clinical use. However, incorporation of this information into treatment algorithms awaits further investigation. Immunohistochemical markers CD10, BCL6, and IRF4/MUM1 have been reported to recapitulate gene expression profiling, separating patients into tumors derived from germinal center origin (CD10+, or BCL6+, IRF4/MUM1–) and non–germinal center origin (CD10–, IRF4/MUM1+ or BCL6–, IRF4/MUM1–).271 However, the validity of this classification scheme has been questioned. An improved immunohistochemical algorithm has been proposed that includes GCET1, FOXP1, BCL6, IFR4/MUM1, and CD10.272 Further work is necessary to validate the robustness of this new algorithm to distinguish between germinal center vs. non–germinal center subtypes.
The typical immunophenotype is CD20+, CD45+, and CD3–. The recommended immunophenotyping panel includes CD20, CD3, CD5, CD10, CD45, BCL2, BCL6, and IRF4/MUM1. When available, GCET1 and FOXP1 can provide information necessary for the Choi immunohistochemical cell of origin algorithm. Additional markers such as CD138, cyclin D1, ALK1, EBV, and HTLV may be useful under certain circumstances to establish the subtype.
Workup
The staging workup is designed to identify all sites of known disease and determine prognosis based on known clinical risk factors. Risk factors used by the International Prognostic Index (IPI) include age, disease stage, serum LDH level, performance status, and number of extranodal disease sites. In patients aged 60 years or younger, these factors include tumor stage, performance status, and serum LDH level. The IPI and age-adjusted IPI can be used to identify specific groups of patients who are more or less likely to be cured with standard therapy.273
PET or PET-CT scans have a more clear-cut role in selected cases of DLBCL than in other lymphoid neoplasms. PET scans are particularly informative during initial staging, in which upstaging results in altered therapy approximately 9% of the time, and in response evaluation after treatment because they can distinguish residual fibrotic masses from masses containing viable tumor. PET scans have now been incorporated into the response criteria, and availability of a baseline study is necessary for optimal interpretation of the posttreatment study. In some centers, β2-microglobulin is considered a major determinant of risk (category 2B). Lumbar puncture is indicated in patients with one or more of the following sites of involvement: paranasal sinus, testicular, epidural, or bone marrow (with large cells), or with HIV-associated lymphoma, or who have 2 or more extranodal sites. Diagnostic yield is improved by flow cytometric analysis of cerebrospinal fluid.
Treatment Options by Clinical Stage
Treatment options for DLBCL differ between patients with localized (Ann Arbor stage I–II) and advanced (Ann Arbor stage III–IV) disease. Prognosis is extremely good for patients with no adverse risk factors (elevated LDH, stage II bulky disease, > 60 years, or ECOG performance status ≥ 2). Patients with advanced disease should be enrolled in clinical trials whenever possible.
Stages I and II: In the SWOG 8736 study, 3 cycles of CHOP followed by IFRT produced significantly better progression-free (5-year estimated progression-free survival: 77% vs. 64% for CHOP alone) and overall survival rates (82% vs. 72% for CHOP alone) than 8 cycles of CHOP alone in patients with localized aggressive NHL274; however, this difference disappeared with further follow-up. The benefit of CHOP (3 cycles) followed by IFRT (5-year overall survival rate of 95%) in patients with limited-stage DLBCL (aged ≤ 60 years with no adverse risk factors) was also confirmed in a series from British Columbia Cancer Agency.275 Another randomized trial (ECOG 1484) showed that the addition of RT to CHOP (8 cycles) prolonged disease-free survival in patients with limited stage DLBCL who had experienced a complete response to CHOP alone (6-year disease-free survival was 73% for IFRT and 56% for observation).276 In the GELA study (LNH 93-4), the addition of RT to 4 cycles of CHOP did not provide any advantage over 4 cycles of CHOP alone for the treatment of elderly patients with low-risk localized aggressive lymphoma. The estimated 5-year event-free survival rates were not different between the groups (61% and 64%, respectively) and the 5-year estimated overall survival rates were 68% and 72%, respectively.277 However, administration of RT was markedly delayed and 12% of patients in the RT arm did not receive RT.
The efficacy of the addition of rituximab to CHOP (3 cycles) and IFRT has also been reported in patients with limited-stage DLBCL. In the SWOG 0014 study, with the median follow-up of 5 years, the 2- and 4-year progression-free survival rates were 93% and 88%, respectively, for patients with at least one stage modified. The corresponding overall survival rates were 95% and 92%, respectively.278 In historical comparison, these results were better than the survival rates for patients treated without rituximab (4-year progression-free and overall survival rates were 78% and 88%, respectively). In the GELA study (LNH 93-1), intensified chemotherapy (ACVBP [doxorubicin, cyclophosphamide, vindesine, bleomycin, and prednisone] followed by consolidation with methotrexate, etoposide, ifosfamide, and cytarabine) with or without rituximab was found to be superior to CHOP with or without rituximab (3 cycles) plus RT in patients with low-risk early-stage disease.279,280 However, this regimen is also associated with significant toxicity and includes vindesine, which is not available in the United States.
NCCN Recommendations: R-CHOP (3 cycles) with IFRT or R-CHOP (6 cycles) with or without IFRT is recommended for patients with nonbulky disease (< 10 cm). IFRT is recommended for patients who are not candidates for chemotherapy. The addition of RT to a full course of 6 cycles of R-CHOP for patients with no adverse factors is included as a category 2B recommendation. Patients with bulky disease (≥ 10 cm) may be more effectively treated with 6 cycles of R-CHOP with or without locoregional RT (category 1).
Stages III and IV: R-CHOP-21 chemotherapy has been the standard treatment for patients with advanced-stage DLBCL based on the results of the GELA study (LNH 98-5), which showed that the addition of rituximab to CHOP-21 improved progression-free and overall survivals in elderly patients with advanced DLBCL. In this study, 399 elderly patients (age 60–80 years) were randomized to receive 8 cycles of R-CHOP or CHOP.281–283 Long-term follow-up of this study showed that progression-free (36.5% vs. 20%), disease-free (64% vs. 43%), and overall survival rates (43.5% vs. 28%) were statistically significant in favor of R-CHOP at a median follow-up of 10 years.284 These findings have been confirmed in 3 additional randomized trials, including the MabThera International Trial (MInT; 6 cycles of R-CHOP or CHOP), which extended the findings to young patients with 0 or 1 risk factors according to the IPI285,286; the Dutch HOVON and Nordic Lymphoma group study (8 cycles of R-CHOP-14 or CHOP-14); and the ECOG/CALGB study, which confirmed the findings in patients older than 60 years.287,288 The ECOG/CALGB study also showed that maintenance rituximab significantly prolonged failure-free survival in older patients whose disease responded to CHOP chemotherapy, but it did not show any clinical benefit for those whose disease responded to R-CHOP as induction therapy.288
Six cycles of dose-dense CHOP (CHOP-14) as first-line therapy was found to be superior to 6 cycles of CHOP-21.289–291 In the RICOVER 60 trial, the addition of rituximab to CHOP-14 improved clinical outcomes in elderly patients compared with CHOP-14 alone.292 In this study, overall survival significantly favored 6 cycles of R-CHOP-14 over 8 cycles (78% and 72.5%, respectively) because of late, non–cancer-related deaths. In patients with a partial response after 4 cycles of chemotherapy, 8 cycles were not better than 6. Ongoing randomized studies are evaluating the role of R-CHOP-14 versus R-CHOP-21.293,294
Dose-adjusted EPOCH (etoposide, prednisone, vincristine, cyclophosphamide, and doxorubicin) plus rituximab has shown significant activity in untreated patients with DLBCL.295,296 An ongoing phase III randomized study is evaluating dose-adjusted R-EPOCH versus R-CHOP in untreated patients with DLBCL. NCCN Recommendations: Patients with advanced disease are treated with 6 cycles of R-CHOP (category 1). In selected cases, RT to bulky sites may be beneficial (category 2B). Involvement of paranasal sinus, testis, bone marrow, or 2 or more extranodal sites is associated with a higher risk of central nervous system relapse.297–300 Central nervous system prophylaxis with 4 to 8 doses of intrathecal methotrexate and/or cytarabine should be given during the course of treatment, although prophylactic therapy has been suggested to be of value.
R-CHOP with rituximab is preferable because of reduced toxicities; however, other comparable anthracycline-based regimens are acceptable. Suggested alternate options include dose-dense R-CHOP-14 or dose-adjusted EPOCH plus rituximab, both of which are listed as category 2B recommendations. The NCCN Guidelines have included the following regimens as first-line therapy for patients with poor left ventricular function (category 2B):
-
CEPP (cyclophosphamide, etoposide, prednisone, and procarbazine) plus rituximab301
-
CDOP (cyclophosphamide, liposomal doxorubicin, vincristine, and prednisone) plus rituximab302–304
-
CNOP (cyclophosphamide, mitoxantrone, vincristine, and prednisone) plus rituximab305–308
-
Dose-adjusted EPOCH plus rituximab
-
CEOP (cyclophosphamide, etoposide, vincristine, and prednisone) plus rituximab309
Participation in clinical trials of new regimens is recommended if available. In patients with bulky disease or impaired renal function, initial therapy should include monitoring and prophylaxis for TLS.
Response Assessment and Follow-Up Therapy
Interim restaging is performed to identify patients whose disease has not responded or has progressed despite induction therapy. PET scans may be particularly useful in determining whether residual masses represent fibrosis or a viable tumor. A negative PET scan after 2 to 4 cycles of chemotherapy has been associated with an excellent outcome.310 Patients with negative PET scans have significantly higher 5-year event-free survival rates than those with positive scans (80% vs. 36%, respectively) after induction therapy with CHOP with or without rituximab.311 However, interim PET scan can produce false-positive results, and some patients treated with chemoimmunotherapy have a favorable long-term outcome despite a positive interim PET scan. A prospective study from Memorial Sloan-Kettering Cancer Center (MSKCC) evaluated the significance of interim PET scans by obtaining biopsies from patients with an interim positive PET. The biopsy showed persistent disease in only 5 of 37 of these scans; progression-free survival was identical for patients with positive interim PET scans and negative biopsy results and those with negative interim PET scans.312 Therefore, an interim PET scan is not recommended outside of a clinical trial. If it is used, a repeat biopsy of residual masses is necessary before changing the treatment course. Patients who are undergoing induction therapy should undergo evaluation before receiving RT, including all positive studies, after 3 to 4 cycles of chemotherapy. End-of-treatment restaging is performed on completion of treatment. Although the optimal time to end-of-treatment restaging is unknown, the panel recommends waiting 6 to 8 weeks after completion of therapy before repeating PET scans.
Interim and End-of-Treatment Response Evaluation for Stages I and II: When the treatment plan involves RT after a short course of chemoimmunotherapy, restaging should be undertaken beforehand, including repeat PET scan, because the dose of RT will be influenced by the result (see Principles of Radiation Therapy, available online, in these guidelines, at www.NCCN.org [NHODG-E]).
If the interim restaging shows a complete response, the planned course of treatment is completed. If the interim restaging shows a partial response, completing the planned course of treatment with a higher dose of RT is appropriate (see Principles of Radiation Therapy, available online, in these guidelines, at www.NCCN.org [NHODG-E]). Alternatively, a repeat biopsy can be obtained and, if positive, the patient can proceed to second-line therapy followed by HDT/ASCR. It is appropriate to enroll patients with an interim partial response in a clinical trial. The choice between these options is often made on clinical grounds. RT is appropriate for patients not eligible for HDT/ASCR. Higher-dose RT is also a reasonable choice in the presence of a very good partial response. Patients with refractory or primarily progressive disease are managed as refractory or relapsed disease.
End-of-treatment restaging is performed on completion of treatment. After end-of-treatment restaging, follow-up at regular intervals is recommended for patients with complete response. Patients with a partial response and those with no response to treatment or progressive disease are treated as described for relapsed or refractory disease.
Patients are then followed up at regular intervals (every 3–6 months for 5 years, and then annually or as clinically indicated). Repeat imaging is performed as clinically indicated.
Interim and End-of-Treatment Response Evaluation for Stages III and IV: After interim staging, the planned course of treatment (R-CHOP to a total of 6 cycles) is completed for patients with complete and partial responses. End-of-treatment restaging is performed on completion of treatment. Observation is preferred for patients with complete response. RT to initially bulky disease or first-line consolidation with HDT/ASCR can be considered (category 2B). Patients with a partial response and those with no response to treatment or progressive disease are treated as described for relapsed or refractory disease.
Relapsed or Refractory Disease
The role of HDT/ASCR in patients with relapsed or refractory disease was shown in an international randomized phase III trial (PARMA study).313 In this study, 109 patients with relapsed or refractory DLBCL responding to first-line chemotherapy were randomized to either chemotherapy plus RT (54 patients) or RT plus HDT/ASCR. At 5 years, the event-free and overall survival rates for the transplant group were 46% and 53%, respectively, compared with 12% and 32% for the nontransplant group.
The efficacy of second-line therapy is predicted by the second-line age-adjusted IPI. Furthermore, pretransplantation PET scans have been identified as predictive factors after HDT/ASCR.314–318 PET positivity before transplant and chemoresistance are also associated with a poor outcome.319,320 The results of the studies from the GEL-TAMO group and Autologous Blood and Bone Marrow Transplant (ABMTR) showed that HDT/ASCR should be considered for patients who never experience a complete remission but are still chemotherapy-sensitive.321–323
Several chemotherapy regimens have been used as second-line therapy before HDT/ASCR.324–328 However, none of these has emerged as a preferred regimen. Rituximab as a single agent was modestly active in patients with relapsed or refractory DLBCL and is reserved for frail, elderly patients.329 In a phase II study, rituximab in combination with ifosfamide, carboplatin, and etoposide (R-ICE) produced a complete response rate of 53% in patients with relapsed or refractory DLBCL, which is significantly better in historical comparison with the response rates observed for these patients treated with ICE alone (27%).330 In an outpatient setting, R-ICE produced an overall response rate of 71% (25% complete response, and 46% partial response) and an estimated 1-year event-free survival rate of 60% in patients with refractory B-cell lymphoma.331 Rituximab with other regimens has also been effective in patients with relapsed or refractory DLBCL.332–338
An international Intergroup study (CORAL study) compared R-ICE and DHAP (dexamethasone, cisplatin, and cytarabine; R-DHAP) followed by ASCR in patients with relapsed DLBCL.339 No significant differences were seen in the 3-year event-free (26% vs. 35%) and overall survival rates (47% vs. 51%) between R-ICE and R-DHAP. However, patients refractory to upfront rituximab-based chemotherapy or those with early relapse have a poor response rate and prognosis. Similar findings were reported in the retrospective analysis conducted by the GEL-TAMO study group.340
Lenalidomide is active in heavily pretreated patients with relapsed or refractory DLBCL. In a subset analysis of the NHL-003 study, which evaluated lenalidomide in patients with relapsed or refractory aggressive NHL among 103 patients with DLBCL, an overall response rate of 30% was seen (7% with a complete response and 23% with a partial response).341 Recent data suggest that the activity of lenalidomide in DLBCL is restricted to the activated B-cell phenotype.342
NCCN Recommendations: HDT/ASCR is the preferred treatment for patients with relapsed or refractory disease. These patients who are candidates for HDT/ASCR should be treated with second-line chemotherapy with or without rituximab. Suggested regimens (with or without rituximab) include the following:
-
DHAP (dexamethasone, cisplatin, and cytarabine)
-
ESHAP (methylprednisolone, etoposide, cytarabine, and cisplatin)
-
GDP (gemcitabine, dexamethasone, and cisplatin)
-
GemOX (gemcitabine and oxaliplatin)
-
ICE (ifosfamide, carboplatin, and etoposide)
-
MINE (mitoxantrone, ifosfamide, mesna, and etoposide)
Patients with a complete or partial response to a second-line chemotherapy regimen should be considered for further consolidation with HDT/ASCR (category 1) with or without RT. IFRT before HDT/ASCR has been shown to result in good local control and an improved outcome.343 Additional RT can be given before or after stem cell rescue to sites with prior positive disease. Pertinent clinical trials that include the option of ASCR is another option.
Patients who are not eligible for HDT/ASCR should be treated in the context of a clinical trial. Alternatively, they can be treated with single-agent rituximab or lenalidomide (in patients with nongerminal center DLBCL) or multiagent chemotherapy regimens (with or without rituximab), such as dose-adjusted EPOCH,344 CEPP,301 GDP, (gemcitabine, dexamethasone, and cisplatin) or GemOx.
Patients with disease relapse after HDT/ASCR should be treated in the context of a clinical trial or individually. However, those with progressive disease after 3 successive regimens are unlikely to derive additional benefit from currently available chemotherapy regimens, except for those with a long disease-free interval.
PMBL
PMBL is a distinct subtype of NHL that can be histologically indistinguishable from DLBCL. It tends to occur in young adults with a median age of 35 years, with a female predominance.345 PMBL arises from thymic B cells, with initial locoregional spread to supraclavicular, cervical, and hilar nodes and into the mediastinum and lung.345 Widespread metastatic disease is uncommon at initial diagnosis, but can be more common at recurrence.346 Clinical symptoms related to rapid growth of mediastinal mass include superior vena cava syndrome, and pericardial and pleural effusions.
Gene expression profiling has indicated that PMBL is distinct from DLBCL; the pattern of gene expression in PMBL is more similar to classical Hodgkin lymphoma.347,348 PMBL expresses B-cell antigens and lacks surface immunoglobulins. PMBL is CD19+, CD20+, CD22+, CD21–, IRF4/MUM1+, and CD23+, with variable expression of BCL2 and BCL6. CD30 is weakly and heterogeneously expressed in more than 80% of cases, and CD15 is occasionally present.346 CD10 positivity is seen in 8% to 32% of cases. PMBL is also characterized by a low expression of human leukocyte antigen class I or II molecules. Rare cases of mediastinal gray zone lymphomas with combined features of PMBL and classical Hodgkin lymphoma have been reported. Cytogenetic and oncogene abnormalities that are common in PMBL include gains in chromosome 9p24 (involving JAK2 in 50%–75% of patients) and chromosome 2p15 (involving c-REL, encoding a member of the NF-κB family of transcription factors), and losses in chromosomes 1p, 3p, 13q, 15q, and 17p.346 The IPI is of limited value in determining the prognosis of PMBL at diagnosis.345,349 In a retrospective analysis of 141 patients from MSKCC, 2 or more extranodal sites and initial therapy received were predictors of event-free survival, whereas only the initial therapy received was a predictor for overall survival.349
In retrospective analyses, intensive chemotherapy regimens have been more effective than CHOP,350–352 and the addition of IFRT improved outcomes.353,354 The role of RT must be confirmed in randomized trials. In a retrospective study, the addition of rituximab to MACOP-B (methotrexate, doxorubicin, cyclophosphamide, vincristine, prednisone, and bleomycin) or VACOP-B (etoposide, doxorubicin, cyclophosphamide, vincristine, prednisone, and bleomycin) did not seem to result in significant differences in clinical outcomes.350,355 Larger studies are needed to confirm this, especially in light of 2 different studies reporting that the addition of rituximab to CHOP plus RT356 or dose-adjusted EPOCH without RT357 improved response rates and event-free survival rates. Sequential dose-dense R-CHOP followed by ICE consolidation (without RT) was also highly effective in patients with PMBL. At a median follow-up for surviving patients at 3 years, the overall and progression-free survival rates were 88% and 78%, respectively.358
However, in the absence of randomized trials, no optimal treatment has been established for patients with PMBL. R-CHOP-21 is widely used at NCCN Member Institutions based on data in DLBCL. PET-CT is essential posttreatment. If PET-CT is negative at the end of treatment, patients may be observed. Residual mediastinal masses are common. If PET-CT is positive, biopsy is recommended if additional treatment is contemplated.
Cutaneous B-Cell Lymphomas
Cutaneous B-cell lymphomas (CBCLs) are a group of B-cell lymphomas originating in and usually confined to the skin. CBCLs are estimated to represent approximately 20% to 25% of all primary cutaneous lymphomas.359,360 In the United States, the SEER data from the NCI indicated that the incidence of cutaneous T-cell lymphomas accounted for 71%, whereas CBCLs accounted for 29% from 2001 to 2005.361 The new WHO-EORTC classification for cutaneous lymphomas distinguishes 3 main types of CBCL360:
-
Primary cutaneous marginal zone B-cell lymphoma (PCMZL)
-
PCFCL
-
PCDLBCL-LT
PCFCL is the most common type of CBCL, whereas PCDLBCL-LT is rare. PCMZL and PCFCL are indolent or slow-growing, whereas PCDLBCL-LT is an aggressive lymphoma with a generally poorer prognosis.362,363 In an Italian series of 467 patients with CBCL, PCFCL and PCMZL accounted for 57% and 31% of cases, respectively. PCDLBCL-LT was reported in only 11% of patients.363 Although the various types of CBCL can occur anywhere on the skin, PCFCL is more prevalent on the scalp and forehead, whereas the trunk and extremities are the most common sites for PCMZL. The leg remains the most common site for PCDLBCL. Extracutaneous involvement is more frequent with PCDLBCL-LT. In the same large Italian series, extracutaneous involvement eventually developed in 6% of patients with PCMZL, 11% with PCFCL, and 17% with PCDLBCL-LT. In patients with PCMZL and PCFCL, patients with single lesions had higher disease-free and overall survival rates than those with regional or disseminated lesions (62% vs. 44% and 97% vs. 85%, respectively), whereas in patients with PCDLBCL-LT the differences were only of borderline significance (55% vs. 44% and 79% vs. 67%, respectively).363 In another series of 145 patients with CBCL, Grange et al.364 also identified locations on the leg and multiple skin lesions as independent poor prognostic factors for patients with CBCLs.
Diagnosis
Adequate biopsy of the lesions should be obtained and the slides reviewed by a pathologist with expertise in the diagnosis of primary CBCLs (PCBCLs). Incisional, excisional, or punch biopsy is preferred to shave biopsy, because CBCLs have primarily dermal infiltrates, often deep, which are less well sampled and can even be missed with a shave biopsy. Adequate immunophenotyping with a panel that evaluates B- and T-cell markers is recommended to establish the diagnosis of the exact subtype of CBCL. The panel should include CD20, CD79a, CD3, CD5, CD10, BCL2, BCL6, Ki-67, kappa/lambda, and IRF4/MUM1. PCFCL is consistently BCL6+, whereas CD10 and BCL2 are expressed in only a few cases with a follicular growth pattern. PCMZLs are always negative for BCL6 and CD10, but are often BCL2+.365
Although the diagnosis of PCMZL is generally straightforward and reproducible among pathologists, it is more difficult to distinguish from PCFCL and PCDLBCL-LT. Part of the difficulty is that cell size (large vs. small) is not a defining feature like it is in nodal B-cell lymphomas. Most of the patients with PCFCL have lesions with a germinal center phenotype, whereas most with PCDLBCL-LT have an activated B-cell phenotype.366 In nodal DLBCL, the germinal center phenotype is associated with a better prognosis than the activated B-cell phenotype. Both PCFCL and PCDLBCL are CD20+ and BCL6+. BCL2 is usually negative in PCFCL but highly expressed in PCDLBCL-LT. In addition, PCFCL is usually MUM/IRF4–, whereas PCDLBCL-LT is usually IRF4/MUM1+ and shows strong expression of FOXP1.367 IRF4/MUM1 and FOXP1 may serve as additional diagnostic markers in the differential diagnosis of PCFCL and PCDLBCL. Assessment of surface IgM and IgD expression may be helpful in distinguishing PCDLBCL-LT from PCFCL.368
The t(14;18) translocation occurs only rarely in PCBCLs. Therefore, detection of a t(14;18) translocation in CBCL suggests the presence of systemic disease.369 Molecular genetic analysis to detect TCR gene rearrangements, PCR to detect IGHV rearrangements, and cytogenetics or FISH to detect t(14;18) may be useful in selected circumstances. In selected cases, the use of cyclin D1 may be useful to differentiate PCMZL (negative for CD5 and cyclin D1) from MCL (positive for CD5 and cyclin D1). MCL is not a primary cutaneous lymphoma, and finding it in the skin requires a careful search for extracutaneous disease.
Workup
Initial workup is geared toward evaluating extent of disease on the skin and seeking extracutaneous disease. The absence of extracutaneous disease at diagnosis is part of the definition of PCBCL. The initial workup includes a complete physical examination, comprehensive skin examination, and CT scans of the chest, abdomen, and pelvis. PET-CT may have higher sensitivity in finding otherwise occult systemic disease, but this is not validated and the higher rates of false-positive findings can create confusion. Bone marrow biopsy is essential for PCDLBCL-LT, whereas its role is unclear for PCFCL and PCMZL. Senff et al.30 evaluated 275 patients with histologic features consistent with MZL (n = 82) or FCL (n = 193) first presenting in the skin.30 Bone marrow involvement was seen in approximately 11% of patients in the FCL group compared with less than 1% in the MZL group. Patients with follicle center lymphoma (FCL) and skin lesions and positive bone marrow biopsy results had a significantly worse prognosis than those with PCFCL. The 5-year overall survival rate was 44% and 84%, respectively.
The International Society of Cutaneous Lymphomas (ISCL) and the EORTC task force recommend that bone marrow biopsy be required for cutaneous lymphomas with intermediate to aggressive behaviors, and should be considered for cutaneous lymphomas with indolent behavior in the presence of any evidence of extracutaneous disease, as indicated by other staging assessments (e.g., radiographic evidence or serologic clues, such as elevated monoclonal or polyclonal immunoglobulins).29 The guidelines recommend considering bone marrow biopsy for patients with PCFCL. It is optional for patients with PCMZL. Peripheral blood flow cytometry will be useful in selected cases when CBC shows lymphocytosis.
Treatment
Primary CBCLs have a different clinical course and prognosis that distinguish them from their nodal counterparts. Treatment options for CBCLs depend on the histology and stage of the disease. Most commonly used therapies include excision, RT, rituximab, or systemic chemotherapy.359,360
In a large retrospective analysis by the Italian Study Group for Cutaneous Lymphomas involving 467 patients with PCBCL, the complete response, 5-year overall survival, and 10-year overall survival rates for all patients with PCFCL and PCMZL who received treatment were 90%, 96%, and 90%, respectively.363 The relapse rate was 44% and extracutaneous spread was observed in 6% to 11% of patients. Relapse rate did not vary by type of initial therapy.
In patients with PCDLBCL-LT, the complete response, 5-year overall survival, and 10-year overall survival rates were 82%, 73%, and 47%, respectively. PCDLBCL-LT is also associated with higher relapse rates (55%) and incidences of extracutaneous spread (17%). Higher relapse rates were confirmed for patients with single or regional lesions treated with RT and those with disseminated cutaneous involvement treated with chemotherapy as first-line treatment.
RT is very effective when used as initial local therapy and for cutaneous relapses in most patients with indolent CBCLs.370–372 In patients with indolent histologies, RT and excision were associated with higher response rates than chemotherapy (96%, 97%, and 79%, respectively) but are generally used for those with more limited disease.363 However, most patients with regional or disseminated disease will experience relapse after any type of treatment. Relapses are generally confined to the skin.
In a retrospective of 34 patients with CBCL treated with RT, the 5-year relapse-free survival rate ranged from 62% to 73% for PCFCL and PCMZL but was only 33% for PCDLBCL-LT.372 Five-year overall survival was 100% for PCFCL and PCMZL but was 67% for PCDLBCL-LT. Senff et al.371 evaluated the outcome of 153 patients with CBCL (25 with PCMZL, 101 with PCFCL, and 27 with PCDLBCL) who were initially treated with RT with a curative intent. Overall, 45% of patients had single lesions, and localized or disseminated lesions were seen in 43% and 12% of patients, respectively. Complete remission was experienced by 151 of 153 patients (99%). Relapse rates for PCMZL, PCFCL, and PCDLBCL-LT were 60%, 29%, and 64%, and the 5-year disease-specific survival rates were 95%, 97%, and 59%, respectively. The PCFCLs presenting on the legs had a higher relapse rate (63%) and a lower 5-year disease-specific survival rate (44%) than PCFCLs at other sites (25% and 99%, respectively).371
Thus, local therapy is suitable for patients with indolent histologies, whereas patients with PCDLBCL-LT, which has a more unfavorable clinical course, are generally treated with more aggressive treatment modalities, often combined modality therapy.373
NCCN Recommendations
Because of the lack of data from randomized clinical trials, the treatment recommendations included in the NCCN Guidelines are derived from the management of patients with CBCL at NCCN Member Institutions based on the limited data from retrospective analyses and studies involving small cohorts of patients.
PCFCL and PCMZL: Initial Treatment: The guidelines recommend local RT or excision as the initial treatment options for patients with solitary lesions or regional disease (T1–2). Selected patients with local disease that is not amenable to local therapy (e.g., lesions on the scalp or forehead) can be observed.
For patients presenting with generalized skin lesions (T3), several treatment options are available. Chlorambucil has been shown to be effective in the treatment of PCMZL with multifocal skin lesions.374 In patients presenting with PCFCL, multiagent chemotherapy and RT were equally effective for multifocal skin lesions.375–377 Rituximab has been effective as a first-line treatment option for patients with indolent CBCLs with multiple lesions for which local therapy is not effective.378–382 In a series of 16 patients with PCBCL, 14 patients (87.5%) experienced complete remission, with 35% of these patients experiencing relapse between 6 and 37 months.382 In another retrospective analysis of 15 patients with indolent CBCLs, the overall objective response rate was 87% (60% with a complete response, 27% with a partial response) with a median follow-up of 36 months.381 The overall response rate was 100% for patients with PCFCL and 60% for those with PCMZL. Median time to response, duration of response, and time to progression were 30 days, 24 months, and 24 months, respectively. There are case reports showing the efficacy of topical therapy with using steroids, imiquimod, and nitrogen mustard or bexarotene gel.375,383–386
For patients presenting with generalized disease, the guidelines have included observation, rituximab, topical therapy, local RT, intralesional steroids, or systemic therapy (chlorambucil or CVP) with or without rituximab as options. In patients with very extensive or symptomatic disease, other chemotherapy regimens recommended for the treatment of FL may be used.
Patients presenting with extracutaneous disease should be managed according to the FL guidelines.
Treatment for Relapsed or Refractory Disease: Although most patients respond to initial therapy, relapses do commonly occur. Patients with regional or localized relapse should undergo additional therapy (excision, intralesional steroids, local RT, or topical therapy using steroids, imiquimod, nitrogen mustard, or bexarotene gel), and those with generalized disease relapse confined to the skin should undergo additional therapy with treatment options recommended for generalized disease at presentation.
Patients with a partial response or persistent progressive disease after additional treatment should be treated with the other options included in the initial treatment to improve response before starting treatment for refractory disease. Patients with extracutaneous or cutaneous relapse that is not responding to any of the initial treatment options should be managed according to the FL guidelines.
PCDLBCL-LT: Initial Treatment: PCDLBCL-LT has a poorer prognosis than other CBCL, particularly in patients with multiple tumors on the legs. RT alone is less-often effective in patients with PCDLBCL. Although these lesions do respond to radiation, remissions are often short-lived, and higher rates of dissemination to extracutaneous sites occur. In a retrospective multicenter study from the French Study Group on 60 patients with PCDLBCL-LT, although no particular therapy (RT or multiagent chemotherapy with or without rituximab) was significantly associated with improved survival, patients treated with anthracycline-containing chemotherapy and rituximab had a more favorable short-term outcome.362 Among 12 patients treated with anthracycline-based chemotherapy with rituximab, the complete response rate was 92% compared with 62% among those who underwent other therapies. The 2-year survival rates for these groups were 81% and 59%, respectively.
For patients with localized disease, the NCCN Guidelines recommend local RT alone or in combination with R-CHOP. RT alone can be used in elderly patients or those unable to tolerate systemic therapy. In patients with generalized disease, R-CHOP with or without RT is recommended. Extracutaneous disease should be managed according to the DLBCL guidelines. The guidelines recommend enrollment in clinical trials for all patients with PCDLBCL-LT.
Treatment for Relapsed or Refractory Disease: In patients with regional relapses, R-CHOP is recommended if they have not received prior chemotherapy. Patients who have received prior chemotherapy should be treated with local RT or second-line chemotherapy regimens recommended for relapsed or refractory DLBCL. Local RT or second-line chemotherapy regimens recommended for relapsed or refractory DLBCL are options for patients with generalized relapse. In a pilot study of 10 patients, radioimmunotherapy with yttrium 90Y ibritumomab tiuxetan was effective in patients with relapsed CBCLs.387 The guidelines have included radioimmunotherapy as one of the treatment options for patients with relapsed disease.
Individual Disclosures for the NCCN Guidelines Panel for Non-Hodgkin's Lymphomas


Please Note
The NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines™) are a statement of consensus of the authors regarding their views of currently accepted approaches to treatment. Any clinician seeking to apply or consult the NCCN Guidelines™ is expected to use independent medical judgment in the context of individual clinical circumstances to determine any patient's care or treatment. The National Comprehensive Cancer Network® (NCCN®) makes no representation or warranties of any kind regarding their content, use, or application and disclaims any responsibility for their applications or use in any way.
© National Comprehensive Cancer Network, Inc. 2011, All rights reserved. The NCCN Guidelines and the illustrations herein may not be reproduced in any form without the express written permission of NCCN.
Disclosures for the NCCN Guidelines Panel for Non-Hodgkin's Lymphomas
At the beginning of each NCCN Guidelines panel meeting, panel members disclosed any financial support they have received from industry. Through 2008, this information was published in an aggregate statement in JNCCN and online. Furthering NCCN's commitment to public transparency, this disclosure process has now been expanded by listing all potential conflicts of interest respective to each individual expert panel member.
Individual disclosures for the NCCN Guidelines for Non-Hodgkin's Lymphomas panel members can be found on page 560. (The most recent version of these guidelines and accompanying disclosures, including levels of compensation, are available on the NCCN Web site at www.NCCN.org.)
These guidelines are also available on the Internet. For the latest update, please visit www.NCCN.org.
References
- 2↑
Groves FD, Linet MS, Travis LB, Devesa SS. Cancer surveillance series: non-Hodgkin’s lymphoma incidence by histologic subtype in the United States from 1978 through 1995. J Natl Cancer Inst 2000;92:1240–1251.
- 3↑
Rappaport H. Tumors of the hematopoietic system. In: Atlas of Tumor Pathology Series (ed I). Washington, DC: Armed Forces Institute of Pathology; 1966.
- 4↑
Hicks EB, Rappaport H, Winter WJ. Follicular lymphoma; a reevaluation of its position in the scheme of malignant lymphoma, based on a survey of 253 cases. Cancer 1956;9:792–821.
- 5↑
Bennetta MH, Farrer-Brown G, Henry K et al.. Classification of non-Hodgkin’s lymphomas. Lancet 1974;2:405–408.
- 6
Lennert K. Malignant lymphomas other than Hodgkin’s disease. New York: Springer-Verlag; 1978.
- 7↑
Lennert K, Feller A. Histopathology of non-Hodgkin’s lymphomas, 2nd ed. Berlin, Germany: Springer-Verlag; 1992.
- 8↑
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 1982;49:2112–2135.
- 9↑
Classification of non-Hodgkin’s lymphomas. Reproducibility of major classification systems. NCI non-Hodgkin’s Classification Project Writing Committee. Cancer 1985;55:91–95.
- 10↑
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 1994;84:1361–1392.
- 11↑
A clinical evaluation of the International Lymphoma Study Group classification of non-Hodgkin’s lymphoma. The Non-Hodgkin’s Lymphoma Classification Project. Blood 1997;89:3909–3918.
- 12↑
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 1998;16:2780–2795.
- 13↑
Harris NL, Jaffe ES, Diebold J et al.. World Health Organization classification of neoplastic diseases of the hematopoietic and lymphoid tissues: report of the Clinical Advisory Committee meeting-Airlie House, Virginia, November 1997. J Clin Oncol 1999;17:3835–3849.
- 14↑
Jaffe ES, Harris NL, Stein H, Vardiman JW. WHO classification of tumours. Pathology and genetics of tumours of haematopoietic and lymphoid tissues. Lyon, France: IARC; 2001.
- 15↑
Vose J, Armitage J, Weisenburger D. International peripheral T-cell and natural killer/T-cell lymphoma study: pathology findings and clinical outcomes. J Clin Oncol 2008;26:4124–4130.
- 16↑
Jaffe ES, Harris NL, Stein H, Isaacson PG. Classification of lymphoid neoplasms: the microscope as a tool for disease discovery. Blood 2008;112:4384–4399.
- 17↑
Swerdlow SH, Campo E, Harris NL et al.. WHO classification of tumours of haematopoietic and lymphoid tissues. Lyon, France: IARC; 2008.
- 18↑
Cheson BD, Horning SJ, Coiffier B et al.. Report of an international workshop to standardize response criteria for non-Hodgkin’s lymphomas. J Clin Oncol 1999;17:1244–1253.
- 19↑
Cheson BD, Pfistner B, Juweid ME et al.. Revised response criteria for malignant lymphoma. J Clin Oncol 2007;25:579–586.
- 20↑
Hehn ST, Grogan TM, Miller TP. Utility of fine-needle aspiration as a diagnostic technique in lymphoma. J Clin Oncol 2004;22:3046–3052.
- 21↑
Meda BA, Buss DH, Woodruff RD et al.. Diagnosis and subclassification of primary and recurrent lymphoma. The usefulness and limitations of combined fine-needle aspiration cytomorphology and flow cytometry. Am J Clin Pathol 2000;113:688–699.
- 22↑
Dong HY, Harris NL, Preffer FI, Pitman MB. Fine-needle aspiration biopsy in the diagnosis and classification of primary and recurrent lymphoma: a retrospective analysis of the utility of cytomorphology and flow cytometry. Mod Pathol 2001;14:472–481.
- 23
Jeffers MD, Milton J, Herriot R, McKean M. Fine needle aspiration cytology in the investigation on non-Hodgkin’s lymphoma. J Clin Pathol 1998;51:189–196.
- 24↑
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 2004;102:55–65.
- 25↑
Dunphy CH. Applications of flow cytometry and immunohistochemistry to diagnostic hematopathology. Arch Pathol Lab Med 2004;128:1004–1022.
- 26↑
Howell SJ, Shalet SM. Fertility preservation and management of gonadal failure associated with lymphoma therapy. Curr Oncol Rep 2002;4:443–452.
- 27↑
Conlan MG, Bast M, Armitage JO, Weisenburger DD. Bone marrow involvement by non-Hodgkin’s lymphoma: the clinical significance of morphologic discordance between the lymph node and bone marrow. Nebraska Lymphoma Study Group. J Clin Oncol 1990;8:1163–1172.
- 28↑
Lim ST, Tao M, Cheung YB et al.. Can patients with early-stage diffuse large B-cell lymphoma be treated without bone marrow biopsy? Ann Oncol 2005;16:215–218.
- 29↑
Kim YH, Willemze R, Pimpinelli N et al.. TNM classification system for primary cutaneous lymphomas other than mycosis fungoides and Sezary syndrome: a proposal of the International Society for Cutaneous Lymphomas (ISCL) and the Cutaneous Lymphoma Task Force of the European Organization of Research and Treatment of Cancer (EORTC). Blood 2007;110:479–484.
- 30↑
Senff N, Kluin-Nelemans H, Willemze R. Results of bone marrow examination in 275 patients with histological features that suggest an indolent type of cutaneous B-cell lymphoma. Br J Haematol 2008;142:52–56.
- 31↑
Juneja SK, Wolf MM, Cooper IA. Value of bilateral bone marrow biopsy specimens in non-Hodgkin’s lymphoma. J Clin Pathol 1990;43:630–632.
- 32↑
Seam P, Juweid ME, Cheson BD. The role of FDG-PET scans in patients with lymphoma. Blood 2007;110:3507–3516.
- 33↑
Isasi CR, Lu P, Blaufox MD. A metaanalysis of 18F-2-deoxy-2-fluoro-D-glucose positron emission tomography in the staging and restaging of patients with lymphoma. Cancer 2005;104:1066–1074.
- 34↑
Trotman J, Fournier M, Lamy T et al.. Result of FDG PET-CT imaging after immunochemotherapy induction is a powerful and independent prognostic indicator of outcome for patients with follicular lymphoma: an analysis from the PRIMA study [abstract]. Blood 2010;116:Abstract 855.
- 35↑
Feeney J, Horwitz S, Gonen M, Schoder H. Characterization of T-cell lymphomas by FDG PET/CT. AJR Am J Roentgenol 2010;195:333–340.
- 36↑
Hoffmann M, Kletter K, Becherer A et al.. 18F-fluorodeoxyglucose positron emission tomography (18F-FDG-PET) for staging and follow-up of marginal zone B-cell lymphoma. Oncology 2003;64:336–340.
- 37↑
Rodriguez-Vigil B, Gomez-Leon N, Pinilla I et al.. PET/CT in lymphoma: prospective study of enhanced full-dose PET/CT versus unenhanced low-dose PET/CT. J Nucl Med 2006;47:1643–1648.
- 38↑
Schaefer NG, Hany TF, Taverna C et al.. Non-Hodgkin lymphoma and Hodgkin disease: coregistered FDG PET and CT at staging and restaging—do we need contrast-enhanced CT? Radiology 2004;232:823–829.
- 39↑
Lau GK. Hepatitis B reactivation after chemotherapy: two decades of clinical research. Hepatol Int 2008;2:152–162.
- 40↑
Ludwig E, Mendelsohn RB, Taur Y et al.. Prevalence of hepatitis B surface antigen and hepatitis B core antibody in a population initiating immunosuppressive therapy [abstract]. J Clin Oncol 2010;28:Abstract 9009.
- 41↑
Hwang J, Fisch M, Zhang H et al.. Hepatitis B screening and positivity prior to chemotherapy [abstract]. J Clin Oncol 2010;28:Abstract 9008.
- 42↑
Lok AS, Liang RH, Chiu EK et al.. Reactivation of hepatitis B virus replication in patients receiving cytotoxic therapy. Report of a prospective study. Gastroenterology 1991;100:182–188.
- 43↑
Lalazar G, Rund D, Shouval D. Screening, prevention and treatment of viral hepatitis B reactivation in patients with haematological malignancies. Br J Haematol 2007;136:699–712.
- 44↑
Yeo W, Chan PK, Zhong S et al.. Frequency of hepatitis B virus reactivation in cancer patients undergoing cytotoxic chemotherapy: a prospective study of 626 patients with identification of risk factors. J Med Virol 2000;62:299–307.
- 45↑
Targhetta C, Cabras MG, Mamusa AM et al.. Hepatitis B virus-related liver disease in isolated anti-hepatitis B-core positive lymphoma patients receiving chemo- or chemo-immune therapy. Haematologica 2008;93:951–952.
- 46↑
Loomba R, Rowley A, Wesley R et al.. Systematic review: the effect of preventive lamivudine on hepatitis B reactivation during chemotherapy. Ann Intern Med 2008;148:519–528.
- 47
Tsutsumi Y, Kawamura T, Saitoh S et al.. Hepatitis B virus reactivation in a case of non-Hodgkin’s lymphoma treated with chemotherapy and rituximab: necessity of prophylaxis for hepatitis B virus reactivation in rituximab therapy. Leuk Lymphoma 2004;45:627–629.
- 48↑
Tsutsumi Y, Tanaka J, Kawamura T et al.. Possible efficacy of lamivudine treatment to prevent hepatitis B virus reactivation due to rituximab therapy in a patient with non-Hodgkin’s lymphoma. Ann Hematol 2004;83:58–60.
- 49↑
Lau GK, Yiu HH, Fong DY et al.. Early is superior to deferred preemptive lamivudine therapy for hepatitis B patients undergoing chemotherapy. Gastroenterology 2003;125:1742–1749.
- 50↑
Carson KR, Evens AM, Richey EA et al.. Progressive multifocal leukoencephalopathy after rituximab therapy in HIV-negative patients: a report of 57 cases from the Research on Adverse Drug Events and Reports project. Blood 2009;113:4834–4840.
- 51↑
Coiffier B, Altman A, Pui CH et al.. Guidelines for the management of pediatric and adult tumor lysis syndrome: an evidence-based review. J Clin Oncol 2008;26:2767–2778.
- 52↑
Cairo MS, Bishop M. Tumour lysis syndrome: new therapeutic strategies and classification. Br J Haematol 2004;127:3–11.
- 53↑
Krakoff IH, Meyer RL. Prevention of hyperuricemia in leukemia and lymphoma: use of alopurinol, a xanthine oxidase inhibitor. JAMA 1965;193:1–6.
- 54↑
Bosly A, Sonet A, Pinkerton CR et al.. Rasburicase (recombinant urate oxidase) for the management of hyperuricemia in patients with cancer: report of an international compassionate use study. Cancer 2003;98:1048–1054.
- 55↑
Coiffier B, Mounier N, Bologna S et al.. Efficacy and safety of rasburicase (recombinant urate oxidase) for the prevention and treatment of hyperuricemia during induction chemotherapy of aggressive non-Hodgkin’s lymphoma: results of the GRAAL1 (Groupe d’Etude des Lymphomes de l’Adulte Trial on Rasburicase Activity in Adult Lymphoma) study. J Clin Oncol 2003;21:4402–4406.
- 56↑
Cortes J, Moore JO, Maziarz RT et al.. Control of plasma uric acid in adults at risk for tumor lysis syndrome: efficacy and safety of rasburicase alone and rasburicase followed by allopurinol compared with allopurinol alone-results of a multicenter phase III study. J Clin Oncol 2010;28:4207–4213.
- 57↑
Tsimberidou AM, Wen S, O’Brien S et al.. Assessment of chronic lymphocytic leukemia and small lymphocytic lymphoma by absolute lymphocyte counts in 2,126 patients: 20 years of experience at the University of Texas M.D. Anderson Cancer Center. J Clin Oncol 2007;25:4648–4656.
- 58↑
Dohner H, Stilgenbauer S, Benner A et al.. Genomic aberrations and survival in chronic lymphocytic leukemia. N Engl J Med 2000;343:1910–1916.
- 59↑
Neilson JR, Auer R, White D et al.. Deletions at 11q identify a subset of patients with typical CLL who show consistent disease progression and reduced survival. Leukemia 1997;11:1929–1932.
- 60↑
Austen B, Skowronska A, Baker C et al.. Mutation status of the residual ATM allele is an important determinant of the cellular response to chemotherapy and survival in patients with chronic lymphocytic leukemia containing an 11q deletion. J Clin Oncol 2007;25:5448–5457.
- 61↑
Tsimberidou AM, Tam C, Abruzzo LV et al.. Chemoimmunotherapy may overcome the adverse prognostic significance of 11q deletion in previously untreated patients with chronic lymphocytic leukemia. Cancer 2009;115:373–380.
- 62↑
Rossi D, Cerri M, Deambrogi C et al.. The prognostic value of TP53 mutations in chronic lymphocytic leukemia is independent of Del17p13: implications for overall survival and chemorefractoriness. Clin Cancer Res 2009;15:995–1004.
- 63↑
Zenz T, Mohr J, Edelmann J et al.. Treatment resistance in chronic lymphocytic leukemia: the role of the p53 pathway. Leuk Lymphoma 2009;50:510–513.
- 64↑
Tam CS, Shanafelt TD, Wierda WG et al.. De novo deletion 17p13.1 chronic lymphocytic leukemia shows significant clinical heterogeneity: the M. D. Anderson and Mayo Clinic experience. Blood 2009;114:957–964.
- 65↑
Zenz T, Krober A, Scherer K et al.. Monoallelic TP53 inactivation is associated with poor prognosis in chronic lymphocytic leukemia: results from a detailed genetic characterization with long-term follow-up. Blood 2008;112:3322–3329.
- 66↑
Zenz T, Eichhorst B, Busch R et al.. TP53 mutation and survival in chronic lymphocytic leukemia. J Clin Oncol 2010;28:4473–4479.
- 67↑
Zenz T, Hoth P, Busch R et al.. TP53 mutations and outcome after fludarabine and cyclophosphamide (FC) or FC plus rituximab (FCR) in the CLL8 trial of the GCLLSG [abstract]. Blood 2009;114:Abstract 1267.
- 68↑
Byrd JC, Gribben JG, Peterson BL et al.. Select high-risk genetic features predict earlier progression following chemoimmunotherapy with fludarabine and rituximab in chronic lymphocytic leukemia: justification for risk-adapted therapy. J Clin Oncol 2006;24:437–443.
- 69↑
Grever MR, Lucas DM, Dewald GW et al.. Comprehensive assessment of genetic and molecular features predicting outcome in patients with chronic lymphocytic leukemia: results from the US Intergroup phase III trial E2997. J Clin Oncol 2007;25:799–804.
- 70↑
Catovsky D, Richards S, Matutes E et al.. Assessment of fludarabine plus cyclophosphamide for patients with chronic lymphocytic leukaemia (the LRF CLL4 Trial): a randomised controlled trial. Lancet 2007;370:230–239.
- 71↑
Moreno C, Montserrat E. New prognostic markers in chronic lymphocytic leukemia. Blood Rev 2008;22:211–219.
- 72↑
Tsimberidou AM, Tam C, Wierda W et al.. Beta-2 microglobulin (B2M) is an independent prognostic factor for clinical outcomes in patients with CLL treated with frontline fludarabine, cyclophosphamide, and rituximab (FCR) regardless of age, creatinine clearance (CrCl) [abstract]. J Clin Oncol 2007;25:Abstract 7034.
- 73↑
Hamblin TJ, Davis Z, Gardiner A et al.. Unmutated Ig V(H) genes are associated with a more aggressive form of chronic lymphocytic leukemia. Blood 1999;94:1848–1854.
- 74↑
Hamblin TJ, Davis ZA, Oscier DG. Determination of how many immunoglobulin variable region heavy chain mutations are allowable in unmutated chronic lymphocytic leukaemia—long-term follow up of patients with different percentages of mutations. Br J Haematol 2008;140:320–323.
- 75↑
Tobin G, Thunberg U, Johnson A et al.. Somatically mutated Ig V(H)3-21 genes characterize a new subset of chronic lymphocytic leukemia. Blood 2002;99:2262–2264.
- 76↑
Ghia P, Guida G, Stella S et al.. The pattern of CD38 expression defines a distinct subset of chronic lymphocytic leukemia (CLL) patients at risk of disease progression. Blood 2003;101:1262–1269.
- 77
Damle RN, Wasil T, Fais F et al.. Ig V gene mutation status and CD38 expression as novel prognostic indicators in chronic lymphocytic leukemia. Blood 1999;94:1840–1847.
- 78↑
Thornton PD, Fernandez C, Giustolisi GM et al.. CD38 expression as a prognostic indicator in chronic lymphocytic leukaemia. Hematol J 2004;5:145–151.
- 79↑
Del Principe MI, Del Poeta G, Buccisano F et al.. Clinical significance of ZAP-70 protein expression in B-cell chronic lymphocytic leukemia. Blood 2006;108:853–861.
- 80
Rassenti LZ, Huynh L, Toy TL et al.. ZAP-70 compared with immunoglobulin heavy-chain gene mutation status as a predictor of disease progression in chronic lymphocytic leukemia. N Engl J Med 2004;351:893–901.
- 81
Wiestner A, Rosenwald A, Barry TS et al.. ZAP-70 expression identifies a chronic lymphocytic leukemia subtype with unmutated immunoglobulin genes, inferior clinical outcome, and distinct gene expression profile. Blood 2003;101:4944–4951.
- 82↑
Crespo M, Bosch F, Villamor N et al.. ZAP-70 expression as a surrogate for immunoglobulin-variable-region mutations in chronic lymphocytic leukemia. N Engl J Med 2003;348:1764–1775.
- 83↑
D’Arena G, Tarnani M, Rumi C et al.. Prognostic significance of combined analysis of ZAP-70 and CD38 in chronic lymphocytic leukemia. Am J Hematol 2007;82:787–791.
- 84
Del Giudice I, Morilla A, Osuji N et al.. Zeta-chain associated protein 70 and CD38 combined predict the time to first treatment in patients with chronic lymphocytic leukemia. Cancer 2005;104:2124–2132.
- 85
Morilla A, Gonzalez de Castro D, Del Giudice I et al.. Combinations of ZAP-70, CD38 and IGHV mutational status as predictors of time to first treatment in CLL. Leuk Lymphoma 2008;49:2108–2115.
- 86↑
Schroers R, Griesinger F, Trumper L et al.. Combined analysis of ZAP-70 and CD38 expression as a predictor of disease progression in B-cell chronic lymphocytic leukemia. Leukemia 2005;19:750–758.
- 87↑
Wierda W, O’Brien S, Wang X et al.. Prognostic nomogram and index for overall survival in previously untreated patients with chronic lymphocytic leukemia. Blood 2007;109:4679–4685.
- 88↑
Rai KR, Sawitsky A, Cronkite EP et al.. Clinical staging of chronic lymphocytic leukemia. Blood 1975;46:219–234.
- 89↑
Binet J, Auquier A, Dighiero G et al.. A new prognostic classification of chronic lymphocytic leukemia derived from a multivariate survival analysis. Cancer 1981;48:198–206.
- 90↑
Rawstron AC. Monoclonal B-cell lymphocytosis. Hematology Am Soc Hematol Educ Program 2009:430–439.
- 91↑
Rawstron AC, Bennett FL, O’Connor SJ et al.. Monoclonal B-cell lymphocytosis and chronic lymphocytic leukemia. N Engl J Med 2008;359:575–583.
- 92↑
Dicker F, Schnittger S, Haferlach T et al.. Immunostimulatory oligonucleotide-induced metaphase cytogenetics detect chromosomal aberrations in 80% of CLL patients: a study of 132 CLL cases with correlation to FISH, IgVH status, and CD38 expression. Blood 2006;108:3152–3160.
- 93↑
Put N, Konings P, Rack K et al.. Improved detection of chromosomal abnormalities in chronic lymphocytic leukemia by conventional cytogenetics using CpG oligonucleotide and interleukin-2 stimulation: a Belgian multicentric study. Genes Chromosomes Cancer 2009;48:843–853.
- 94↑
Struski S, Gervais C, Helias C et al.. Stimulation of B-cell lymphoproliferations with CpG-oligonucleotide DSP30 plus IL-2 is more effective than with TPA to detect clonal abnormalities. Leukemia 2009;23:617–619.
- 95↑
Heerema NA, Byrd JC, Cin PD et al.. Karyotype results from CpG oligodeoxynucleotide stimulated chronic lymphocytic leukemia (CLL) cultures are consistent among laboratories: a CLL Research Consortium (CRC) study [abstract]. Blood 2009;114:Abstract 1614.
- 96↑
Cheson BD, Bennett JM, Grever M et al.. National Cancer Institute-sponsored Working Group guidelines for chronic lymphocytic leukemia: revised guidelines for diagnosis and treatment. Blood 1996;87:4990–4997.
- 97↑
Hallek M, Cheson BD, Catovsky D et al.. Guidelines for the diagnosis and treatment of chronic lymphocytic leukemia: a report from the International Workshop on Chronic Lymphocytic Leukemia updating the National Cancer Institute-Working Group 1996 guidelines. Blood 2008;111:5446–5456.
- 98↑
Raphael B, Andersen JW, Silber R et al.. Comparison of chlorambucil and prednisone versus cyclophosphamide, vincristine, and prednisone as initial treatment for chronic lymphocytic leukemia: long-term follow-up of an Eastern Cooperative Oncology Group randomized clinical trial. J Clin Oncol 1991;9:770–776.
- 99↑
Rai KR, Peterson BL, Appelbaum FR et al.. Fludarabine compared with chlorambucil as primary therapy for chronic lymphocytic leukemia. N Engl J Med 2000;343:1750–1757.
- 100↑
Eichhorst B, Goede V, Hallek M. Treatment of elderly patients with chronic lymphocytic leukemia. Leuk Lymphoma 2009;50:171–178.
- 101↑
Leporrier M, Chevret S, Cazin B et al.. Randomized comparison of fludarabine, CAP, and ChOP in 938 previously untreated stage B and C chronic lymphocytic leukemia patients. Blood 2001;98:2319–2325.
- 102↑
Flinn IW, Neuberg DS, Grever MR et al.. Phase III trial of fludarabine plus cyclophosphamide compared with fludarabine for patients with previously untreated chronic lymphocytic leukemia: US Intergroup Trial E2997. J Clin Oncol 2007;25:793–798.
- 103↑
Eichhorst BF, Busch R, Hopfinger G et al.. Fludarabine plus cyclophosphamide versus fludarabine alone in first-line therapy of younger patients with chronic lymphocytic leukemia. Blood 2006;107:885–891.
- 104↑
Byrd JC, Peterson BL, Morrison VA et al.. Randomized phase 2 study of fludarabine with concurrent versus sequential treatment with rituximab in symptomatic, untreated patients with B-cell chronic lymphocytic leukemia: results from Cancer and Leukemia Group B 9712 (CALGB 9712). Blood 2003;101:6–14.
- 105↑
Woyach JA, Ruppert AS, Heerema NA et al.. Chemoimmunotherapy with fludarabine and rituximab produces extended overall survival and progression-free survival in chronic lymphocytic leukemia: long-term follow-up of CALGB Study 9712. J Clin Oncol 2011;29:1349–1355.
- 106↑
Byrd JC, Rai K, Peterson BL et al.. Addition of rituximab to fludarabine may prolong progression-free survival and overall survival in patients with previously untreated chronic lymphocytic leukemia: an updated retrospective comparative analysis of CALGB 9712 and CALGB 9011. Blood 2005;105:49–53.
- 107↑
Keating MJ, O’Brien S, Albitar M et al.. Early results of a chemoimmunotherapy regimen of fludarabine, cyclophosphamide, and rituximab as initial therapy for chronic lymphocytic leukemia. J Clin Oncol 2005;23:4079–4088.
- 108↑
Tam CS, O’Brien S, Wierda W et al.. Long-term results of the fludarabine, cyclophosphamide, and rituximab regimen as initial therapy of chronic lymphocytic leukemia. Blood 2008;112:975–980.
- 109↑
Parikh SA, Wierda WG, Badoux X et al.. Comparison of fludarabine (F) plus cyclophosphamide (C) versus FC plus rituximab (R) in previously untreated Rai stage III/IV chronic lymphocytic leukemia (CLL) [abstract]. J Clin Oncol 2010;28(Suppl 1):Abstract 6519.
- 110↑
Hallek M, Fischer K, Fingerle-Rowson G et al.. Addition of rituximab to fludarabine and cyclophosphamide in patients with chronic lymphocytic leukaemia: a randomised, open-label, phase 3 trial. Lancet 2010;376:1164–1174.
- 111↑
Kay NE, Geyer SM, Call TG et al.. Combination chemoimmunotherapy with pentostatin, cyclophosphamide, and rituximab shows significant clinical activity with low accompanying toxicity in previously untreated B chronic lymphocytic leukemia. Blood 2007;109:405–411.
- 112↑
Kay NE, Wu W, Kabat B et al.. Pentostatin and rituximab therapy for previously untreated patients with B-cell chronic lymphocytic leukemia. Cancer 2010;116:2180–2187.
- 113↑
Knauf WU, Lissichkov T, Aldaoud A et al.. Phase III randomized study of bendamustine compared with chlorambucil in previously untreated patients with chronic lymphocytic leukemia. J Clin Oncol 2009;27:4378–4384.
- 114↑
Knauf WU, Lissitchkov T, Aldaoud A et al.. Bendamustine induces higher remission rates, prolongs progression free survival as well as time to next treatment, and improves overall survival for patients in complete remission without compromising quality of life when compared to chlorambucil in first line treatment of chronic lymphocytic leukemia [abstract]. Blood 2010;116:Abstract 2449.
- 115↑
Fischer K, Cramer P, Stilgenbauer S et al.. Bendamustine combined with rituximab (BR) in first-line therapy of advanced CLL: a multicenter phase II trial of the German CLL Study Group (GCLLSG) [abstract]. Blood 2009;114:Abstract 205.
- 116↑
Lundin J, Kimby E, Bjorkholm M et al.. Phase II trial of subcutaneous anti-CD52 monoclonal antibody alemtuzumab (Campath-1H) as first-line treatment for patients with B-cell chronic lymphocytic leukemia (B-CLL). Blood 2002;100:768–773.
- 117↑
Hillmen P, Skotnicki AB, Robak T et al.. Alemtuzumab compared with chlorambucil as first-line therapy for chronic lymphocytic leukemia. J Clin Oncol 2007;25:5616–5623.
- 118↑
Wierda W, O’Brien S, Wen S et al.. Chemoimmunotherapy with fludarabine, cyclophosphamide, and rituximab for relapsed and refractory chronic lymphocytic leukemia. J Clin Oncol 2005;23:4070–4078.
- 119↑
Robak T, Dmoszynska A, Solal-Celigny P et al.. Rituximab plus fludarabine and cyclophosphamide prolongs progression-free survival compared with fludarabine and cyclophosphamide alone in previously treated chronic lymphocytic leukemia. J Clin Oncol 2010;28:1756–1765.
- 120↑
Lamanna N, Kalaycio M, Maslak P et al.. Pentostatin, cyclophosphamide, and rituximab is an active, well-tolerated regimen for patients with previously treated chronic lymphocytic leukemia. J Clin Oncol 2006;24:1575–1581.
- 121↑
Weiss MA, Maslak PG, Jurcic JG et al.. Pentostatin and cyclophosphamide: an effective new regimen in previously treated patients with chronic lymphocytic leukemia. J Clin Oncol 2003;21:1278–1284.
- 122↑
Tsimberidou AM, Wierda WG, Plunkett W et al.. Phase I-II study of oxaliplatin, fludarabine, cytarabine, and rituximab combination therapy in patients with Richter’s syndrome or fludarabine-refractory chronic lymphocytic leukemia. J Clin Oncol 2008;26:196–203.
- 123↑
Tsimberidou AM, Wierda WG, Wen S et al.. Results of a phase I-II clinical trial of oxaliplatin, fludarabine, cytarabine, and rituximab (OFAR) combination therapy in patients with aggressive, relapsed/refractory chronic lymphocytic leukemia (CLL) and Richter syndrome (RS) [abstract]. Blood 2010;116:Abstract 923.
- 124↑
Fischer K, Stilgenbauer S, Schweighofer CD et al.. Bendamustine in combination with rituximab (BR) for patients with relapsed chronic lymphocytic leukemia (CLL): a multicentre phase II trial of the German CLL Study Group (GCLLSG) [abstract]. Blood 2008;112:Abstract 330.
- 125↑
Bowen DA, Call TG, Jenkins GD et al.. Methylprednisolone-rituximab is an effective salvage therapy for patients with relapsed chronic lymphocytic leukemia including those with unfavorable cytogenetic features. Leuk Lymphoma 2007;48:2412–2417.
- 126↑
Castro JE, James DF, Sandoval-Sus JD et al.. Rituximab in combination with high-dose methylprednisolone for the treatment of chronic lymphocytic leukemia. Leukemia 2009;23:1779–1789.
- 127↑
Dungarwalla M, Evans SO, Riley U et al.. High dose methylprednisolone and rituximab is an effective therapy in advanced refractory chronic lymphocytic leukemia resistant to fludarabine therapy. Haematologica 2008;93:475–476.
- 128↑
Keating MJ, Flinn I, Jain V et al.. Therapeutic role of alemtuzumab (Campath-1H) in patients who have failed fludarabine: results of a large international study. Blood 2002;99:3554–3561.
- 129↑
Stilgenbauer S, Dohner H. Campath-1H-induced complete remission of chronic lymphocytic leukemia despite p53 gene mutation and resistance to chemotherapy. N Engl J Med 2002;347:452–453.
- 130
Lozanski G, Heerema NA, Flinn IW et al.. Alemtuzumab is an effective therapy for chronic lymphocytic leukemia with p53 mutations and deletions. Blood 2004;103:3278–3281.
- 131↑
Osuji NC, Del Giudice I, Matutes E et al.. The efficacy of alemtuzumab for refractory chronic lymphocytic leukemia in relation to cytogenetic abnormalities of p53. Haematologica 2005;90:1435–1436.
- 132↑
Cortelezzi A, Pasquini MC, Sarina B et al.. A pilot study of low-dose subcutaneous alemtuzumab therapy for patients with chemotherapy-refractory chronic lymphocytic leukemia. Haematologica 2005;90:410–412.
- 133
Karlsson C, Lundin J, Kimby E et al.. Phase II study of subcutaneous alemtuzumab without dose escalation in patients with advanced-stage, relapsed chronic lymphocytic leukaemia. Br J Haematol 2009;144:78–85.
- 134
Stilgenbauer S, Zenz T, Winkler D et al.. Subcutaneous alemtuzumab in fludarabine-refractory chronic lymphocytic leukemia: clinical results and prognostic marker analyses from the CLL2H study of the German Chronic Lymphocytic Leukemia Study Group. J Clin Oncol 2009;27:3994–4001.
- 135↑
Varghese AM, Sayala HA, Moreton P et al.. Long term survival report of the UKCLL02 trial: a phase II study of subcutaneous alemtuzumab in patients with fludarabine refractory CLL (on behalf of the NCRI CLL Trials Sub-Group) [abstract]. Blood 2010;116:Abstract 922.
- 136↑
Fiegl M, Erdel M, Tinhofer I et al.. Clinical outcome of pretreated B-cell chronic lymphocytic leukemia following alemtuzumab therapy: a retrospective study on various cytogenetic risk categories. Ann Oncol 2010;21:2410–2419.
- 137↑
Elter T, Borchmann P, Schulz H et al.. Fludarabine in combination with alemtuzumab is effective and feasible in patients with relapsed or refractory B-cell chronic lymphocytic leukemia: results of a phase II trial. J Clin Oncol 2005;23:7024–7031.
- 138↑
Kennedy B, Rawstron A, Carter C et al.. Campath-1H and fludarabine in combination are highly active in refractory chronic lymphocytic leukemia. Blood 2002;99:2245–2247.
- 139↑
Faderl S, Thomas DA, O’Brien S et al.. Experience with alemtuzumab plus rituximab in patients with relapsed and refractory lymphoid malignancies. Blood 2003;101:3413–3415.
- 140↑
Nabhan C, Patton D, Gordon L et al.. A pilot trial of rituximab and alemtuzumab combination therapy in patients with relapsed and/or refractory chronic lymphocytic leukemia (CLL). Leuk Lymphoma 2004;45:2269–2273.
- 141↑
Badoux XC, Keating M, O’Brien S et al.. Chemoimmunotherapy with cyclophosphomide, fludarabine, alemtuzumab and rituximab (cfar) is effective in relapsed patients with chronic lymphocytic leukemia (CLL) [abstract]. Blood 2009;114:Abstract 3431.
- 142↑
Coiffier B, Lepretre S, Pedersen LM et al.. Safety and efficacy of ofatumumab, a fully human monoclonal anti-CD20 antibody, in patients with relapsed or refractory B-cell chronic lymphocytic leukemia: a phase 1-2 study. Blood 2008;111:1094–1100.
- 143↑
Wierda WG, Kipps TJ, Mayer J et al.. Ofatumumab as single-agent CD20 immunotherapy in fludarabine-refractory chronic lymphocytic leukemia. J Clin Oncol 2010;28:1749–1755.
- 144↑
Wierda WG, Kipps TJ, Mayer J et al.. Final analysis from the international trial of single-agent ofatumumab in patients with fludarabine-refractory chronic lymphocytic leukemia. Blood 2010;116:Abstract 921.
- 145↑
Gribben JG, Zahrieh D, Stephans K et al.. Autologous and allogeneic stem cell transplantations for poor-risk chronic lymphocytic leukemia. Blood 2005;106:4389–4396.
- 146
Khouri IF, Keating MJ, Saliba RM, Champlin RE. Long-term follow-up of patients with CLL treated with allogeneic hematopoietic transplantation. Cytotherapy 2002;4:217–221.
- 147
Sorror ML, Storer BE, Sandmaier BM et al.. Five-year follow-up of patients with advanced chronic lymphocytic leukemia treated with allogeneic hematopoietic cell transplantation after nonmyeloablative conditioning. J Clin Oncol 2008;26:4912–4920.
- 148
Khouri IF, Saliba RM, Admirand J et al.. Graft-versus-leukaemia effect after non-myeloablative haematopoietic transplantation can overcome the unfavourable expression of ZAP-70 in refractory chronic lymphocytic leukaemia. Br J Haematol 2007;137:355–363.
- 149
Moreno C, Villamor N, Colomer D et al.. Allogeneic stem-cell transplantation may overcome the adverse prognosis of unmutated VH gene in patients with chronic lymphocytic leukemia. J Clin Oncol 2005;23:3433–3438.
- 150↑
Schetelig J, van Biezen A, Brand R et al.. Allogeneic hematopoietic stem-cell transplantation for chronic lymphocytic leukemia with 17p deletion: a retrospective European Group for Blood and Marrow Transplantation analysis. J Clin Oncol 2008;26:5094–5100.
- 151↑
Dreger P, Stilgenbauer S, Boettcher S et al.. Prognostic factors for outcome of nonmyeloablative allogeneic stem cell transplantation (NST) in poor-risk chronic lymphocytic leukemia (CLL): final results from a prospective multicenter trial (GCLLSG CLL3X study) [abstract]. Blood 2008;112:Abstract 565.
- 152↑
Tsimberidou AM, Keating MJ. Treatment of fludarabine-refractory chronic lymphocytic leukemia. Cancer 2009;115:2824–2836.
- 153↑
Salvi F, Miller MD, Grilli A et al.. A manual of guidelines to score the modified cumulative illness rating scale and its validation in acute hospitalized elderly patients. J Am Geriatr Soc 2008;56:1926–1931.
- 154↑
Reynolds C, Di Bella N, Lyons RM et al.. Phase III trial of fludarabine, cyclophosphamide, and rituximab vs. pentostatin, cyclophosphamide, and rituximab in B-cell chronic lymphocytic leukemia. Blood 2008;112:Abstract 327.
- 155↑
Keating MJ, Wierda WG, Tam CS et al.. Long term outcome following treatment failure of FCR chemoimmunotherapy as initial therapy for chronic lymphocytic leukemia [abstract]. Blood 2009;114:Abstract 2381.
- 156↑
Rossi D, Gaidano G. Richter syndrome: molecular insights and clinical perspectives. Hematol Oncol 2009;27:1–10.
- 157
Tsimberidou AM, Keating MJ. Richter syndrome: biology, incidence, and therapeutic strategies. Cancer 2005;103:216–228.
- 158↑
Tsimberidou AM, O’Brien S, Kantarjian HM et al.. Hodgkin transformation of chronic lymphocytic leukemia: the M. D. Anderson Cancer Center experience. Cancer 2006;107:1294–1302.
- 159↑
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 2006;24:2343–2351.
- 160↑
Rodriguez J, Keating MJ, O’Brien S et al.. Allogeneic haematopoietic transplantation for Richter’s syndrome. Br J Haematol 2000;110:897–899.
- 161↑
Tsimberidou AM, Kantarjian HM, Cortes J et al.. Fractionated cyclophosphamide, vincristine, liposomal daunorubicin, and dexamethasone plus rituximab and granulocyte-macrophage-colony stimulating factor (GM-CSF) alternating with methotrexate and cytarabine plus rituximab and GM-CSF in patients with Richter syndrome or fludarabine-refractory chronic lymphocytic leukemia. Cancer 2003;97:1711–1720.
- 162↑
Morrison VA. Infectious complications of chronic lymphocytic leukaemia: pathogenesis, spectrum of infection, preventive approaches. Best Pract Res Clin Haematol 2010;23:145–153.
- 163↑
Tsai HT, Caporaso NE, Kyle RA et al.. Evidence of serum immunoglobulin abnormalities up to 9.8 years before diagnosis of chronic lymphocytic leukemia: a prospective study. Blood 2009;114:4928–4932.
- 164↑
Perkins JG, Flynn JM, Howard RS, Byrd JC. Frequency and type of serious infections in fludarabine-refractory B-cell chronic lymphocytic leukemia and small lymphocytic lymphoma. Cancer 2002;94:2033–2039.
- 165↑
Chapel H, Dicato M, Gamm H et al.. Immunoglobulin replacement in patients with chronic lymphocytic leukaemia: a comparison of two dose regimes. Br J Haematol 1994;88:209–212.
- 166
Intravenous immunoglobulin for the prevention of infection in chronic lymphocytic leukemia. A randomized, controlled clinical trial. Cooperative Group for the Study of Immunoglobulin in Chronic Lymphocytic Leukemia. N Engl J Med 1988;319:902–907.
- 167
Boughton BJ, Jackson N, Lim S, Smith N. Randomized trial of intravenous immunoglobulin prophylaxis for patients with chronic lymphocytic leukaemia and secondary hypogammaglobulinaemia. Clin Lab Haematol 1995;17:75–80.
- 168
Molica S, Musto P, Chiurazzi F et al.. Prophylaxis against infections with low-dose intravenous immunoglobulins (IVIG) in chronic lymphocytic leukemia. Results of a crossover study. Haematologica 1996;81:121–126.
- 169↑
Raanani P, Gafter-Gvili A, Paul M et al.. Immunoglobulin prophylaxis in chronic lymphocytic leukemia and multiple myeloma: systematic review and meta-analysis. Leuk Lymphoma 2009;50:764–772.
- 170↑
Sinisalo M, Vilpo J, Itala M et al.. Antibody response to 7-valent conjugated pneumococcal vaccine in patients with chronic lymphocytic leukaemia. Vaccine 2007;26:82–87.
- 171↑
Sinisalo M, Aittoniemi J, Kayhty H, Vilpo J. Vaccination against infections in chronic lymphocytic leukemia. Leuk Lymphoma 2003;44:649–652.
- 172↑
Van der Velden AM, Van Velzen-Blad H, Claessen AM et al.. The effect of ranitidine on antibody responses to polysaccharide vaccines in patients with B-cell chronic lymphocytic leukaemia. Eur J Haematol 2007;79:47–52.
- 173↑
Jurlander J, de Nully Brown P, Skov PS et al.. Improved vaccination response during ranitidine treatment, and increased plasma histamine concentrations, in patients with B cell chronic lymphocytic leukemia. Leukemia 1995;9:1902–1909.
- 174↑
Nguyen DD, Cao TM, Dugan K et al.. Cytomegalovirus viremia during Campath-1H therapy for relapsed and refractory chronic lymphocytic leukemia and prolymphocytic leukemia. Clin Lymphoma 2002;3:105–110.
- 175↑
Laurenti L, Piccioni P, Cattani P et al.. Cytomegalovirus reactivation during alemtuzumab therapy for chronic lymphocytic leukemia: incidence and treatment with oral ganciclovir. Haematologica 2004;89:1248–1252.
- 176↑
Visani G, Mele A, Guiducci B et al.. An observational study of once weekly intravenous ganciclovir as CMV prophylaxis in heavily pre-treated chronic lymphocytic leukemia patients receiving subcutaneous alemtuzumab. Leuk Lymphoma 2006;47:2542–2546.
- 177↑
O’Brien SM, Keating MJ, Mocarski ES. Updated guidelines on the management of cytomegalovirus reactivation in patients with chronic lymphocytic leukemia treated with alemtuzumab. Clin Lymphoma Myeloma 2006;7:125–130.
- 178↑
Dearden C. Disease-specific complications of chronic lymphocytic leukemia. Hematology Am Soc Hematol Educ Program 2008;2008:450–456.
- 179↑
Ding W, Zent CS. Diagnosis and management of autoimmune complications of chronic lymphocytic leukemia/small lymphocytic lymphoma. Clin Adv Hematol Oncol 2007;5:257–261.
- 180↑
Borthakur G, O’Brien S, Wierda WG et al.. Immune anaemias in patients with chronic lymphocytic leukaemia treated with fludarabine, cyclophosphamide and rituximab—incidence and predictors. Br J Haematol 2007;136:800–805.
- 181↑
Barcellini W, Capalbo S, Agostinelli R et al.. Relationship between autoimmune phenomena and disease stage and therapy in B-cell chronic lymphocytic leukemia. Haematologica 2006;91:1689–1692.
- 182
Zanotti R, Frattini F, Ghia P et al.. ZAP-70 expression is associated with increased risk of autoimmune cytopenias in CLL patients. Am J Hematol 2010;85:494–498.
- 183↑
Moreno C, Hodgson K, Ferrer G et al.. Autoimmune cytopenias in chronic lymphocytic leukemia: prevalence, clinical associations, and prognostic significance. Blood 2010;116:4771–4776.
- 184↑
Visco C, Ruggeri M, Laura Evangelista M et al.. Impact of immune thrombocytopenia on the clinical course of chronic lymphocytic leukemia. Blood 2008;111:1110–1116.
- 185↑
Cortes J, O’Brien S, Loscertales J et al.. Cyclosporin A for the treatment of cytopenia associated with chronic lymphocytic leukemia. Cancer 2001;92:2016–2022.
- 186↑
D’Arena G, Laurenti L, Capalbo S et al.. Rituximab therapy for chronic lymphocytic leukemia-associated autoimmune hemolytic anemia. Am J Hematol 2006;81:598–602.
- 187
Gupta N, Kavuru S, Patel D et al.. Rituximab-based chemotherapy for steroid-refractory autoimmune hemolytic anemia of chronic lymphocytic leukemia. Leukemia 2002;16:2092–2095.
- 188
Berentsen S. Rituximab for the treatment of autoimmune cytopenias. Haematologica 2007;92:1589–1596.
- 189
Godeau B, Porcher R, Fain O et al.. Rituximab efficacy and safety in adult splenectomy candidates with chronic immune thrombocytopenic purpura: results of a prospective multicenter phase 2 study. Blood 2008;112:999–1004.
- 190
Hegde UP, Wilson WH, White T, Cheson BD. Rituximab treatment of refractory fludarabine-associated immune thrombocytopenia in chronic lymphocytic leukemia. Blood 2002;100:2260–2262.
- 191
Shanafelt TD, Madueme HL, Wolf RC, Tefferi A. Rituximab for immune cytopenia in adults: idiopathic thrombocytopenic purpura, autoimmune hemolytic anemia, and Evans syndrome. Mayo Clin Proc 2003;78:1340–1346.
- 192↑
Ghazal H. Successful treatment of pure red cell aplasia with rituximab in patients with chronic lymphocytic leukemia. Blood 2002;99:1092–1094.
- 193↑
Bussel JB, Kuter DJ, Newland A et al.. Long-term efficacy and safety of romiplostim for the treatment of patients with chronic immune thrombocytopenia (ITP): 5-year update from an open-label extension study [abstract]. Blood 2009;114:Abstract 681.
- 194
Kuter DJ, Bussel JB, Lyons RM et al.. Efficacy of romiplostim in patients with chronic immune thrombocytopenic purpura: a double-blind randomised controlled trial. Lancet 2008;371:395–403.
- 195↑
Bussel JB, Cheng G, Saleh MN et al.. Eltrombopag for the treatment of chronic idiopathic thrombocytopenic purpura. N Engl J Med 2007;357:2237–2247.
- 196↑
Dearden C, Wade R, Else M et al.. The prognostic significance of a positive direct antiglobulin test in chronic lymphocytic leukemia: a beneficial effect of the combination of fludarabine and cyclophosphamide on the incidence of hemolytic anemia. Blood 2008;111:1820–1826.
- 197↑
Koster A, Tromp HA, Raemaekers JM et al.. The prognostic significance of the intra-follicular tumor cell proliferative rate in follicular lymphoma. Haematologica 2007;92:184–190.
- 198↑
Wang SA, Wang L, Hochberg EP et al.. Low histologic grade follicular lymphoma with high proliferation index: morphologic and clinical features. Am J Surg Pathol 2005;29:1490–1496.