B-Cell Lymphoma

Back

Practice Essentials

Non-Hodgkin lymphoma (NHL) is a collective term for a heterogeneous group of lymphoproliferative malignancies with differing patterns of behavior and responses to treatment.[1] NHL usually originates in the lymphoid tissues and can spread to other organs Most NHLs (80-85%) are of B-cell origin.

NHL must be distinguished from Hodgkin lymphoma with certainty before therapy is initiated. Compared with Hodgkin lymphoma, NHL is much less predictable and has a far greater predilection to disseminate to extranodal sites.The prognosis depends on the histologic type, stage, and treatment.

Background

NHL can be divided into two general prognostic groups: indolent lymphomas and aggressive lymphomas. Indolent lymphomas carry a relatively good prognosis, with median survival as long as 10 years, but they are not usually curable in advanced stages. Early-stage (stage I and II) indolent NHL can be treated effectively with radiation therapy alone. Most of the indolent types are nodular (or follicular) in morphology. The aggressive type of NHL has a shorter natural history, but a significant number of cases are curable with combination chemotherapy regimens.

With modern treatment of patients with NHL, the overall survival rate at 5 years is approximately 70%.[2] Most relapses occur in the first 2 years after therapy. The risk of late relapse is higher in patients with a divergent histology of both indolent and aggressive disease.

Whereas indolent NHL is responsive to radiation therapy and chemotherapy, a continuous rate of relapse is usually observed in advanced stages. However, patients can often be retreated with considerable success as long as the disease histology remains low grade.

Patients who present with aggressive forms of NHL, or whose disease converts to an aggressive form, may achieve complete remission with combination chemotherapy regimens, with or without aggressive high-dose consolidation therapy with marrow or stem cell support. Aggressive lymphomas are increasingly observed in patients who are HIV positive, and treatment of these patients requires special consideration.

Classification

B-cell and T/natural killer (NK)-cell neoplasms are clonal tumors of mature and immature B cells, T cells, or NK cells at various stages of differentiation. B-cell neoplasms tend to mimic stages of normal B-cell differentiation, and the resemblance to normal cell stages is a major basis for their classification and nomenclature.[3]

Per the 2008 revised World Health Organization (WHO) classification, B-cell malignancies are divided into two broad categories[3] : (1) precursor B-cell neoplasms, which include B lymphoblastic leukemia/lymphoma with or without recurrent genetic abnormalities, and (2) mature B-cell neoplasms.

The following classification deals with only the mature B-cell neoplasms. Both lymphomas and lymphoid leukemias are included in this classification because both solid and circulating phases are present in many lymphoid neoplasms and distinction between them is artificial. For instance, B-cell chronic lymphocytic leukemia (CLL) and B-cell small lymphocytic lymphoma (SLL) are different manifestations of the same neoplasm, as are lymphoblastic lymphomas and T-cell acute lymphocytic leukemias.

The WHO classification of mature B-cell neoplasms is as follows[3] :

Pathophysiology and Etiology

Any of the following may play a role in the etiology of non-Hodgkin lymphoma (NHL) in a particular patient:

Chromosomal translocations and molecular rearrangements

Non-random chromosomal and molecular rearrangements (see the table below) play an important role in the pathogenesis of many lymphomas and correlate with histology and immunophenotype.

Table 1. Chromosomal Abnormalities in B-Cell Non-Hodgkin Lymphoma



View Table

See Table

The most common chromosomal abnormality associated with NHL is the t(14;18)(q32;q21) translocation, which is found in 85% of follicular lymphomas and 25-30% of intermediate-grade NHLs. This translocation results in the juxtaposition of the bcl-2 apoptotic inhibitor oncogene at band 18q21 to the heavy-chain region of the immunoglobulin (Ig) locus within band 14q32, resulting in its overexpression.

The t(11;14)(q13;q32) translocation results in overexpression of bcl -1 (cyclin-D1/PRAD1), a cell cycle control gene on band 11q13, and is diagnostic of mantle cell lymphoma.

For the workup of any high-grade B-cell lymphoma of germinal center origin, fluorescence in situ hybridization (FISH) studies for evaluation of myc, bcl-6, and bcl-2 rearrangements should typically be ordered, in order to establish classification of "double hit" lymphomas. Double hit B-cell lymphomas are a specific subtype of diffuse large B-cell lymphoma (DLBCL) that have a much more aggressive clinical course and are associated with translocations of two of the three oncogenes (myc, bcl-2, and bcl-6).

Environmental factors

The risk of NHL is increased in certain workers, including the following:

Chemicals that have been linked to the development of NHL include the following:

Patients who receive cancer chemotherapy, radiation therapy, or both are at increased risk of developing NHL.

Viruses

Several viruses have been implicated in the pathogenesis of NHL. Epstein-Barr virus (EBV) is linked to Burkitt lymphoma (especially in endemic areas of Africa), sinonasal lymphoma in Asia and South America, and lymphomas in immunocompromised patients. Human T-lymphotropic virus 1 (HTLV-1) is linked to adult T-cell lymphoma/leukemia. Human herpesvirus 8 (HHV-8) is implicated i n body cavity–based lymphomas in patients with HIV infection.

Results of an Italian study support a role for chronic hepatitis C virus infection in NHL, and suggest involvement of hepatitis BV infection. Associations were clearest for B-cell NHL and diffuse large B-cell lymphoma.[4]

Immunodeficiency states

Immunodeficiency states that seem to predispose to NHL include congenital immunodeficiency states (eg, ataxia telangiectasia, Wiskott-Aldrich syndrome, common variable hypogammaglobulinemia, and severe combined immunodeficiency), as well as acquired immunodeficiency states (eg, HIV infection and iatrogenic immunosuppression for solid organ or bone marrow transplant recipients).[5]

Connective-tissue disorders

Connective-tissue disorders associated with an increased risk of NHL include the following:

Other disease states

Increased incidence of gastrointestinal (GI) lymphomas is observed in patients with celiac sprue and inflammatory bowel disease. Gastric mucosa-associated lymphoid tissue (MALT) lymphoma is observed most frequently, but not exclusively, in association with Helicobacter pylori infection.

Epidemiology

United States statistics

After a striking increase in incidence rates between 1970 and 1995 (which may in part have reflected improved diagnosis), the rates of new non-Hodgkin lymphoma (NHL) cases stabilzed. From 2007-2016, rates of new cases fell on average 0.9% each year and death rates fell on average 2.2% each year.[2]

The lifetime risk of being diagnosed with NHL is 2.1%. It has been estimated that in 2019, 74,200 new cases of NHL will be diagnosed, with 19,970 deaths. NHL is the seventh most common cancer in the US, accounting for 4.2% of new cancers and 3.3% of all cancer-related deaths.[2]

International statistics

Perry and colleagues identified the following significant differences in the epidemiology of NHL in developing countries, compared with the developed countries[6] :

In developing regions, diffuse large B-cell lymphoma was the most common subtype (42.5%) Other subtypes seen more frequently were Burkitt lymphoma (2.2%), precursor B- and T-lymphoblastic leukemia/lymphoma (1.1% and 2.9%) and extranodal NK/T-cell lymphoma (2.2%).[6]

In Europe, NHL is the 11th most common cancer, representing 3% of the total cancer diagnoses in 2012.[7]

Certain endemic geographical factors appear to influence the development of NHL in specific areas. For example, in equatorial Africa, a variant of Burkitt lymphoma associated with Epstein-Barr virus (EBV) is the most common childhood malignancy.

In the Middle East, heavy-chain disease (alpha) is a disorder of B-lymphoid cells that is characterized by diffuse thickening of the small intestine caused by a lymphoplasmacytic infiltrate with secretion of incomplete immunoglobulin A (IgA) heavy chains. This clinicopathologic entity is rarely encountered in individuals who are not of Mediterranean ethnicity.

Follicular lymphomas are more common in North America and Europe but are rare in the Caribbean, Africa, China, Japan, and the Middle East.

Age-, sex-, and race-associated differences in incidence

NHL is most frequently diagnosed in  persons 65-74 years old. High-grade lymphoblastic and small noncleaved-cell lymphomas are the only subtypes of B-cell NHL that are observed more commonly in children and young adults.

NHL is more common in men: the reported incidence is 23.7 cases per 100,000 population in men, compared with 16.0 cases per 100,000 population in women. However, the incidence of NHL in some anatomic sites, such as the thyroid, may be higher in women.[2]

The incidence of NHL is highest in whites: it is reported to be 24.8 cases per 100,000 population in white men, compared with 20.7 and 17.6 cases per 100,000 population in Hispanic and African-American men, respectively. The incidence rates are lowest among Asians, Pacific Islanders, and Native Americans.[2]

Prognosis

Overall, the prognosis for non-Hodgkin lymphoma (NHL) varies with the histology, the stage of disease at diagnosis, the response of disease to therapy, and other factors as listed in the International Prognostic Index (IPI) score.

 The IPI was originally designed as a prognostic factor model for aggressive NHL, but can be useful for predicting the outcome of patients with low-grade lymphoma. This index is also used to identify patients at high risk of relapse based on specific sites of involvement, including bone marrow, central nervous system, liver, testis, lung, and spleen.[8] Separate indices have been developed for follicular and mantle cell lymphoma.[9, 10, 11]

The IPI includes the following risk factors (for each factor that is present, the patient receives 1 point)[8] :

Based on the IPI score, patients can be categorized as follows:

With this model, relapse-free and overall survival rates at 5 years are as follows:

History and Physical Examination

Lymphadenopathy is the most common manifestation of lymphoma. Lymphadenopathy may wax and wane. Spontaneous remissions have been documented, most commonly in patients with low-grade lymphomas.

Systemic symptoms known to be associated with adverse prognosis include the following:

Organ-specific symptoms that may lead to identification of specific sites of involvement include the following:

Typical physical examination findings include the following:

Findings in patients with an advanced high-grade lymphoma may include the following:

Examples of findings that might direct further investigations and subsequent therapy include the following:

Approach Considerations

The initial evaluation of the patient with newly diagnosed non-Hodgkin lymphoma (NHL) must establish the precise histologic subtype, the extent and sites of disease (localized or advanced; nodal or extranodal), and the patient's general health status. These studies help to determine the appropriate therapy and the patient's prognosis.

Tissue Biopsy

Incisional or excisional biopsy is preferred for the initial diagnosis; core needle biopsy is discouraged except for cases in which a lymph node is not easily accessible. Fine-needle aspiration is not an acceptable diagnostic tool; however, in selected circumstances it can be used in combination with immunohistochemistry and flow cytometry.

Flow cytometry and immunohistochemical stains of the biopsied material should be performed to confirm the diagnosis and for accurate subtyping

Laboratory Studies

Laboratory studies are as follows:

Other Tests

Immunoglobulin gene rearrangement

Immunoglobulin (Ig) gene rearrangement is useful for differentiating a B-cell lymphoproliferative process from a monoclonal or reactive proliferation of lymphocytes. This technique not only provides a specific marker for B cells but also is a true marker for monoclonality.

Polymerase chain reaction

Polymerase chain reaction (PCR) is used to assess minimal residual disease. This technique has generally been applied to the t(14;18) translocation and the associated bcl -2 gene.

Whereas PCR positivity for bcl-2 gene rearrangements can be found in healthy individuals, patients with lymphoma in remission who show positive results for bcl -2 gene rearrangements by PCR in the blood or bone marrow are more likely to experience relapse than patients who do not have this abnormality. However, the correlation between relapse and PCR positivity is not perfect, and, at present, PCR is still considered a research tool.

Imaging Studies

Fluorodeoxyglucose (FDG) positron emission tomography with computed tomography (PET/CT) is the preferred imaging modality for staging FDG-avid nodal lymphomas, while CT alone is preferred for FDG-nonavid and variably FDG-avid histologies.[1]

For these purposes, essentially all nodal lymphoma histologies are considered FDG-avid except the following:

Although PET/CT is generally not useful for staging these more indolent histologies, it may be helpful in some circumstances (eg, to identify a preferred biopsy site if aggressive transformation is suspected).  

A dedicated contrast-enhanced CT scan may be required in addition to the PET/CT to define the extent of disease in special situations, such as in the setting of lymphadenopathy close to bowel or if there is compression or thrombosis of blood vessels. Additional tests may be helpful in the evaluation of specific disease sites. 

EVALUATION OF SPECIFIC SITES — Specific evaluation of the gastrointestinal tract, liver, spleen, central nervous system, skeleton, or genitourinary tract is reserved for patients with symptoms or those at particularly high risk for involvement of these sites.

Histologic Findings

The histologic findings in B-cell non-Hodgkin lymphoma (NHL) are varied. The salient features of the most common subtypes are described below.

Chronic lymphocytic leukemia/small lymphocytic lymphoma

Chronic lymphocytic leukemia (CLL)/small lymphocytic leukemia (SLL) is a neoplasm composed of monomorphic small, round to slightly irregular B lymphocytes in the peripheral blood, bone marrow, spleen, and lymph nodes, admixed with prolymphocytes and paraimmunoblasts forming proliferation centers in tissue infiltrates.

The lymph node architecture is effaced, with a pseudofollicular pattern of regularly distributed pale areas corresponding to proliferation centers containing larger cells in a dark background of small cells. The predominant cell type is the small lymphocyte. Mitotic activity is usually low. The size of proliferation centers and the number of paraimmunoblasts vary from case to case, but there is no correlation between lymph node histology and clinical course.

On peripheral blood smears and bone marrow aspirate smears the CLL/SLL cells are small lymphocytes with clumped chromatin and scant cytoplasm. Smudge or basket cells are typically seen in peripheral blood smears.

Extranodal marginal zone lymphoma of mucosa-associated lymphoid tissue

Mucosa-associated lymphoid tissue (MALT) lymphoma is an extranodal lymphoma composed of morphologically heterogeneous small B cells, including marginal zone (centrocytelike) cells, small lymphocytes, and scattered immunoblast and centroblastlike cells.

The lymphoma cells infiltrate around reactive B-cell follicles, external to a preserved follicle mantle, in a marginal zone distribution and spread out to form larger confluent areas that eventually overrun some or most of the follicles. There is plasma cell differentiation in a proportion of the cases. In epithelial tissues, the neoplastic cells typically infiltrate the epithelium, forming lymphoepithelial lesions.

Nodal marginal zone lymphoma

Nodal marginal zone lymphoma (NMZL) is a primary nodal B-cell neoplasm that morphologically resembles lymph nodes involved by marginal zone lymphoma of extranodal or splenic types, but without evidence of extranodal or splenic disease.

Follicular lymphoma

Follicular lymphoma is a neoplasm composed of follicle center B cells (typically both centrocytes and centroblasts) that usually has at least a partially follicular pattern. It is graded by counting or estimating the absolute number of centroblasts in 10 neoplastic follicles, expressed per 40× high-power microscopic field (hpf). Grade 1 and grade 2 cases have a marked predominance of centrocytes and only a few centroblasts (grade 1, 0-5 centroblasts/hpf; grade 2, 6-15 centroblasts/hpf; grade 3, >15 centroblasts/hpf).



View Image

Neoplastic follicles comprising cleaved cells (centrocytes) and larger cells with vesicular nuclei and prominent 2-3 nucleoli (centroblasts).

If diffuse areas of any size composed predominantly or entirely of blastic cells are present in any case of follicular lymphoma, a diagnosis of diffuse large B-cell lymphoma (DLBCL) is also made. Peripheral blood involvement sufficient to produce lymphocytosis (usually < 20,000/µL) is observed in about 10% of patients. Bone marrow involvement occurs in 65% of patients and characteristically takes the form of paratrabecular lymphoid aggregates. Splenic white pulp and hepatic portal triads are also frequently involved.

Mantle cell lymphoma

Mantle cell lymphoma is a B-cell neoplasm generally composed of monomorphic small to medium-sized lymphoid cells with irregular nuclear contours and a cyclin-D1 translocation (see the image below). Neoplastic transformed cells resembling centroblasts, immunoblasts, or paraimmunoblasts and proliferation centers are absent. Hyalinized small vessels are commonly seen.



View Image

Mantle cell lymphoma. Small lymphoid cells with oval to slightly irregular nuclei and clumped chromatin and rare admixed pink histiocytes.

A spectrum of morphologic variants is recognized. The blastoid and pleomorphic variants are considered to have poorer prognosis and to be of important clinical significance. Further evaluation of the proliferation fraction, either by counting mitotic figures or estimating the proportion of Ki67-positive nuclei, is important because of its prognostic impact.

Diffuse large B-cell lymphoma

The common morphologic features that unite the various forms of DLBCL are the relatively large cell size (usually 4-5 times that of a small lymphocyte) and a diffuse pattern of growth.[12] In other respects, a fair degree of morphologic variation exists.

In most cases, the tumor cells have a round or oval nucleus that appears vesicular because of margination of chromatin at the nuclear membrane, but large multilobed or cleaved nuclei predominate in some cases (see the image below). Nucleoli may be 2-3 in number and located adjacent to the nuclear membrane, or they may be single and centrally placed. Cytoplasm is usually present in moderate abundance and may be pale or basophilic.



View Image

Diffuse large B-cell non-Hodgkin lymphoma. Large cells with abundant cytoplasm and large round-ovoid nuclei with thick nuclear membrane and multiple p....

Other more anaplastic tumors may contain multinucleated cells with large inclusionlike nucleoli that closely resemble Reed-Sternberg cells, and phenotyping is often necessary to distinguish these 2 entities.

An immunophenotypical subdivision of DLBCL has been proposed that uses a combination of antibodies to CD10, bcl-6, and MUM-1 to divide DLBCL into germinal center B cell-like (GCB) and nongerminal center B cell-like (non-GCB) varieties. Cases that are CD10 positive or CD10 negative and bcl-6 positive but MUM-1 negative are regarded as GCB type, whereas all others are regarded as non-GCB or activated B-cell (ABC) type.

This immunophenotypical subdivision does not completely correlate with gene expression-based subgrouping of DLBCL. In the non-GCB or the ABC, bcl-2 expression is known to be associated with poor patient outcome.

DLBCL may be classified in several different ways. Common morphologic variants of DLBCL, not otherwise specified, include centroblastic, immunoblastic, anaplastic, and rare morphologic variants. Molecular subgroups include GCB and ABC. Immunohistochemical subgroups include CD5-positive DLBCL, GCB, and non-GCB.

DLBCL subtypes include the following:

Other lymphomas of large B cells include the following:

Borderline cases include the following:

Burkitt lymphoma

Burkitt lymphoma is a B-cell lymphoma with an extremely short doubling time that often presents in extranodal sites or as an acute leukemia. Involved tissues are effaced by a diffuse infiltrate of monomorphic, medium-sized (nuclei similar or smaller to those of histiocytes) transformed lymphoid cells.

The nuclei are round with finely clumped and dispersed chromatin, with multiple basophilic medium-sized paracentrically situated nucleoli. The cytoplasm is deeply basophilic and usually contains lipid vacuoles (see the images below). A high mitotic index is typical, as is apoptotic tumor cell death, accounting for the presence of numerous tissue macrophages with their ingested tissue debris. These macrophages are often surrounded by a clear space, creating the characteristic starry sky pattern.



View Image

Burkitt lymphoma. Normal architecture is entirely replaced by lymphoma cells and evenly dispersed macrophages, starry sky (250×).



View Image

Burkitt lymphoma cells with round noncleaved nuclei and strongly basophilic cytoplasm (1000×).

Primary mediastinal (thymic) large B-cell lymphoma

Primary mediastinal large B-cell lymphoma is a diffuse large B-cell lymphoma that arises in the mediastinum from a putative thymic B-cell origin and has distinctive clinical, phenotypic, and genotypic features. Histologically, it is commonly associated with compartmentalizing alveolar fibrosis (see the image below). The cells are medium-sized to large and have abundant clear/pale cytoplasm and round to oval nuclei. A few cases have multilobated and pleomorphic nuclei resembling the Reed-Sternberg cells of Hodgkin lymphoma.



View Image

Compartmentalizing fibrosis and infiltrate of medium-sized to large lymphoid cells.

Immunophenotypic Analysis

Flow cytometry analysis can help provide accurate diagnosis of some categories of B-cell lymphoma. Characteristic immunophenotypes are associated with major types of lymphoma (see the table below).

Table 2. Characteristic Immunophenotypes of Major Subtypes of Lymphoma



View Table

See Table

Staging

The Ann Arbor staging system has commonly been used for patients with non-Hodgkin lymphoma (NHL).[13] This system divides NHL into four stages and subclassifies it into A and B categories. The B designation is applied to individuals with any of the following well-defined generalized symptoms:

In 2014, the International Conference on Malignant Lymphomas (a multidisciplinary team of researchers representing major lymphoma clinical trial groups and cancer centers from North America, Europe, Japan, and Australasia) met in Lugano, Italy and created a modification of the Ann Arbor system, the Lugano classification.[14] The 2015 NCCN Non-Hodgkin’s Lymphomas guidelines adopted the Lugano classification.[15]

The Lugano classification (see Table 3, below) includes four stages. However, the A and B subclassifications are not required in NHL since they are not prognostic.

Table 3. Lugano Modification of the Ann Arbor Staging System.



View Table

See Table

Other staging considerations

The following factors are not a part of the Ann Arbor staging system, but have important prognostic value in B-cell NHL:

To identify subgroups of patients most likely to relapse, the International Prognostic Index (IPI) was compiled for 2031 patients with aggressive NHL.[8] The IPI has been validated by several cancer centers and has been incorporated into new trials designed by various cooperative groups. (See Prognosis.)

Approach Considerations

Treatment of B-cell lymphoma varies according to the disease type. See Guidelines.

In addition, in October 2017 the US Food and Drug Administration approved chimeric antigen receptor (CAR) T-cell therapy with axicabtagene ciloleucel (Yescarta) for use in adults with diffuse large B-cell lymphoma (DLBCL) who have not responded to or who have relapsed after at least two other kinds of treatment. Approved uses include DLBCL, primary mediastinal large B-cell lymphoma, high-grade B-cell lymphoma and DLBCL arising from follicular lymphoma. Axicabtagene ciloleucel is not indicated for the treatment of primary central nervous system lymphoma.[16]

In May 2018, a second CAR T-cell therapy, tisagenlecleucel (Kymriah), was approved for adults with relapsed or refractory DLBCL who are ineligible for or relapsed after autologous stem cell transplantation.[17]

Complications

Complications arising from therapy include the following:

Consultations

Radiation oncologists are often consulted because radiation plays a role in management of B-cell lymphoma; it is considered the treatment of choice in early stage indolent lymphoma and has a role in consolidation of treatment in localized aggressive lymphoma.

A surgical consultation is indicated when central venous access devices are employed, especially when stem cell or bone marrow transplantation is considered.

Transfer

Patients with lymphoma who are being managed at peripheral health care facilities may require their care to be transferred to a tertiary care or research institute for access to clinical trials and investigational agents.

Patients who are profoundly ill or are experiencing significant complications from either the disease or its therapy also warrant transfer to a facility that is better equipped to handle such emergencies.

Diet

Typically, no dietary restrictions exist for patients with non-Hodgkin lymphoma (NHL). Patients with prolonged neutropenia following chemotherapy, especially those undergoing high-dose chemotherapy with hematopoietic precursor cell rescue, are provided with a reduced-bacteria diet. Patients with high tumor burden undergoing chemotherapy may be at risk for tumor lysis syndrome and sometimes require a diet low in uric acid and potassium.

Staging

In 2014, the International Conference on Malignant Lymphomas (a multidisciplinary team of researchers representing major lymphoma clinical trial groups and cancer centers from North America, Europe, Japan, and Australasia) published guidelines for the evaluation, staging, and response assessment of patients with malignant lymphomas. This staging system is known as the Lugano Modification of the Ann Arbor staging system.[14]  In 2015, the National Comprehensive Cancer Network (NCCN) adopted this system.[15]

The revised recommendations for staging include the following[14] :

In addition, these guidelines offered consensus on further modifications to the Ann Arbor staging classification, as shown in Table 1, below[15, 14] :

  Table 4. Non-Hodgkin lymphoma staging.



View Table

See Table

*Stage II bulky disease is considered limited or advanced; this distinction is made on the basis of histology and a number of prognostic factors.

Suffixes A and B are not required. X for bulky disease replaced with documenting of largest tumor diameter. Definition of “bulky” disease varies, depending on lymphoma histology.

Risk Stratification

The International Prognostic Index (IPI), which was originally designed as a prognostic factor model for aggressive non-Hodgkin lymphoma (NHL), also appears to be useful for predicting the outcome of patients with low-grade lymphoma. This index is also used to identify patients at high risk of relapse, based on specific sites of involvement, including bone marrow, central nervous system, liver, testis, lung, and spleen.[8]  Separate indices have been developed for follicular and mantle cell lymphoma.[9, 10, 11]

The IPI includes the following risk factors[8] :

Each factor is worth 1 point. Based on the IPI score, patients can be categorized as follows[8] :

With this model, relapse-free and overall survival rates at 5 years are as follows:

Follicular Lymphoma

World Health Organization Classification

In 2001, the WHO classification called for grading of follicular lymphoma (FL) from grades 1-3 based on the number of centroblasts per high-power field (hpf). However, the 2008 update consolidated cases with few centroblasts as FL grade 1-2 (low grade) and divided FL grade 3 into 3A (presence) and 3B (absence) of centrocytes.[3, 18]

National Comprehensive Cancer Network (NCCN) guidelines note that that grade 3B is commonly treated as diffuse large B-cell lymphoma (DLBCL), whereas opinion is divided on whether to treat grade 3A as FL or DLBCL. FL of any grade that is found to contain an area of DLBCL should be managed as a DLBCL.[19]

Prior to 2008, primary cutaneous follicle center lymphoma (PCFCL) was classified as a variant of FL. In the 2008 update, its classification was changed to that of a distinct entity. PCFCL may contain a high proportion of large B cells, including large centrocytes and centroblasts. Dissemination beyond the skin is rare.[3]

Diagnosis

In addition to its general guidance on diagnosis of lymphoma, the NCCN recommends the following studies to establish a diagnosis of FL[15] :

Risk stratification

The Groupe d’Etude des Lymphomes Folliculaires (GELF) recommends the following criteria for identifying patients in whom immediate therapy is necessary[20] :

The NCCN recommends both the GELF criteria and the 2004 Follicular Lymphoma International Prognostic Index (FLIPI) for risk stratification. The FLIPI includes the following risk factors[10] :

For each factor, the patient receives 1 point. Based on the FLIPI score, patients can be categorized as follows[10] :

In 2009, the International Follicular Lymphoma Prognostic Factor Project published an updated score, FLIPI2. The FLIPI2 includes the following risk factors (as with FLIPI1, each factor is worth 1 point)[9] :

Based on the FLIPI2 score, patients can be categorized as low, intermediate, or high risk.[9]

Treatment

The NCCN guidelines recommend the following regimens as preferred first-line therapy (in alphabetical order)[19] :

Rituximab (375 mg/m2 weekly for 4 doses) may be considered for patients with a low tumor burden.

Rituximab-abbs may be substituted for rituximab at the start of a course of treatment.

For elderly or infirm patients who, in the opinion of treating physician, are unlikely to tolerate most of the above regimens, suggested options for first-line, second-line, and subsequent therapy include the following:

First-line regimens for consolidation or extended dosing (optional) include the following:

Preferred regimens for second-line and subsequent therapy are as follows:

Options for second-line consolidation or extended dosing (optional) include the following:

Marginal Zone Lymphomas of MALT Type

Mucosa-associated lymphoid tissue (MALT) lymphomas are usually localized and are most often observed in the stomach, lung, salivary gland, thyroid, orbit, conjunctiva, and lacrimal gland. An association of MALT lymphomas with autoimmune disease is well known.

The development of gastric MALT lymphomas has been attributed to antigenic stimulation associated with chronic Helicobacter pylori gastritis. Indeed, both the NCCN and ESMO guidelines recommend antibiotic eradication of H pylori as first-line treatment for H pylori–positive patients, including t(11;18)-positive patients. After eradication of H pylori, t(11;18) patients have a < 5% response rate to the tumor. They will require involved-site radiation therapy (ISRT) or rituximab. In H pylori –negative patients, involved-site radiation therapy (ISRT), 30 Gy, is the preferred treatment option. Rituximab is an option if radiation therapy is contraindicated.[19]

For non-gastric MALT lymphomas, radiotherapy is preferred but surgery is also an appropriate treatment (and in most cases is required to make the diagnosis). Further treatment may not be needed. When the tumor is incompletely excised, radiotherapy should be considered. In the minority of patients presenting with a more widespread disease, systemic chemotherapy is effective. The majority of stage III-IV MALT lymphomas respond to treatment similar to that for follicular lymphoma.[19]

Therapy for gastric MALToma

European Society for Medical Oncology (ESMO) guidelines for treatment of gastric marginal zone lymphoma of the MALT type include the following recommendations[21] :

 

Mantle Cell Lymphoma

Diagnosis

Mantle cell lymphoma (MCL) is diagnosed in accordance with the World Health Organization criteria for hematological neoplasms and detection of Cyclin D1 expression or the t(11;14) translocation along with mature B-cell proliferation.[21] The National Comprehensive Cancer Network (NCCN) recommends the following studies to establish a diagnosis of MCL[19] :

Risk stratification

The European Society for Medical Oncology (ESMO) recommends the 2008 MCL International Prognostic Index (MIPI) for risk stratification.[22]  The MIPI includes the following risk factors[11] :

Based on the MIPI score, patients can be categorized as follows[11] :

In addition, both the NCCN and ESMO recommend assaying Ki-67 proliferative antigen to evaluate cell proliferation. Low Ki-67 (< 30%) is associated with a more favorable prognosis; however, it is not used to guide treatment decisions.[15, 22]

Treatment

The NCCN and ESMO offer similar treatment recommendations, as follows[15, 22]

Induction therapy

For induction therapy, aggressive regimens include the following:

The CALGB regimen comprises 5 treatments; all but Treatment 5 are accompanied by granulocyte colony-stimulating factor (G-CSF) therapy and infection prophylaxis. The 5 treatments are as follows:

Less aggressive regimens include the following:

Consolidation therapy

In patients who are candidates for it, high-dose therapy with autologous stem cell rescue plus rituximab maintenance (category 1 for rituximab maintenance) is first-line therapy. In patients who are not candidates, first-line therapy is rituximab maintenance (category 1 following R-CHOP). Second-line consolidation therapy is with allogeneic stem cell transplantation (nonmyeloablative or myeloablative).

Second-line therapy

Second-line therapy regimens include the following:

Diffuse Large B-Cell Lymphoma

Diagnosis

In addition to its general guidance on diagnosis of lymphoma, the National Comprehensive Cancer Network (NCCN) recommends the following studies to establish a diagnosis of diffuse large B-cell lymphoma (DLBCL)[15] :

IHC should include adequate markers to differentiate the two subtypes of DLBCL: activated B-cell type (ABC) and germinal center B-cell type (GCB). The subtypes are genetically different diseases, and survival in patients with the ABC subtype is worse than in those with GCB.[23]

Risk stratification

The NCCN and the European Society for Medical Oncology (ESMO) recommend use of the International prognostic index (IPI) for all patients. Age-adjusted International Prognostic Index (aa-IPI) should be used for risk stratification of patients aged 60 years and younger.[15, 24]

The IPI includes the following risk factors:

Each risk factor is worth 1 point. On the basis of the IPI score, patients can be categorized as follows:

The aa-IPI includes the following risk factors (1 point is allotted for each factor)[8] :

Based on the aa-IPI score, patients can be categorized as follows[8] :

Treatment

National Comprehensive Cancer Network recommendations [19]

First-line therapy (in alphabetical order) options are as follows:

First-line therapy options for patients who have poor left ventricular function or are very frail are as follows:

Regimens for patients >80 years of age with comorbidities are as follows:

First-line consolidation (optional) regimens are as follows:

Regimens for concurrent presentation with CNS disease are as follows:

Second-line and subsequent therapy (intention to proceed to high-dose therapy) regimens are as follows:

Second-line and subsequent therapy regimens, for patients who are not candidates for high-dose therapy, include the following:

Follow-up

A PET-CT scan should be performed to confirm complete remission (CR). Patients in CR should receive clinical follow-up with a history and physical examination and laboratory studies (eg, complete blood cell count, comprehensive metabolic panel, lactate dehydrogenase level) every 3-6 months for 5 years and then yearly or as clinically indicated. Imaging studies (CT scan) should be performed no more often than every 6 mo for 2 years after the completion of treatment, and then only as clinically indicated.[19]

Burkitt Lymphoma

Classification

The 2008 World Health Organization classification identifies the following three clinical variants of Burkitt lymphoma (BL)3:

Diagnosis

In addition to its general guidance on diagnosis of lymphoma, the National Comprehensive Cancer Network (NCCN) recommends the following studies to establish a diagnosis of Burkitt lymphoma[19] :

Risk stratification

A prognostic scoring system was developed in 2013 using the Surveillance, Epidemiology, and End Results (SEER) database. Risk factors and points assigned are as follows32:

The four risk groups based on the scoring system are as follows:

With this model, relative survival rates at 5 years are as follows:

Treatment

Induction therapy for low-risk Burkitt lymphoma includes the following:

Induction therapy for high-risk Burkitt lymphoma includes the following:

• CODOX-M (original or modified) (cyclophosphamide, doxorubicin, vincristine with intrathecal methotrexate and cytarabine followed by high-dose systemic methotrexate) alternating with IVAC (ifosfamide, cytarabine, etoposide, and intrathecal methotrexate) ± rituximab

• Dose-adjusted EPOCH (etoposide, prednisone, vincristine, cyclophosphamide, doxorubicin) + rituximab (for high-risk patients not able to tolerate aggressive treatments) (regimen includes intrathecal methotrexate) (Data are for patients without CNS disease.)

• HyperCVAD (cyclophosphamide, vincristine, doxorubicin, and dexamethasone) alternating with high-dose methotrexate and cytarabine + rituximab (regimen includes intrathecal therapy)

Second-line therapy (for patients with dsease relapse >6 mo after appropriate first-line therapy)

While no definitive second-line therapies exist, limited data support the following regimens:

Follow-up

The NCCN recommends follow up every 2-3 months for the first year after complete response, then every 3 months for the next year, and every 6 months thereafter. Relapse is rare after 2 years.12

Primary Cutaneous B-Cell Lymphomas

In addition to the National Comprehensive Cancer Network (NCCN), the European Organization for Research and Treatment of Cancer (EORTC) and the International Society for Cutaneous Lymphoma (ISCL) published guidelines for the management of primary cutaneous B-cell lymphomas (CBCL).[25]

Classification

The NCCN and EORTC/ISCL guidelines recommend use of the WHO-EORTC classification for cutaneous B-cell lymphomas (CBCL), which distinguishes the following three main types[26] :

Of note, a germinal (or follicle) center phenotype and large cells in a skin lesion is not equivalent to diffuse large B-cell lymphoma (DLBCL) but is consistent with primary cutaneous germinal/follicle center lymphoma.

Diagnosis

In addition to its general guidance on diagnosis of lymphoma, the NCCN recommends the following studies to establish a diagnosis of CBCL[15] :

Staging

The 2007 TNM classification system of the ISCL/EORTC is used for staging, as shown in Table 3, below.[27]

Table 3. International Society for Cutaneous Lymphoma/European Organization for Research and Treatment of Cancer tumor-node-metastasis classification for cutaneous B-cell lymphoma



View Table

See Table

Treatment

PC-FCL and PC-MZL

Both the NCCN and EORTC/ISCL guidelines recommend local radiation therapy or excision for T1-2 PC-FCL and PC-MZL.[15, 25]  The NCCN recommends intralesional steroids or topical therapy including steroids, imiquimod, nitrogen mustard, and bexarotene as alternative treatment options.[15]

For T3 disease, the NCCN recommends radiation therapy; chlorambucil; or cyclophosphamide, vincristine, and prednisone (CVP) with or without rituximab. Extracutaneous disease should be managed using the treatment guidelines for follicular lymphoma.[15]

EORTC/ISCL guidelines recommend systemic rituximab as the first choice of treatment for patients with extensive skin lesions. Combination chemotherapy (eg, R-COP, R-CHOP) should be considered only in exceptional cases, such as in patients with progressive disease not responding to rituximab or patients developing extracutaneous disease.[25]

PC-DBCL, LT

Both guidelines caution that radiation therapy is less effective in PC-DBCL, LT. R-CHOP with local radiation therapy is recommended as first line of treatment for all stages of DBCL, LT.[15, 25] . Because of the lack of studies on relapsed disease, EORTC/ISCL recommend that treatment protocols for relapsed DBCL be followed.[25] .

Medication Summary

The goals of pharmacotherapy are to induce remission, reduce morbidity, and prevent complications.

Pegfilgrastim (Neulasta)

Clinical Context:  Pegfilgrastim is a long-acting filgrastim created by the covalent conjugate of recombinant granulocyte colony-stimulating factor (ie, filgrastim) and monomethoxypolyethylene glycol. As with filgrastim, it acts on hematopoietic cells by binding to specific cell surface receptors, thereby activating and stimulating the production, maturation, migration, and cytotoxicity of neutrophils.

Epoetin alfa (Epogen, Procrit)

Clinical Context:  This agent is a purified glycoprotein produced from mammalian cells modified with gene coding for human erythropoietin (EPO). Its amino acid sequence is identical to that of endogenous EPO. The biological activity of epoetin alfa mimics human urinary EPO, which stimulates division and differentiation of committed erythroid progenitor cells and induces the release of reticulocytes from bone marrow into the blood stream.

Class Summary

These agents can induce an increase in reticulocyte counts, with a subsequent increase in hematocrit and hemoglobin levels.

Ibritumomab tiuxetan (Zevalin Y-90)

Clinical Context:  A murine monoclonal antibody that targets the CD20 antigen, ibritumomab tiuxetan is chelated to the radioisotopes indium-111 or yttrium-90. It is used in conjunction with rituximab to treat B-cell NHL or rituximab-refractory follicular NHL. The regimen consists of two low doses of rituximab, an imaging dose, two-three whole-body scans, and a therapeutic dose, all of which are delivered in an outpatient setting over 8 days.

Ofatumumab (Arzerra)

Clinical Context:  Ofatumumab is an anti-CD20 human monoclonal antibody that inhibits B-cell activation in early stages. It is indicated for chronic lymphocytic leukemia refractory to fludarabine and alemtuzumab.

Alemtuzumab (Lemtrada, Campath)

Clinical Context:  Alemtuzumab is a monoclonal antibody against CD52, an antigen found on B cells, T cells, and almost all chronic lymphocytic leukemia cells. It binds to the CD52 receptor of the lymphocytes, which slows the proliferation of leukocytes.

Rituximab (Rituxan)

Clinical Context:  Rituximab is a genetically engineered chimeric murine/human monoclonal antibody directed against the CD20 antigen found on the surface of normal and malignant B lymphocytes. The antibody is an immunoglobulin G1 (IgG1) kappa immunoglobulin containing murine light- and heavy-chain variable region sequences and human constant region sequences.

Class Summary

The agents in this class target specific antigens in carcinoma cells and induce cytotoxicity.

Dexamethasone (Decadron, Dexamethasone intensol)

Clinical Context:  A component of the m-BACOD (methotrexate, bleomycin, doxorubicin [Adriamycin], cyclophosphamide, Oncovin, and dexamethasone) regimen, dexamethasone is a glucocorticoid that acts as an immunosuppressant by stimulating the synthesis of enzymes needed to decrease the inflammatory response. It also acts as an anti-inflammatory agent by inhibiting the recruitment of leukocytes and monocyte-macrophages into affected areas via inhibition of chemotactic factors and factors that increase capillary permeability.

Dexamethasone is readily absorbed via the GI tract and metabolized in the liver. Inactive metabolites are excreted via the kidneys. Most of the adverse effects of corticosteroids are dose dependent or duration dependent.

Prednisone (Prednisone Intensol, Rayos, Deltasone)

Clinical Context:  A component of several regimens, such as CHOP, prednisone is a glucocorticoid that acts as an immunosuppressant by stimulating the synthesis of enzymes needed to decrease the inflammatory response. It also acts as an anti-inflammatory agent by inhibiting the recruitment of leukocytes and monocyte-macrophages into affected areas via inhibition of chemotactic factors and factors that increase capillary permeability.

Prednisone is readily absorbed via the GI tract and metabolized in the liver. Inactive metabolites are excreted via the kidneys. Most of the adverse effects of corticosteroids are dose dependent or duration dependent.

Class Summary

These drugs have anti-inflammatory properties and cause profound and varied metabolic effects. Corticosteroids modify the body's immune response to diverse stimuli.

Chlorambucil (Leukeran)

Clinical Context:  Chlorambucil alkylates and cross-links strands of DNA, inhibiting DNA replication and RNA transcription. It is used mainly to treat indolent lymphomas, particularly chronic lymphocytic leukemia (CLL) and Waldenstrom macroglobulinemia. This agent may be preferable for elderly patients with serious comorbid medical problems who require treatment for lymphoma. It is well absorbed orally.

Cyclophosphamide

Clinical Context:  Cyclophosphamide is chemically related to nitrogen mustards. As an alkylating agent, the mechanism of action of the active metabolites may involve cross-linking of DNA, which may interfere with growth of normal and neoplastic cells. This agent can be used alone but is mostly used as a component of multiple combination chemotherapy regimens.

Bendamustine (Bendeka, Treanda)

Clinical Context:  Bendamustine is an alkylating agent that is a bifunctional mechlorethamine derivative. It forms covalent bonds with electron-rich nucleophilic moieties that can lead to cell death. It is active against both quiescent and dividing cells and is indicated for chronic lymphocytic leukemia (CLL).

Cisplatin

Clinical Context:  Cisplatin is a platinum-containing compound that exerts its antineoplastic effect by covalently binding to DNA, with preferential binding to the N-7 position of guanine and adenosine. It can react with 2 different sites on DNA to cause cross-links. The platinum complex also can bind to the nucleus and to cytoplasmic protein. A bifunctional alkylating agent, once cisplatin is activated to the aquated form in the cell, it binds to DNA, resulting in interstrand and intrastrand cross-linking and denaturation of the double helix.

Doxorubicin (Adriamycin)

Clinical Context:  An anthracycline antibiotic that can intercalate with DNA, doxorubicin affects many of the functions of DNA, including synthesis. It forms DNA-cleavable complexes by interaction with topoisomerase II, which is responsible for the cytocidal activity of the drug. Doxorubicin is administered IV and distributes widely into bodily tissues, including the heart, kidneys, lungs, liver, and spleen. It does not cross the blood-brain barrier and is excreted primarily in bile. It forms an important part of multiple chemotherapeutic regimens for lymphomas, including cyclophosphamide, hydroxydaunomycin (doxorubicin), vincristine (Oncovin), and prednisone (CHOP).

Vincristine (Vincasar PFS)

Clinical Context:  The mechanism of action of vincristine is uncertain. It may involve a decrease in reticuloendothelial cell function or an increase in platelet production; however, neither of these mechanisms fully explains the effect in thrombocytopenic purpura and hemolytic-uremic syndrome. Vincristine is used in hematologic and nonhematologic malignancies. It is a component of CHOP and other regimens for lymphoma.

Fludarabine

Clinical Context:  Fludarabine is a purine analogue that interferes with DNA synthesis by inhibiting ribonucleotide reductase. It is also incorporated into RNA, causing inhibition of RNA and protein synthesis; however, its primary effect may result from activation of apoptosis.

Nelarabine (Arranon)

Clinical Context:  Nelarabine is a prodrug of the deoxyguanosine analogue 9-beta-D-arabinofuranosylguanine (ara-G). It is converted to the active 5'-triphosphate, ara-GTP, a T-cell–selective nucleoside analog. Leukemic blast cells accumulate ara-GTP. This allows for incorporation into DNA, leading to inhibition of DNA synthesis and cell death

Cytarabine

Clinical Context:  Cytarabine is converted intracellularly to the active compound cytarabine-5'-triphosphate, which inhibits DNA polymerase. It is cell cycle S phase specific and it blocks the progression from the G1 to the S phase, in turn killing cells that undergo DNA synthesis in the S phase of the cell proliferation cycle.

Gemcitabine (Gemzar)

Clinical Context:  Gemcitabine is a cytidine analog. It is metabolized intracellularly to an active nucleotide. It inhibits ribonucleotide reductase and competes with deoxycytidine triphosphate for incorporation into DNA. It is cell-cycle specific for the S phase. Gemcitabine is indicated as first-line treatment for locally advanced (nonresectable stage II or stage III) or metastatic (stage IV) pancreatic adenocarcinoma.

Etoposide (Toposar)

Clinical Context:  Etoposide is a glycosidic derivative of podophyllotoxin that exerts its cytotoxic effect through stabilization of the normally transient covalent intermediates formed between DNA substrate and topoisomerase II, leading to single- and double-strand DNA breaks. This causes cell proliferation to arrest in the late S or early G2 portion of the cell cycle.

Mitoxantrone

Clinical Context:  Related to anthracyclines. Mitoxantrone intercalates into DNA. This results in cross-links and strand breaks. Binds to nucleic acids and inhibits DNA and RNA synthesis by template disordering and steric obstruction. Replication is decreased by binding to DNA topoisomerase II. Active through entire cell cycle.

Carboplatin

Clinical Context:  Carboplatin is an analog of cisplatin. This is a heavy metal coordination complex that exerts its cytotoxic effect by platination of DNA, a mechanism analogous to alkylation, leading to interstrand and intrastrand DNA cross-links and inhibition of DNA replication. It binds to protein and other compounds containing the SH group. Cytotoxicity can occur at any stage of the cell cycle, but the cell is most vulnerable to action of these drugs in the G1 and S phases. It has same efficacy as cisplatin, but with a better toxicity profile. The main advantages over cisplatin include less nephrotoxicity and ototoxicity, the lack of a need for extensive prehydration, and a smaller likelihood of inducing nausea and vomiting; however, it is more likely to induce myelotoxicity.

Bleomycin

Clinical Context:  This agent is composed of a group of glycopeptides extracted from Streptomyces species. Each molecule has a planar end and an amine end; different glycopeptides of the group differ in their terminal amine moieties. The planar end intercalates with DNA, while the amine end facilitates oxidation of bound ferrous ions to ferric ions, thereby generating free radicals, which subsequently cleave DNA, acting specifically at purine-G-C-pyrimidine sequences.

Vorinostat (Zolinza)

Clinical Context:  Vorinostat is a histone deacetylase (HDAC) inhibitor. HDAC inhibition results in hypoacetylation of core nucleosomal histones, it condenses the chromatin structure, and it represses gene transcription. It is indicated for the treatment of progressive, persistent, or recurrent cutaneous T-cell lymphoma.

Belinostat (Beleodaq)

Clinical Context:  Belinostat is a histone deacetylase (HDAC) inhibitor. HDACs catalyze the removal of acetyl groups from the lysine residues of histones and some nonhistone proteins. Inhibiting this action induces cell cycle arrest and/or apoptosis.

Class Summary

These agents inhibit cell growth and proliferation and lead to cell death.

Ibrutinib (Imbruvica)

Clinical Context:  Bruton’s tyrosine kinase (BTK) inhibitor forms a covalent bond with a cysteine residue in the BTK active site, leading to inhibition of BTK enzymatic activity. BTK is a signaling molecule of the B-cell antigen receptor (BCR) and cytokine receptor pathways.

BTK’s role in signaling through the B-cell surface receptors results in activation of pathways necessary for B-cell trafficking, chemotaxis, and adhesion. It is indicated for mantle cell lymphoma in patients who have received at least 1 previous therapy. It also indicated for marginal zone lymphoma (MZL) in patients who require systemic therapy and have received at least 1 prior anti-CD20-based therapy.

Class Summary

Ibrutinib received accelerated approval from the FDA for marginal zone lymphoma following analysis of phase 2 data in 60 patients who had a median follow-up of 19.4 months. Of these patients, 3% (n=2) treated with ibrutinib achieved complete responses (CR) and 45% (n=27) achieved partial responses (PR). Overall, 78% of patients experienced tumor reduction. Median progression-free survival was 14.2 months.[28]

Copanlisib (Aliqopa)

Clinical Context:  Pan class I phosphatidylinositol-3-kinase (PI3K) inhibitor with predominant inhibitory activity against PI3K-alpha and PI3K-delta isoforms expressed in malignant B cells. By inhibiting several key cell-signaling pathways may induce apoptosis and inhibition of proliferation of premalignant B cells and in turn cause tumor cell death. It is indicated for relapsed follicular lymphoma (FL) in patients who have received at least 2 prior systemic therapies.

Idelalisib (Zydelig)

Clinical Context:  Idelalisib induces apoptosis and inhibits proliferation in cell lines derived from malignant B cells and in primary tumor cells; also inhibits several cell- signaling pathways, including B cell receptor (BCR) signaling and the CXCR4 and CXCR5 signaling, which are involved in trafficking and homing of B cells to the lymph nodes and bone marrow. It gained accelerated approval by the FDA (ie, confirmatory clinical trials in progress) in July 2014 for relapsed follicular B-cell non-Hodgkin lymphoma (FL) and relapsed small lymphocytic lymphoma (SLL) in patients who have received at least 2 prior systemic therapies.

Class Summary

This drug class inhibits one or more of the phosphoinositide 3-kinase enzymes, which are part of the PI3K/AKT/mTOR pathway, an important signalling pathway for many cellular functions such as growth control, metabolism and translation initiation. Within this pathway there are many components, inhibition of which may result in tumor suppression.

Lenalidomide (Revlimid)

Clinical Context:  Thalidomide analogue; inhibits TNF-alpha production, stimulates T cells, reduces serum levels of the cytokines vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF), and inhibits angiogenesis. This agent also promotes G1 cell cycle arrest and apoptosis of malignant cells. Indicated for in combination with rituximab product for the treatment of previously treated follicular lymphoma or marginal zone lymphoma.

Class Summary

These agents antagonize or inhibit the development of new blood vessels.

Pembrolizumab (Keytruda)

Clinical Context:  Indicated for the treatment of adult and pediatric patients with refractory primary mediastinal large B-cell lymphoma (PMBCL), or who have relapsed after 2 or more prior lines of therapy.

Class Summary

Monoclonal antibodies that bind the programmed cell death-1 protein (PD-1) ligands, PD-L1 and PD-L2, to the PD-1 receptor found on T cells, inhibits T cell proliferation and cytokine production.

Axicabtagene ciloleucel (Yescarta)

Clinical Context:  CD19-directed genetically modified autologous T cell immunotherapy, binds to CD19-expressing cancer cells and normal B cells. Studies demonstrated that following the binding anti-CD19 CAR T cells with target cells, the CD28 and CD3-zeta co-stimulatory domains activate downstream signaling cascades which eventually leads to killing of CD19-expressing cells. It is indicated for relapsed or refractory large B-cell lymphoma after two or more lines of systemic therapy, including diffuse large B-cell lymphoma (DLBCL) not otherwise specified, primary mediastinal large B-cell lymphoma, high grade B-cell lymphoma, and DLBCL arising from follicular lymphoma.

Tisagenlecleucel (Kymriah)

Clinical Context:  CD19-directed genetically modified autologous T-cell immunotherapy that involves reprogramming a patient’s own T cells with a transgene encoding a chimeric antigen receptor (CAR) to identify and eliminate CD19-expressing malignant and normal cells. It is indicated in adults with relapsed or refractory large B-cell lymphoma (r/rDLBCL) including DLBCL not otherwise specified, high grade B-cell lymphoma, and DLBCL arising from follicular lymphoma after ≥2 lines of systemic therapy.

Class Summary

Chimeric antigen receptor (CAR) T cell therapy is a form of adoptive T-cell therapy in which T cells are genetically engineered to express a CAR. CAR T cells preparation begins with obtaining a blood sample from the patient. The CAR molecule is introduced into the patient’s T cells through viral or nonviral approaches. The cells undergo a brief round of expansion in the laboratory and are then infused back into the patient. T cells become activated when they recognize the target antigen on the surface of the tumor, in this case, CD19. When T cells are activated, they undergo massive expansion in the body. The cells start to produce multiple different cytokines and proliferate. These cytokines improve the T cells’ function, help them traffic to the tumor site, and start killing the tumor cells by expressing cytotoxic molecules (eg, granzymes and perforins).

In October 2017, FDA approved axicabtagene ciloleucel (Yescarta) for large B-cell lymphoma after at least two other kinds of treatment have failed. Axicabtagene ciloleucel is not indicated for the treatment of patients with primary central nervous system lymphoma.

FDA approval is based on results from the ZUMA-1 study, an open-label, multicenter trial enrolling 111 patients from 22 institutions. Patients in ZUMA-1 received the target dose of axicabtagene ciloleucel (2 x 106cells/kg) after low-dose conditioning with cyclophosphamide and fludarabine for 3 days. The modified intention-to-treat population involved 101 patients who received axicabtagene ciloleucel. In adults with relapsed/refractory DLBCL, the response rates are approximately 60-80%, with complete responses seen in 40-70 %t of patients. Following axicabtagene ciloleucel, 40% of patients maintain their complete response at six-month follow-up. The trial had a median survival follow-up of 8.7 months.[29]

In May 2018, a second CAR T-cell therapy, tisagenlecleucel (Kymriah), was approved for adults with relapsed or refractory DLBCL who are ineligible for or relapsed after ASCT. Approval was based on a single-arm, open-label, multicenter, phase 2 JULIET trial in adults with relapsed or refractory DLBCL and DLBCL after transformation from follicular lymphoma. Eligible patients must have been treated with at least 2 prior lines of therapy, including an anthracycline and rituximab, or relapsed following ASCT. Patients received a single infusion of tisagenlecleucel following completion of lymphodepleting chemotherapy. The ORR for the 68 evaluable patients was 50% (95% CI: 37.6, 62.4) with a CR rate of 32% (95% CI: 21.5, 44.8). With a median follow-up time of 9.4 months, the duration of response (DOR) was longer in patients with a best overall response of CR, as compared to a best overall response of partial response (PR). Among patients achieving CR, the estimated median DOR was not reached (95% CI: 10.0 months, not estimable [NE]). The estimated median response duration among patients in PR was 3.4 months (95% CI: 1.0, NE).[30, 31]

 

What is the relationship between non-Hodgkin lymphoma (NHL) and B-cell lymphoma?What is the difference between non-Hodgkin lymphoma and Hodgkin lymphoma?What is the prognosis of NHL/B-cell lymphoma?What is the survival rate of NHL/B-cell lymphoma?What is the survival rate of indolent non-Hodgkin lymphoma/B-cell lymphoma?How is complete remission achieved in aggressive forms of NHL/B-cell lymphoma?What are B-cell and T/natural killer (NK)-cell neoplasms?What are the WHO categories of B-cell malignancies in B-cell lymphoma?What is the WHO classification of mature B-cell neoplasms in B-cell lymphoma?What are the etiologic factors in B-cell lymphoma?What is the pathogenesis of B-cell lymphoma?What is the most common chromosomal abnormality in NHL/B-cell lymphoma?Which tests are indicated in the workup of B-cell lymphoma of germinal center origin?Which occupations are associated with a high risk of NHL/B-cell lymphoma?Which chemicals have been linked to NHL/B-cell lymphoma?Which cancer treatments are associated with an increased risk for NHL/B-cell lymphoma?Which viruses are linked to the pathogenesis of NHL/B-cell lymphoma?Which immunodeficiencies are associated with an increased risk of NHL/B-cell lymphoma?Which connective-tissue disorders are associated with an increased risk of NHL/B-cell lymphoma?Which GI conditions predispose patients to developing NHL/B-cell lymphoma?What is the mortality rate of NHL/B-cell lymphoma in the US?What is the risk of NHL/B-cell lymphoma in the US?How does the epidemiology of NHL/B-cell lymphoma differ between developing countries and developed countries?What are the most common types of B-cell lymphoma in developing countries?How common is NHL/B-cell lymphoma in Europe?What endemic factors are associated with NHL/B-cell lymphoma?Which condition is associated with NHL/B-cell lymphoma in individuals of Mediterranean ethnicity?What is the geographic distribution of follicular NHL/B-cell lymphoma?What is the age-related incidence of NHL/B-cell lymphoma?Is NHL/B-cell lymphoma more common in men or women?Does NHL/B-cell lymphoma have a racial predilection?What are the prognostic factors of NHL/B-cell lymphoma?What is the role of the International Prognostic Index (IPI) in the prognosis of NHL/B-cell lymphoma?Which risk factors for NHL/B-cell lymphoma are included in the International Prognostic Index (IPI)?What are the prognostic risk categories for NHL/B-cell lymphoma in the International Prognostic Index (IPI)?What are the 5-year survival rates for NHL/B-cell lymphoma according to the International Prognostic Index (IPI)?What is the most common manifestation of B-cell lymphoma?Which systemic symptoms are poor prognostic factors for B-cell lymphoma?Which organ-specific symptoms are associated with specific sites of involvement of B-cell lymphoma?What are the typical physical exam findings of B-cell lymphoma?Which physical findings are associated with advanced high-grade B-cell lymphoma?Which findings suggest additional workup is indicated in patients with B-cell lymphoma?What are the diagnostic considerations of NHL/B-cell lymphoma?What are the differential diagnoses for B-Cell Lymphoma?What are the approach considerations in the workup of NHL/B-cell lymphoma?Which procedure is preferred for the initial diagnosis of NHL/B-cell lymphoma?Which studies are indicated to confirm a diagnosis of NHL/B-cell lymphoma?Which lab studies are indicated in the workup of NHL/B-cell lymphoma?What is the role of an immunoglobulin (Ig) gene rearrangement in the workup of NHL/B-cell lymphoma?How is polymerase chain reaction (PCR) used in the workup of NHL/B-cell lymphoma?What is the role of polymerase chain reaction (PCR) in the workup of NHL/B-cell lymphoma?What are the preferred imaging studies for staging NHL/B-cell lymphoma?Which nodal lymphoma histologies are indicated in the workup of NHL/B-cell lymphoma?What is the role of PET/CT scanning in the workup of B-cell lymphoma?What is the role of contrast-enhanced CT scanning in the workup of NHL/B-cell lymphoma?When is evaluation of specific sites indicated in the workup of NHL/B-cell lymphoma?How is follicular lymphoma characterized in the histologic findings of NHL/B-cell lymphoma?How is Burkitt lymphoma characterized in the histologic findings of NHL/B-cell lymphoma?What are the histologic findings of NHL/B-cell lymphoma?How is chronic lymphocytic leukemia (CLL)/small lymphocytic leukemia (SLL) characterized in the histologic findings of NHL/B-cell lymphoma?How is mucosa-associated lymphoid tissue (MALT) lymphoma characterized in the histologic findings of NHL/B-cell lymphoma?How is nodal marginal zone lymphoma (NMZL) characterized in the histologic findings of NHL/B-cell lymphoma?How is diffuse large B-cell lymphoma (DLBCL) characterized in the histologic findings of follicular lymphoma?How is mantle cell lymphoma characterized in the histologic findings of B-cell lymphoma?What are the common morphologic features of diffuse large B-cell lymphoma (DLBCL)?What are the histologic findings in diffuse large B-cell lymphoma (DLBCL)?What are the immunophenotypic subdivisions of diffuse large B-cell lymphoma (DLBCL)?How is diffuse large B-cell lymphoma (DLBCL) classified?What are the subtypes of diffuse large B-cell lymphoma (DLBCL)?What are the types of large B-cell lymphoma?What is primary mediastinal (thymic) large B-cell lymphoma?What is the role of flow cytometry analysis in the workup of NHL/B-cell lymphoma?How does the Ann Arbor staging system classify NHL/B-cell lymphoma?How does the Lugano classification stage NHL/B-cell lymphoma?What other factors have important prognostic value in B-cell NHL?How is the International Prognostic Index (IPI) used in NHL/B-cell lymphoma?What are the approach considerations in the treatment of NHL/B-cell lymphoma?What complications are associated with treatment for NHL/B-cell lymphoma?Which specialist consultations are indicated in the treatment of NHL/B-cell lymphoma?When is transfer indicated for patients with NHL/B-cell lymphoma?What are the dietary recommendations for patients with NHL/B-cell lymphoma?What are the staging guidelines for NHL/B-cell lymphoma?What is the role of the International Prognostic Index (IPI) in low grade B-cell lymphoma?Which risk factors are included in the International Prognostic Index (IPI) in the prognosis of NHL/B-cell lymphoma?What are the risk categories of NHL/B-cell lymphoma in the International Prognostic Index (IPI) score?What are the survival rates of NHL/B-cell lymphoma based on the International Prognostic Index (IPI) score?What is the WHO classification for grading NHL/B-cell follicular lymphoma (FL)?What do the NCCN guidelines note about the treatment of grade 3 follicular lymphoma (FL) and diffuse large B-cell lymphoma (DLBCL)?How is primary cutaneous follicle center lymphoma (PCFCL) classified in the WHO guidelines on NHL/B-cell follicular lymphoma?Which studies does the NCCN recommend for the diagnosis of B-cell follicular lymphoma (FL)?What are the GELF criteria for immediate therapy in NHL/B-cell follicular lymphoma?What are the FLIPI risk stratification factors for NHL/B-cell follicular lymphoma?What are the FLIPI2 risk stratification factors for NHL/B-cell follicular lymphoma?What are the NCCN guidelines on first-line therapy in the treatment of NHL/B-cell follicular lymphoma?What are the first-line regimens for consolidation or extended dosing in the treatment of B-cell follicular lymphoma?What are the preferred regimens for second-line and subsequent therapy in the treatment of B-cell follicular lymphoma?What are the options for second-line consolidation or extended dosing in the treatment of B-cell follicular lymphoma?What are B-cell mucosa-associated lymphoid tissue (MALT) lymphomas?What causes B-cell gastric MALT lymphomas?What is the preferred treatment for B-cell non-gastric MALT lymphomas?How are B-cell gastric MALT lymphomas treated?How is B-cell mantle cell lymphoma (MCL) diagnosed?What is the risk stratification for B-cell mantle cell lymphoma (MCL)?What are the MIPI categories for risk stratification in B-cell mantle cell lymphoma (MCL)?What is the role of the Ki-67 proliferative assay in in the workup of B-cell mantle cell lymphomas (MCL)?What are the treatment recommendations for B-cell mantle cell lymphoma (MCL)?What are the induction therapy regimens for the treatment of B-cell mantle cell lymphoma (MCL)?What is the CALGB regimen for the treatment of B-cell mantle cell lymphoma (MCL)?What are the less aggressive regimens for the treatment of B-cell mantle cell lymphoma (MCL)?What is consolidation therapy for the treatment of B-cell mantle cell lymphoma (MCL)?What are the second-line therapy regimens for the treatment of B-cell mantle cell lymphoma (MCL)?Which studies are recommended by the NCCN to establish a diagnosis of diffuse large B-cell lymphoma (DLBCL)?What are the IPI risk factors for diffuse large B-cell lymphoma (DLBCL)?What are the IPI risk categories for diffuse large B-cell lymphoma (DLBCL)?What are the Ann Arbor IPI risk factors for diffuse large B-cell lymphoma (DLBCL)?What are the Ann Arbor IPI risk categories for diffuse large B-cell lymphoma (DLBCL)?What is the first-line therapy for the treatment of diffuse large B-cell lymphoma (DLBCL)?What is the first-line therapy for the treatment of diffuse large B-cell lymphoma (DLBCL) in patients who have poor left ventricular functions or are very frail?Which treatment regimens for diffuse large B-cell lymphoma (DLBCL) are indicated in elderly patients with comorbidities?What are the first-line consolidation/optional treatment regimens for diffuse large B-cell lymphoma (DLBCL)?What are the treatment regimens for diffuse large B-cell lymphoma (DLBCL) with concurrent CNS disease?What are the second-line and subsequent treatment regimens for diffuse large B-cell lymphoma (DLBCL)?What are the second-line and subsequent treatment regimens for diffuse large B-cell lymphoma (DLBCL) in patients who are not candidates for high-dose therapy?What is the follow-up for patients with diffuse large B-cell lymphoma (DLBCL) who are in complete remission?How does WHO classify the clinical variants of B-cell Burkitt lymphoma (BL)?Which studies are recommended to establish a diagnosis of B-cell Burkitt lymphoma (BL)?What are the SEER risk factors for B-cell Burkitt lymphoma (BL)?What are the SEER risk groups for B-cell Burkitt lymphoma (BL)?What are the SEER 5-year survival rates for B-cell Burkitt lymphoma (BL)?What induction and second-line therapy regimens are used in the treatment of Burkitt lymphoma?What are the NCCN recommendations for follow-up in patients with B-cell Burkitt lymphoma?Which organizations have issued treatment guidelines for cutaneous B-cell lymphomas (CBCL)?What are the main types of cutaneous B-cell lymphomas (CBCL)?Which studies are recommended to establish a diagnosis of cutaneous B-cell lymphoma (CBCL)?What are the TNM staging classifications for cutaneous B-cell lymphomas (CBCL)?What is the recommended treatment for cutaneous B-cell lymphomas (CBCL)?What are the goals of drug treatment for B-cell lymphoma?Which medications in the drug class PD-1/PD-L1 Inhibitors are used in the treatment of B-Cell Lymphoma?Which medications in the drug class Antineoplastics, Angiogenesis inhibitor are used in the treatment of B-Cell Lymphoma?Which medications in the drug class Antineoplastics, PI3K Delta Inhibitors are used in the treatment of B-Cell Lymphoma?Which medications in the drug class Antineoplastics, Tyrosine Kinase Inhibitor are used in the treatment of B-Cell Lymphoma?Which medications in the drug class Antineoplastic Agents are used in the treatment of B-Cell Lymphoma?Which medications in the drug class Corticosteroids are used in the treatment of B-Cell Lymphoma?Which medications in the drug class Monoclonal Antibodies are used in the treatment of B-Cell Lymphoma?Which medications in the drug class Hematopoietic Growth Factors are used in the treatment of B-Cell Lymphoma?Which medications in the drug class CAR T cell Therapy are used in the treatment of B-Cell Lymphoma?

Author

Mohammad Muhsin Chisti, MD, FACP, Assistant Professor of Hematology and Oncology, Medical Director of Research, Karmanos Cancer Institute, Wayne State University School of Medicine

Disclosure: Nothing to disclose.

Coauthor(s)

Haresh Kumar, MBBS, Resident Physician, Department of Internal Medicine, St Joseph Mercy Oakland Hospital

Disclosure: Nothing to disclose.

Specialty Editors

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

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

Chief Editor

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

Disclosure: Nothing to disclose.

Additional Contributors

Ajeet Gajra, MD, Associate Professor of Medicine, Director of Hematology/Oncology Fellowship Program, State University of New York Upstate Medical University; Consulting Staff, Department of Internal Medicine, Division of Hematology and Oncology, Veterans Affairs Medical Center

Disclosure: Nothing to disclose.

Neerja Vajpayee, MD, Associate Professor, Department of Pathology, State University of New York Upstate Medical University

Disclosure: Nothing to disclose.

Acknowledgements

The authors and editors of Medscape Reference gratefully acknowledge the contributions of previous authors Sara J Grethlein, MD, and Uzma Athar, MD, to the development and writing of the source article.

References

  1. Armitage JO. Treatment of non-Hodgkin's lymphoma. N Engl J Med. 1993 Apr 8. 328(14):1023-30. [View Abstract]
  2. SEER Cancer Statistics Factsheets: Non-Hodgkin Lymphoma. National Cancer Institute: Surveillance, Epidemiology, and End Results Program. Available at http://seer.cancer.gov/statfacts/html/nhl.html. Accessed: June 12, 2019.
  3. Swerdlow SH, Campo E, Harris NL, Jaffe ES, Pileri SA, Stein H, et al. WHO Classification of Tumors of Haematopoietic and Lymphoid Tissues. 4th Edition. Lyon, France: IARC; 2008.
  4. Taborelli M, Polesel J, Montella M, Libra M, Tedeschi R, Battiston M, et al. Hepatitis B and C viruses and risk of non-Hodgkin lymphoma: a case-control study in Italy. Infect Agent Cancer. 2016. 11:27. [View Abstract]
  5. Tanyildiz HG, Dincaslan H, Yavuz G, Unal E, Ikinciogulları A, Dogu F, et al. Lymphoma Secondary to Congenital and Acquired Immunodeficiency Syndromes at a Turkish Pediatric Oncology Center. J Clin Immunol. 2016 Aug 4. [View Abstract]
  6. Perry AM, Diebold J, Nathwani BN, MacLennan KA, Müller-Hermelink HK, Bast M, et al. Non-Hodgkin lymphoma in the developing world: review of 4539 cases from the International Non-Hodgkin Lymphoma Classification Project. Haematologica. 2016 Jun 27. [View Abstract]
  7. Ferlay J, Steliarova-Foucher E, Lortet-Tieulent J, Rosso S, Coebergh JW, Comber H, et al. Cancer incidence and mortality patterns in Europe: estimates for 40 countries in 2012. Eur J Cancer. 2013 Apr. 49 (6):1374-403. [View Abstract]
  8. International Non-Hodgkin's Lymphoma Prognostic Factors Project. A predictive model for aggressive non-Hodgkin's lymphoma. The International Non-Hodgkin's Lymphoma Prognostic Factors Project. N Engl J Med. 1993 Sep 30. 329(14):987-94. [View Abstract]
  9. Federico M, Bellei M, Marcheselli L, Luminari S, Lopez-Guillermo A, Vitolo U, et al. Follicular lymphoma international prognostic index 2: a new prognostic index for follicular lymphoma developed by the international follicular lymphoma prognostic factor project. J Clin Oncol. 2009 Sep 20. 27 (27):4555-62. [View Abstract]
  10. Solal-Celigny P, Roy P, Colombat P, et al. Follicular lymphoma international prognostic index. Blood. 2004 Sep 1. 104(5):1258-65. [View Abstract]
  11. Hoster E, Dreyling M, Klapper W, et al. A new prognostic index (MIPI) for patients with advanced-stage mantle cell lymphoma. Blood. 2008 Jan 15. 111(2):558-65. [View Abstract]
  12. Choi WW, Weisenburger DD, Greiner TC, et al. A new immunostain algorithm classifies diffuse large B-cell lymphoma into molecular subtypes with high accuracy. Clin Cancer Res. 2009 Sep 1. 15(17):5494-502. [View Abstract]
  13. American Joint Committee on Cancer, American Cancer Society, American College of Surgeons. Non-Hodgkin's lymphoma. Fleming ID, Cooper JS, Henson DE, Hutter RVP, Kennedy BJ, Murphy GP, O'Sullivan B, Sobin LH, Yarbro JW, eds. AJCC Cancer Staging Manual. 5th ed. Philadelphia, Pa: Lippincott-Raven; 1997. 289-294.
  14. Cheson BD, Fisher RI, Barrington SF, Cavalli F, Schwartz LH, et al. Recommendations for initial evaluation, staging, and response assessment of Hodgkin and non-Hodgkin lymphoma: the Lugano classification. J Clin Oncol. 2014 Sep 20. 32 (27):3059-68. [View Abstract]
  15. [Guideline] NCCN Clinical Practice Guidelines in Oncology. Non-Hodgkin's lymphomas. National Comprehensive Cancer Network. Available at http://www.nccn.org/professionals/physician_gls/pdf/nhl.pdf. Version 3.2016; Accessed: September 17, 2016.
  16. FDA approves CAR-T cell therapy to treat adults with certain types of large B-cell lymphoma. U.S. Food & Drug Administration. Available at https://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm581216.htm. October 18, 2017; Accessed: October 20, 2017.
  17. FDA approves tisagenlecleucel for adults with relapsed or refractory large B-cell lymphoma. U.S. Food and Drug Administration. 2018 May 03. Available at https://www.fda.gov/Drugs/InformationOnDrugs/ApprovedDrugs/ucm606540.htm?utm_campaign=Oncology%205%2F1%2F2018_Kymriah&utm_medium=email&utm_source=Eloqua&elqTrackId=5578022d641d45d5a723adaf982a493c&elq=933c3d8ba491426680135d5ae119dcc6&elqaid=3339&elqat=1&e
  18. Jaffe ES. The 2008 WHO classification of lymphomas: implications for clinical practice and translational research. Hematology Am Soc Hematol Educ Program. 2009. 523-31. [View Abstract]
  19. [Guideline] NCCN Clinical Practice Guidelines in Oncology. B-cell Lymphomas. National Comprehensive Cancer Network. Available at https://www.nccn.org/professionals/physician_gls/pdf/b-cell.pdf. Version 3.2019 — May 6, 2019; Accessed: June 12, 2019.
  20. Brice P, Bastion Y, Lepage E, Brousse N, Haïoun C, Moreau P, et al. Comparison in low-tumor-burden follicular lymphomas between an initial no-treatment policy, prednimustine, or interferon alfa: a randomized study from the Groupe d'Etude des Lymphomes Folliculaires. Groupe d'Etude des Lymphomes de l'Adulte. J Clin Oncol. 1997 Mar. 15 (3):1110-7. [View Abstract]
  21. [Guideline] Zucca E, Copie-Bergman C, Ricardi U, Thieblemont C, Raderer M, Ladetto M, et al. Gastric marginal zone lymphoma of MALT type: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2013 Oct. 24 Suppl 6:vi144-8. [View Abstract]
  22. [Guideline] Dreyling M, Geisler C, Hermine O, Kluin-Nelemans HC, Le Gouill S, Rule S, et al. Newly diagnosed and relapsed mantle cell lymphoma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2014 Sep. 25 Suppl 3:iii83-92. [View Abstract]
  23. Rosenwald A, Wright G, Chan WC, Connors JM, Campo E, et al. The use of molecular profiling to predict survival after chemotherapy for diffuse large-B-cell lymphoma. N Engl J Med. 2002 Jun 20. 346 (25):1937-47. [View Abstract]
  24. [Guideline] Tilly H, Gomes da Silva M, Vitolo U, Jack A, Meignan M, Lopez-Guillermo A, et al. Diffuse large B-cell lymphoma (DLBCL): ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2015 Sep. 26 Suppl 5:v116-25. [View Abstract]
  25. [Guideline] Senff NJ, Noordijk EM, Kim YH, Bagot M, Berti E, Cerroni L, et al. European Organization for Research and Treatment of Cancer and International Society for Cutaneous Lymphoma consensus recommendations for the management of cutaneous B-cell lymphomas. Blood. 2008 Sep 1. 112 (5):1600-9. [View Abstract]
  26. Willemze R, Jaffe ES, Burg G, et al. WHO-EORTC classification for cutaneous lymphomas. Blood. 2005 May 15. 105 (10):3768-85. [View Abstract]
  27. Kim YH, Willemze R, Pimpinelli N, Whittaker S, Olsen EA, Ranki A, 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 Jul 15. 110 (2):479-84. [View Abstract]
  28. Noy, A, et al. Single-Agent Ibrutinib Demonstrates Efficacy and Safety in Patients with Relapsed/Refractory Marginal Zone Lymphoma: A Multicenter, Open-Label, Phase 2 Study (abstract 1213). Presented at the 58th Annual American Society of Hematology (ASH) Meeting, San Diego, CA. 2016 Dec 5. Available at https://ash.confex.com/ash/2016/webprogram/Paper89393.html
  29. Frederick L, et al. Primary results from ZUMA-1: a pivotal trial of axicabtagene ciloleucel (axicel; KTE-C19) in patients with refractory aggressive non-Hodgkin lymphoma (NHL) [abstract]. Proceedings of the American Association for Cancer Research Annual Meeting 2017. 2017 Apr 1-5. Available at http://cancerres.aacrjournals.org/content/77/13_Supplement/CT019
  30. Schuster S, et al. Primary Analysis of Juliet: A Global, Pivotal, Phase 2 Trial of CTL019 in Adult Patients with Relapsed or Refractory Diffuse Large B-Cell Lymphoma. 59th American Society of Hematology Annual Meeting and Exposition. Abstract #577. Atlanta, GA. December 9-12, 2017.
  31. Schuster SJ, Svoboda J, Chong EA, Nasta SD, Mato AR, Anak Ö, et al. Chimeric Antigen Receptor T Cells in Refractory B-Cell Lymphomas. N Engl J Med. 2017 Dec 28. 377 (26):2545-2554. [View Abstract]
  32. Sehn LH, Chua N, Mayer J, Dueck G, Trněný M, Bouabdallah K, et al. Obinutuzumab plus bendamustine versus bendamustine monotherapy in patients with rituximab-refractory indolent non-Hodgkin lymphoma (GADOLIN): a randomised, controlled, open-label, multicentre, phase 3 trial. Lancet Oncol. 2016 Jun 23. [View Abstract]
  33. Barta SK, Li H, Hochster HS, Hong F, Weller E, Gascoyne RD, et al. Randomized phase 3 study in low-grade lymphoma comparing maintenance anti-CD20 antibody with observation after induction therapy: A trial of the ECOG-ACRIN Cancer Research Group (E1496). Cancer. 2016 Jun 28. [View Abstract]
  34. Kansara R, Connors JM, Savage KJ, Gerrie AS, Scott DW, Slack GW, et al. Maintenance rituximab following induction R-CHOP chemotherapy in patients with composite or discordant, indolent and aggressive, B-cell non-Hodgkin lymphomas. Haematologica. 2016 Jun 16. [View Abstract]
  35. [Guideline] Dreyling M, Ghielmini M, Marcus R, Salles G, Vitolo U, Ladetto M, et al. Newly diagnosed and relapsed follicular lymphoma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2014 Sep. 25 Suppl 3:iii76-82. [View Abstract]
  36. Colombat P, Salles G, Brousse N, et al. Rituximab (anti-CD20 monoclonal antibody) as single first-line therapy for patients with follicular lymphoma with a low tumor burden: clinical and molecular evaluation. Blood. 2001 Jan 1. 97(1):101-6. [View Abstract]
  37. Hainsworth JD, Litchy S, Shaffer DW, Lackey VL, Grimaldi M, Greco FA. Maximizing therapeutic benefit of rituximab: maintenance therapy versus re-treatment at progression in patients with indolent non-Hodgkin's lymphoma--a randomized phase II trial of the Minnie Pearl Cancer Research Network. J Clin Oncol. 2005 Feb 20. 23 (6):1088-95. [View Abstract]
  38. Stiff PJ, Unger JM, Cook JR, Constine LS, Couban S, Stewart DA, et al. Autologous transplantation as consolidation for aggressive non-Hodgkin's lymphoma. N Engl J Med. 2013 Oct 31. 369(18):1681-90. [View Abstract]
  39. Hambley B, Caimi PF, William BM. Bortezomib for the treatment of mantle cell lymphoma: an update. Ther Adv Hematol. 2016 Aug. 7 (4):196-208. [View Abstract]
  40. Robak T, Huang H, Jin J, Zhu J, Liu T, Samoilova O, et al. Bortezomib-based therapy for newly diagnosed mantle-cell lymphoma. N Engl J Med. 2015 Mar 5. 372 (10):944-53. [View Abstract]
  41. Zhou Z, Sehn LH, Rademaker AW, Gordon LI, Lacasce AS, Crosby-Thompson A, et al. An enhanced International Prognostic Index (NCCN-IPI) for patients with diffuse large B-cell lymphoma treated in the rituximab era. Blood. 2014 Feb 6. 123 (6):837-42. [View Abstract]
  42. Castillo JJ, Winer ES, Olszewski AJ. Population-based prognostic factors for survival in patients with Burkitt lymphoma: an analysis from the Surveillance, Epidemiology, and End Results database. Cancer. 2013 Oct 15. 119 (20):3672-9. [View Abstract]
  43. WHO's cancer pain ladder for adults. World Health Organization. Available at http://www.who.int/cancer/palliative/painladder/en/. Accessed: August 10, 2016.
  44. 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 May 15. 49 (10):2112-35. [View Abstract]
  45. Harris NL, Jaffe ES, Stein H, Banks PM, Chan JK, Cleary ML, et al. A revised European-American classification of lymphoid neoplasms: a proposal from the International Lymphoma Study Group. Blood. 1994 Sep 1. 84 (5):1361-92. [View Abstract]
  46. Rohatiner A, d'Amore F, Coiffier B, Crowther D, Gospodarowicz M, Isaacson P, et al. Report on a workshop convened to discuss the pathological and staging classifications of gastrointestinal tract lymphoma. Ann Oncol. 1994 May. 5 (5):397-400. [View Abstract]
  47. Ruskoné-Fourmestraux A, Dragosics B, Morgner A, Wotherspoon A, De Jong D. Paris staging system for primary gastrointestinal lymphomas. Gut. 2003 Jun. 52 (6):912-3. [View Abstract]

Neoplastic follicles comprising cleaved cells (centrocytes) and larger cells with vesicular nuclei and prominent 2-3 nucleoli (centroblasts).

Mantle cell lymphoma. Small lymphoid cells with oval to slightly irregular nuclei and clumped chromatin and rare admixed pink histiocytes.

Diffuse large B-cell non-Hodgkin lymphoma. Large cells with abundant cytoplasm and large round-ovoid nuclei with thick nuclear membrane and multiple prominent nucleoli.

Burkitt lymphoma. Normal architecture is entirely replaced by lymphoma cells and evenly dispersed macrophages, starry sky (250×).

Burkitt lymphoma cells with round noncleaved nuclei and strongly basophilic cytoplasm (1000×).

Compartmentalizing fibrosis and infiltrate of medium-sized to large lymphoid cells.

Neoplastic follicles comprising cleaved cells (centrocytes) and larger cells with vesicular nuclei and prominent 2-3 nucleoli (centroblasts).

Mantle cell lymphoma. Small lymphoid cells with oval to slightly irregular nuclei and clumped chromatin and rare admixed pink histiocytes.

Diffuse large B-cell non-Hodgkin lymphoma. Large cells with abundant cytoplasm and large round-ovoid nuclei with thick nuclear membrane and multiple prominent nucleoli.

Burkitt lymphoma. Normal architecture is entirely replaced by lymphoma cells and evenly dispersed macrophages, starry sky (250×).

Burkitt lymphoma cells with round noncleaved nuclei and strongly basophilic cytoplasm (1000×).

Compartmentalizing fibrosis and infiltrate of medium-sized to large lymphoid cells.

Lymphoma cells show strong membranous positivity with CD20 indicative of B-cell origin.

Cytogenetic Abnormality Histology Antigen Rearrangement Oncogene Expression
t(14;18)(q32;q21)Follicular, diffuse large cellIgH*bcl- 2
t(11;14)(q13;q32)Mantle cellIgH cyclin-D1/bcl-1
t(1;14)(p22;q32)MALT lymphomaIgHbcl- 10
t(11;18)(q21;q21)MALT lymphomaIgHUnknown
t(9;14)(p13;q32)Lymphoplasmacytic lymphomaIgHPAX-5
8q24 translocations



t(8;14)(q24;q32)



t(2;8)(p11-12;q24)



t(8;22)(q24;q11)



Burkitt lymphomaIgH



Ig-8



Ig-6



c-myc
Trisomy 12, deletion 11q22-23, 17p13, and 6q21CLL
*Immunoglobulin H (IgH)



MALT = Mucosa-associated lymphoid tissue.



Lymphoma Immunophenotype
FollicularCD20+, CD3-, CD10+, CD5-, BCL2+, CD23-/+, CD43-, cyclinD-1-, BCL6+
Small lymphocyticCD 20+, CD3-, CD10-, CD5+, CD23+,
MALTCD20+, CD3-, CD 10-, CD5-, CD23-/+, BCL2+
Marginal zoneCD20+, CD3-, CD 10-, CD5-, CD23-/+, CD43-/+, cyclinD-1-, BCL2
Mantle cellCD20+, CD23-/+, CD10-/+, CD5+, CD43-/+, cyclinD-1+
Mediastinal large B-cellCD20+, CD30+, MUM-1+
Burkittslg+,CD20+, Tdt-, CD10+, BCL2--, BCL6+, Ki-67+ (≥95%)
B-lymphoblasticslg-, CD19+, CD10-/+, CD20-/+, TdT+
MALT = mucosa-associated lymphoid tissue.
Stage Area of Involvement Extranodal (E) Status
ISingle node or adjacent group of nodesSingle extranodal lesions without nodal involvement
IIMultiple lymph node groups on same side of diaphragmStage I or II by nodal extent with limited contiguous extranodal involvement
II bulky*Multiple lymph node groups on same side of diaphragm with “bulky disease”.N/A
IIIMultiple lymph node groups on both sides of diaphragm; nodes above the diaphragm with spleen involvementN/A
IVMultiple noncontiguous extranodal sitesN/A
*Stage II bulky disease considered limited or advanced as determined by histology and a number of prognostic factors.



Suffixes A and B are not required



X for bulky disease replaced with documenting of largest tumor diameter



Definition of “Bulky” disease varies depending on lymphoma histology.



Stage Area of Involvement Extranodal (E) Status
ISingle node or adjacent group of nodesSingle extranodal lesions without nodal involvement
IIMultiple lymph node groups on same side of diaphragmStage I or II by nodal extent with limited contiguous extranodal involvement
II bulky*Multiple lymph node groups on same side of diaphragm with “bulky disease”N/A
IIIMultiple lymph node groups on both sides of diaphragm; nodes above the diaphragm with spleen involvementN/A
IVMultiple noncontiguous extranodal sitesN/A
Tumor Involvement Node Involvement Metastatic Spread Involvement
T1Solitary skin involvement



T1a:  ≤5 cm diameter



T1b:  >5 cm diameter



N0No lymph node involvementM0No evidence of extracutaneous non-lymph node disease
T2Multiple lesions limited to one body region or two contiguous body regions



T2a: all-disease in a < 15-cm diameter



T2b: all-disease in a >15- and < 30-cm diameter



T2c: all-disease in a >30-cm diameter



N1Involvement of one peripheral lymph node regionM1Evidence of extracutaneous non-lymph node disease
T3Generalized skin involvement



T3a: multiple lesions involving two noncontiguous body regions



T3b: multiple lesions involving three body regions



N2Involvement of two or more peripheral lymph node regions or involvement of any lymph node region that does not drain an area of current or prior skin involvement  
  N3Central lymph nodes involvement