U.S. flag

An official website of the United States government

NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.

StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2024 Jan-.

Cover of StatPearls

StatPearls [Internet].

Show details

Chronic Lymphocytic Leukemia

; ; ; .

Author Information and Affiliations

Last Update: March 7, 2023.

Continuing Education Activity

Chronic lymphocytic leukemia is a chronic lymphoproliferative disorder characterized by monoclonal B cell proliferation. It is the most common adult leukemia in Western populations and comprises 25 to 30 percent of leukemias in the United States. This activity reviews the evaluation and management of chronic lymphocytic leukemia and highlights the role of interprofessional team members in collaborating to provide well-coordinated care and enhance outcomes for affected patients.

Objectives:

  • Outline some of the epidemiological factors associated with the development of chronic lymphocytic leukemia.
  • Describe exam findings consistent with chronic lymphocytic leukemia.
  • Explain management considerations for chronic lymphocytic leukemia.
  • Describe how coordination of the interprofessional team can lead to more rapid diagnosis of chronic lymphocytic leukemia and subsequently decrease associated morbidity and mortality in affected patients.
Access free multiple choice questions on this topic.

Introduction

Chronic lymphocytic leukemia (CLL) or small lymphocytic lymphoma (SLL) is an indolent malignancy characterized by increased production of mature but dysfunctional B lymphocytes. CLL/SLL is defined as a monoclonal lymphoproliferative disease characterized by the proliferation and accumulation of morphologically mature but immunologically dysfunctional B-cell lymphocytes that are smudge cells, as noted on peripheral smear. The primary disease sites include peripheral blood, spleen, lymph nodes, and bone marrow. CLL and SLL are identical from a pathologic and immunophenotypic standpoint. Both CLL and SLL originate from B-cell lymphocytes but present with different manifestations depending on where the abnormal cells are found.  Usually, the initial leukemic phase represents CLL, where the cells are present in the blood.  This eventually progresses to the lymphoma phase, representing SLL, where the cells are found in the lymph nodes. The term SLL is commonly used to represent the lymphoproliferative process limited to the lymph nodes. 

Etiology

The exact etiology of CLL is unknown. Genetic factors, rather than environmental factors, are the most likely cause of CLL. However, few known risk factors for CLL include occupational causes by exposure to certain chemicals, radiation exposure, and tobacco users.  Reports of farmers working around rubber manufacturing industries and workers with exposure to benzene and heavy solvent have shown an increased risk of CLL/SLL. However, these associations have not yet been proven. In atomic bomb survivors, no known increase in the incidence of CLL/SLL has been noted. However, there has been an increased risk of other types of leukemia. The uranium miner population, who suffer exposure to ionizing and non-ionizing radiation, has shown increased CLL incidence. Tobacco users and cigarette smokers show a significantly elevated risk of CLL compared to non-tobacco users. The Veterans Affairs recognizes CLL as having a relationship to exposure to Agent Orange or other herbicides during military service.[1][2][3][4][5][6][7][8][9][10]

Epidemiology

CLL comprises 25 to 30% of total leukemias in the United States. According to the American Cancer Society, there will be approximately 21,040 new CLL cases and about 4,060 deaths in the year of 2020. Worldwide, 191,000 cases and 61,000 deaths are attributed to CLL/SLL every year. CLL can affect adults as young as 30 years of age. However, it is mostly seen in adults with an average age of 70 years. CLL is extremely rare in children. The incidence is known to rapidly increase with increasing age. CLL has a slightly higher incidence in male populations than female populations (1.3 to 1 to 1.7 to 1). However, studies have shown that women can have a more aggressive form of the disease than men.[11][12][13]

The incidence of CLL varies by geographic location and race. CLL is most commonly seen in adults of the Western population. It is high amongst the Caucasian population compared to the Asian Pacific Islanders or the African-American population. The incidence of CLL in Western countries is similar to that of the United States but is rarely seen in Asian countries (China and Japan).  CLL is common amongst the Jews of Eastern European descent. It is most commonly seen in Non-Hispanic Whites and least common in Asians. The incidence in African Americans is in between the Caucasian and the Asian ethnicity groups.[12][14][15][16][17][18][19]

CLL is reported to have a genetic basis and is known to run in families (familial CLL). The age at diagnosis of the second-generation offspring is nearly 20 years younger as compared to the parent. First-degree relatives (siblings, children, or parents) of CLL patients have double the risk for CLL. Moreover, 17% of first-degree family members of CLL patients had monoclonal B cell lymphocytosis, which is a precursor of CLL. Ultimately only a small percentage of patients with monoclonal B-cell lymphocytosis (MBL) will develop into CLL.[20][21][22]

Pathophysiology

The pathogenesis of CLL/SLL is a two-step process that leads to the clonal replication of malignant B lymphocytes.  The first step is the development of MBL cells secondary to multiple factors such as antigenic stimulation, genetic mutations, and cytogenetic abnormalities.  The second step is the progression of MBL to CLL/SLL by the further insult to the B-cell clone, either due to additional genetic abnormalities or changes in the bone marrow microenvironment. B-cell antigen receptor (BCR) expression induces antigen-independent cell-autonomous signaling, which is an important step in the pathogenesis of CLL.[23][24]

In CLL, CD5+ B cells are continuously activated by mutations leading to MBL. The accumulation of genetic abnormalities in more mature B cells causes a clonal division of the neoplastic B-cell within the lymph nodes. In CLL/SLL, the most common abnormal physical examination findings are lymphadenopathy, which is seen in 50 to 90% of patients. Cervical, supraclavicular, and axillary lymph nodes are the most commonly affected sites. These increased B-cell lymphocytes eventually spill into the peripheral blood leading to the detection of lymphocytosis on a CBC.[25][26][27]

These neoplastic B cells escape apoptosis and continue to divide over time within the lymph nodes. They then infiltrate the spleen and bone marrow, causing splenomegaly and hypercellular bone marrow (on bone marrow biopsy).  The splenomegaly leads to increase sequestration of RBCs and platelets, leading to anemia and thrombocytopenia by decreasing the RBCs and platelets.  Patients are more susceptible to autoimmune hemolytic anemia (positive Coombs test) and autoimmune thrombocytopenia. These B cells eventually spread throughout the body, causing systemic symptoms such as fever, night sweats, unintentional weight loss, fatigue, and early satiety.  Lack of functional B cells decreases the body's ability to produce antibodies for immune responses leading to hypogammaglobinemia, which eventually leads to an increased risk of infection.[28][29][30][31][32]

Skin is the most commonly affected nonlymphoid tissue in patients with CLL.  Leukemia cutis (skin lesions) mainly involves the face and manifests as papules, macules, plaques, ulcers, blisters, or nodules.  Skin biopsy can help confirm the diagnosis of CLL.  Nonspecific secondary cutaneous lesions may occur due to bleeding, vasculitis, and infection.  Exaggerated reactions to insect bites have been reported in patients as well.[33][34][35][36]

Histopathology

The first and foremost laboratory abnormality found in CLL is lymphocytosis in peripheral blood and bone marrow. The peripheral blood smear shows leukemic cells, which are small, mature lymphocytes with a darkly stained nucleus, condensed chromatin, and indistinguishable nucleoli with a narrow rim of basophilic cytoplasm. Classical "smudge" cells ("basket" cells) are also seen on peripheral blood smear; these are pathognomic of CLL. The smudge cells are more fragile than normal lymphocytes that are disrupted during the process of being spread on a glass slide.[25][37]

Immunophenotypic analysis of the peripheral circulating lymphocytes by peripheral blood flow cytometry can be performed, which can help to diagnose CLL. Most cases of CLL can be identified using antibody specific panel for CD5, CD19, CD20, CD23, and immunoglobulin light chains. Characteristic CLL/SLL lymphocyte phenotype features include the following: low levels of immunoglobulins (most often IgM immunoglobulin and sometimes both IgM and IgD); expression of B-cell associated antigens (CD19, CD20, CD21, CD23, and/or CD24); and expression of CD5 which is a T-cell associated antigen.  The most common immunophenotype expression of CLL/SLL is the coexpression of CD5, CD19, and CD23. As for the light chain immunoglobulins, only one type of light chain is seen, suggesting the monoclonality of the lymphocytes. However, in some cases, biclonal CLL expressing both light chains or different levels of expression of other immunophenotypic antigens is also seen.[38][39][25][40][41][42][43]

Fluorescence in situ hybridization (FISH) is a highly sensitive test used to detect chromosomal abnormalities in patients with CLL/SLL. Cytogenetics evaluation of the peripheral blood smear with FISH for 17p. deletion, 11q. deletion, 13q. deletion and trisomy 12 is routine pretreatment evaluation of patients with CLL. Though all patients with symptomatic and advanced-stage CLL/SLL are treated similarly, patients with 17p. deletion or 11q. deletion requires special consideration.[44][45][46]17p. deletion causes CLL by TP53 mutation.  TP53 is a tumor suppressor gene which is located on the short arm of chromosome 17. The normal "wild–type" TP53 gene activation occurs in response to damaged DNA and/or other stressors such as hypoxia, leading to cellular apoptosis. The deletion/point mutation of TP53 leads to CLL. 11q chromosome contains the ataxia-telangiectasia mutated (ATM) gene.  ATM kinase is responsible for the delayed progression of the cell cycle in the presence of DNA damage, allowing the cell to repair the damage.  ATM kinase phosphorylates tumor suppressor p53 protein in the presence of DNA damage.  The phosphorylation of p53 protein ultimately leads to cell cycle arrest/apoptosis of cells with DNA damage.  In the absence of ATM kinase in 11q. deletion, phosphorylation of p53 does not occur, which hence cannot prevent the cell cycle arrest in DNA damaged cells.[44][47][48][49][50]

History and Physical

Patients with CLL are often asymptomatic at the initial presentation, when a routine CBC reveals abnormal lymphocytosis, leading to CLL diagnosis. Approximately 5 to 10% of patients with CLL are symptomatic with B symptoms such as Fever of > 100.5 degrees F for > 2 weeks with no evidence of infection; unintentional weight loss >/= 10% of body weight over the last 6 months; drenching night sweats with no evidence of infection; extreme fatigue; and early satiety.[25]

Upon physical examination, 50 to 90% of the patients with CLL present with localized/generalized lymphadenopathy.  The most common sites include cervical, supraclavicular, and axillary lymph nodes. On palpation, the nodes are firm, non-tender, round, and freely mobile.  Splenomegaly is the second most commonly enlarged lymphoid organ, seen in 25 to 55% of cases. On palpation, it is painless on non-tender with a smooth, firm surface.  Hepatomegaly is seen in 15 to 25% cases.  The liver is mildly enlarged and palpated 2-6 cm below the right costal margin.  On palpation, the liver is firm and non-tender with a smooth surface.[26][27]

Skin examination is an important part of physical examination because skin cancers are a relatively frequent CLL complication. Skin is the most common non-lymphoid tissue involved in patients with CLL. Leukemia cutis (skin lesions) mainly involve the face and manifest as papules, macules, plaques, ulcers, blisters, or nodules.  Skin biopsy can help confirm the diagnosis of CLL. Nonspecific secondary cutaneous lesions may occur due to bleeding, vasculitis, and infection.  Exaggerated reactions to insect bites have been reported in for patients as well.[33][34][35][36]

Infiltration of the spleen and bone marrow with neoplastic B cells causes splenomegaly and hypercellular bone marrow. The splenomegaly leads to increased sequestration of RBCs and platelets, leading to anemia and thrombocytopenia by decreasing the RBCs and platelets. Patients with anemia are symptomatic with fatigue and shortness of breath; patients with thrombocytopenia easily bleed/bruises, and petechiae can be seen on physical examination. Lack of functional B cells decreases the body's ability to produce antibodies for immune responses leading to hypogammaglobinemia, which eventually leads to an increased risk of infection.[28][29][30][31][32]

Evaluation

The first step in the diagnosis of CLL is a peripheral blood smear. The peripheral blood smear shows an absolute lymphocyte count of greater than 5000/mcL and smudge cells that confirm CLL. Although the diagnostic criteria for CLL are >=5000/mcL B lymphocytes on peripheral smear, a large proportion of patients present with an absolute lymphocyte count > 100,000/mcL. In patients with SLL, peripheral blood smear shows an absolute lymphocyte count of less than 5000/mcL along with lymphadenopathy but without cytopenias. However, lymph node biopsy is required to confirm the diagnosis of SLL.[25][51][25]

Immunophenotypic analysis of the peripheral circulating lymphocytes by peripheral blood flow cytometry can be performed, which can help confirm the clonality of circulating B cells in CLL patients. Flow cytometry can be performed on both peripheral blood and bone marrow aspirate to look for the classical immunophenotypic markers of CLL. To reiterate, characteristic CLL/SLL lymphocyte phenotype features include low levels of immunoglobulins (most often IgM immunoglobulin and sometimes both IgM and IgD); expression of B-cell associated antigens (CD19, CD20, CD21, CD23, and/or CD24); and expression of CD5 which is a T-cell associated antigen. The most common immunophenotype expression of CLL/SLL is the coexpression of CD5, CD19, and CD23. However, different levels of expression of other immunophenotypic antigens are also seen. Serum immunoglobulins and free-light chains are measured at baseline to assess the immunodeficiency and during treatment, to look at immune reconstitution, especially with the newer generation B-cell receptor signaling drugs. As for the light chain immunoglobulins, only one type of light chain suggesting the monoclonality of the lymphocytes. Rarely, some patients express both kappa and lambda light chains, known as "biclonal CLL."[38][39][25][40][41][42][43][25]

FISH is a highly sensitive test used to detect chromosomal abnormalities in patients with CLL/SLL.

Although not needed for diagnosis, bone marrow aspiration and biopsy are often done as a part of a diagnostic workup or before treatment. If the biopsy specimen demonstrates greater than 30% lymphocytes of all nucleated cells in a normocellular/hypercellular bone marrow aspirate, this confirms the diagnosis of CLL. The reduction of lymphocytic infiltration to less than 30% on treatment indicates a complete response. Furthermore, three infiltrative patterns of lymphocytes have been recognized in the bone marrow biopsy specimens: nodular, interstitial, and diffuse. A biopsy sample can demonstrate a mixture of nodular and interstitial, or nodular and diffuse patterns. Studies have shown that patients with a diffuse pattern on biopsy tend to have advanced disease with poor prognosis, whereas nodular and interstitial patterns ("non-diffuse" category) have a better prognosis.[52][53][54]

Excisional lymph node histology demonstrates diffuse effacement of nodal architecture along with some scattered residual likely germinal centers. These lymph node infiltrates are predominantly composed of small lymphocytes. However, large lymphoid cells, such as pro-lymphocytes, are always present in clusters and are known as " pseudo-follicles" (proliferation centers), a pathognomic finding, and CLL/SLL patients. Spleen histology demonstrates the infiltration of red and white pulp with a more prominent white pulp involvement compared to red pulp. From an imaging standpoint, a CT scan helps in evaluation to see the degree of lymphadenopathy and organ infiltration in the form of spleen and liver sizes.[25]

Diagnoses of complications of CLL such as autoimmune hemolytic anemia involves a positive direct antiglobulin (Coombs) test, increased reticulocyte count, elevated serum LDH, reduced haptoglobin, and elevated serum indirect bilirubin. Diagnosis of further complications such as pure red cell aplasia and thrombocytopenia can be made by a peripheral blood smear and a bone marrow aspiration and biopsy. Hypogammaglobulinemia (less than 15% of cases), elevated uric acid level and elevated hepatic enzymes are other findings seen in CLL. Important lab studies include serum lactate dehydrogenase (LDH) and beta-2 microglobulin (read with creatinine because it can normally be elevated in patients with elevated creatinine), which correlate with disease activity.[55]

Treatment / Management

All patients diagnosed with CLL will not require treatment because CLL is a heterogeneous disease. Certain patients receiving no treatment have survival rates similar to that of a normal population; current treatment regimens cannot cure CLL except for allogeneic hematopoietic stem cell transplantation (HCT). Randomized control trials between immediate versus delayed treatments found no significant improvement in long-term survival with early treatment initiation.[56][57]

According to the International Workshop on Chronic Lymphocytic Leukemia (iwCLL) criteria for "active disease," indications for treatment include [25] :

  • Constitutional symptoms in patients such as fevers >100.5 degrees F or 38.0 degrees C for ≥2 weeks without evidence of infection, night sweats for ≥1 month without evidence of infection, unintentional weight loss of ≥10% within the previous 6 months, extreme fatigue, and early satiety.  
  • Progressive lymphocytosis with >50% increase in lymphocytes over a 2-month period or lymphocyte doubling time (LDT) of <6 months. LDT is obtained by linear regression extrapolation of absolute lymphocyte counts at every 2 weeks interval period over a time span of 2 to 3 months. Patients with initial blood lymphocyte counts of <30,000/mcL may need to be observed for a longer time period to help determine the LDT. Also, other lymphocytosis/lymphadenopathy contributing factors (e.g., infection) other than CLL should be excluded.
  • Patients with rapid disease progression such as massive (i.e., ≥6 cm below the left costal margin)/progressive/symptomatic splenomegaly or massive nodes (i.e., ≥10 cm in longest diameter)/progressive/symptomatic lymphadenopathy.
  • Autoimmune hemolytic anemia and/or autoimmune thrombocytopenia that is poorly responsive or unresponsive to corticosteroids. 
  • Evidence of progressive bone marrow failure by developing/worsening/symptomatic anemia and/or thrombocytopenia.
  • Patients with recurrent infections.

Treatment of CLL is further classified into; 

A. Patients without "active disease": This is further divided into early-stage asymptomatic CLL and localized SLL.

1.  Early-stage asymptomatic CLL

  • Initial observation as standard care: CLL patients who are asymptomatic, or patients with early-stage disease (e.g., Rai stage <3, Binet stage A or B), the standard of care is observation, as they do not require treatment. During this observation period, the clinical examination of the patient and blood counts should occur every 3 months.  Based on these evaluations, at the end of 12 months, a decision is to be made whether the patient requires aggressive treatment or to continue observation of the patient.  Several randomized control trials support observation over aggressive treatment in patients with asymptomatic/early-stage CLL.[57][58]
  • International Prognostic Score for Early-stage CLL (IPS-E): Some early-stage CLL patients may require treatment within the first few years of diagnosis, while others remain asymptomatic without treatment for many years. The International Prognostic Score for Early-stage CLL (IPS-E) uses three variables (unmutated IGHV, lymphocytes >15,000/mcL, palpable lymph nodes) to further divide patients with early-stage CLL into three groups based on the number of risk factors to evaluate treatment requirement at one and five years:[59]
    • Low risk (no risk factors) – <1 percent treated at 1 year; 8 percent treated at 5 years
    • Intermediate risk (one risk factor) – 3 percent treated at 1 year; 28 percent treated at 5 years
    • High risk (two or three risk factors) – 14 percent treated at 1 year; 61 percent treated at 5 years 

2.  Localized SLL

  • Some patients present with small lymphocytic lymphoma (SLL), which involves a single lymph node site.  First-line management for localized SLL is similar to patients with early-stage asymptomatic CLL, which is observation unless there is clear evidence of "active disease" for which radiation therapy (RT) is used. In patients with stage I SLL contemplating radiation therapy, extensive pretreatment staging evaluation with imaging, and bone marrow biopsy is required to rule out an extensive disease involvement.  Limited data is available from small retrospective studies regarding the use of radiation therapy and localized SLL.[60]

B. Patients with Active Disease

The main purpose of systemic therapy is for symptomatic relief, prolonged remission, and prolonged survival in patients. Symptomatic CLL is treated, and despite many recent advances, it remains an incurable disease. For those patients for whom treatment is considered, a pre-treatment assessment should be done, including gathering information about age and general health, other features such as TP53 abnormalities or adverse cytogenetics, relapsed disease, or treatment-naive status. The selection of initial therapy in patients with symptomatic/advanced CLL is based on the patient, patient preference, tumor characteristics, and therapy goals.  Various treatment approaches for CLL use different combinations of the following agents. These agents significantly differ in their rates of time to progression, measurable residual disease (MRD), complete remission, and toxicities of drugs. There have been studies comparing different treatment modalities, and an individualized approach needs to be considered. Hence, patient preference plays a vital role in the ultimate treatment decision. Various drugs utilized in the management of CLL include;

  • Bruton tyrosine kinase inhibitors (ibrutinib, acalabrutinib)
  • BCL-2 inhibitor (venetoclax)
  • Purine analogs (fludarabine, pentostatin)
  • Alkylating agents (cyclophosphamide, chlorambucil, bendamustine)
  • Monoclonal antibodies (rituximab, ofatumumab, obinutuzumab)
  • PI3K inhibitor (idelalisib)

Choice of Therapy: The preferred initial treatment choice is based upon genetic risk stratification of the tumor along with patient fitness assessment.

1. Genetic risk stratification is described as following:

  1. Very high-risk disease: 17p deletion and/or TP53 mutations
  2. High-risk disease: IGHV unmutated (without 17p deletion and TP53 mutation)
  3. Standard-risk disease: IGHV mutated (without 17p deletion and TP53 mutation)

2. Fitness Assessment:  includes an assessment of comorbidities and the impact of these comorbidities on general function, liver function, and kidney function. Tools used for this assessment include the Eastern Cooperative Oncology Group performance status (ECOG PS), Cumulative illness rating scale (CIRS), geriatric assessment tools, and creatinine clearance to estimate renal function. Eligibility for targeted agents varies, depending upon the toxicity of the agent being used and the predicted tolerability based upon the patient's general fitness and comorbidities. Eligibility is more restrictive for chemoimmunotherapy. Patients are usually considered unfit for chemoimmunotherapy with intense regimens such as fludarabine, cyclophosphamide, and rituximab (FCR) if they have one or more of the following findings: ECOG PS of >/=2, CIRS >6, significant hepatic impairment (Child-Pugh class B or C), and creatinine clearance (CrCl) <70 mL/min.[61]

a. Very high-risk disease: 17p deletion and/or TP53 mutations:  These patients are at high risk of not responding to initial treatment with chemoimmunotherapy, or relapsing immediately after achieving remission. Initial treatment options include:

  • Single-agent ibrutinib until progression (all ages)–is preferred based upon good tolerability of the drug, longer follow-up, and high response rates.
  • Ibrutinib plus rituximab until progression (younger patients)– studies have shown that the addition of rituximab improved outcomes in younger adults compared to older adults.[62]
  • Fixed duration venetoclax plus obinutuzumab for one year (all ages)[63][64][63]
  • Single-agent venetoclax until progression (all ages)
  • Single-agent acalabrutinib until progression (all ages)

b. High-risk disease: IGHV unmutated (without 17p deletion and TP53 mutation): These patients do respond to initial treatment with chemoimmunotherapy; however, targeted agents are preferred due to an improvement in progression-free survival (PFS) and overall survival (OS) benefit compared to other chemoimmunotherapy. Treatment of choice is strongly based on patient preference and patient comorbidities. Initial treatment options include:

  • Single-agent ibrutinib until progression (all ages)
  • Ibrutinib plus rituximab until progression (younger patients)
  • Fixed duration venetoclax plus obinutuzumab (all ages, use in younger patients extrapolated from trials in older adults)
  • Acalabrutinib until progression (all ages)

c. Standard-risk disease: IGHV mutated (without 17p deletion and TP53 mutation):  In these patients, PFS and OS appear to be similar regardless of targeted therapy or chemoimmunotherapy being used in the patient. These patients are further categorized into Fit and Unfit based upon their Fitness assessment.

Fit Patients: Clinically fit patients with IGHV-mutated CLL and without 17p deletion or TP53 mutation may either choose targeted therapy or chemoimmunotherapy depending upon patient preference and Fitness assessment. Intensive Chemoimmunotherapy (FCR or BR) may be preferred by patients desiring a long treatment-free interval. Options include:

  • Fludarabine, cyclophosphamide, rituximab (FCR for six cycles (younger patients).
  • Bendamustine plus rituximab (BR) for six cycles (older patients).
  • Ibrutinib as a single agent (all ages) or in combination with rituximab or obinutuzumab (younger patients).
  • Fixed duration venetoclax plus obinutuzumab (all ages).
  • Acalabrutinib as a single agent (all ages).

Unfit Patients: The management of patients with limited functional status is informed by the genetic features of the tumor (17p deletion, TP53 mutation, and IGHV mutation status). Many clinically unfit patients with comorbidities may benefit from treatment with single-agent ibrutinib, fixed duration venetoclax plus obinutuzumab, single-agent acalabrutinib, and/or the combination of bendamustine plus rituximab. Chlorambucil plus obinutuzumab is a treatment option for IGHV-mutated CLL in older patients with comorbidities.

Initial Treatment Options

1. Targeted Agents

Bruton tyrosine kinase (BTK) inhibitors

  • Single-agent ibrutinib is an effective treatment for older patients with CLL. It has demonstrated improvement in both overall survival (OS) and progression-free survival (PFS) when compared to chlorambucil in older patients and when compared to the combination of bendamustine plus rituximab (BR). In younger patients, ibrutinib plus rituximab or obinutuzumab has demonstrated an overall PFS and OS benefit when compared to the combination of fludarabine plus cyclophosphamide and rituximab. Acalabrutinib is another BTK inhibitor that improves PFS when compared to chlorambucil plus obinutuzumab.[65][66][67][65]
  • A combination of venetoclax plus obinutuzumab is particularly used in patients in whom ibrutinib is contraindicated, such as patients with multiple comorbidities (example, history of severe bleeding, hepatic impairment, or atrial fibrillation) or if on any anticoagulation medications.  Studies have shown that the combination of venetoclax plus obinutuzumab demonstrates higher PFS and OS when compared with chlorambucil plus obinutuzumab.[63][64]

2. Chemoimmunotherapy

  • Fludarabine, cyclophosphamide, and rituximab (FCR) is an initial treatment option in younger patients who can tolerate chemotherapy without 17p deletion/TP53 mutation but with IGHV mutated CLL.  In patients with IGHV unmutated CLL, targeted therapy with ibrutinib is preferred over FCR due to improved OS and PFS. FCR is contraindicated in older patients due to the high risk of infections, adverse events, and high risk of immunosuppression.[62][68][62]
  • Bendamustine plus rituximab (BR) as a treatment of choice for older patients (> 65 years of age) without 17p deletion/TP53 mutation but with IGHV mutated CLL.  In patients with IGHV unmutated CLL, targeted therapy with ibrutinib is preferred over BR due to improved OS and PFS.[65]
  • Chlorambucil based therapy is usually not used as an initial treatment for CLL as other targeted therapy (example, ibrutinib, venetoclax plus obinutuzumab) has demonstrated higher efficacy in patients with 17p deletion, TP53 mutation, or IGHV unmutated CLL.  Chlorambucil plus obinutuzumab is a combination that could be used in elderly patients with IGHV mutated CLL, and recently it was found that the combination of chlorambucil with obinutuzumab is superior to rituximab plus chlorambucil in achieving a complete response and prolonging progression-free survival. This combination has demonstrated similar efficacy to ibrutinib based therapy and venetoclax plus obinutuzumab combination therapy in patients with IGHV mutated CLL.  Adverse effects of chlorambucil include significant anemia, neutropenia, and/or thrombocytopenia.  Less common adverse effects include hepatotoxicity, drug hypersensitivity, seizures, interstitial pneumonia, pulmonary fibrosis, and infertility.[64][66][64]

C. Management of complications of CLL and its therapy

CLL/SLL is characterized by hypo/agammaglobulinemia, which increases the risk of infections, especially Staphylococcus, H. influenza, Pneumococcus, and herpes. These infections can be managed with antimicrobials for bacterial, fungal, or viral infections. For refractory infections in patients with hypogammaglobinemia, or for 2 or more severe infections within a timeframe of 6-month, monthly IVIG infusions are considered.

CLL/SLL also produces autoantibodies directed against RBCs and platelets, leading to immune-mediated hemolytic anemia and thrombocytopenia.  These can be acutely managed with packed RBC transfusion for anemia and platelet transfusion for bleeding secondary to thrombocytopenia.  The cytopenias can be eventually treated with prednisone.

D. Relapsed/Refractory CLL

It is important to confirm relapsed/refractory CLL histologically prior to restarting treatment.  Recurrent CLL in asymptomatic patients should be monitored for symptoms requiring treatment. Richter's transformation should specifically be excluded.  The treatment of choice at relapse depends upon the initial treatment used and the initial duration of response to that treatment.

Ibrutinib is the first-line therapy to improve progression-free survival and response to treatment in relapsed/refractory CLL.  Other drugs of choice for relapsed CLL include idelalisib (a phosphoinositide 3'-kinase [PI3K] delta inhibitor), Alemtuzumab (a monoclonal antibody directed against CD52, a cellular marker found in CLL) and venetoclax (Bcl-2 inhibitor). Venetoclax is actively used in refractory CLL patients with 17p deletion. Venetoclax induces brisk apoptosis and occasionally induces tumor lysis syndrome. Anti-CD19 chimeric antigen receptor (CAR) T-cell therapy has also been active in refractory cases. Anti-CD20 monoclonal antibody (rituximab, ofatumumab, obinutuzumab) may briefly alleviate symptoms in relapsed/refractory CLL patients. 

Allogeneic Hematopoietic Stem Cell Transplantation is rarely performed in CLL but has been used in refractory cases in appropriate patients. Those with a very aggressive form of the disease, such as patients with 17p deletion, especially in younger patients, HLA-matched donors, should be recommended to get bone marrow transplant. Those without a matched donor or older in age can be given a trial of ibrutinib. Palliative radiation therapy can be given to chemotherapy-resistant areas of lymphadenopathy, liver, and spleen.  Total body irradiation occasionally helps relieve symptoms temporarily. 

Differential Diagnosis

  • Acute Lymphoblastic Leukemia (ALL)
  • Acute promyelocytic leukemia
  • Diffuse large cell lymphoma
  • Follicular lymphoma
  • Hairy cell leukemia
  • Lymphoblastic lymphoma
  • Mantle cell lymphoma
  • Non-Hodgkin lymphoma
  • Monoclonal B-cell lymphocytosis (MBL)
  • Prolymphocytic lymphoma (PLL)
  • Lymphoplasmacytic lymphoma
  • Histologic transformation — CLL/SLL can convert to more aggressive histology (Richter transformation), either diffuse large B cell lymphoma or Hodgkin lymphoma.

Staging

Two staging systems are used in CLL patients. The modified Rai-Sawitsky staging in the United States and the Binet staging in Europe.

      1. RAI Classification

  • Stage 0: Absolute lymphocytosis of > 10,000/mcL in peripheral blood and >/= 30% lymphocytes in bone marrow
  • Stage I: Stage 0 plus lymphadenopathy (enlarged lymph nodes)
  • Stage II: Stage 0 plus hepatomegaly or splenomegaly
  • Stage III: Stage 0 plus anemia with hemoglobin < 11 g/dL (< 110 g/L) due to bone marrow infiltration of tumor cells.
  • Stage IV: Stage 0 plus thrombocytopenia with platelet counts <100,000/mcL due to bone marrow infiltration of tumor cells.

     2. Binet Classification 

  • Stage A:
    • Absolute lymphocytosis of > 10,000/mcL in blood and ≥ 30% lymphocytes in bone marrow
    • Hemoglobin ≥ 10 g/dL ( ≥ 100 g/L)
    • Platelets ≥100,000/mcL
    • ≤ 2 involved sites*
  • Stage B: As for stage A, but 3–5 involved sites
  • Stage C : As for stage A or B, but hemoglobin < 10 g/dL (< 100 g/L) or platelets < 100,000/mcL.

* Sites considered: Cervical, axillary, and inguinal lymph nodes; liver; and spleen.

Prognosis

In patients with CLL, the survival period could range from 2 to >20 years with a median survival of 10 years. The patients who present as Rai stage 0-II may survive for 5 to 20 years with no treatment. Lymphocyte doubling time is a prognostic factor of CLL, defined as the number of months it takes to double the absolute lymphocyte count. A more aggressive expression of CLL is seen in untreated patients with a lymphocyte doubling time of <12 months. Favorable prognostic factors include mutated Ig heavy chain variable region, 13 q deletion, low expression of ZAP-70, and low CD38 levels on flow cytometry. Unfavorable prognostic factors include specific high-risk cytogenetics abnormalities such as 17p deletion and 11q deletion. The patients with multiple chain lymphadenopathy, hepatosplenomegaly, anemia, and thrombocytopenia have a worse prognosis. One of the commonly used tools to predict CLL related outcomes includes CLL-IPI (International Prognostic Index for Chronic Lymphocytic Leukemia).[69]

Complications

Complications accompanying CLL may include increased susceptibility to infections, particularly of the respiratory tract, progression to diffuse large B-cell lymphoma (Richter syndrome), an elevated risk of other malignancies (e.g., cancers of the skin, lungs, and GI tract), and immune system issues, where the immune system attacks red blood cells or platelets, although this is rare.[70]

Deterrence and Patient Education

Patients will CLL need to understand the nature of their disease, the staging criteria, and where they stand within that staging, the treatments available, and their prognosis, given the precise nature of their disease. All treatment and management decisions should involve the patient and their family, and in late-stage disease, psychological and emotional support from qualified mental health professionals is in order.

Pearls and Other Issues

  • CLL is an indolent malignancy characterized by increased production of mature but dysfunctional B lymphocytes. The primary disease sites include peripheral blood, spleen, lymph node, and bone marrow. Signs and symptoms are absent in many patients, but when present, include B symptoms (fevers, night sweats, unintentional weight loss), fatigue, early satiety, hepatomegaly, splenomegaly, and lymphadenopathy. CLL can be diagnosed by peripheral blood smear and confirmed by flow cytometry and immunophenotyping of peripheral blood. Treatment, including chemotherapy/immunotherapy, is generally not curative and is given only in symptomatic patients. Asymptomatic patients are generally observed. Current first-line treatment for CLL now includes BTK and BCL-2 inhibitors. Chemoimmunotherapy decreases symptoms and prolongs survival in symptomatic patients.
  • About 2 to 10% of CLL patients undergo Richter's transformation, where CLL evolved into an aggressive lymphoma, most commonly diffuse large B-cell lymphoma, which presents with fever, rapid enlargement of previously stable nodal disease, and severely rising LDH levels.  CLL can also transform into high-grade non-Hodgkin's lymphoma by involving into B-cell prolymphocytic leukemia.
  • It is important to differentiate CLL from other causes of lymphocytosis, including other B-cell disorders, which can present similarly, such as infectious causes of lymphocytosis. Certain infections can present with lymphocytoses such as pertussis and infectious mononucleosis. However, unlike CLL, it is transient and is not monoclonal.
  • Monoclonal B-cell lymphocytosis (MBL): is defined as an absolute lymphocyte count of less than 5000/mcL with evidence of clonality on flow cytometry. CLL is a monoclonal expansion of B cells preceded by an oligoclonal expansion of B cells termed monoclonal B-cell lymphocytosis. It is said to have a genetic basis, and certain genetic polymorphisms are known to predispose individuals to develop CLL. Patients with MBL are asymptomatic, present with no constitutional symptoms, no anemia, no thrombocytopenia, no organomegaly, and lymph nodes are less than 1.5 cm in size.  Patients with MBL are managed by observation. 
  • Other B cell disorders: Mantle cell lymphoma, splenic marginal zone lymphoma, follicular lymphoma, hairy cell leukemia. Even though these disorders can mimic CLL clinically, a careful look at the immunophenotypic markers, morphology, or specific genetic abnormalities such as translocation t(14;18) for follicular lymphoma can lead to the diagnosis.

Enhancing Healthcare Team Outcomes

CLL is a complex disorder best managed by an interprofessional team that includes an internist/general practitioner, hematologist/oncologist, hematopathologist, and radiologist.

Given that most patients with CLL are asymptomatic, treatment is not recommended for everyone. The recommendation for treatment depends on severe disease symptoms or rapidly progressing disease. Symptoms such as severe fatigue interfering with daily activities, B-symptoms, recurrent infections, or increased tumor burden indicate early treatment. Rapidly progressing disease such as an absolute lymphocyte count doubling time of fewer than 12 months is also an indication for early treatment.

For patients who are not considered for treatment, regular follow-up at 3-month intervals is recommended. If the patient has developed increasingly abnormal laboratory finds or is symptomatically worse, further care should be discussed with the oncologist.  At the 12-month mark, depending on the symptoms and pace of disease, a decision on whether treatment is needed can be made.

Symptomatic CLL is treated, and despite many recent advances, it remains an incurable disease. However, the use of newer biological agents appears to be making some improvement in lifespan. For symptomatic patients, the outlook is dependent on various disease characteristics and underlying comorbidities, but those who are asymptomatic may have a normal life span.[71][72] [Level 2]

Review Questions

References

1.
Blair A, White DW. Leukemia cell types and agricultural practices in Nebraska. Arch Environ Health. 1985 Jul-Aug;40(4):211-4. [PubMed: 4051575]
2.
Burmeister LF, Van Lier SF, Isacson P. Leukemia and farm practices in Iowa. Am J Epidemiol. 1982 May;115(5):720-8. [PubMed: 7081203]
3.
Talibov M, Auvinen A, Weiderpass E, Hansen J, Martinsen JI, Kjaerheim K, Tryggvadottir L, Pukkala E. Occupational solvent exposure and adult chronic lymphocytic leukemia: No risk in a population-based case-control study in four Nordic countries. Int J Cancer. 2017 Sep 15;141(6):1140-1147. [PubMed: 28571111]
4.
Preston DL, Kusumi S, Tomonaga M, Izumi S, Ron E, Kuramoto A, Kamada N, Dohy H, Matsuo T, Matsui T [corrected to Matsuo T] Cancer incidence in atomic bomb survivors. Part III. Leukemia, lymphoma and multiple myeloma, 1950-1987. Radiat Res. 1994 Feb;137(2 Suppl):S68-97. [PubMed: 8127953]
5.
Schubauer-Berigan MK, Daniels RD, Fleming DA, Markey AM, Couch JR, Ahrenholz SH, Burphy JS, Anderson JL, Tseng CY. Chronic lymphocytic leukaemia and radiation: findings among workers at five US nuclear facilities and a review of the recent literature. Br J Haematol. 2007 Dec;139(5):799-808. [PubMed: 17922878]
6.
Arp EW, Wolf PH, Checkoway H. Lymphocytic leukemia and exposures to benzene and other solvents in the rubber industry. J Occup Med. 1983 Aug;25(8):598-602. [PubMed: 6886869]
7.
Monson RR, Fine LJ. Cancer mortality and morbidity among rubber workers. J Natl Cancer Inst. 1978 Oct;61(4):1047-53. [PubMed: 279710]
8.
Brown LM, Gibson R, Blair A, Burmeister LF, Schuman LM, Cantor KP, Fraumeni JF. Smoking and risk of leukemia. Am J Epidemiol. 1992 Apr 01;135(7):763-8. [PubMed: 1595675]
9.
Khalade A, Jaakkola MS, Pukkala E, Jaakkola JJ. Exposure to benzene at work and the risk of leukemia: a systematic review and meta-analysis. Environ Health. 2010 Jun 28;9:31. [PMC free article: PMC2903550] [PubMed: 20584305]
10.
Marwick C. Link found between Agent Orange and chronic lymphocytic leukaemia. BMJ. 2003 Feb 01;326(7383):242. [PMC free article: PMC1169216] [PubMed: 12560261]
11.
Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA Cancer J Clin. 2020 Jan;70(1):7-30. [PubMed: 31912902]
12.
Hernández JA, Land KJ, McKenna RW. Leukemias, myeloma, and other lymphoreticular neoplasms. Cancer. 1995 Jan 01;75(1 Suppl):381-94. [PubMed: 8001009]
13.
Smith A, Howell D, Patmore R, Jack A, Roman E. Incidence of haematological malignancy by sub-type: a report from the Haematological Malignancy Research Network. Br J Cancer. 2011 Nov 22;105(11):1684-92. [PMC free article: PMC3242607] [PubMed: 22045184]
14.
Yamamoto JF, Goodman MT. Patterns of leukemia incidence in the United States by subtype and demographic characteristics, 1997-2002. Cancer Causes Control. 2008 May;19(4):379-90. [PubMed: 18064533]
15.
Wu SJ, Huang SY, Lin CT, Lin YJ, Chang CJ, Tien HF. The incidence of chronic lymphocytic leukemia in Taiwan, 1986-2005: a distinct increasing trend with birth-cohort effect. Blood. 2010 Nov 25;116(22):4430-5. [PubMed: 20713960]
16.
Miranda-Filho A, Piñeros M, Ferlay J, Soerjomataram I, Monnereau A, Bray F. Epidemiological patterns of leukaemia in 184 countries: a population-based study. Lancet Haematol. 2018 Jan;5(1):e14-e24. [PubMed: 29304322]
17.
Fleming AF. The epidemiology of lymphomas and leukaemias in Africa--an overview. Leuk Res. 1985;9(6):735-40. [PubMed: 2989624]
18.
Oloo AJ, Ogada TA. Chronic lymphocytic leukaemia (CLL): clinical study at Kenyatta National Hospital (KNH). East Afr Med J. 1984 Nov;61(11):797-801. [PubMed: 6535701]
19.
Little MP, Wakeford R, Borrego D, French B, Zablotska LB, Adams MJ, Allodji R, de Vathaire F, Lee C, Brenner AV, Miller JS, Campbell D, Pearce MS, Doody MM, Holmberg E, Lundell M, Sadetzki S, Linet MS, Berrington de González A. Leukaemia and myeloid malignancy among people exposed to low doses (<100 mSv) of ionising radiation during childhood: a pooled analysis of nine historical cohort studies. Lancet Haematol. 2018 Aug;5(8):e346-e358. [PMC free article: PMC6130888] [PubMed: 30026010]
20.
Casola S, Perucho L, Tripodo C, Sindaco P, Ponzoni M, Facchetti F. The B-cell receptor in control of tumor B-cell fitness: Biology and clinical relevance. Immunol Rev. 2019 Mar;288(1):198-213. [PubMed: 30874349]
21.
PDQ Adult Treatment Editorial Board. PDQ Cancer Information Summaries [Internet]. National Cancer Institute (US); Bethesda (MD): Dec 8, 2023. Chronic Lymphocytic Leukemia Treatment (PDQ®): Health Professional Version. [PubMed: 26389470]
22.
Patrussi L, Capitani N, Baldari CT. Abnormalities in chemokine receptor recycling in chronic lymphocytic leukemia. Cell Mol Life Sci. 2019 Aug;76(16):3249-3261. [PubMed: 30830241]
23.
Ghia P, Caligaris-Cappio F. Monoclonal B-cell lymphocytosis: right track or red herring? Blood. 2012 May 10;119(19):4358-62. [PubMed: 22422819]
24.
Dühren-von Minden M, Übelhart R, Schneider D, Wossning T, Bach MP, Buchner M, Hofmann D, Surova E, Follo M, Köhler F, Wardemann H, Zirlik K, Veelken H, Jumaa H. Chronic lymphocytic leukaemia is driven by antigen-independent cell-autonomous signalling. Nature. 2012 Sep 13;489(7415):309-12. [PubMed: 22885698]
25.
Hallek M, Cheson BD, Catovsky D, Caligaris-Cappio F, Dighiero G, Döhner H, Hillmen P, Keating M, Montserrat E, Chiorazzi N, Stilgenbauer S, Rai KR, Byrd JC, Eichhorst B, O'Brien S, Robak T, Seymour JF, Kipps TJ. iwCLL guidelines for diagnosis, indications for treatment, response assessment, and supportive management of CLL. Blood. 2018 Jun 21;131(25):2745-2760. [PubMed: 29540348]
26.
Rai KR, Sawitsky A, Cronkite EP, Chanana AD, Levy RN, Pasternack BS. Clinical staging of chronic lymphocytic leukemia. Blood. 1975 Aug;46(2):219-34. [PubMed: 1139039]
27.
Binet JL, Auquier A, Dighiero G, Chastang C, Piguet H, Goasguen J, Vaugier G, Potron G, Colona P, Oberling F, Thomas M, Tchernia G, Jacquillat C, Boivin P, Lesty C, Duault MT, Monconduit M, Belabbes S, Gremy F. A new prognostic classification of chronic lymphocytic leukemia derived from a multivariate survival analysis. Cancer. 1981 Jul 01;48(1):198-206. [PubMed: 7237385]
28.
Diehl LF, Ketchum LH. Autoimmune disease and chronic lymphocytic leukemia: autoimmune hemolytic anemia, pure red cell aplasia, and autoimmune thrombocytopenia. Semin Oncol. 1998 Feb;25(1):80-97. [PubMed: 9482530]
29.
Dearden C, Wade R, Else M, Richards S, Milligan D, Hamblin T, Catovsky D., UK National Cancer Research Institute (NCRI). Haematological Oncology Clinical Studies Group. NCRI CLL Working Group. 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 Feb 15;111(4):1820-6. [PubMed: 18055869]
30.
Mauro FR, Foa R, Cerretti R, Giannarelli D, Coluzzi S, Mandelli F, Girelli G. Autoimmune hemolytic anemia in chronic lymphocytic leukemia: clinical, therapeutic, and prognostic features. Blood. 2000 May 01;95(9):2786-92. [PubMed: 10779422]
31.
Visco C, Ruggeri M, Laura Evangelista M, Stasi R, Zanotti R, Giaretta I, Ambrosetti A, Madeo D, Pizzolo G, Rodeghiero F. Impact of immune thrombocytopenia on the clinical course of chronic lymphocytic leukemia. Blood. 2008 Feb 01;111(3):1110-6. [PubMed: 17986663]
32.
Parikh SA, Leis JF, Chaffee KG, Call TG, Hanson CA, Ding W, Chanan-Khan AA, Bowen D, Conte M, Schwager S, Slager SL, Van Dyke DL, Jelinek DF, Kay NE, Shanafelt TD. Hypogammaglobulinemia in newly diagnosed chronic lymphocytic leukemia: Natural history, clinical correlates, and outcomes. Cancer. 2015 Sep 01;121(17):2883-91. [PMC free article: PMC4545721] [PubMed: 25931291]
33.
Agnew KL, Ruchlemer R, Catovsky D, Matutes E, Bunker CB. Cutaneous findings in chronic lymphocytic leukaemia. Br J Dermatol. 2004 Jun;150(6):1129-35. [PubMed: 15214899]
34.
Robak E, Robak T. Skin lesions in chronic lymphocytic leukemia. Leuk Lymphoma. 2007 May;48(5):855-65. [PubMed: 17487727]
35.
WEED RI. EXAGGERATED DELAYED HYPERSENSITIVITY TO MOSQUITO BITES IN CHRONIC LYMPHOCYTIC LEUKEMIA. Blood. 1965 Sep;26:257-68. [PubMed: 14332055]
36.
Grandi V, Maglie R, Antiga E, Vannucchi M, Delfino C, Lastrucci I, Gunnella S, Ciolli S, Quintarelli L, Massi D, Caproni M, Pimpinelli N. Eosinophilic dermatosis of hematologic malignancy: A retrospective cohort of 37 patients from an Italian center. J Am Acad Dermatol. 2019 Jul;81(1):246-249. [PubMed: 30528498]
37.
Nowakowski GS, Hoyer JD, Shanafelt TD, Geyer SM, LaPlant BR, Call TG, Jelinek DF, Zent CS, Kay NE. Using smudge cells on routine blood smears to predict clinical outcome in chronic lymphocytic leukemia: a universally available prognostic test. Mayo Clin Proc. 2007 Apr;82(4):449-53. [PubMed: 17418074]
38.
Rawstron AC, Kreuzer KA, Soosapilla A, Spacek M, Stehlikova O, Gambell P, McIver-Brown N, Villamor N, Psarra K, Arroz M, Milani R, de la Serna J, Cedena MT, Jaksic O, Nomdedeu J, Moreno C, Rigolin GM, Cuneo A, Johansen P, Johnsen HE, Rosenquist R, Niemann CU, Kern W, Westerman D, Trneny M, Mulligan S, Doubek M, Pospisilova S, Hillmen P, Oscier D, Hallek M, Ghia P, Montserrat E. Reproducible diagnosis of chronic lymphocytic leukemia by flow cytometry: An European Research Initiative on CLL (ERIC) & European Society for Clinical Cell Analysis (ESCCA) Harmonisation project. Cytometry B Clin Cytom. 2018 Jan;94(1):121-128. [PMC free article: PMC5817234] [PubMed: 29024461]
39.
Rawstron AC, Villamor N, Ritgen M, Böttcher S, Ghia P, Zehnder JL, Lozanski G, Colomer D, Moreno C, Geuna M, Evans PA, Natkunam Y, Coutre SE, Avery ED, Rassenti LZ, Kipps TJ, Caligaris-Cappio F, Kneba M, Byrd JC, Hallek MJ, Montserrat E, Hillmen P. International standardized approach for flow cytometric residual disease monitoring in chronic lymphocytic leukaemia. Leukemia. 2007 May;21(5):956-64. [PubMed: 17361231]
40.
Potter KN, Mockridge CI, Neville L, Wheatley I, Schenk M, Orchard J, Duncombe AS, Packham G, Stevenson FK. Structural and functional features of the B-cell receptor in IgG-positive chronic lymphocytic leukemia. Clin Cancer Res. 2006 Mar 15;12(6):1672-9. [PubMed: 16551848]
41.
Geisler CH, Larsen JK, Hansen NE, Hansen MM, Christensen BE, Lund B, Nielsen H, Plesner T, Thorling K, Andersen E. Prognostic importance of flow cytometric immunophenotyping of 540 consecutive patients with B-cell chronic lymphocytic leukemia. Blood. 1991 Oct 01;78(7):1795-802. [PubMed: 1717071]
42.
Fournier S, Delespesse G, Rubio M, Biron G, Sarfati M. CD23 antigen regulation and signaling in chronic lymphocytic leukemia. J Clin Invest. 1992 Apr;89(4):1312-21. [PMC free article: PMC442993] [PubMed: 1532590]
43.
Freedman AS, Boyd AW, Bieber FR, Daley J, Rosen K, Horowitz JC, Levy DN, Nadler LM. Normal cellular counterparts of B cell chronic lymphocytic leukemia. Blood. 1987 Aug;70(2):418-27. [PubMed: 3496927]
44.
Döhner H, Stilgenbauer S, Benner A, Leupolt E, Kröber A, Bullinger L, Döhner K, Bentz M, Lichter P. Genomic aberrations and survival in chronic lymphocytic leukemia. N Engl J Med. 2000 Dec 28;343(26):1910-6. [PubMed: 11136261]
45.
Reddy KS. Chronic lymphocytic leukaemia profiled for prognosis using a fluorescence in situ hybridisation panel. Br J Haematol. 2006 Mar;132(6):705-22. [PubMed: 16487171]
46.
Knuutila S, Elonen E, Teerenhovi L, Rossi L, Leskinen R, Bloomfield CD, de la Chapelle A. Trisomy 12 in B cells of patients with B-cell chronic lymphocytic leukemia. N Engl J Med. 1986 Apr 03;314(14):865-9. [PubMed: 3081810]
47.
Kastan MB, Onyekwere O, Sidransky D, Vogelstein B, Craig RW. Participation of p53 protein in the cellular response to DNA damage. Cancer Res. 1991 Dec 01;51(23 Pt 1):6304-11. [PubMed: 1933891]
48.
Kuerbitz SJ, Plunkett BS, Walsh WV, Kastan MB. Wild-type p53 is a cell cycle checkpoint determinant following irradiation. Proc Natl Acad Sci U S A. 1992 Aug 15;89(16):7491-5. [PMC free article: PMC49736] [PubMed: 1323840]
49.
Khanna KK, Keating KE, Kozlov S, Scott S, Gatei M, Hobson K, Taya Y, Gabrielli B, Chan D, Lees-Miller SP, Lavin MF. ATM associates with and phosphorylates p53: mapping the region of interaction. Nat Genet. 1998 Dec;20(4):398-400. [PubMed: 9843217]
50.
Ambrose M, Gatti RA. Pathogenesis of ataxia-telangiectasia: the next generation of ATM functions. Blood. 2013 May 16;121(20):4036-45. [PMC free article: PMC3709651] [PubMed: 23440242]
51.
Swerdlow SH, Campo E, Pileri SA, Harris NL, Stein H, Siebert R, Advani R, Ghielmini M, Salles GA, Zelenetz AD, Jaffe ES. The 2016 revision of the World Health Organization classification of lymphoid neoplasms. Blood. 2016 May 19;127(20):2375-90. [PMC free article: PMC4874220] [PubMed: 26980727]
52.
Lipshutz MD, Mir R, Rai KR, Sawitsky A. Bone marrow biopsy and clinical staging in chronic lymphocytic leukemia. Cancer. 1980 Sep 15;46(6):1422-7. [PubMed: 7417943]
53.
Pangalis GA, Boussiotis VA, Kittas C. B-chronic lymphocytic leukemia. Disease progression in 150 untreated stage A and B patients as predicted by bone marrow pattern. Nouv Rev Fr Hematol (1978). 1988;30(5-6):373-5. [PubMed: 3222147]
54.
Rozman C, Montserrat E, Rodríguez-Fernández JM, Ayats R, Vallespí T, Parody R, Ríos A, Prados D, Morey M, Gomis F. Bone marrow histologic pattern--the best single prognostic parameter in chronic lymphocytic leukemia: a multivariate survival analysis of 329 cases. Blood. 1984 Sep;64(3):642-8. [PubMed: 6466871]
55.
Borthakur G, O'Brien S, Wierda WG, Thomas DA, Cortes JE, Giles FJ, Kantarjian HM, Lerner S, Keating MJ. Immune anaemias in patients with chronic lymphocytic leukaemia treated with fludarabine, cyclophosphamide and rituximab--incidence and predictors. Br J Haematol. 2007 Mar;136(6):800-5. [PubMed: 17341265]
56.
Effects of chlorambucil and therapeutic decision in initial forms of chronic lymphocytic leukemia (stage A): results of a randomized clinical trial on 612 patients. The French Cooperative Group on Chronic Lymphocytic Leukemia. Blood. 1990 Apr 01;75(7):1414-21. [PubMed: 2180492]
57.
Chemotherapeutic options in chronic lymphocytic leukemia: a meta-analysis of the randomized trials. CLL Trialists' Collaborative Group. J Natl Cancer Inst. 1999 May 19;91(10):861-8. [PubMed: 10340906]
58.
Hoechstetter MA, Busch R, Eichhorst B, Bühler A, Winkler D, Eckart MJ, Vehling-Kaiser U, Schimke H, Jäger U, Hurtz HJ, Hopfinger G, Hartmann F, Fuss H, Abenhardt W, Blau I, Freier W, Müller L, Goebeler M, Wendtner CM, Bahlo J, Fischer K, Bentz M, Emmerich B, Döhner H, Hallek M, Stilgenbauer S. Early, risk-adapted treatment with fludarabine in Binet stage A chronic lymphocytic leukemia patients: results of the CLL1 trial of the German CLL study group. Leukemia. 2017 Dec;31(12):2833-2837. [PubMed: 28804126]
59.
Condoluci A, Terzi di Bergamo L, Langerbeins P, Hoechstetter MA, Herling CD, De Paoli L, Delgado J, Rabe KG, Gentile M, Doubek M, Mauro FR, Chiodin G, Mattsson M, Bahlo J, Cutrona G, Kotaskova J, Deambrogi C, Smedby KE, Spina V, Bruscaggin A, Wu W, Moia R, Bianchi E, Gerber B, Zucca E, Gillessen S, Ghielmini M, Cavalli F, Stussi G, Hess MA, Baumann TS, Neri A, Ferrarini M, Rosenquist R, Forconi F, Foà R, Pospisilova S, Morabito F, Stilgenbauer S, Döhner H, Parikh SA, Wierda WG, Montserrat E, Gaidano G, Hallek M, Rossi D. International prognostic score for asymptomatic early-stage chronic lymphocytic leukemia. Blood. 2020 May 21;135(21):1859-1869. [PubMed: 32267500]
60.
Morrison WH, Hoppe RT, Weiss LM, Picozzi VJ, Horning SJ. Small lymphocytic lymphoma. J Clin Oncol. 1989 May;7(5):598-606. [PubMed: 2651577]
61.
Goede V, Fischer K, Busch R, Engelke A, Eichhorst B, Wendtner CM, Chagorova T, de la Serna J, Dilhuydy MS, Illmer T, Opat S, Owen CJ, Samoylova O, Kreuzer KA, Stilgenbauer S, Döhner H, Langerak AW, Ritgen M, Kneba M, Asikanius E, Humphrey K, Wenger M, Hallek M. Obinutuzumab plus chlorambucil in patients with CLL and coexisting conditions. N Engl J Med. 2014 Mar 20;370(12):1101-10. [PubMed: 24401022]
62.
Shanafelt TD, Wang XV, Kay NE, Hanson CA, O'Brien S, Barrientos J, Jelinek DF, Braggio E, Leis JF, Zhang CC, Coutre SE, Barr PM, Cashen AF, Mato AR, Singh AK, Mullane MP, Little RF, Erba H, Stone RM, Litzow M, Tallman M. Ibrutinib-Rituximab or Chemoimmunotherapy for Chronic Lymphocytic Leukemia. N Engl J Med. 2019 Aug 01;381(5):432-443. [PMC free article: PMC6908306] [PubMed: 31365801]
63.
Tausch E, Schneider C, Robrecht S, Zhang C, Dolnik A, Bloehdorn J, Bahlo J, Al-Sawaf O, Ritgen M, Fink AM, Eichhorst B, Kreuzer KA, Tandon M, Humphrey K, Jiang Y, Schary W, Bullinger L, Mertens D, Lurà MP, Kneba M, Döhner H, Fischer K, Hallek M, Stilgenbauer S. Prognostic and predictive impact of genetic markers in patients with CLL treated with obinutuzumab and venetoclax. Blood. 2020 Jun 25;135(26):2402-2412. [PubMed: 32206772]
64.
Fischer K, Al-Sawaf O, Bahlo J, Fink AM, Tandon M, Dixon M, Robrecht S, Warburton S, Humphrey K, Samoylova O, Liberati AM, Pinilla-Ibarz J, Opat S, Sivcheva L, Le Dû K, Fogliatto LM, Niemann CU, Weinkove R, Robinson S, Kipps TJ, Boettcher S, Tausch E, Humerickhouse R, Eichhorst B, Wendtner CM, Langerak AW, Kreuzer KA, Ritgen M, Goede V, Stilgenbauer S, Mobasher M, Hallek M. Venetoclax and Obinutuzumab in Patients with CLL and Coexisting Conditions. N Engl J Med. 2019 Jun 06;380(23):2225-2236. [PubMed: 31166681]
65.
Woyach JA, Ruppert AS, Heerema NA, Zhao W, Booth AM, Ding W, Bartlett NL, Brander DM, Barr PM, Rogers KA, Parikh SA, Coutre S, Hurria A, Brown JR, Lozanski G, Blachly JS, Ozer HG, Major-Elechi B, Fruth B, Nattam S, Larson RA, Erba H, Litzow M, Owen C, Kuzma C, Abramson JS, Little RF, Smith SE, Stone RM, Mandrekar SJ, Byrd JC. Ibrutinib Regimens versus Chemoimmunotherapy in Older Patients with Untreated CLL. N Engl J Med. 2018 Dec 27;379(26):2517-2528. [PMC free article: PMC6325637] [PubMed: 30501481]
66.
Moreno C, Greil R, Demirkan F, Tedeschi A, Anz B, Larratt L, Simkovic M, Samoilova O, Novak J, Ben-Yehuda D, Strugov V, Gill D, Gribben JG, Hsu E, Lih CJ, Zhou C, Clow F, James DF, Styles L, Flinn IW. Ibrutinib plus obinutuzumab versus chlorambucil plus obinutuzumab in first-line treatment of chronic lymphocytic leukaemia (iLLUMINATE): a multicentre, randomised, open-label, phase 3 trial. Lancet Oncol. 2019 Jan;20(1):43-56. [PubMed: 30522969]
67.
Burger JA, Barr PM, Robak T, Owen C, Ghia P, Tedeschi A, Bairey O, Hillmen P, Coutre SE, Devereux S, Grosicki S, McCarthy H, Simpson D, Offner F, Moreno C, Dai S, Lal I, Dean JP, Kipps TJ. Long-term efficacy and safety of first-line ibrutinib treatment for patients with CLL/SLL: 5 years of follow-up from the phase 3 RESONATE-2 study. Leukemia. 2020 Mar;34(3):787-798. [PMC free article: PMC7214263] [PubMed: 31628428]
68.
Polizzotto MN, Tam CS, Milner A, Januszewicz EH, Prince HM, Westerman D, Wolf MM, Seymour JF. The influence of increasing age on the deliverability and toxicity of fludarabine-based combination chemotherapy regimens in patients with indolent lymphoproliferative disorders. Cancer. 2006 Aug 15;107(4):773-80. [PubMed: 16847886]
69.
Molica S, Shanafelt TD, Giannarelli D, Gentile M, Mirabelli R, Cutrona G, Levato L, Di Renzo N, Di Raimondo F, Musolino C, Angrilli F, Famà A, Recchia AG, Chaffee KG, Neri A, Kay NE, Ferrarini M, Morabito F. The chronic lymphocytic leukemia international prognostic index predicts time to first treatment in early CLL: Independent validation in a prospective cohort of early stage patients. Am J Hematol. 2016 Nov;91(11):1090-1095. [PMC free article: PMC5072993] [PubMed: 27465919]
70.
Iskierka-Jażdżewska E, Robak T. Minimizing and managing treatment-associated complications in patients with chronic lymphocytic leukemia. Expert Rev Hematol. 2020 Jan;13(1):39-53. [PubMed: 31747803]
71.
O'Brien SM, Byrd JC, Hillmen P, Coutre S, Brown JR, Barr PM, Barrientos JC, Devereux S, Robak T, Reddy NM, Kipps TJ, Tedeschi A, Cymbalista F, Ghia P, Chang S, Ninomoto J, James DF, Burger JA. Outcomes with ibrutinib by line of therapy and post-ibrutinib discontinuation in patients with chronic lymphocytic leukemia: Phase 3 analysis. Am J Hematol. 2019 May;94(5):554-562. [PMC free article: PMC6593416] [PubMed: 30767298]
72.
Gohil SH, Wu CJ. Dissecting CLL through high-dimensional single-cell technologies. Blood. 2019 Mar 28;133(13):1446-1456. [PMC free article: PMC6440295] [PubMed: 30728142]

Disclosure: Shiva Kumar Mukkamalla declares no relevant financial relationships with ineligible companies.

Disclosure: Alankrita Taneja declares no relevant financial relationships with ineligible companies.

Disclosure: Dhatri Malipeddi declares no relevant financial relationships with ineligible companies.

Disclosure: Samip Master declares no relevant financial relationships with ineligible companies.

Copyright © 2024, StatPearls Publishing LLC.

This book is distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0) ( http://creativecommons.org/licenses/by-nc-nd/4.0/ ), which permits others to distribute the work, provided that the article is not altered or used commercially. You are not required to obtain permission to distribute this article, provided that you credit the author and journal.

Bookshelf ID: NBK470433PMID: 29261864

Views

  • PubReader
  • Print View
  • Cite this Page

Related information

  • PMC
    PubMed Central citations
  • PubMed
    Links to PubMed

Similar articles in PubMed

See reviews...See all...

Recent Activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...