ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Treatment of relapsed or refractory chronic lymphocytic leukemia

Treatment of relapsed or refractory chronic lymphocytic leukemia
Literature review current through: Jan 2024.
This topic last updated: Dec 20, 2023.

INTRODUCTION — Chronic lymphocytic leukemia (CLL) is a chronic lymphoproliferative disorder (lymphoid neoplasm) characterized by a progressive accumulation of functionally incompetent lymphocytes, which are usually monoclonal in origin.

CLL is considered to be identical (ie, one disease with different manifestations) to the mature (peripheral) B cell neoplasm small lymphocytic lymphoma (SLL), a clinically indolent non-Hodgkin lymphoma. The term CLL is used when the disease manifests primarily in the bone marrow and blood while the term SLL is used when involvement is primarily nodal. (See "Clinical features and diagnosis of chronic lymphocytic leukemia/small lymphocytic lymphoma".)

Most patients will have a complete or partial response to initial therapy. However, conventional therapy for CLL is not curative and most patients experience relapse. In addition, many patients will require a change in therapy due to intolerance. The choice of subsequent therapy for patients with progressive disease or intolerance to initial treatment will be reviewed here. The initial treatment of CLL, the use of allogeneic hematopoietic cell transplantation in CLL, and the management of complications of CLL are discussed separately.

(See "Overview of the treatment of chronic lymphocytic leukemia".)

(See "Hematopoietic cell transplantation in chronic lymphocytic leukemia".)

(See "Overview of the complications of chronic lymphocytic leukemia".)

(See "Risk of infections in patients with chronic lymphocytic leukemia".)

(See "Prevention of infections in patients with chronic lymphocytic leukemia".)

EVALUATION PRIOR TO CHANGE IN THERAPY

Patients with BTK inhibitor intolerance — Drug intolerance leads to treatment discontinuation in a sizeable minority of patients on continuous therapy with a Bruton tyrosine kinase (BTK) inhibitor (eg, acalabrutinib, zanubrutinib, ibrutinib) [1]. The management of treatment-emergent adverse effects is individualized. Acalabrutinib and zanubrutinib appear to be better tolerated than ibrutinib.

General guidance for drug holidays and dose reductions is provided in the drug package inserts and can be used to maximize drug benefit. Practical recommendations for the management of adverse events have also been proposed by some expert groups [2,3]. Before switching therapy due to BTK intolerance, we consider the following:

BTK inhibitors can be continued without dose reductions for most grade 1 or 2 nonhematologic toxicities and for hematologic toxicities less than grade 4. More severe toxicities may respond to a brief drug holiday followed by retreatment at the same dose or a lower dose.

We usually continue therapy in the setting of bruising or minor bleeding, low grade arthralgia or myalgia, rash, and hypertension that can be controlled with antihypertensive medications.

Symptom-directed management may improve tolerability and ameliorate these toxicities (eg, topical steroids and/or antihistamines for pruritic rash; acetaminophen for arthralgias/myalgias/headaches; loperamide for diarrhea; antihypertensives for hypertension).

BTK inhibitors have numerous drug interactions that may necessitate dose adjustments. Consultation with a clinical pharmacist may help with identifying and managing interactions. For more detailed information on potential drug-drug interactions, refer to the drug interactions program within UpToDate.

Development of atrial fibrillation does not always necessitate BTK inhibitor discontinuation. Patients with atrial fibrillation should be managed in conjunction with a cardiologist for initial rate and rhythm control with particular attention to drug interactions. BTK inhibitors should be used with caution in patients on anticoagulation, especially warfarin.

Patients with disease progression

Confirming progression — Progressive disease may be suspected in a patient with CLL who experiences a rising lymphocyte count, enlargement of lymph nodes and/or spleen, and/or worsening cytopenias (table 1). The evaluation of such patients depends in part on the context of these findings:

Lymphocytosis is commonly seen in patients treated with BTK inhibitors (ibrutinib, acalabrutinib, zanubrutinib) and phosphoinositide 3'-kinase (PI3K) inhibitors (idelalisib, duvelisib) and reflects a redistribution of leukemia cells from lymphoid tissues. Lymphocytosis during the first months of therapy must not be confused with disease progression.

Lymphocytosis flare and/or small nodal enlargement may be seen when a BTK inhibitor is held, and usually reverses with reinstitution of therapy [4].

Lymphocytosis and/or nodal enlargement may be seen in the setting of infection.

Transformation to a more aggressive histologic subtype (Richter transformation) should be suspected in patients with rapid progression of lymphadenopathy, infiltration of uncommon extranodal sites (excluding the bone marrow), development of systemic symptoms (eg, fever, weight loss, night sweats), and/or elevated serum lactate dehydrogenase. (See "Richter transformation in chronic lymphocytic leukemia/small lymphocytic lymphoma".)

Before changing therapy for progressive disease, the diagnosis of CLL must be reconfirmed. This evaluation should rule out disorders that may mimic CLL such as the leukemic phase of mantle cell lymphoma.

Cytogenetic and molecular studies – Most cases of CLL can be confirmed by a complete blood count with differential, flow cytometry of the peripheral blood to determine the immunophenotype of circulating lymphocytes, and examination of the peripheral smear. (See "Clinical features and diagnosis of chronic lymphocytic leukemia/small lymphocytic lymphoma".)

Prior to a change in therapy, we perform fluorescence in situ hybridization (FISH) of the peripheral blood to look for del17p (TP53). We also perform mutation analysis of TP53 in all patients. The decision to test for other mutations depends on prior therapy received and whether the patient is a candidate for retreatment. It is important to repeat this testing, as clonal evolution does occur. In contrast, immunoglobulin heavy chain variable (IGHV) mutational status will remain unchanged at relapse. (See 'Evaluate exposure to prior therapies and response' below.)

Imaging and/or lymph node biopsy – Imaging is not mandatory unless there is a concern for transformation or need for a symptom-directed investigation. Imaging with 18F-2-deoxy-2-fluoro-D-glucose labelled positron emission tomography (FDG PET) may also help to select a lymph node for biopsy to confirm suspected histologic transformation. (See "Richter transformation in chronic lymphocytic leukemia/small lymphocytic lymphoma".)

Bone marrow aspiration and biopsy – A bone marrow aspiration and biopsy may be indicated if cytopenias are present. Patients with anemia should also be evaluated for autoimmune hemolytic anemia. (See "Overview of the complications of chronic lymphocytic leukemia", section on 'Diagnosis'.)

Indications for treatment — Patients with asymptomatic recurrent/progressive CLL do not necessarily require an immediate change in treatment but should be followed closely for the development of symptomatic disease (ie, "active disease" by International Workshop on CLL [iwCLL] criteria) (table 2). In general, the same indications used for first-line therapy are used at the time of relapse or progressive disease. These are presented separately. (See "Overview of the treatment of chronic lymphocytic leukemia", section on 'Indications for treatment ("active disease")'.)

For patients on continuous therapy, such as a BTK inhibitor, we continue treatment as long as there appears to be a clinical benefit and adjust monitoring parameters based on disease tempo. As an example, we would continue treatment with standard monitoring in patients with stable disease. In contrast, continued treatment with more frequent monitoring would be used for those with a small increase in lymph node size or those with small increases in absolute lymphocyte count without thrombocytopenia or anemia. A change in therapy is indicated for those with "active disease" by iwCLL criteria.

CHOICE OF THERAPY — Our choice of subsequent therapy for patients with progressive disease or intolerance to initial treatment is individualized and takes into account multiple factors including prior therapy, response and its duration, and reason for discontinuation; patient and tumor characteristics; patient preference; and goals of therapy (algorithm 1 and table 3).

While we offer guidance, experts differ in their preferred approach and there is no single agreed-upon standard. We always encourage patients to enroll in a well-conducted clinical trial. (See 'Investigational therapies' below.)

Treatment may incorporate one or more of the following targeted agents, often administered as combinations (table 4):

A Bruton tyrosine kinase (BTK) inhibitor (eg, ibrutinib, acalabrutinib, zanubrutinib, pirtobrutinib)

The BCL2 inhibitor venetoclax

A phosphoinositide 3'-kinase (PI3K) inhibitor (eg, idelalisib, duvelisib)

An anti-CD20 monoclonal antibody (eg, rituximab, ofatumumab, obinutuzumab)

With the development of targeted agents, there has been a shift away from the use of traditional chemotherapy agents such as purine analogs (eg, fludarabine, pentostatin) and alkylating agents (eg, chlorambucil, cyclophosphamide, bendamustine).

The following general concepts apply:

Intolerance versus progression – Symptom-directed management may improve tolerability to maximize duration and therefore benefit of therapy. Patients who discontinue a therapy due to intolerance may be candidates for treatment with a different agent from the same class (eg, acalabrutinib after ibrutinib). Some toxicities are due to off-target effects that may be unique to the particular agent. (See 'Patients with BTK inhibitor intolerance' above.)

Time-limited versus continuous therapy – Retreatment may be an option for patients who experienced a prolonged treatment-free interval after time-limited therapy (eg, five years after treatment with venetoclax plus obinutuzumab as front-line treatment). It is not an option for progression that occurs on continuous therapy (eg, single-agent BTK inhibitor). For patients on a continuous therapy, aim for a treatment-free interval as short as possible since progression may accelerate when continuous targeted therapies are discontinued.

Genetics of the CLL clone – del17p, TP53 mutation, and IGHV mutation are associated with inferior outcomes with all therapies. Outcomes in this group are particularly poor with chemoimmunotherapy. Mutations of specific genes involved in the relevant pathways (ie, BTK and PLCg2 for covalent BTK inhibitors, BCL2 for BCL2 inhibitors) are associated with resistance towards the respective agent and should be tested for, in particular when re-exposure is considered.

Expected toxicities and patient comorbidities/fitness – Treatment regimens differ significantly in their burden of administration and associated toxicities. When there are several available treatment options, the choice among them is primarily made based on these factors, which are highly dependent upon patient values and preferences (table 4). (See 'Consider treatment burdens and patient comorbidities' below.)

Candidacy for allogeneic hematopoietic cell transplantation (HCT) – Most patients with CLL are not candidates for allogeneic HCT. However, HCT is a potentially curative treatment option for selected young patients with clinically aggressive relapsed or refractory CLL or for those with high-risk genetic factors. Observational studies suggest that patients in complete remission (CR) at the time of HCT will have a better prognosis. As such, even patients who plan to undergo HCT should be initially treated at the time of relapse to achieve a remission prior to transplant. (See 'Hematopoietic cell transplantation' below.)

Encourage clinical trials – Most commonly, there is no better therapy to offer a patient than enrollment onto a well-designed, scientifically valid clinical trial. Additional information and instructions for referring a patient to an appropriate research center can be obtained from the United States National Institutes of Health. (See 'Investigational therapies' below.)

Evaluate exposure to prior therapies and response — The management of patients with progressive disease or intolerance should take into account the quality and duration of response to prior therapy (algorithm 1 and table 3). Response duration significantly less than the median progression-free survival (PFS) expected for an individual treatment regimen is a marker of aggressive disease and a predictor of short survival, which should lead to a change in therapy [5].

Estimating the median PFS for a given treatment is complicated given the paucity of direct comparisons in randomized trials and relatively short follow-up of trials evaluating targeted therapies. We generally consider patients with the following to have early relapse requiring a change in treatment:

Progression within five to six years of initial treatment with targeted therapy (eg, ibrutinib, acalabrutinib, or zanubrutinib; venetoclax plus obinutuzumab; ibrutinib plus venetoclax)

Progression within two to three years of treatment with FCR (fludarabine, cyclophosphamide, and rituximab)

Progression within one year of treatment with other chemoimmunotherapy regimens

PFS differs greatly by treatment regimen. Data regarding the median PFS of patients following initial treatment that includes a targeted therapy are immature, but prospective trials have reported PFS rates of up to 70 percent at five years following ibrutinib-based therapy [6]; 78 percent at four years following acalabrutinib [7]; 86 percent at two years following zanubrutinib [8]; 74 percent at four years following venetoclax plus obinutuzumab [9]; and 84 percent at two years following ibrutinib plus venetoclax [10]. These estimates are not direct comparisons and so cannot accurately compare the efficacy of these agents as first-line therapy, in particular since the patient populations varied greatly with regard to age and fitness.

Prior BTK inhibitor — For patients treated initially with a covalent BTK inhibitor (ie, ibrutinib, acalabrutinib, zanubrutinib), our preferred subsequent therapy depends on the reason for discontinuation (algorithm 1 and table 3):

Progression on BTK inhibitor – For patients with CLL progressing on a BTK inhibitor and requiring treatment ("active disease"), we suggest venetoclax-based therapy. Venetoclax is highly active and works through a different mechanism of action (BCL2 inhibition). An alternative covalent BTK inhibitor is unlikely to be effective in this setting as acalabrutinib, zanubrutinib, and ibrutinib share a common mode of action (covalent binding to C481 of BTK); in contrast, the noncovalent BTK inhibitor pirtobrutinib has demonstrated activity in this setting and may be an option for multiply relapsed disease. While idelalisib plus rituximab is approved by regulatory agencies in this setting, its use is limited by toxicities. (See 'BCL2 inhibitors: Venetoclax' below and 'Pirtobrutinib' below.)

After attaining a response, younger patients with a matched donor (related or unrelated) should be evaluated for allogeneic HCT. HCT may be especially attractive in those with higher-risk CLL (eg, del17p, TP53 mutation) and/or progression within five to six years of starting a BTK inhibitor. (See 'Hematopoietic cell transplantation' below.)

Intolerance to BTK inhibitor – For patients who discontinue a BTK inhibitor due to intolerance, options include treatment with an alternative BTK inhibitor or venetoclax-based therapy. Mutation analysis can help guide the choice; those with mutations in BTK and PLCg2 are unlikely to respond to an alternative covalent BTK inhibitor and are treated with venetoclax-based therapy [11-14]; pirtobrutinib is an option for multiply relapsed disease. For those without a mutation in BTK or PLCg2, a choice is made based on comorbidities and patient preference. (See 'Consider treatment burdens and patient comorbidities' below.)

Prior venetoclax plus obinutuzumab — For patients treated initially with venetoclax plus obinutuzumab, our choice of subsequent therapy depends on the timing of relapse (algorithm 1 and table 3):

Early progression – For patients progressing within a relatively short time (eg, three to five years) after initial treatment with venetoclax plus obinutuzumab, we suggest a BTK inhibitor (eg, ibrutinib, acalabrutinib, zanubrutinib). We suggest acalabrutinib or zanubrutinib rather than ibrutinib. If both are available, the choice depends on individual treatment goals and a desire to balance efficacy and tolerability. The two agents have not been compared directly. However, in our practice, if the goal is best efficacy with acceptable tolerability, we offer zanubrutinib. In contrast, if the goal is best tolerability with acceptable efficacy, we offer acalabrutinib.

When compared with ibrutinib, single-agent acalabrutinib has similar efficacy and an overall better tolerability profile (eg, fewer class-associated adverse effects including cardiac toxicity, atrial fibrillation, hypertension, arthralgia, and bleeding). Efficacy can be improved with the addition of obinutuzumab, although at a cost of increased toxicity. (See 'Acalabrutinib' below.)

The improvement in tolerability is more narrow for zanubrutinib (ie, less cardiac toxicity, in particular less atrial fibrillation, but similar rates of other toxicities); however, zanubrutinib is more effective than ibrutinib with deeper responses and improved progression-free survival. (See 'Zanubrutinib' below.)

Experience with acalabrutinib and zanubrutinib is relatively short and ibrutinib remains a reasonable alternative for select patients despite an increased risk for atrial fibrillation, hypertension, bleeding, and arthralgias. (See 'Ibrutinib' below.)

Other experts might offer retreatment with venetoclax-based therapy. Retreatment with venetoclax-based therapy is expected to result in a shorter remission duration as compared to the initial remission, although we do not know what duration of prior remission will allow for meaningful benefit upon retreatment. While idelalisib plus rituximab is approved by regulatory agencies in this setting, its use is limited by toxicities.

After attaining a response, younger patients with a matched donor (related or unrelated) should be evaluated for allogeneic HCT. HCT may be especially attractive in those with higher-risk CLL (eg, del17p, TP53 mutation) and/or progression within five to six years of venetoclax plus obinutuzumab. (See 'Hematopoietic cell transplantation' below.)

Late progression – For patients with progression more than three to five years following venetoclax plus obinutuzumab, options include retreatment with venetoclax plus an anti-CD20 monoclonal antibody (rituximab or obinutuzumab) or single-agent therapy with a BTK inhibitor. Mutation analysis can help guide the choice; those with mutations in BCL2 are unlikely to achieve a durable response following venetoclax-based therapy and are treated with a BTK inhibitor [15-18]. For those without BCL2 mutation, a choice is made based on comorbidities and patient preference. (See 'Consider treatment burdens and patient comorbidities' below.)

Most prospective trials evaluating BTK inhibitors and PI3K inhibitors did not include patients with prior exposure to venetoclax. Small case series and retrospective studies suggest that BTK inhibitors are effective in this population [19-21]. The largest was an international retrospective study of 188 heavily pretreated patients receiving their next treatment after venetoclax, which reported the following overall response rates (ORR) and median PFS according to class of therapy and exposure to therapies before venetoclax [19]:

BTK inhibitor (74 patients) – BTK inhibitor naïve (ORR 84 percent; PFS 32 months); BTK inhibitor intolerant (ORR 70 percent; PFS not reached); BTK inhibitor resistant (ORR 50 percent; PFS 4 months)

Chimeric antigen receptor T cells (18 patients) – ORR 67 percent; PFS 9 months

PI3K inhibitor (17 patients) – ORR 47 percent; PFS 5 months

Anti-CD20 monoclonal antibody (19 patients) – ORR 32 percent; PFS 2 months

Repeat treatment with venetoclax-based therapy was not evaluated in this study as most patients had received continuous venetoclax monotherapy and not time-limited therapy with venetoclax plus obinutuzumab. Evidence supporting retreatment with venetoclax plus obinutuzumab or rituximab is limited to case reports [22]. Further data regarding the efficacy of BTK inhibitors is described in detail below. (See 'Bruton tyrosine kinase inhibitors' below.)

Prior chemoimmunotherapy — Most patients with progression after initial chemoimmunotherapy should be treated with a BTK inhibitor (eg, ibrutinib, acalabrutinib, or zanubrutinib) or venetoclax-based therapy. Targeted therapies are well established in this setting as this population corresponds with that used in the trials demonstrating efficacy of these agents. The choice between targeted therapies is made based on comorbidities and patient preference (algorithm 1 and table 3). (See 'Consider treatment burdens and patient comorbidities' below.)

Consider treatment burdens and patient comorbidities — Treatment regimens differ significantly in their rates of CR and measurable residual disease (MRD), time to progression, and associated toxicities (table 4). The burden of administration is also different (eg, oral versus intravenous medications, continuous versus fixed duration therapy). Given these differences, patient preferences weigh substantially in the final treatment decision.

Targeted therapies have numerous drug interactions that may necessitate dose adjustments. Consultation with a clinical pharmacist may help with identifying and managing interactions. For more detailed information on potential drug-drug interactions, refer to the drug interactions program within UpToDate.

When choosing between multiple acceptable treatment options, we consider the following:

BTK inhibitor (ie, ibrutinib, acalabrutinib, zanubrutinib) – Oral medication administered as continuous therapy. Numerous drug interactions. BTK inhibitors increase the risk for bleeding, atrial fibrillation, and hypertension. Other toxicities include fatigue, rash, infections; myalgia/arthralgia. Acalabrutinib is associated with headaches and diarrhea. When compared with ibrutinib, single-agent acalabrutinib has an overall better tolerability profile with fewer class-associated adverse effects including cardiac toxicity, atrial fibrillation, hypertension, arthralgia, and bleeding [23]. The improvement in tolerability is more narrow for zanubrutinib with less cardiac toxicity, in particular less atrial fibrillation, but similar rates of other toxicities) [24].

May be preferred over venetoclax in patients with impaired creatine clearance and in those on strong CYP3A inhibitors.

Venetoclax – Oral medication either given as continuous therapy (as a single agent) or for a fixed duration (in combination with an anti-CD20 monoclonal antibody). Requires frequent visits at treatment initiation to monitor for tumor lysis syndrome (TLS), which is increased in those with renal insufficiency. Avoid use with nephrotoxic drugs and strong CYP3A inhibitors/inducers as they can alter the metabolism of venetoclax and increase the risk for TLS [25]. Other adverse events include fatigue, rash, diarrhea, and infections.

May be preferred over BTK inhibitors in those with cardiovascular disorders, uncontrolled hypertension, and/or a high risk for bleeding (eg, low platelet counts, anticoagulation).

PI3K inhibitors (ie, idelalisib, duvelisib) – Oral medication administered as continuous therapy. Fatal and/or serious toxicities can be seen, including opportunistic infections, diarrhea or colitis, cutaneous reactions, and pneumonitis. Other toxicities include fatigue and rash. Avoid or dose reduce with strong CYP3A inhibitors/inducers.

Reserved for patients with relapsed/refractory CLL with prior exposure to BTK inhibitors and venetoclax.

SPECIFIC THERAPIES

Bruton tyrosine kinase inhibitors

Acalabrutinib — Acalabrutinib is an oral, selective, irreversible inhibitor of BTK that is active in CLL. In the relapsed setting, acalabrutinib improves PFS when compared with idelalisib plus rituximab or bendamustine plus rituximab. In the treatment-naïve setting, it improves PFS when compared with chlorambucil plus obinutuzumab. In patients with relapsed CLL, acalabrutinib appears to have similar efficacy to ibrutinib, but a better toxicity profile with fewer class-associated adverse effects including cardiac toxicity, atrial fibrillation, hypertension, arthralgia, and bleeding. Extrapolation of studies in previously untreated patients suggests that the addition of obinutuzumab increases efficacy and increases toxicity with higher rates of cytopenias and infections. (See "Selection of initial therapy for symptomatic or advanced chronic lymphocytic leukemia/small lymphocytic lymphoma", section on 'Acalabrutinib'.)

Regulatory approval and administrationAcalabrutinib is approved by the US Food and Drug Administration (FDA) and the European Medicines Agency (EMA) for initial and subsequent therapy for adults with CLL at a dose of 100 mg every 12 hours [26]. Treatment is continued until progression or unacceptable toxicity.

Toxicities – The most common toxicities include anemia, neutropenia, thrombocytopenia, headache, upper respiratory tract infection, and diarrhea. Serious, potentially life-threatening, toxicities include opportunistic infections, bleeding, arrhythmias, and second primary malignancies.

Efficacy in relapsed CLL/SLL – In a multicenter phase 3 trial (ASCEND), patients with relapsed or refractory CLL were randomly assigned to acalabrutinib (155 patients) or to the physician's choice of idelalisib plus rituximab (119 patients) or bendamustine plus rituximab (36 patients) [27,28]. After a median follow-up of 47 months, acalabrutinib improved PFS (4-year PFS 62 versus 19 percent, HR 0.28; 95% CI 0.20-0.38). Median OS had not been reached in any of the arms. Estimated 4-year OS was numerically higher with acalabrutinib but this difference did not reach statistical significance (4-year OS 78 versus 65 percent, HR 0.69; 95% CI 0.46-1.04); 52 percent of patients on the control arm received acalabrutinib at progression.

Acalabrutinib was better tolerated than idelalisib plus rituximab, with lower rates of discontinuation due to toxicity (23 versus 67 percent). The most common toxicities (all grades) with acalabrutinib were neutropenia (24 percent), headache (23 percent), diarrhea (21 percent), upper respiratory tract infection (20 percent), anemia (18 percent), and cough (18 percent). Additional notable toxicities included atrial fibrillation (8 percent), hypertension (8 percent), and major hemorrhage (3 percent).

While these results support the activity of acalabrutinib in CLL, interpretation is limited by the choice of therapy in the control arm, which did not include some of the more active available targeted therapies (eg, venetoclax, ibrutinib).

Comparison with ibrutinib – In a multicenter, open-label phase 3 trial (ELEVATE-RR), 533 patients with relapsed CLL with del(17p) or del(11)(q22.3) were randomly assigned to acalabrutinib or to ibrutinib [23,29]. After a median follow-up of 41 months, the two treatments produced similar PFS (median 38 months; HR 1.00, 95% CI 0.79-1.27), a finding that was maintained across prespecified subgroups. Overall survival data are immature with approximately 75 percent of patients alive at last follow-up.

Acalabrutinib resulted in fewer patients with bleeding events (38 versus 51 percent), diarrhea (35 versus 46 percent), arthralgia (16 versus 23 percent), hypertension (9 versus 23 percent), contusion (12 versus 18 percent), and atrial fibrillation (9 versus 16 percent), and higher rates of headache (35 versus 20 percent) and cough (29 versus 21 percent).

Fewer patients assigned to acalabrutinib discontinued treatment due to toxicity (15 versus 22 percent). These lower rates of adverse events are consistent with what has been seen in other trials of acalabrutinib in CLL/SLL [27,30-32].

Zanubrutinib — Zanubrutinib is an oral, selective, irreversible inhibitor of BTK. Where available, it is one of our preferred treatments for relapsed or refractory BTK-naïve CLL/SLL. When compared with ibrutinib in the relapsed setting, zanubrutinib improves PFS and has less cardiac toxicity, in particular less atrial fibrillation, but similar rates of other toxicities. It has not been directly compared with acalabrutinib in this population.

Regulatory approval and administrationZanubrutinib is approved by the US FDA and EMA for initial and subsequent therapy for adults with CLL/SLL at a dose of either 160 mg twice daily or 320 mg once daily. Treatment is continued until progression or unacceptable toxicity.

Efficacy and comparison with ibrutinib – In a multicenter, open label, phase 3 trial (ALPINE), 652 patients with relapsed or refractory CLL/SLL were randomly assigned to zanubrutinib (160 mg twice daily) or ibrutinib (420 mg once daily) until disease progression [24,33]. After a median follow-up of 29.6 months, zanubrutinib improved overall response rate (84 versus 74 percent) and PFS (78 versus 66 percent at 24 months; HR 0.65, 95% CI 0.49-0.86). OS data are immature. Numerically fewer deaths were reported in the zanubrutinib arm (48 versus 60 deaths), but this difference was not statistically significant (HR for death 0.76, 95% CI 0.51-1.11).

Toxicities – The most common toxicities include infection, neutropenia, hypertension, diarrhea, anemia, and arthralgia. Serious, potentially life-threatening, toxicities include opportunistic infections, bleeding, arrhythmias, and second primary malignancies.

In ALPINE, those assigned to zanubrutinib had fewer adverse events leading to drug discontinuation (15.4 versus 22.2 percent) and similar treatment-related deaths (10.2 versus 11.1 percent) [24,33]. Zanubrutinib resulted in less cardiotoxicity with fewer cardiac disorders (21 versus 30 percent), less atrial fibrillation/flutter (5.2 versus 13.3 percent), and fewer treatment discontinuations due to cardiac disorders (1 versus 14 patients). These lower rates of cardiac events are consistent with what has been seen in other trials of zanubrutinib in CLL/SLL [8].

There were similar rates of hypertension of any grade (24 versus 23 percent) and grade 3 hypertension (15 versus 14 percent).

Zanubrutinib resulted in a higher rate of neutropenia (29 versus 24 percent), yet similar rates of grade 3 or greater infections (27 versus 28 percent). Major bleeding occurred with similar frequency in the two arms (3.7 versus 4.3 percent).

Ibrutinib — Ibrutinib is an oral irreversible inhibitor of Bruton tyrosine kinase (BTK) and an option for patients with relapsed or refractory ibrutinib-naïve CLL. It is well tolerated and produces sustained remissions in a majority of patients [34-39]. While there is some evidence that progression-free survival (PFS) and overall survival (OS) are shorter in subgroups with del(17p) or del(11q) [39-41], for patients with del(17p), the outcomes with ibrutinib treatment are much better than for chemotherapy-based treatment [42]. In patients with relapsed CLL, we prefer acalabrutinib or zanubrutinib since they have at least similar efficacy to ibrutinib, but better toxicity profiles, in particular with less cardiotoxicity. (See 'Acalabrutinib' above and 'Zanubrutinib' above.)

Regulatory approval and administrationIbrutinib is approved by the US FDA and the EMA for both previously treated as well as previously untreated patients with CLL. Treatment is continued until progression or unacceptable toxicity. Administration and use as initial therapy are described separately.

Efficacy and toxicity in relapsed CLL/SLL – The efficacy of ibrutinib in relapsed/refractory CLL was demonstrated in a multicenter, open-label randomized trial (RESONATE) that compared ibrutinib versus ofatumumab in 391 patients with relapsed/refractory CLL/SLL [43-47]. Ibrutinib led to a clear improvement in PFS (59 versus 3 percent at three years, median 44 versus 8.1 months, hazard ratio [HR] 0.15; 95% CI 0.113-0.196) and demonstrated improved OS (median 68 versus 65 months, HR 0.81; 95% CI 0.60-1.09) despite extensive cross-over. Ibrutinib resulted in more diarrhea, pyrexia, and nausea and lower rates of cough and infusion-related reaction. Ibrutinib also had higher than expected rates of hypertension (21 percent), atrial fibrillation (12 percent), and major hemorrhage (10 percent).

In a multicenter, single-arm trial of ibrutinib in 144 patients with relapsed/refractory CLL/SLL with del(17p), the overall response rate (ORR) was 83 percent (10 percent complete) at a median follow-up of 28 months [48]. Estimated rates of PFS and OS at two years were 63 and 75 percent, respectively. Richter transformation occurred in 17 patients, 11 of which transformed within the first six months. Toxicities were similar to those seen in other trials of ibrutinib.

Case reports and a retrospective analysis have also reported high response rates when ibrutinib is used after venetoclax-based therapy [19-21]. (See 'Prior venetoclax plus obinutuzumab' above.)

Impact of adding rituximab – The addition of rituximab may shorten time to response but without improved PFS [49,50]. In a randomized phase 2 trial, 208 patents with relapsed CLL or treatment-naïve high-risk CLL were randomly assigned to receive ibrutinib with or without rituximab [49]. After a median follow-up of 36 months, the two treatment arms had similar ORR (92 percent each), PFS (HR 1.16; 95% CI 0.55-2.44), and OS (HR 0.75; 95% CI 0.28-2.02). The addition of rituximab resulted in a quicker time to complete response (CR; median 12 versus 22 months) and lower levels of minimal residual disease.

Combination therapy – While initial reports suggest improved clinical outcomes when ibrutinib is added to classical chemoimmunotherapy (eg, bendamustine plus rituximab [BR]), it is not known whether this combination improves on outcomes seen with single-agent ibrutinib or whether it allows for the earlier discontinuation of ibrutinib therapy [51-55]. While initial reports suggest deep responses when ibrutinib is given in combination with venetoclax, further study is needed before this combination should be used in routine practice. (See "Selection of initial therapy for symptomatic or advanced chronic lymphocytic leukemia/small lymphocytic lymphoma", section on 'Ibrutinib plus venetoclax'.)

Pirtobrutinib — Pirtobrutinib is an oral, noncovalent (reversible) BTK inhibitor. Initial studies suggest that pirtobrutinib is effective and well tolerated, although it has not been directly compared with other agents.

Since they do not require binding to the BTK C481 residue, noncovalent BTK inhibitors like pirtobrutinib are active in patients with BTK C481 mutations commonly seen in those progressing on a covalent (irreversible) BTK inhibitor, such as ibrutinib, acalabrutinib, or zanubrutinib. In contrast, CLL progressing on a noncovalent BTK inhibitor exhibits other mutations in BTK or PLCg2, some of which also confer resistance to covalent BTK inhibitors, highlighting the importance of this pathway for efficacy [56].

Regulatory approval and administrationPirtobrutinib is approved by the FDA for treatment of adults with CLL/SLL who have received at least two prior lines of therapy, including a BTK inhibitor and a BCL-2 inhibitor. The usual dose is 200 mg once daily. Treatment is continued until progression of unacceptable toxicity.

Efficacy and toxicity – In a phase 1/2 trial of the noncovalent BTK inhibitor pirtobrutinib in 317 patients CLL/SLL (BRUIN), the 247 patients with prior exposure to a BTK inhibitor had an overall response rate (including partial response with lymphocytosis) of 82 percent and median progression-free survival of 19.6 months [57,58]. On subgroup analysis, response rates of 70 to 80 percent were seen in those with prior exposure to a BTK inhibitor plus venetoclax, progression on the prior BTK inhibitor, and BTK C481 mutation. A lower response rate was seen in those with PLCg2 mutation (10 of 18 patients, 56 percent).

Pirtobrutinib was well tolerated, and few patients discontinued therapy due to toxicity (2.8 percent). The most common adverse events were infection (71 percent), bleeding (43 percent), and neutropenia (33 percent). Adverse events of particular interest with BTK inhibitors included hypertension (14 percent), atrial fibrillation and flutter (3.8 percent), and major hemorrhage (2.2 percent).

These results illustrate the activity of a noncovalent BTK inhibitor in CLL/SLL previously exposed to a covalent BTK inhibitor, including CLL/SLL with BTK C481 mutation, and those with refractory disease.

BCL2 inhibitors: Venetoclax — Venetoclax is an oral, selective, small molecule inhibitor of BCL2 and one of our preferred treatments for patients with relapsed or refractory CLL, especially for those progressing after a BTK inhibitor. The combination of venetoclax plus rituximab improved PFS and OS when compared with bendamustine plus rituximab in a randomized trial. The combination of venetoclax plus obinutuzumab offers a time-limited treatment option, as studied in treatment-naïve patients. Deep responses can be achieved by combining a BTK inhibitor with venetoclax. Use of venetoclax plus ibrutinib is discussed in more detail separately. (See "Selection of initial therapy for symptomatic or advanced chronic lymphocytic leukemia/small lymphocytic lymphoma", section on 'Ibrutinib plus venetoclax'.)

Regulatory approvalVenetoclax is approved by the US FDA and EMA for the treatment of patients with CLL [59,60].

Administration and TLS prophylaxisVenetoclax is started at 20 mg daily and increased gradually over five weeks to a final daily dose of 400 mg. If used, anti-CD20 monoclonal antibodies are not started concurrently with venetoclax (ie, obinutuzumab is started before venetoclax, rituximab is started after venetoclax).

The dose escalation schedule, dose modifications for adverse reactions, and important drug interactions are provided in the US prescribing information for venetoclax [61]. With this dose escalation schedule, approximately 6 percent of patients will develop tumor lysis syndrome (TLS) (table 5). The patient's risk of TLS must be estimated prior to the initiation of therapy to guide management:

Hypouricemic agents (allopurinol or rasburicase, depending on TLS risk category) are started two to three days prior to the initiation of venetoclax. All patients require laboratory monitoring for TLS (potassium, uric acid, phosphorus, calcium, creatinine), aggressive hydration, and correction of any electrolyte disturbances and elements of reversible renal failure. The risk of TLS increases with tumor mass and renal dysfunction. Venetoclax has not been studied in patients with severe renal impairment (creatinine clearance <30 mL/min and patients on dialysis).

Patients at low risk for TLS (ie, all lymph nodes <5 cm and absolute lymphocyte count [ALC] <25 x 109/L) may be managed as outpatients with oral hydration (1.5 to 2 liters), allopurinol, and frequent laboratory studies (pre-dose, 6 to 8 hours, and 24 hours).

Patients at medium risk for TLS (ie, any lymph node 5 cm to <10 cm or ALC ≥25 x 109/L) can be managed in either the outpatient or inpatient setting depending on comorbidities. Hospitalization is preferred for patients with creatinine clearance <80 mL/min and allows for frequent monitoring and supplementation of oral intake with intravenous fluids. Allopurinol is used as the hypouricemic agent.

Patients at high risk for TLS (ie, any lymph node ≥10 cm or any lymph node ≥5 cm and an ALC ≥25 x 109/L) should be hospitalized at the time of the first doses at the 20 mg and 50 mg level. This allows for more frequent laboratory studies (pre-dose, 4, 8, 12, and 24 hours) and the administration of both oral (1.5 to 2 liters) and intravenous (150 to 200 mL/hour as tolerated) fluids. While most patients receive allopurinol as the hypouricemic agent, rasburicase is preferred if uric acid is elevated at baseline.

A faster dose escalation has been used for selected patients at experienced centers with close inpatient monitoring [62]. TLS may occur on resumption of venetoclax after treatment interruption. Thus, the risk of TLS should be reassessed if the dose interruption lasted more than one week during the dose escalation phase, or more than two weeks after completion of the dose escalation phase. The diagnosis and management of TLS is presented in more detail separately. (See "Tumor lysis syndrome: Prevention and treatment" and "Tumor lysis syndrome: Pathogenesis, clinical manifestations, definition, etiology and risk factors".)

Decision to add an anti-CD20 monoclonal antibody – Data regarding the efficacy and safety of venetoclax come from single-arm prospective trials that have reported high response rates with single-agent therapy or in combination with rituximab or obinutuzumab, and from a randomized trial that demonstrated a PFS and OS advantage for venetoclax plus rituximab over bendamustine plus rituximab [63-69]. Nonrandomized comparisons have had mixed results with some suggesting higher CR rates when venetoclax is combined with rituximab or obinutuzumab and others reporting no clear benefit from the addition of an anti-CD20 monoclonal antibody [67-70].

As more data accumulate and experience with combination therapy expands, our contributors differ in their use of venetoclax for relapsed CLL. Some prefer to use venetoclax as a single agent, especially for those wanting an all-oral regimen, while others offer venetoclax in combination with rituximab or obinutuzumab based on cross-trial comparisons that suggest deeper responses with combination therapy and the ability to administer a time-limited course (24 months) in patients without TP53 mutation or 17p deletion. We anticipate that our approach will evolve as we gain more experience with combination therapy.

Toxicities – The most common short-term toxicities include neutropenia, thrombocytopenia, anemia, diarrhea, nausea, upper respiratory tract infection, cough, musculoskeletal pain, fatigue, and edema. Serious, potentially life-threatening toxicities include tumor lysis syndrome and infections. Long-term venetoclax exposure may also influence the clonal dynamics of non-CLL cells, which may manifest as cytopenias in the setting of BAX mutations in myeloid cells [71].

Efficacy in relapsed/refractory CLL/SLL

Following ibrutinib – An interim analysis of a multicenter, phase 2, open-label study evaluated the efficacy of venetoclax in 91 patients with heavily pretreated CLL who relapsed after or were refractory to ibrutinib [65]. The ORR was 65 percent (9 percent complete) and similar among patients with and without high-risk genetic abnormalities (eg, del17p or TP53 mutations). After a median follow-up of 14 months, estimated PFS and OS at 12 months were 75 and 91 percent. Median PFS was 25 months. In a separate report of this trial, venetoclax was associated with a similar response rate (67 percent ORR, <1 percent complete) and estimated PFS at 12 months (79 percent) among 36 patients with relapse after idelalisib [72].

Patients with 17p deletion – An international, single-arm phase 2 trial evaluated venetoclax in 153 patients with relapsed or refractory CLL with 17p deletion and 5 patients with previously untreated CLL with 17p deletion [64,66]. Most patients were hospitalized for the initial dose, and all received TLS prophylaxis and a gradual dose escalation as recommended in the package insert. At a median follow-up of 27 months, the ORR was 77 percent (20 percent complete) with a median time to first response of one month (range 0.5 to 4.4 months) and an estimated median duration of response of 33 months. At two years, estimated PFS and OS were 54 and 73 percent, respectively. There were 53 deaths on study; 44 were due to disease progression, one was deemed possible due to treatment, and the rest were deemed unrelated.

Venetoclax plus rituximab superior to bendamustine plus rituximab (BR) – In an international, open-label, phase 3 trial (the MURANO trial), 389 patients with relapsed or refractory CLL were randomly assigned to six months of BR or to two years of venetoclax plus six months of rituximab [22,68,73]. Cross-over to venetoclax was not allowed at the time of progression. After a median follow-up of 59 months, venetoclax plus rituximab improved PFS (median 54 versus 17 months; HR 0.19; 95% CI 0.15-0.26) and OS (82 versus 62 percent at 5 years; HR 0.40; 95% CI 0.26-0.62). The benefit was maintained in several subgroup analyses, including patients with high-risk disease and older adults. The most common toxicities of any grade with venetoclax were neutropenia (61 percent), diarrhea (40 percent), nausea (21 percent), anemia (16 percent), fatigue (18 percent), upper respiratory tract infection (22 percent), and thrombocytopenia (13 percent). Serious adverse events were seen in 46 percent and fatal adverse events occurred in 5 percent.

Patients assigned to venetoclax plus rituximab were more likely to achieve undetectable minimal residual disease (uMRD), a status predictive of superior PFS and OS [22,74]. Of the 83 patients who attained uMRD after venetoclax plus rituximab, 32 (39 percent) remained with uMRD at five years from the start of treatment, while 28 (34 percent) had confirmed MRD conversion without progression and 23 (30 percent) had progressive disease. At first progression, an at least partial response was attained following retreatment with venetoclax-based therapy in 13 of 18 patients (72 percent) and following treatment with ibrutinib in 14 of 14 patients (100 percent) [22]. These longer-term results confirm the PFS and OS benefit of venetoclax plus rituximab in this population and demonstrate prolonged remissions following a time-limited course of therapy.

Nonrandomized studies have reported prolonged treatment-free periods in patients achieving deep remission following limited duration venetoclax plus rituximab with retreatment upon progression [75].

Venetoclax plus obinutuzumab – The combination of venetoclax plus obinutuzumab has demonstrated superior response rates and PFS when compared with chlorambucil plus obinutuzumab in the treatment-naïve population [76]. Small prospective studies have also demonstrated deep responses in patients with relapsed CLL [69]. The use of venetoclax plus obinutuzumab for the initial treatment of CLL is discussed in detail separately. (See "Selection of initial therapy for symptomatic or advanced chronic lymphocytic leukemia/small lymphocytic lymphoma", section on 'Venetoclax plus obinutuzumab'.)

PI3K inhibitors

Idelalisib — Idelalisib is an oral inhibitor of phosphoinositide 3'-kinase (PI3K) delta that has shown activity in patients with relapsed or refractory CLL, although its use may be limited by toxicity [77-79]. We reserve PI3K inhibitors for patients with relapsed/refractory CLL with prior exposure to BTK inhibitors and venetoclax.

Regulatory approval Idelalisib is approved by the US FDA for the treatment of patients with relapsed CLL, in combination with rituximab, for whom rituximab alone would be considered appropriate therapy due to other comorbidities [80]. Trials of idelalisib included some patients with significant comorbidities, including cardiac and renal dysfunction [81]. Approval by the EMA is for patients who have received at least one prior therapy.

Administration and warnings – The starting dose is 150 mg twice daily, and suggested dose reductions for toxicities are provided in the package insert. The following should be noted:

Idelalisib is associated with an increase in opportunistic infections and the manufacturer suggests prophylaxis for Pneumocystis jirovecii pneumonia (PJP) and monitoring for cytomegalovirus. (See "Prevention of infections in patients with chronic lymphocytic leukemia", section on 'Bruton tyrosine kinase and phosphatidylinositol 3-kinase inhibitors' and "Risk of infections in patients with chronic lymphocytic leukemia", section on 'Phosphatidylinositol 3-kinase inhibitors'.)

Idelalisib carries boxed warnings regarding an increased risk of fatal and/or serious hepatotoxicity, diarrhea, colitis, intestinal perforation, and pneumonitis. The package insert also includes warnings about severe cutaneous reactions, hypersensitivity reactions, and neutropenia. Idelalisib has also resulted in hypertriglyceridemia, hyperglycemia, alanine aminotransferase (ALT) elevations, and aspartate aminotransferase (AST) elevations [82]. Hepatic function should be evaluated prior to and during treatment. Hepatotoxicity may be more common among younger patients and those without prior therapy [83].

In addition to early-onset mild diarrhea, there are a few patients with a severe, wasting diarrhea due to histologically proven colitis, which is poorly understood. Colitis usually occurs after more than eight months of therapy and the overall incidence of severe (grade 3 or worse) colitis is low. Management is not clear, but usually treatment is paused, and systemic or local corticosteroids used. After recovery, many patients tolerate restarting idelalisib at a lower dose (eg, 100 mg twice daily).

The lymphocyte count typically increases dramatically within 24 hours of starting idelalisib plus rituximab, peaks during the second week of therapy, and usually resolves by week 12.

When compared with those reported in the trials leading to approval, patients treated in routine practice tend to be older, have more comorbidities, and more complications, especially infectious complications [84]. Idelalisib is administered with rituximab, which may result in the following major toxicities:

Infusion reactions (ie, fevers, rigors, and hypotension). (See "Infusion-related reactions to therapeutic monoclonal antibodies used for cancer therapy".)

Infections related to immunosuppression. (See "Secondary immunodeficiency induced by biologic therapies", section on 'Monoclonal antibodies to B cells'.)

Hepatitis B virus reactivation among patients positive for hepatitis B surface antigen (HBsAg) or antibodies against hepatitis B core antigen (anti-HBc). (See "Hepatitis B virus reactivation associated with immunosuppressive therapy".)

Rarely, JC virus infection can result in potentially fatal progressive multifocal leukoencephalopathy. (See "Progressive multifocal leukoencephalopathy (PML): Epidemiology, clinical manifestations, and diagnosis".)

Efficacy and toxicity in relapsed/refractory CLL/SLL – In randomized phase 3 trials, the addition of idelalisib improved PFS and OS, but increased severe toxicity when added to rituximab [81,85], ofatumumab [86], or the combination of bendamustine plus rituximab [87].

As an example, when compared with rituximab plus placebo, the combination of idelalisib plus rituximab led to a clear improvement in PFS (median 19.4 versus 6.5 months), and a more modest improvement in OS (median 41 versus 35 months; HR 0.8 [95% CI 0.5-1.1]) despite extensive cross-over [81,85]. However, at least one grade 3 or greater toxicity occurred in 74 percent of patients receiving idelalisib, 54 percent of patients receiving placebo, and >90 percent of patients in the extension study, with neutropenia and pneumonia being the most common. Febrile neutropenia was reported in 6 percent. More common, but less severe, toxicities included fever, fatigue, nausea, chills, and diarrhea. The frequency of diarrhea increased with longer exposure to idelalisib. Severe elevations in hepatic aminotransferase levels were described (ALT in 9 percent, AST in 6 percent); most resolved upon holding idelalisib and none resulted in permanent drug discontinuation. There were several cases of PJP pneumonia (5 cases), fungal infection (22 cases), cytomegalovirus (2 cases), and progressive multifocal leukoencephalopathy (1 case). None of the patients with PJP pneumonia had received PJP prophylaxis.

Patients who have received venetoclax may be unlikely to achieve a durable response with idelalisib; in one international retrospective study of patients with relapsed or refractory CLL who discontinued venetoclax-based therapy for any reason, those treated with a PI3K inhibitor had an ORR of 47 percent and a median PFS of five months [19].

Duvelisib — Duvelisib is an oral inhibitor of PI3K delta and gamma isoforms that has shown activity in small nonrandomized studies of patients with multiply relapsed CLL/SLL and superior PFS when compared with ofatumumab in a multicenter randomized trial [88-92]. However, duvelisib has substantial toxicity; increased risk of death and serious side effects were highlighted in an FDA warning in June 2022 [93], and more safety concerns were raised for PI3K inhibitors as a class in April 2022 [94]. We reserve the use of duvelisib for patients with multiply relapsed disease, usually after treatment with ibrutinib and venetoclax, with or without prior chemoimmunotherapy.

Regulatory approvalDuvelisib is approved by the US FDA as a single agent for the treatment of patients with relapsed CLL/SLL who have received at least two prior therapies [88].

Administration – The starting dose is 25 mg administered orally twice a day until progression or unacceptable toxicity. Suggested dose reductions for toxicities are provided in the package insert in addition to dose adjustments for patients taking CYP3A inhibitors/inducers.

Toxicity – Fatal and/or serious toxicities can be seen, including opportunistic infections, diarrhea or colitis, cutaneous reactions, and pneumonitis.

Hepatic function and blood counts must be monitored for hepatotoxicity and neutropenia. The prescribing information recommends prophylaxis for PJP pneumonia and suggests that clinicians consider the use of prophylactic antivirals to prevent cytomegalovirus infection and reactivation. (See "Risk of infections in patients with chronic lymphocytic leukemia", section on 'Duvelisib' and "Prevention of infections in patients with chronic lymphocytic leukemia", section on 'Bruton tyrosine kinase and phosphatidylinositol 3-kinase inhibitors'.)

Lymphocytosis is expected during the first few weeks of therapy and may persist for several weeks.

Efficacy – While duvelisib can achieve partial responses in most patients, survival may be worse due to increased treatment-related deaths.

In a multicenter trial (DUO), 319 patients with relapsed or refractory CLL/SLL were randomly assigned to receive duvelisib (25 mg twice daily until progression) or a finite course of ofatumumab [91,95]. Crossover to the alternative therapy was allowed at the time of progression and more than half of patients assigned to ofatumumab received duvelisib at progression. Duvelisib resulted in a higher ORR (74 versus 45 percent) and superior median PFS (13.3 versus 9.9 months, HR 0.52). A subset analysis demonstrated a benefit in all patient populations, including those with 17p deletion and/or TP53 mutation. In the final OS analysis with a median follow-up of 63 months, OS was worse with duvelisib; however, these findings did not reach statistical significance (median OS 52 versus 63 months; HR 1.09, 95% CI 0.79-1.51) [93]. Patients receiving duvelisib had more deaths due to adverse events, serious adverse events, grade ≥3 adverse events, and treatment modifications due to adverse events.

Duvelisib has not been directly compared with other targeted agents, such as idelalisib (which inhibits PI3K delta and not gamma), ibrutinib (which inhibits BTK), or venetoclax (which inhibits BCL2). While indirect comparisons are complicated by differences in study populations and other biases, indirect data suggest duvelisib has similar toxicities to idelalisib and may result in shorter remissions and different toxicities than ibrutinib and venetoclax.

Hematopoietic cell transplantation — Patients with CLL are generally older adults with a median age greater than 70 years, and due to the relatively benign course of the disease in the majority of patients, only selected patients are candidates for intensive treatments such as HCT. The determination of transplant eligibility should be made based on a risk-benefit assessment and the needs and wishes of the patient. (See "Determining eligibility for allogeneic hematopoietic cell transplantation".)

The ideal timing of HCT in CLL is unknown and transplants should be carried out in the context of a clinical trial whenever possible. Which patients may be candidates for HCT is changing with advances in targeted therapies and our knowledge of who might respond to these therapies. A conceptual framework for determining HCT candidacy has been proposed by the European Research Initiative on CLL and European Society for Blood and Marrow Transplantation [96]. This framework identifies patients with CLL resistant to chemoimmunotherapy and resistant to a BTK inhibitor and/or BCL2 inhibitor as having particularly high-risk disease ("high risk-II" disease) that might be suitable for HCT.

An HCT trial may also be appropriate for young patients with relapsed or refractory CLL already exposed to a BTK inhibitor and venetoclax [97]. HCT may also be considered for patients with histologic transformation to a more aggressive histology. (See "Hematopoietic cell transplantation in chronic lymphocytic leukemia".)

There have been no randomized trials comparing allogeneic HCT versus non-HCT based strategies in CLL. Retrospective studies and small prospective trials have reported outcomes following HCT conducted in highly selected patient populations prior to the widespread use of targeted agents [98-102]. Conditioning regimen, donor source, and other details of care vary. As examples:

A retrospective study of 68 patients with high-risk CLL who underwent reduced intensity conditioning followed by unrelated cord blood transplantation reported three year rates of relapse (16 percent), nonrelapse mortality (39 percent), PFS (45 percent), and OS (54 percent) [101].

Another retrospective study of 117 patients with CLL who underwent haploidentical donor transplantation reported five year cumulative incidences of relapse (26 percent), nonrelapse mortality (44 percent), PFS (31 percent), and OS (38 percent) [102].

A prospective trial of nonmyeloablative conditioning followed by allogeneic HCT in 90 patients with poor-risk CLL as defined by primary refractory disease, initial relapse within 12 months, relapse after autologous HCT, or progressive disease in the presence of unfavorable genetic features reported 10 year rates of relapse (46 percent), nonrelapse mortality (20 percent), and OS (51 percent) [103-105].

Limited data suggest that prior exposure to targeted therapy does not impact outcomes following HCT [106,107]. As an example, a multicenter, retrospective study of 65 patients with CLL and prior exposure to at least one targeted therapy reported two year rates of relapse (27 percent), nonrelapse mortality (13 percent), PFS (63 percent), and OS (81 percent) [106]. This study included patients with prior exposure to ibrutinib, venetoclax, and idelalisib, and a majority also had prior chemoimmunotherapy. In multivariate analysis, HCT comorbidity index score of one or greater was associated with inferior PFS. In contrast, PFS did not appear to be impacted by the number or types of targeted therapies used, the timing of targeted therapy, the depth of response in those with responding disease, or the presence of high-risk disease characteristics (eg, TP53 mutation, del17p).

The decision to proceed with HCT is highly dependent upon patient values and preferences. Patients with different values and preferences will make different choices.

PALLIATION OF SYMPTOMS — Anti-CD20 monoclonal antibodies and corticosteroids may offer a short-lived partial response (PR) with only moderate toxicity. Splenectomy may be useful in patients with splenomegaly and profound cytopenias unresponsive to other therapies. Radiation therapy to the spleen or other bulky lymphoid tissue is an option for patients with bulky disease compromising critical structures. Leukapheresis is seldom used in CLL.

Anti-CD20 monoclonal antibodies — When used as single agents, anti-CD20 monoclonal antibodies (rituximab, ofatumumab, obinutuzumab) are usually well tolerated and can result in short-lived PRs.

Rituximab – Single-agent rituximab produces PRs when used as a single agent for the treatment of refractory CLL; these responses are short lived with a median response time of 20 weeks [108-110]. Rituximab may also be combined with corticosteroids for palliation. A prospective trial of rituximab plus high-dose methylprednisolone in 14 patients with fludarabine-refractory CLL, 21 percent of whom had previously received rituximab, reported an overall response rate (ORR) of 93 percent with a median time to progression of 15 months [111].

Obinutuzumab – Small prospective studies have demonstrated the efficacy of single-agent obinutuzumab in CLL [112,113]. In a phase I/II study of obinutuzumab in 23 patients with relapsed/refractory CLL, 14 patients (61 percent) had a documented response [112]. The estimated median PFS and duration of response were 11 and 9 months, respectively. In a second study, the ORR was 42 percent among the 64 patients with relapsed/refractory CLL treated with single-agent obinutuzumab [114].

Ofatumumab – Single-agent ofatumumab has demonstrated PR rates of approximately 50 percent in patients with relapsed or refractory CLL [115-121]. With appropriate premedication, infusion reactions are seen in approximately 44 percent of patients receiving their first infusion and decreased in number and intensity with subsequent infusions [115,116]. The most common severe (grade 3/4) toxicities include thrombocytopenia (9 percent), neutropenia (6 percent), anemia (3 percent), and infections (9 percent). Although ofatumumab has been withdrawn from the Canadian and European markets and its use in CLL has been decreasing, it is available in the United States.

The major toxicities of anti-CD20 antibodies include:

Infusion reactions (ie, fevers, rigors, and hypotension). (See "Infusion-related reactions to therapeutic monoclonal antibodies used for cancer therapy".)

Infections related to immunosuppression. (See "Secondary immunodeficiency induced by biologic therapies", section on 'Monoclonal antibodies to B cells'.)

Hepatitis B virus reactivation among patients positive for hepatitis B surface antigen (HBsAg) or antibodies against hepatitis B core antigen (anti-HBc). (See "Hepatitis B virus reactivation associated with immunosuppressive therapy".)

Rarely, JC virus infection can result in potentially fatal progressive multifocal leukoencephalopathy. (See "Progressive multifocal leukoencephalopathy (PML): Epidemiology, clinical manifestations, and diagnosis".)

Corticosteroids — Corticosteroids, such as prednisone, have been administered as single agents in CLL, usually in an initial dose of 20 to 60 mg/day by mouth, with dose reduction occurring in a graduated fashion thereafter [122]. With steroid therapy alone, one-third of patients have shrinkage of lymph nodes and reduction in splenic size. An increase in the lymphocyte count often occurs in the first one to two months of treatment, followed by a subsequent decline. A shift of lymphocytes from lymphoid organs and bone marrow into the blood is thought to be responsible for this initial increase in blood lymphocyte count.

The major indication for corticosteroids in CLL is in the management of antibody-mediated (autoimmune) anemia and thrombocytopenia, which improve in approximately two-thirds of patients. Three to six months of treatment are usually required for achievement of clinical response. (See "Overview of the complications of chronic lymphocytic leukemia", section on 'Autoimmune hemolytic anemia'.)

In addition to psychiatric, metabolic (eg, hyperglycemia), gastrointestinal, and musculoskeletal toxicities, a major side effect of chronic corticosteroid therapy in patients with CLL is increased susceptibility to infections. (See "Major adverse effects of systemic glucocorticoids".)

High-dose corticosteroids have been used in CLL as salvage therapy in patients with end-stage or treatment-resistant disease. Usual starting doses of dexamethasone are 40 mg/day orally on days 1 through 4, 9 through 12, and 17 through 20, every 28 days [123,124]. An alternative is methylprednisolone 1 g/m2 daily for five days [125,126]. Clinical responses can occasionally be gratifying, although toxicity in the form of susceptibility to infection is common and troublesome, often requiring dose reduction.

Splenectomy — Most patients with CLL who develop splenomegaly and profound cytopenia will initially respond to chemoimmunotherapy. However, some patients, especially those with advanced disease, will demonstrate marked splenomegaly along with cytopenias refractory to treatment [127-130]. Splenectomy is preferred for the management of these patients since it provides long-term benefit. Splenic irradiation can achieve a temporary improvement in blood counts in patients that are not surgical candidates.

A number of reports have demonstrated the hematologic and survival benefits of splenectomy in patients with splenomegaly, often massive and with mechanical symptoms, and/or cytopenias unresponsive to chemotherapy [127-134]. Platelet count increments on the order of 50,000/microL and hemoglobin concentration increments of >2 g/dL can be achieved in up to 80 to 90 percent of patients and improvement in absolute neutrophil count in 60 percent. Improvements in hemoglobin and neutrophil counts appear to correlate with splenic weight. In addition, a case-control retrospective analysis reported a trend toward improved survival among Rai stage IV patients undergoing splenectomy [132]. Perioperative mortality (9 percent) is more common in patients with a preoperative performance status ≥2 (table 6).

Radiation therapy — The major indication for radiation therapy in CLL is the presence of large, bulky lymphoid masses causing compression symptoms, especially if the CLL has been unresponsive to other treatments [135]. CLL lymphocytes are extremely radiation sensitive [136,137]; treatment usually results in a rapid shrinkage of lymphoid masses. However, in most cases the benefits have been only temporary and/or used in a palliative setting [138].

In one study, 52 patients with stages II to IV CLL were treated with a median splenic irradiation dose of 7 gray (Gy), given in weekly treatments of 1 Gy [139]. Clinical response was noted in 82 percent, with a median remission duration of nine months (range: 3 to 24 months). Since splenic irradiation does not usually result in long-term control, splenectomy is the preferred option if the patient is in a satisfactory clinical state.

Management of complications — Patients with CLL commonly develop complications associated with their intrinsic immune dysfunction resulting in immunodeficiency and autoimmune disorders. The most common complications are infection, anemia, and thrombocytopenia. Rare but potentially life-threatening complications include leukostasis, tumor lysis syndrome, and second cancers. These are discussed in more detail separately. (See "Overview of the complications of chronic lymphocytic leukemia" and "Prevention of infections in patients with chronic lymphocytic leukemia" and "Risk of infections in patients with chronic lymphocytic leukemia".)

INVESTIGATIONAL THERAPIES — Most commonly, there is no better therapy to offer a patient than enrollment onto a well-designed, scientifically valid, peer-reviewed clinical trial. Additional information and instructions for referring a patient to an appropriate research center can be obtained from the United States National Institutes of Health.

Drugs under development — Many agents are under active investigation. These include novel agents (eg, additional noncovalent BTK inhibitors, BTK degraders [140]), combinations of agents already used in CLL, and agents approved for other diseases.

We await results of these studies before incorporating medications not approved for CLL. Specifically, lenalidomide should not be used for patients with CLL outside of a clinical trial. While initial studies reported moderate activity for lenalidomide [141-144], some studies have been terminated due to toxicity concerns and excess deaths [145].

We also do not use the anti-CD52 monoclonal antibody alemtuzumab for patients with CLL. While partial responses may be seen in approximately one-third of patients, use is limited by toxicities that include infusion-related side effects, myelosuppression, and infections [146-158].

Chimeric antigen receptor T cells — Chimeric antigen receptor (CAR)-T cell therapy is an investigational therapy in CLL/SLL that genetically modifies the patient's own T lymphocytes to target leukemia cells. It is used for the treatment of other hematologic malignancies.

Small, prospective trials have evaluated CD19-directed CAR-T cells in CLL/SLL [159-167]. In one early report, all three patients with relapsed or refractory CLL treated with CAR-T cells demonstrated a tumor response, which persisted in one for at least 10 months [159]. While subsequent reports demonstrate lower response rates in a broader CLL/SLL population [167,168], some patients with persistent CAR-T cells have experienced remissions lasting over a decade [169]. In addition, CAR-T cells appear to be effective in patients with CLL/SLL refractory to both Bruton tyrosine kinase (BTK) inhibition and BCL2 inhibition (venetoclax) [168], a population with few therapeutic alternatives.

CAR-T cells are genetically modified ex vivo, expanded in a production facility, and then infused back into the patient as therapy. Prior to reinfusion, patients receive a lymphodepleting chemotherapy preparative/conditioning regimen (ie, fludarabine plus cyclophosphamide). Trials have allowed for additional "bridging" therapy for disease control during the manufacturing process.

Toxicity in CLL/SLL appears to be greater than in diffuse large B cell lymphoma. CAR-T cells can have serious and potentially fatal complications, including neurologic events and cytokine release syndrome (CRS), which is a severe systemic response (eg, high fever, flu-like symptoms, hypotension, mental status changes) to the activation and proliferation of CAR-T cells. This is described in more detail separately. (See "Cytokine release syndrome (CRS)" and "Immune effector cell-associated neurotoxicity syndrome (ICANS)" and "Diffuse large B cell lymphoma (DLBCL): Second or later relapse or patients who are medically unfit", section on 'Chimeric antigen receptor T cell therapy'.)

A multicenter, open-label, single-arm phase 1/2 trial (TRANSCEND CLL 004) evaluated lisocabtagene maraleucel (liso-cel) in patients with relapsed/refractory CLL/SLL after at least two prior lines of therapy (median five), including a BTK inhibitor [168]. Key findings included:

Leukapheresis was performed in 137 patients, and a safety analysis included the 117 patients that received liso-cel. The median time from leukapheresis to infusion was 36 days. Although manufacturing was successful in 96 percent and bridging therapy was allowed, several patients experienced disease progression before infusion, leading to ineligibility or death, resulting in a drop-out rate of 15 percent. Safety and efficacy analyses were performed on an "as treated" population rather than "intention-to-treat".

There were five deaths within 90 days of liso-cel infusion, one of which was attributed to liso-cel (macrophage activation syndrome).

Most patients had CRS with 67 percent requiring tocilizumab and/or corticosteroids. Grade 3 CRS occurred in 9 percent, and there were no grade 4 or 5 CRS.

Neurologic events occurred in 45 percent of patients, and 33 percent required tocilizumab and/or corticosteroids for a neurologic event. Grade 3 or 4 neurologic events occurred in 19 percent, and there were no grade 5 neurologic events.

Other notable adverse events included prolonged cytopenias (54 percent), grade 3 or greater infection (17 percent), hypogammaglobulinemia (15 percent), and tumor lysis syndrome (11 percent).

The primary efficacy analysis focused on the 49 patients refractory to both BTK inhibition and BCL2 inhibition who received a target dose of 100 x 106 CAR-positive viable T-cells (target dose 2). In this population, 9 of 49 patients (18 percent) achieved a complete response (CR) or remission (including with incomplete marrow recovery), and the median duration of response was 35.5 months (not reached in those with a CR or remission). An 18 percent CR or remission rate was also reported for the broader population of 96 patients treated with liso-cel and evaluable for efficacy.

These results suggest that CAR-T cell therapy is modestly effective in this difficult to treat population with CLL/SLL refractory to both BTK inhibition and BCL2 inhibition. Further follow-up is needed to assess response duration in those with remission.

Another cohort included 32 evaluable patients with relapsed or refractory CLL/SLL enrolled on a clinical trial of CD19-directed CAR-T cells [167]. Patients had received a median of 3.5 prior therapies; nine had received a BTK inhibitor, and one had received venetoclax. A majority (72 percent) had unmutated IGHV, and nine patients (28 percent) had mutated TP53 or 17p deletion. Key findings included:

Grade 3 or higher CRS developed in nine patients, and grade 3 or higher neurotoxicity developed in three patients. There were no treatment-related deaths.

The overall response rate and CR rate were 44 and 28 percent, respectively. After a median follow-up of 32 months, the median overall survival was 64 months. Median progression-free survival was longer in those attaining CR (40 versus 1 month). Outcomes did not differ by patient age, number of prior therapies, or genetic findings.

Further follow-up of these patients and larger trials are needed to better define the efficacy of this approach and how it might be incorporated into patient care.

The logistical and clinical complications of autologous CAR-T cells have led to efforts to create off-the-shelf CAR products. As an example, CAR-NK cell therapy derived from umbilical cord blood (UCB) is an investigational allogeneic product that does not require full histocompatibility (HLA) matching. In a phase 1/2 study of UCB-derived CAR-NK cells, infusion was not associated with cytokine release syndrome, neurotoxicity, or graft-versus-host disease [170]. Three of five patients with CLL/SLL achieved a CR.

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Chronic lymphocytic leukemia/small lymphocytic lymphoma".)

INFORMATION FOR PATIENTS — UpToDate offers two types of patient education materials, "The Basics" and "Beyond the Basics." The Basics patient education pieces are written in plain language, at the 5th to 6th grade reading level, and they answer the four or five key questions a patient might have about a given condition. These articles are best for patients who want a general overview and who prefer short, easy-to-read materials. Beyond the Basics patient education pieces are longer, more sophisticated, and more detailed. These articles are written at the 10th to 12th grade reading level and are best for patients who want in-depth information and are comfortable with some medical jargon.

Here are the patient education articles that are relevant to this topic. We encourage you to print or e-mail these topics to your patients. (You can also locate patient education articles on a variety of subjects by searching on "patient education" and the keyword(s) of interest.)

Basics topics (see "Patient education: Chronic lymphocytic leukemia (CLL) (The Basics)" and "Patient education: Tumor lysis syndrome (The Basics)")

Beyond the Basics topics (see "Patient education: Chronic lymphocytic leukemia (CLL) in adults (Beyond the Basics)")

SUMMARY AND RECOMMENDATIONS

Evaluation prior to change in therapy – Most patients with chronic lymphocytic leukemia (CLL) will have a response to initial therapy. However, conventional therapy is not curative, and most patients experience relapse. In addition, many patients will require a change in therapy due to intolerance.

Drug intolerance leads to treatment discontinuation in a sizeable minority of patients on continuous therapy with a Bruton tyrosine kinase (BTK) inhibitor (eg, ibrutinib, acalabrutinib, zanubrutinib). Before switching therapy due to BTK intolerance, we consider drug holidays, dose adjustments, and symptom-directed management in an effort to maximize drug benefit. (See 'Patients with BTK inhibitor intolerance' above.)

Progressive disease may be suspected due to a rising lymphocyte count, enlargement of lymph nodes and/or spleen, and/or worsening cytopenias (table 1). Before changing therapy for progressive CLL, the diagnosis must be reconfirmed. Most cases of relapsed CLL can be confirmed by a complete blood count with differential, flow cytometry of the peripheral blood to determine the immunophenotype of circulating lymphocytes, and examination of the peripheral smear. (See 'Confirming progression' above.)

Indications for treatment – Patients with recurrent/progressive CLL do not necessarily require immediate treatment but should be followed closely for the development of symptomatic disease (ie, "active disease") (table 2), which necessitates intervention. For patients on continuous therapy, such as a BTK inhibitor, we continue treatment as long as there appears to be a clinical benefit and adjust monitoring parameters based on disease tempo. (See 'Indications for treatment' above.)

Choice of therapy – Our choice of subsequent therapy for patients with progressive disease or intolerance to initial treatment is individualized and takes into account prior therapy, response, and reason for discontinuation; patient and tumor characteristics; patient preference; and goals of therapy (algorithm 1 and table 3). Patients with CLL experience serial relapses and many will be treated with all available agents at some point during their disease course (table 4). A preferred order for their use has not been established. (See 'Choice of therapy' above.)

Prior BTK inhibitor – For patients treated initially with a BTK inhibitor (ie, ibrutinib, acalabrutinib, zanubrutinib), our preferred subsequent therapy depends on the reason for discontinuation (see 'Prior BTK inhibitor' above):

For patients with CLL progressing on a BTK inhibitor and requiring treatment (ie, with "active disease"), we suggest venetoclax-based therapy rather than an alternative BTK inhibitor or a phosphoinositide 3'-kinase (PI3K) inhibitor (Grade 2C). Venetoclax is highly active and works through a different mechanism of action (BCL2 inhibition). (See 'BCL2 inhibitors: Venetoclax' above.)

After attaining a response, younger patients with a matched donor (related or unrelated) should be evaluated for allogeneic hematopoietic cell transplantation (HCT). (See 'Hematopoietic cell transplantation' above.)

For patients who discontinue a BTK inhibitor due to intolerance, options include treatment with an alternative BTK inhibitor or venetoclax-based therapy. Mutation analysis can help guide the choice; those with mutations in BTK and PLCg2 are unlikely to respond to an alternative covalent BTK inhibitor and are treated with venetoclax-based therapy; in contrast, the noncovalent BTK inhibitor pirtobrutinib has demonstrated activity in this setting and may be an option for multiply relapsed disease. For those without a mutation in BTK or PLCg2, a choice is made based on comorbidities and patient preference. (See 'Consider treatment burdens and patient comorbidities' above.)

Prior venetoclax plus obinutuzumab – For patients treated initially with venetoclax plus obinutuzumab, our choice of subsequent therapy depends on the timing of relapse (see 'Prior venetoclax plus obinutuzumab' above):

For patients progressing within three to five years after initial treatment with venetoclax plus obinutuzumab, we suggest a BTK inhibitor rather than retreatment with venetoclax-based therapy or a PI3K inhibitor (Grade 2C). Where available, we suggest acalabrutinib or zanubrutinib rather than ibrutinib (Grade 2B). When compared with ibrutinib, single-agent acalabrutinib has similar efficacy and an overall better tolerability profile (eg, fewer class-associated adverse effects including cardiac toxicity, atrial fibrillation, hypertension, arthralgia, and bleeding). The improvement in tolerability is more narrow for zanubrutinib (ie, less cardiac toxicity, in particular less atrial fibrillation, but similar rates of other toxicities); however, zanubrutinib is more effective than ibrutinib with deeper responses and improved progression-free survival. (See 'Bruton tyrosine kinase inhibitors' above.)

After attaining a response, younger patients with a matched donor (related or unrelated) should be evaluated for allogeneic HCT. (See 'Hematopoietic cell transplantation' above.)

For patients with progression more than three to five years following venetoclax plus obinutuzumab, options include retreatment with venetoclax plus an anti-CD20 monoclonal antibody (rituximab or obinutuzumab) or single-agent therapy with a BTK inhibitor. Mutation analysis can help guide the choice; those with mutations in BCL2 are unlikely to achieve a durable response following venetoclax-based therapy and are treated with a BTK inhibitor. For those without BCL2 mutation, a choice is made based on comorbidities and patient preference. (See 'Consider treatment burdens and patient comorbidities' above.)

Prior chemoimmunotherapy – Most patients with progression after initial chemoimmunotherapy should be treated with a BTK inhibitor or venetoclax-based therapy; a choice between these is made based on comorbidities and patient preference.

Other therapies – We reserve PI3K inhibitors for patients with relapsed/refractory CLL with prior exposure to BTK inhibitors and venetoclax. (See 'PI3K inhibitors' above.)

For those seeking palliation of symptoms, anti-CD20 monoclonal antibodies and corticosteroids may offer short-lived partial responses with only moderate toxicity. Splenectomy, obtained either surgically or via splenic irradiation, may be useful in patients with splenomegaly and profound cytopenias unresponsive to chemotherapy. Radiation therapy is an option for patients with bulky disease compromising critical structures. (See 'Palliation of symptoms' above.)

ACKNOWLEDGMENT — The UpToDate editorial staff acknowledges Michael J Keating, MD, who contributed to earlier versions of this topic review.

  1. Mato AR, Nabhan C, Thompson MC, et al. Toxicities and outcomes of 616 ibrutinib-treated patients in the United States: a real-world analysis. Haematologica 2018; 103:874.
  2. Gribben JG, Bosch F, Cymbalista F, et al. Optimising outcomes for patients with chronic lymphocytic leukaemia on ibrutinib therapy: European recommendations for clinical practice. Br J Haematol 2018; 180:666.
  3. Stephens DM, Byrd JC. How I manage ibrutinib intolerance and complications in patients with chronic lymphocytic leukemia. Blood 2019; 133:1298.
  4. Barnea Slonim L, Ma S, Behdad A, Chen Q. Pseudo-Richter transformation of chronic lymphocytic leukaemia/small lymphocytic lymphoma following ibrutinib interruption: a diagnostic pitfall. Br J Haematol 2020; 191:e22.
  5. Ahn IE, Farber CM, Davids MS, et al. Early progression of disease as a predictor of survival in chronic lymphocytic leukemia. Blood Adv 2017; 1:2433.
  6. Burger JA, Barr PM, Robak T, et al. 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; 34:787.
  7. Sharman JP, Egyed M, Jurczak W, et al. Efficacy and safety in a 4-year follow-up of the ELEVATE-TN study comparing acalabrutinib with or without obinutuzumab versus obinutuzumab plus chlorambucil in treatment-naïve chronic lymphocytic leukemia. Leukemia 2022; 36:1171.
  8. Tam CS, Brown JR, Kahl BS, et al. Zanubrutinib versus bendamustine and rituximab in untreated chronic lymphocytic leukaemia and small lymphocytic lymphoma (SEQUOIA): a randomised, controlled, phase 3 trial. Lancet Oncol 2022; 23:1031.
  9. Al-Sawaf O, Zhang C, Lu T, et al. Minimal Residual Disease Dynamics after Venetoclax-Obinutuzumab Treatment: Extended Off-Treatment Follow-up From the Randomized CLL14 Study. J Clin Oncol 2021; 39:4049.
  10. Kater AP, Owen C, Moreno C, et al. Fixed-Duration Ibrutinib-Venetoclax in Patients with Chronic Lymphocytic Leukemia and Comorbidities. NEJM Evid 2022; 1.
  11. Woyach JA, Ruppert AS, Guinn D, et al. BTK(C481S)-Mediated Resistance to Ibrutinib in Chronic Lymphocytic Leukemia. J Clin Oncol 2017; 35:1437.
  12. Woyach JA, Furman RR, Liu TM, et al. Resistance mechanisms for the Bruton's tyrosine kinase inhibitor ibrutinib. N Engl J Med 2014; 370:2286.
  13. Furman RR, Cheng S, Lu P, et al. Ibrutinib resistance in chronic lymphocytic leukemia. N Engl J Med 2014; 370:2352.
  14. Ahn IE, Underbayev C, Albitar A, et al. Clonal evolution leading to ibrutinib resistance in chronic lymphocytic leukemia. Blood 2017; 129:1469.
  15. Tausch E, Close W, Dolnik A, et al. Venetoclax resistance and acquired BCL2 mutations in chronic lymphocytic leukemia. Haematologica 2019; 104:e434.
  16. Blombery P, Anderson MA, Gong JN, et al. Acquisition of the Recurrent Gly101Val Mutation in BCL2 Confers Resistance to Venetoclax in Patients with Progressive Chronic Lymphocytic Leukemia. Cancer Discov 2019; 9:342.
  17. Lucas F, Larkin K, Gregory CT, et al. Novel BCL2 mutations in venetoclax-resistant, ibrutinib-resistant CLL patients with BTK/PLCG2 mutations. Blood 2020; 135:2192.
  18. Blombery P, Thompson ER, Nguyen T, et al. Multiple BCL2 mutations cooccurring with Gly101Val emerge in chronic lymphocytic leukemia progression on venetoclax. Blood 2020; 135:773.
  19. Mato AR, Roeker LE, Jacobs R, et al. Assessment of the Efficacy of Therapies Following Venetoclax Discontinuation in CLL Reveals BTK Inhibition as an Effective Strategy. Clin Cancer Res 2020; 26:3589.
  20. Anderson MA, Tam C, Lew TE, et al. Clinicopathological features and outcomes of progression of CLL on the BCL2 inhibitor venetoclax. Blood 2017; 129:3362.
  21. Lin VS, Lew TE, Handunnetti SM, et al. BTK inhibitor therapy is effective in patients with CLL resistant to venetoclax. Blood 2020; 135:2266.
  22. Seymour JF, Kipps TJ, Eichhorst BF, et al. Enduring undetectable MRD and updated outcomes in relapsed/refractory CLL after fixed-duration venetoclax-rituximab. Blood 2022; 140:839.
  23. Byrd JC, Hillmen P, Ghia P, et al. Acalabrutinib Versus Ibrutinib in Previously Treated Chronic Lymphocytic Leukemia: Results of the First Randomized Phase III Trial. J Clin Oncol 2021; 39:3441.
  24. Brown JR, Eichhorst B, Hillmen P, et al. Zanubrutinib or Ibrutinib in Relapsed or Refractory Chronic Lymphocytic Leukemia. N Engl J Med 2023; 388:319.
  25. Freise KJ, Shebley M, Salem AH. Quantitative Prediction of the Effect of CYP3A Inhibitors and Inducers on Venetoclax Pharmacokinetics Using a Physiologically Based Pharmacokinetic Model. J Clin Pharmacol 2017; 57:796.
  26. Acalabrutinib. United States prescribing information. Revised November 2019. US Food and Drug Administration. https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/210259s006s007lbl.pdf (Accessed on November 22, 2019).
  27. Ghia P, Pluta A, Wach M, et al. ASCEND: Phase III, Randomized Trial of Acalabrutinib Versus Idelalisib Plus Rituximab or Bendamustine Plus Rituximab in Relapsed or Refractory Chronic Lymphocytic Leukemia. J Clin Oncol 2020; 38:2849.
  28. Ghia P, Pluta A, Wach M, et al. Acalabrutinib Versus Investigator's Choice in Relapsed/Refractory Chronic Lymphocytic Leukemia: Final ASCEND Trial Results. Hemasphere 2022; 6:e801.
  29. Seymour JF, Byrd JC, Ghia P, et al. Detailed safety profile of acalabrutinib vs ibrutinib in previously treated chronic lymphocytic leukemia in the ELEVATE-RR trial. Blood 2023; 142:687.
  30. Sharman JP, Egyed M, Jurczak W, et al. Acalabrutinib with or without obinutuzumab versus chlorambucil and obinutuzmab for treatment-naive chronic lymphocytic leukaemia (ELEVATE TN): a randomised, controlled, phase 3 trial. Lancet 2020; 395:1278.
  31. Byrd JC, Wierda WG, Schuh A, et al. Acalabrutinib monotherapy in patients with relapsed/refractory chronic lymphocytic leukemia: updated phase 2 results. Blood 2020; 135:1204.
  32. Sharman J, Egyed M, Jurczak W, et al. Acalabrutinib +/- obinutuzumab vs obinutuzumab + chlorambucil in treatment-naive chronic lymphocytic leukemia: ELEVATE-TN 4-year follow-up (abstract S148). Hemasphere 2021.
  33. Hillmen P, Eichhorst B, Brown JR, et al. Zanubrutinib Versus Ibrutinib in Relapsed/Refractory Chronic Lymphocytic Leukemia and Small Lymphocytic Lymphoma: Interim Analysis of a Randomized Phase III Trial. J Clin Oncol 2023; 41:1035.
  34. Advani RH, Buggy JJ, Sharman JP, et al. Bruton tyrosine kinase inhibitor ibrutinib (PCI-32765) has significant activity in patients with relapsed/refractory B-cell malignancies. J Clin Oncol 2013; 31:88.
  35. O'Brien S, Furman RR, Coutre SE, et al. Ibrutinib as initial therapy for elderly patients with chronic lymphocytic leukaemia or small lymphocytic lymphoma: an open-label, multicentre, phase 1b/2 trial. Lancet Oncol 2014; 15:48.
  36. Byrd JC, Furman RR, Coutre SE, et al. Three-year follow-up of treatment-naïve and previously treated patients with CLL and SLL receiving single-agent ibrutinib. Blood 2015; 125:2497.
  37. Byrd JC, Furman RR, Coutre SE, et al. Targeting BTK with ibrutinib in relapsed chronic lymphocytic leukemia. N Engl J Med 2013; 369:32.
  38. O'Brien S, Furman RR, Coutre S, et al. Single-agent ibrutinib in treatment-naïve and relapsed/refractory chronic lymphocytic leukemia: a 5-year experience. Blood 2018; 131:1910.
  39. Byrd JC, Furman RR, Coutre SE, et al. Ibrutinib Treatment for First-Line and Relapsed/Refractory Chronic Lymphocytic Leukemia: Final Analysis of the Pivotal Phase Ib/II PCYC-1102 Study. Clin Cancer Res 2020; 26:3918.
  40. Burger JA, Keating MJ, Wierda WG, et al. Safety and activity of ibrutinib plus rituximab for patients with high-risk chronic lymphocytic leukaemia: a single-arm, phase 2 study. Lancet Oncol 2014; 15:1090.
  41. Farooqui MZ, Valdez J, Martyr S, et al. Ibrutinib for previously untreated and relapsed or refractory chronic lymphocytic leukaemia with TP53 aberrations: a phase 2, single-arm trial. Lancet Oncol 2015; 16:169.
  42. Jones J, Mato A, Coutre S, et al. Evaluation of 230 patients with relapsed/refractory deletion 17p chronic lymphocytic leukaemia treated with ibrutinib from 3 clinical trials. Br J Haematol 2018; 182:504.
  43. Byrd JC, Brown JR, O'Brien S, et al. Ibrutinib versus ofatumumab in previously treated chronic lymphoid leukemia. N Engl J Med 2014; 371:213.
  44. Brown JR, Hillmen P, O'Brien S, et al. Extended follow-up and impact of high-risk prognostic factors from the phase 3 RESONATE study in patients with previously treated CLL/SLL. Leukemia 2018; 32:83.
  45. Barrientos JC, O'Brien S, Brown JR, et al. Improvement in Parameters of Hematologic and Immunologic Function and Patient Well-being in the Phase III RESONATE Study of Ibrutinib Versus Ofatumumab in Patients With Previously Treated Chronic Lymphocytic Leukemia/Small Lymphocytic Lymphoma. Clin Lymphoma Myeloma Leuk 2018; 18:803.
  46. Byrd JC, Hillmen P, O'Brien S, et al. Long-term follow-up of the RESONATE phase 3 trial of ibrutinib vs ofatumumab. Blood 2019; 133:2031.
  47. Munir T, Brown JR, O'Brien S, et al. Final analysis from RESONATE: Up to six years of follow-up on ibrutinib in patients with previously treated chronic lymphocytic leukemia or small lymphocytic lymphoma. Am J Hematol 2019; 94:1353.
  48. O'Brien S, Jones JA, Coutre SE, et al. Ibrutinib for patients with relapsed or refractory chronic lymphocytic leukaemia with 17p deletion (RESONATE-17): a phase 2, open-label, multicentre study. Lancet Oncol 2016; 17:1409.
  49. Burger JA, Sivina M, Jain N, et al. Randomized trial of ibrutinib vs ibrutinib plus rituximab in patients with chronic lymphocytic leukemia. Blood 2019; 133:1011.
  50. Woyach JA, Ruppert AS, Heerema NA, et al. Ibrutinib Regimens versus Chemoimmunotherapy in Older Patients with Untreated CLL. N Engl J Med 2018; 379:2517.
  51. Brown JR, Barrientos JC, Barr PM, et al. The Bruton tyrosine kinase inhibitor ibrutinib with chemoimmunotherapy in patients with chronic lymphocytic leukemia. Blood 2015; 125:2915.
  52. Chanan-Khan A, Cramer P, Demirkan F, et al. Ibrutinib combined with bendamustine and rituximab compared with placebo, bendamustine, and rituximab for previously treated chronic lymphocytic leukaemia or small lymphocytic lymphoma (HELIOS): a randomised, double-blind, phase 3 study. Lancet Oncol 2016; 17:200.
  53. Fraser G, Cramer P, Demirkan F, et al. Updated results from the phase 3 HELIOS study of ibrutinib, bendamustine, and rituximab in relapsed chronic lymphocytic leukemia/small lymphocytic lymphoma. Leukemia 2019; 33:969.
  54. Fraser GAM, Chanan-Khan A, Demirkan F, et al. Final 5-year findings from the phase 3 HELIOS study of ibrutinib plus bendamustine and rituximab in patients with relapsed/refractory chronic lymphocytic leukemia/small lymphocytic lymphoma. Leuk Lymphoma 2020; 61:3188.
  55. Pleyer C, Tian X, Rampertaap S, et al. A phase II study of ibrutinib and short-course fludarabine in previously untreated patients with chronic lymphocytic leukemia. Am J Hematol 2020; 95:E310.
  56. Wang E, Mi X, Thompson MC, et al. Mechanisms of Resistance to Noncovalent Bruton's Tyrosine Kinase Inhibitors. N Engl J Med 2022; 386:735.
  57. Mato AR, Shah NN, Jurczak W, et al. Pirtobrutinib in relapsed or refractory B-cell malignancies (BRUIN): a phase 1/2 study. Lancet 2021; 397:892.
  58. Mato AR, Woyach JA, Brown JR, et al. Pirtobrutinib after a Covalent BTK Inhibitor in Chronic Lymphocytic Leukemia. N Engl J Med 2023; 389:33.
  59. Venetoclax tablets. United States prescribing information. Revised May 2019. US Food and Drug Administration. https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/208573s013lbl.pdf (Accessed on May 16, 2019).
  60. https://www.ema.europa.eu/en/documents/overview/venclyxto-epar-medicine-overview_en.pdf (Accessed on May 16, 2019).
  61. Venetoclax tablets. United States prescribing information. Revised June 2018. US Food and Drug Administration. https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/208573s004s005lbl.pdf?et_cid=40352090&et_rid=1004920403&linkid=https%3a%2f%2fwww.accessdata.fda.gov%2fdrugsatfda_docs%2flabel%2f2018%2f208573s004s005lbl.pdf&et_cid=40352378&et_rid=931330620&linkid=https%3a%2f%2fwww.accessdata.fda.gov%2fdrugsa (Accessed on June 08, 2018).
  62. Koenig KL, Huang Y, Dotson EK, et al. Safety of venetoclax rapid dose escalation in CLL patients previously treated with B-cell receptor signaling antagonists. Blood Adv 2020; 4:4860.
  63. Roberts AW, Davids MS, Pagel JM, et al. Targeting BCL2 with Venetoclax in Relapsed Chronic Lymphocytic Leukemia. N Engl J Med 2016; 374:311.
  64. Stilgenbauer S, Eichhorst B, Schetelig J, et al. Venetoclax in relapsed or refractory chronic lymphocytic leukaemia with 17p deletion: a multicentre, open-label, phase 2 study. Lancet Oncol 2016; 17:768.
  65. Jones JA, Mato AR, Wierda WG, et al. Venetoclax for chronic lymphocytic leukaemia progressing after ibrutinib: an interim analysis of a multicentre, open-label, phase 2 trial. Lancet Oncol 2018; 19:65.
  66. Stilgenbauer S, Eichhorst B, Schetelig J, et al. Venetoclax for Patients With Chronic Lymphocytic Leukemia With 17p Deletion: Results From the Full Population of a Phase II Pivotal Trial. J Clin Oncol 2018; 36:1973.
  67. Seymour JF, Ma S, Brander DM, et al. Venetoclax plus rituximab in relapsed or refractory chronic lymphocytic leukaemia: a phase 1b study. Lancet Oncol 2017; 18:230.
  68. Seymour JF, Kipps TJ, Eichhorst B, et al. Venetoclax-Rituximab in Relapsed or Refractory Chronic Lymphocytic Leukemia. N Engl J Med 2018; 378:1107.
  69. Flinn IW, Gribben JG, Dyer MJS, et al. Phase 1b study of venetoclax-obinutuzumab in previously untreated and relapsed/refractory chronic lymphocytic leukemia. Blood 2019; 133:2765.
  70. Mato AR, Roeker LE, Eyre TA, et al. A retrospective comparison of venetoclax alone or in combination with an anti-CD20 monoclonal antibody in R/R CLL. Blood Adv 2019; 3:1568.
  71. Blombery P, Lew TE, Dengler MA, et al. Clonal hematopoiesis, myeloid disorders and BAX-mutated myelopoiesis in patients receiving venetoclax for CLL. Blood 2022; 139:1198.
  72. Coutre S, Choi M, Furman RR, et al. Venetoclax for patients with chronic lymphocytic leukemia who progressed during or after idelalisib therapy. Blood 2018; 131:1704.
  73. Kater AP, Wu JQ, Kipps T, et al. Venetoclax Plus Rituximab in Relapsed Chronic Lymphocytic Leukemia: 4-Year Results and Evaluation of Impact of Genomic Complexity and Gene Mutations From the MURANO Phase III Study. J Clin Oncol 2020; 38:4042.
  74. Kater AP, Seymour JF, Hillmen P, et al. Fixed Duration of Venetoclax-Rituximab in Relapsed/Refractory Chronic Lymphocytic Leukemia Eradicates Minimal Residual Disease and Prolongs Survival: Post-Treatment Follow-Up of the MURANO Phase III Study. J Clin Oncol 2019; 37:269.
  75. Ma S, Seymour JF, Brander DM, et al. Efficacy of venetoclax plus rituximab for relapsed CLL: 5-year follow-up of continuous or limited- duration therapy. Blood 2021; 138:836.
  76. Fischer K, Al-Sawaf O, Bahlo J, et al. Venetoclax and Obinutuzumab in Patients with CLL and Coexisting Conditions. N Engl J Med 2019; 380:2225.
  77. Hoellenriegel J, Meadows SA, Sivina M, et al. The phosphoinositide 3'-kinase delta inhibitor, CAL-101, inhibits B-cell receptor signaling and chemokine networks in chronic lymphocytic leukemia. Blood 2011; 118:3603.
  78. O'Brien SM, Lamanna N, Kipps TJ, et al. A phase 2 study of idelalisib plus rituximab in treatment-naïve older patients with chronic lymphocytic leukemia. Blood 2015; 126:2686.
  79. Brown JR, Byrd JC, Coutre SE, et al. Idelalisib, an inhibitor of phosphatidylinositol 3-kinase p110δ, for relapsed/refractory chronic lymphocytic leukemia. Blood 2014; 123:3390.
  80. Idelalisib. United States prescribing information. Revised July 2014. US Food and Drug Administration. http://www.accessdata.fda.gov/drugsatfda_docs/label/2014/206545lbl.pdf?et_cid=34165715&et_rid=789337055&linkid=http%3a%2f%2fwww.accessdata.fda.gov%2fdrugsatfda_docs%2flabel%2f2014%2f206545lbl.pdf (Accessed on July 23, 2014).
  81. Furman RR, Sharman JP, Coutre SE, et al. Idelalisib and rituximab in relapsed chronic lymphocytic leukemia. N Engl J Med 2014; 370:997.
  82. Lampson BL, Kasar SN, Matos TR, et al. Idelalisib given front-line for treatment of chronic lymphocytic leukemia causes frequent immune-mediated hepatotoxicity. Blood 2016; 128:195.
  83. Brown JR, Zelenetz A, Furman R, et al. Risk factors for grade 3/4 transaminase elevation in patients with chronic lymphocytic leukemia treated with idelalisib. Leukemia 2020; 34:3404.
  84. Bird ST, Tian F, Flowers N, et al. Idelalisib for Treatment of Relapsed Follicular Lymphoma and Chronic Lymphocytic Leukemia: A Comparison of Treatment Outcomes in Clinical Trial Participants vs Medicare Beneficiaries. JAMA Oncol 2020; 6:248.
  85. Sharman JP, Coutre SE, Furman RR, et al. Final Results of a Randomized, Phase III Study of Rituximab With or Without Idelalisib Followed by Open-Label Idelalisib in Patients With Relapsed Chronic Lymphocytic Leukemia. J Clin Oncol 2019; 37:1391.
  86. Jones JA, Robak T, Brown JR, et al. Efficacy and safety of idelalisib in combination with ofatumumab for previously treated chronic lymphocytic leukaemia: an open-label, randomised phase 3 trial. Lancet Haematol 2017; 4:e114.
  87. Zelenetz AD, Barrientos JC, Brown JR, et al. Idelalisib or placebo in combination with bendamustine and rituximab in patients with relapsed or refractory chronic lymphocytic leukaemia: interim results from a phase 3, randomised, double-blind, placebo-controlled trial. Lancet Oncol 2017; 18:297.
  88. Duvelisib. United States prescribing information. Revised September 2018. US Food and Drug Administration. https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/211155s000lbl.pdf (Accessed on September 26, 2018).
  89. O'Brien S, Patel M, Kahl BS, et al. Duvelisib, an oral dual PI3K-δ,γ inhibitor, shows clinical and pharmacodynamic activity in chronic lymphocytic leukemia and small lymphocytic lymphoma in a phase 1 study. Am J Hematol 2018; 93:1318.
  90. Flinn IW, O'Brien S, Kahl B, et al. Duvelisib, a novel oral dual inhibitor of PI3K-δ,γ, is clinically active in advanced hematologic malignancies. Blood 2018; 131:877.
  91. Flinn IW, Hillmen P, Montillo M, et al. The phase 3 DUO trial: duvelisib vs ofatumumab in relapsed and refractory CLL/SLL. Blood 2018; 132:2446.
  92. Flinn IW, Miller CB, Ardeshna KM, et al. DYNAMO: A Phase II Study of Duvelisib (IPI-145) in Patients With Refractory Indolent Non-Hodgkin Lymphoma. J Clin Oncol 2019; 37:912.
  93. FDA warns about possible increased risk of death and serious side effects with cancer drug Copiktra (duvelisib). US Food and Drug Administration. Available at: https://www.fda.gov/media/159559/download (Accessed on July 07, 2022).
  94. https://www.fda.gov/media/157762/download (Accessed on April 25, 2022).
  95. Davids MS, Kuss BJ, Hillmen P, et al. Efficacy and Safety of Duvelisib Following Disease Progression on Ofatumumab in Patients with Relapsed/Refractory CLL or SLL in the DUO Crossover Extension Study. Clin Cancer Res 2020; 26:2096.
  96. Dreger P, Ghia P, Schetelig J, et al. High-risk chronic lymphocytic leukemia in the era of pathway inhibitors: integrating molecular and cellular therapies. Blood 2018; 132:892.
  97. Lew TE, Lin VS, Cliff ER, et al. Outcomes of patients with CLL sequentially resistant to both BCL2 and BTK inhibition. Blood Adv 2021; 5:4054.
  98. Kharfan-Dabaja MA, Pidala J, Kumar A, et al. Comparing efficacy of reduced-toxicity allogeneic hematopoietic cell transplantation with conventional chemo-(immuno) therapy in patients with relapsed or refractory CLL: a Markov decision analysis. Bone Marrow Transplant 2012; 47:1164.
  99. Herth I, Dietrich S, Benner A, et al. The impact of allogeneic stem cell transplantation on the natural course of poor-risk chronic lymphocytic leukemia as defined by the EBMT consensus criteria: a retrospective donor versus no donor comparison. Ann Oncol 2014; 25:200.
  100. Poon ML, Fox PS, Samuels BI, et al. Allogeneic stem cell transplant in patients with chronic lymphocytic leukemia with 17p deletion: consult-transplant versus consult- no-transplant analysis. Leuk Lymphoma 2015; 56:711.
  101. Xavier E, Cornillon J, Ruggeri A, et al. Outcomes of Cord Blood Transplantation Using Reduced-Intensity Conditioning for Chronic Lymphocytic Leukemia: A Study on Behalf of Eurocord and Cord Blood Committee of Cellular Therapy and Immunobiology Working Party, Chronic Malignancies Working Party of the European Society for Blood and Marrow Transplantation, and the Societé Française de Greffe de Moelle et Therapie Cellulaire. Biol Blood Marrow Transplant 2015; 21:1515.
  102. van Gorkom G, van Gelder M, Eikema DJ, et al. Outcomes of haploidentical stem cell transplantation for chronic lymphocytic leukemia: a retrospective study on behalf of the chronic malignancies working party of the EBMT. Bone Marrow Transplant 2018; 53:255.
  103. Dreger P, Döhner H, Ritgen M, et al. Allogeneic stem cell transplantation provides durable disease control in poor-risk chronic lymphocytic leukemia: long-term clinical and MRD results of the German CLL Study Group CLL3X trial. Blood 2010; 116:2438.
  104. Dreger P, Schnaiter A, Zenz T, et al. TP53, SF3B1, and NOTCH1 mutations and outcome of allotransplantation for chronic lymphocytic leukemia: six-year follow-up of the GCLLSG CLL3X trial. Blood 2013; 121:3284.
  105. Krämer I, Stilgenbauer S, Dietrich S, et al. Allogeneic hematopoietic cell transplantation for high-risk CLL: 10-year follow-up of the GCLLSG CLL3X trial. Blood 2017; 130:1477.
  106. Roeker LE, Dreger P, Brown JR, et al. Allogeneic stem cell transplantation for chronic lymphocytic leukemia in the era of novel agents. Blood Adv 2020; 4:3977.
  107. Kim HT, Shaughnessy CJ, Rai SC, et al. Allogeneic hematopoietic cell transplantation after prior targeted therapy for high-risk chronic lymphocytic leukemia. Blood Adv 2020; 4:4113.
  108. O'Brien SM, Kantarjian H, Thomas DA, et al. Rituximab dose-escalation trial in chronic lymphocytic leukemia. J Clin Oncol 2001; 19:2165.
  109. Huhn D, von Schilling C, Wilhelm M, et al. Rituximab therapy of patients with B-cell chronic lymphocytic leukemia. Blood 2001; 98:1326.
  110. Byrd JC, Murphy T, Howard RS, et al. Rituximab using a thrice weekly dosing schedule in B-cell chronic lymphocytic leukemia and small lymphocytic lymphoma demonstrates clinical activity and acceptable toxicity. J Clin Oncol 2001; 19:2153.
  111. Castro JE, Sandoval-Sus JD, Bole J, et al. Rituximab in combination with high-dose methylprednisolone for the treatment of fludarabine refractory high-risk chronic lymphocytic leukemia. Leukemia 2008; 22:2048.
  112. Cartron G, de Guibert S, Dilhuydy MS, et al. Obinutuzumab (GA101) in relapsed/refractory chronic lymphocytic leukemia: final data from the phase 1/2 GAUGUIN study. Blood 2014; 124:2196.
  113. Byrd JC, Flynn JM, Kipps TJ, et al. Randomized phase 2 study of obinutuzumab monotherapy in symptomatic, previously untreated chronic lymphocytic leukemia. Blood 2016; 127:79.
  114. Leblond V, Aktan M, Ferra Coll CM, et al. Safety of obinutuzumab alone or combined with chemotherapy for previously untreated or relapsed/refractory chronic lymphocytic leukemia in the phase IIIb GREEN study. Haematologica 2018; 103:1889.
  115. Coiffier B, Lepretre S, Pedersen LM, et al. Safety and efficacy of ofatumumab, a fully human monoclonal anti-CD20 antibody, in patients with relapsed or refractory B-cell chronic lymphocytic leukemia: a phase 1-2 study. Blood 2008; 111:1094.
  116. Coiffier B, Losic N, Rønn BB, et al. Pharmacokinetics and pharmacokinetic/pharmacodynamic associations of ofatumumab, a human monoclonal CD20 antibody, in patients with relapsed or refractory chronic lymphocytic leukaemia: a phase 1-2 study. Br J Haematol 2010; 150:58.
  117. Wierda WG, Kipps TJ, Mayer J, et al. Ofatumumab as single-agent CD20 immunotherapy in fludarabine-refractory chronic lymphocytic leukemia. J Clin Oncol 2010; 28:1749.
  118. Wierda WG, Padmanabhan S, Chan GW, et al. Ofatumumab is active in patients with fludarabine-refractory CLL irrespective of prior rituximab: results from the phase 2 international study. Blood 2011; 118:5126.
  119. van Oers MH, Kuliczkowski K, Smolej L, et al. Ofatumumab maintenance versus observation in relapsed chronic lymphocytic leukaemia (PROLONG): an open-label, multicentre, randomised phase 3 study. Lancet Oncol 2015; 16:1370.
  120. Doubek M, Brychtova Y, Panovska A, et al. Ofatumumab added to dexamethasone in patients with relapsed or refractory chronic lymphocytic leukemia: Results from a phase II study. Am J Hematol 2015; 90:417.
  121. Miklos U, Strugov V, Lewerin C, et al. Five-year survival follow-up of a phase III randomised trial comparing ofatumumab versus physicians' choice for bulky fludarabine-refractory chronic lymphocytic leukaemia: a short report. Br J Haematol 2020; 189:689.
  122. Ezdinli EZ, Stutzman L, Aungst CW, Firat D. Corticosteroid therapy for lymphomas and chronic lymphocytic leukemia. Cancer 1969; 23:900.
  123. Molica S. High-dose dexamethasone in refractory B-cell chronic lymphocytic leukemia patients. Am J Hematol 1994; 47:334.
  124. Friedenberg WR, Anderson J, Wolf BC, et al. Modified vincristine, doxorubicin, and dexamethasone regimen in the treatment of resistant or relapsed chronic lymphocytic leukemia. An Eastern Cooperative Oncology Group study. Cancer 1993; 71:2983.
  125. Thornton PD, Matutes E, Bosanquet AG, et al. High dose methylprednisolone can induce remissions in CLL patients with p53 abnormalities. Ann Hematol 2003; 82:759.
  126. Xu W, Miao KR, Hong M, et al. High-dose methylprednisolone can induce remissions in patients with fludarabine-refractory chronic lymphocytic leukaemia. Eur J Cancer 2010; 46:2145.
  127. Coad JE, Matutes E, Catovsky D. Splenectomy in lymphoproliferative disorders: a report on 70 cases and review of the literature. Leuk Lymphoma 1993; 10:245.
  128. Majumdar G, Singh AK. Role of splenectomy in chronic lymphocytic leukaemia with massive splenomegaly and cytopenia. Leuk Lymphoma 1992; 7:131.
  129. Neal TF Jr, Tefferi A, Witzig TE, et al. Splenectomy in advanced chronic lymphocytic leukemia: a single institution experience with 50 patients. Am J Med 1992; 93:435.
  130. Thiruvengadam R, Piedmonte M, Barcos M, et al. Splenectomy in advanced chronic lymphocytic leukemia. Leukemia 1990; 4:758.
  131. Pegourie B, Sotto JJ, Hollard D, et al. Splenectomy during chronic lymphocytic leukemia. Cancer 1987; 59:1626.
  132. Seymour JF, Cusack JD, Lerner SA, et al. Case/control study of the role of splenectomy in chronic lymphocytic leukemia. J Clin Oncol 1997; 15:52.
  133. Delpero JR, Gastaut JA, Letreut YP, et al. The value of splenectomy in chronic lymphocytic leukemia. Cancer 1987; 59:340.
  134. Ferrant A, Michaux JL, Sokal G. Splenectomy in advanced chronic lymphocytic leukemia. Cancer 1986; 58:2130.
  135. Keating MJ. Immunosuppression with purine analogues--the flip side of the gold coin. Ann Oncol 1993; 4:347.
  136. Rossier C, Schick U, Miralbell R, et al. Low-dose radiotherapy in indolent lymphoma. Int J Radiat Oncol Biol Phys 2011; 81:e1.
  137. Chan EK, Fung S, Gospodarowicz M, et al. Palliation by low-dose local radiation therapy for indolent non-Hodgkin lymphoma. Int J Radiat Oncol Biol Phys 2011; 81:e781.
  138. Jóhannsson J, Specht L, Mejer J, Jensen BA. Phase II study of palliative low-dose local radiotherapy in disseminated indolent non-Hodgkin's lymphoma and chronic lymphocytic leukemia. Int J Radiat Oncol Biol Phys 2002; 54:1466.
  139. Chisesi T, Capnist G, Dal Fior S. Splenic irradiation in chronic lymphocytic leukemia. Eur J Haematol 1991; 46:202.
  140. A Study of NX-2127 in Adults With Relapsed/​Refractory B-cell Malignancies. Clinicaltrials.gov. Available at: https://www.clinicaltrials.gov/study/NCT04830137?term=NCT04830137&rank=1 (Accessed on July 06, 2023).
  141. Ferrajoli A, Lee BN, Schlette EJ, et al. Lenalidomide induces complete and partial remissions in patients with relapsed and refractory chronic lymphocytic leukemia. Blood 2008; 111:5291.
  142. Chanan-Khan A, Miller KC, Musial L, et al. Clinical efficacy of lenalidomide in patients with relapsed or refractory chronic lymphocytic leukemia: results of a phase II study. J Clin Oncol 2006; 24:5343.
  143. Witzig TE, Wiernik PH, Moore T, et al. Lenalidomide oral monotherapy produces durable responses in relapsed or refractory indolent non-Hodgkin's Lymphoma. J Clin Oncol 2009; 27:5404.
  144. Badoux XC, Keating MJ, Wen S, et al. Phase II study of lenalidomide and rituximab as salvage therapy for patients with relapsed or refractory chronic lymphocytic leukemia. J Clin Oncol 2013; 31:584.
  145. http://newsroom.celgene.com/press-release/corporate/celgene-will-discontinue-phase-iii-origin-trial-previously-untreated-elderly (Accessed on July 19, 2013).
  146. Lozanski G, Heerema NA, Flinn IW, et al. Alemtuzumab is an effective therapy for chronic lymphocytic leukemia with p53 mutations and deletions. Blood 2004; 103:3278.
  147. Keating MJ, Flinn I, Jain V, et al. Therapeutic role of alemtuzumab (Campath-1H) in patients who have failed fludarabine: results of a large international study. Blood 2002; 99:3554.
  148. http://www.campath.com/index.html (Accessed on September 27, 2012).
  149. Rai KR, Freter CE, Mercier RJ, et al. Alemtuzumab in previously treated chronic lymphocytic leukemia patients who also had received fludarabine. J Clin Oncol 2002; 20:3891.
  150. O'Brien SM, Kantarjian HM, Thomas DA, et al. Alemtuzumab as treatment for residual disease after chemotherapy in patients with chronic lymphocytic leukemia. Cancer 2003; 98:2657.
  151. Ravandi F, O'brien S. Alemtuzumab in CLL and other lymphoid neoplasms. Cancer Invest 2006; 24:718.
  152. Osterborg A, Dyer MJ, Bunjes D, et al. Phase II multicenter study of human CD52 antibody in previously treated chronic lymphocytic leukemia. European Study Group of CAMPATH-1H Treatment in Chronic Lymphocytic Leukemia. J Clin Oncol 1997; 15:1567.
  153. Karlsson C, Lundin J, Kimby E, et al. Phase II study of subcutaneous alemtuzumab without dose escalation in patients with advanced-stage, relapsed chronic lymphocytic leukaemia. Br J Haematol 2009; 144:78.
  154. Stilgenbauer S, Zenz T, Winkler D, et al. Subcutaneous alemtuzumab in fludarabine-refractory chronic lymphocytic leukemia: clinical results and prognostic marker analyses from the CLL2H study of the German Chronic Lymphocytic Leukemia Study Group. J Clin Oncol 2009; 27:3994.
  155. Fraser G, Smith CA, Imrie K, et al. Alemtuzumab in chronic lymphocytic leukemia. Curr Oncol 2007; 14:96.
  156. Moreton P, Kennedy B, Lucas G, et al. Eradication of minimal residual disease in B-cell chronic lymphocytic leukemia after alemtuzumab therapy is associated with prolonged survival. J Clin Oncol 2005; 23:2971.
  157. Montillo M, Schinkoethe T, Elter T. Eradication of minimal residual disease with alemtuzumab in B-cell chronic lymphocytic leukemia (B-CLL) patients: the need for a standard method of detection and the potential impact of bone marrow clearance on disease outcome. Cancer Invest 2005; 23:488.
  158. Montillo M, Tedeschi A, Miqueleiz S, et al. Alemtuzumab as consolidation after a response to fludarabine is effective in purging residual disease in patients with chronic lymphocytic leukemia. J Clin Oncol 2006; 24:2337.
  159. Porter DL, Levine BL, Kalos M, et al. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N Engl J Med 2011; 365:725.
  160. Brentjens RJ, Rivière I, Park JH, et al. Safety and persistence of adoptively transferred autologous CD19-targeted T cells in patients with relapsed or chemotherapy refractory B-cell leukemias. Blood 2011; 118:4817.
  161. Koehler P, Schmidt P, Hombach AA, et al. Engineered T cells for the adoptive therapy of B-cell chronic lymphocytic leukaemia. Adv Hematol 2012; 2012:595060.
  162. Kochenderfer JN, Dudley ME, Feldman SA, et al. B-cell depletion and remissions of malignancy along with cytokine-associated toxicity in a clinical trial of anti-CD19 chimeric-antigen-receptor-transduced T cells. Blood 2012; 119:2709.
  163. Cruz CR, Micklethwaite KP, Savoldo B, et al. Infusion of donor-derived CD19-redirected virus-specific T cells for B-cell malignancies relapsed after allogeneic stem cell transplant: a phase 1 study. Blood 2013; 122:2965.
  164. Brudno JN, Somerville RP, Shi V, et al. Allogeneic T Cells That Express an Anti-CD19 Chimeric Antigen Receptor Induce Remissions of B-Cell Malignancies That Progress After Allogeneic Hematopoietic Stem-Cell Transplantation Without Causing Graft-Versus-Host Disease. J Clin Oncol 2016; 34:1112.
  165. Turtle CJ, Hay KA, Hanafi LA, et al. Durable Molecular Remissions in Chronic Lymphocytic Leukemia Treated With CD19-Specific Chimeric Antigen Receptor-Modified T Cells After Failure of Ibrutinib. J Clin Oncol 2017; 35:3010.
  166. Siddiqi T, Soumerai JD, Dorritie KA, et al. Phase 1 TRANSCEND CLL 004 study of lisocabtagene maraleucel in patients with relapsed/refractory CLL or SLL. Blood 2022; 139:1794.
  167. Frey NV, Gill S, Hexner EO, et al. Long-Term Outcomes From a Randomized Dose Optimization Study of Chimeric Antigen Receptor Modified T Cells in Relapsed Chronic Lymphocytic Leukemia. J Clin Oncol 2020; 38:2862.
  168. Siddiqi T, Maloney DG, Kenderian SS, et al. Lisocabtagene maraleucel in chronic lymphocytic leukaemia and small lymphocytic lymphoma (TRANSCEND CLL 004): a multicentre, open-label, single-arm, phase 1-2 study. Lancet 2023; 402:641.
  169. Melenhorst JJ, Chen GM, Wang M, et al. Decade-long leukaemia remissions with persistence of CD4+ CAR T cells. Nature 2022; 602:503.
  170. Liu E, Marin D, Banerjee P, et al. Use of CAR-Transduced Natural Killer Cells in CD19-Positive Lymphoid Tumors. N Engl J Med 2020; 382:545.
Topic 4520 Version 143.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟