Physical Address
304 North Cardinal St.
Dorchester Center, MA 02124
Disclosures: FTA has provided consultancy to: Genentech, Astrazeneca, Abbvie, Janssen, Pharmacyclics, Gilead sciences, Kite pharma, Celgene, Karyopharm, MEI Pharma, Verastem, Incyte, Beigene, Johnson and Johnson, Dava Oncology, BMS, Merck, Cardinal Health, ADCT therapeutics, Epizyme. PRG has provided consultancy to: Pharmacyclics LLC, Kite pharma, BMS, Rafael Pharma and Cellectar Biosciences
Chronic lymphocytic leukemia (CLL) is the most prevalent adult leukemia in the West, with a median age at diagnosis of 71 years and a male preponderance. Roughly a third of patients are under 65 years of age, ~10% are under 55 years of age, and <2% are under 45 years of age at diagnosis. The disease is characterized by an accumulation of monoclonal, CD5+ B-cells in the bone marrow, peripheral blood, and secondary lymphoid organs. CLL is a very heterogeneous disease with a highly variable clinical course, ranging from indolent disease not needing therapy (in approximately 30% of patients) to aggressive presentations with progressive B-symptoms, lymphocytosis, cytopenias, lymphadenopathy, hepatosplenomegaly, recurrent infections, or autoimmune manifestations. Staging criteria for CLL like the Rai (stages 0–IV) and Binet (stages A–C) were developed in the era of chemotherapy and estimated median life expectancy ranging from 13-, 8- and 2-years for early-, intermediate- and advanced-stage disease, respectively. These have since been replaced by the CLL-International Prognostic Index (IPI) that incorporates fluorescence in situ hybridization and molecular markers and is a better predictor for overall outcomes and time to treatment.
There have been significant advances in our understanding of the molecular biology of CLL in the past three decades, and this has translated into the development initially of chemoimmunotherapy (CIT) replacing chemotherapy and, more recently, novel targeted drugs that have changed the therapeutic landscape for the disease. These U.S. Food and Drug Administration (FDA)-approved novel agents include drugs that target kinases downstream of the B-cell receptor like the Bruton tyrosine kinase inhibitors (BTKi), ibrutinib and acalabrutinib, phosphoinositide-3 kinase inhibitors (PI-3ki), idelalisib and duvelisib, and the BCL2 antagonist, venetoclax. Several large randomized clinical trials have demonstrated a survival benefit for these targeted drugs over CIT in both treatment-naïve and relapsed/refractory CLL.
Despite these major strides, CLL remains an incurable disease with CIT and targeted agents, and allogeneic hematopoietic cell transplantation (allo-HCT) remains the only potential curative treatment option, and still has a role in patients with high-risk disease characteristics. Newer cellular immunotherapies, including chimeric antigen receptor T-cell therapy (CAR-T-cell), are showing promise across the spectrum of B-cell malignancies and are being actively investigated in CLL. In this chapter, we provide a comprehensive review of the evolving role of HCT and other cellular therapies for CLL in the modern era.
In the CIT era, the following disease features have shown to predict inferior outcomes in CLL and include cytogenetic abnormalities like del(17p), del(11q), complex karyotype (≥ 3 chromosomal abnormalities), TP53 mutations, unmutated immunoglobulin heavy chain variable genes (U- IGHV ), overexpression of CD38, CD49, ZAP-70, serum beta-2 microglobulin, and gene mutations like NOTCH1 and SF3B1 . These features usually portend a shorter time to initial treatment and shorter remissions after chemotherapy-based treatment, than low-risk CLL features like del(13q), trisomy 12 or mutated IGHV ( M- IGHV) . The CLL-IPI categorizes risk to low, intermediate, high, or very high based on risk factors [del(17p), Tp53 mutation, serum beta-2 microglobulin (<3.5 or ≥3.5 mg/L), age and clinical stage]. The median time to initial therapy from diagnosis ranges from 7 years in low- and intermediate-risk disease (75% patients) compared to 2 years in high/very-high risk disease. However, in recent trials of targeted agents like ibrutinib and venetoclax, the presence of some of these traditional unfavorable features like del(11q) and U-IGHV have lost their prognostic significance with only Tp53 mutation or del(17p) still predictive of inferior outcomes.
In 2007, a consensus paper from the European Research Initiative on CLL (ERIC) and the European Society for Blood and Marrow Transplantation (EBMT) defined “high-risk” CLL for which HCT should be considered as disease refractory to purine analogs, relapsing disease within 2 years after combination therapy with purine analogs, and/or disease with del(17p)/Tp53 mutations. In the modern era of novel agents, the outcomes of patients with Tp53 abnormalities and early relapse post-CIT have improved and the 4-year survival probability has increased from <40% to >80% with ibrutinib. Hence, two new high-risk CLL categories were defined in 2018 ( Table 17.1 ).
CLL high-risk-I, CIT-resistant: This group includes patients with Tp53 abnormalities who have failed CIT but are responding to a first-targeted therapy (BTKi or BCL2i). Cellular therapies like HCT should be considered in selected patients with low procedure-related risk.
CLL high-risk-II, CIT- and novel-therapy resistant: This group includes patients who, independent of Tp53 status, have failed both CIT and a first-targeted agent (BTKi or BCL2i), even if responding to an alternate novel agent. Cellular therapies are to be strongly considered for eligible patients in this group.
Therapy CLL Refractory to | TP53 Lesion Present (del 17p/TP53 Mutation) | High-Risk Category |
---|---|---|
CIT only | Yes | High-Risk-I (CIT resistant but sensitive to BTKi and BCL2 antagonist) |
|
Yes or No | High-Risk-II (CIT and novel agent resistant) |
Thus, currently, younger patients (<70 years) with CLL should be evaluated for allo-HCT if their disease has del(17p)/TP53 mutations/complex karyotype and they have failed either a BTKi or venetoclax. Patients whose disease has progressed on both a BTKi and BCL2i should be evaluated for cellular therapies regardless of karyotype/Tp53 status with eventual plans for allo-HCT.
A 48-year-old lady was diagnosed in 2016 with symptomatic CLL/small lymphocytic lymphoma (SLL) with bulky lymphadenopathy, and on cytogenetics showed del(11q). She was treated with ibrutinib, which she tolerated well and attained a good partial response. Her disease progressed after 4 years, and the disease did not show a del (17p) or TP53 mutation. She had no human leukocyte antigen (HLA)-matched sibling donors and did not want to proceed with an allo-HCT at this juncture. She was started on venetoclax with obinutuzumab and achieved a complete remission.
BTKis and PI3K-inhibitors are targeted agents that have shown impressive response rates and durable remissions and have changed the natural history of CLL in the current era.
CLL cells expresses high levels of BTK, a nonreceptor tyrosine kinase that is triggered through the B-cell receptor prosurvival pathway and is activated by upstream kinases LYN and SYK. Ibrutinib was the first-in-class irreversible BTKi that forms a covalent bond to cysteine 481 in BTK. In the phase I trial, ibrutinib showed very durable responses, with a 7-year progression-free survival (PFS) of 83% in treatment-naïve and 34% in previously treated patients with CLL. The RESONATE trial showed a survival benefit for ibrutinib compared to the anti-CD20 monoclonal antibody (MAB) ofatumumab and led to its approval in 2014 for relapsed/refractory (R/R) CLL. The RESONATE-2 trial established its efficacy as front-line therapy with 5-year PFS of 70%. Recent practice-changing trials like the ALLIANCE A041202 (versus Bendamustine and Rituximab) and E1912 (versus Fludarabine, Cyclophosphamide, and Rituximab) have shown improved PFS with an ibrutinib-based upfront regimen compared to CIT in CLL. Side effects associated with ibrutinib therapy include bleeding, cardiac arrythmias like atrial fibrillation, arthralgia, risk of infections, and hypertension, and these have led to drug discontinuation rates of up to 25% in some series.
Acalabrutinib is a second-generation BTKi that has higher BTK selectivity compared to ibrutinib. It does not inhibit other kinases like epidermal growth factor receptor (EGFR), interleukin-2 inducible T-cell kinase (ITK), or TEC, and hence has a better safety profile compared to ibrutinib. The drug has been approved for both front-line and relapsed CLL therapy based on the ELEVATE-TN and ASCEND trials. Acalabrutinib has been shown to be noninferior to ibrutinib the R/R CLL setting and with a significant reduction in adverse events such as atrial fibrillation and bleeding. Other BTKi including zanubrutinib and noncovalent third-generation BTKi are being explored in clinical trials. Response rates to BTKi in the upfront setting are not significantly affected by Tp53 lesions compared to CIT.
Resistance to BTKi has been described and is usually caused by acquired mutations in the BTK binding site (C481S), activating mutations in downstream PLCG2 leading to BTK-independent pathway activation or both. Noncovalent BTKi and other combination strategies are being explored as treatment options in acquired BTKi resistance.
Idelalisib, an oral reversible inhibitor of PI3Kδ, inhibits BCR signaling and homing receptors for CLL, and has been approved in combination with rituximab or ofatumumab for CLL therapy. In a randomized study, idelalisib plus rituximab significantly improved PFS and overall survival (OS) (93% at 24 weeks, median, 20.3 months) compared to placebo/rituximab. Its efficacy was, however, limited by autoimmune side effects (hepatitis, colitis, pneumonitis) and infectious side effects (pneumocystis pneumonia and cytomegalovirus infection). Its efficacy is shorter than BTKi in the R/R CLL setting but is a valuable alternative in BTKi intolerant scenarios. Duvelisib is a dual PI3K inhibitor targeting PI3Kδ and PI3Kγ. The DUO trial showed an improved response rates and PFS (13.3 vs. 9.9 months) with duvelisib versus ofatumumab and led to its FDA approval for R/R CLL.
Venetoclax is an oral, potent selective inhibitor of BCL2, an antiapoptotic molecule that has significant therapeutic activity in CLL. In the phase I trial, venetoclax resulted in a median PFS of 25 months in R/R CLL. Tumor lysis syndrome (TLS) and neutropenia were noted as side effects with the drug. Thus, slow ramp-up weekly dosing with close monitoring of TLS is how the drug is administered. In the MURANO trial for R/R CLL, fixed-duration therapy with venetoclax for 24 cycles (rituximab given for first 6 cycles) significantly improved PFS compared to bendamustine-rituximab (BR) (2-year PFS 84.9% vs. 36.3%). The CLL14 trial from the German CLL study group for elderly, treatment-naïve CLL also showed a major survival benefit with fixed duration of 12 cycles of venetoclax (with obinutuzumab for first 6 cycles) compared to chlorambucil and obinutuzumab. Based on these trials, venetoclax in combination with anti-CD20 MABs received approval for treatment of front-line and relapsed CLL. A significant proportion of patients (20%–30%) treated with venetoclax achieve minimal residual disease (MRD)-negative remissions. It is also active in patients with resistance to kinase inhibitors with an overall response rate (ORR) in 65% and a median PFS of 24.7 months.
Venetoclax resistance has been linked to BCL2 mutations (G101V) that reduce its binding to the target, B-cell translocation gene ( BTG1 ) mutations and aberrations of CDKN2A or CDKN2B (cyclin-dependent kinase genes), and overexpression of BCL-XL and MCL-1.
There has been no data suggesting superiority of either a BTKi or BCL2 antagonist for CLL treatment, and hence current sequencing of therapies is determined based on patient and disease characteristics. The concept of limited-duration therapy with “doublet” or “triplet” combinations including a BTKi, BCL2 antagonist, and a CD20 MAB are being explored in clinical trials. In a phase II trial, 2-year fixed-duration therapy with ibrutinib and venetoclax led to an impressive complete response (CR) rate of 88% and 61% patients with undetectable MRD at 12 months.
Thus, the advent of these pathway inhibitors has changed the treatment landscape of CLL. However, concerns of drug toxicity, financial toxicity caused by the need for indefinite therapy (especially with BTKi), acquired resistance, Richter’s transformation (RT), and inability to attain a “cure” with novel agents remain unaddressed issues. The optimal sequencing and combination strategies are being explored.
The historical and current role of HCT and other cellular therapies will be reviewed in this chapter in light of these major treatment advances in CLL.
Become a Clinical Tree membership for Full access and enjoy Unlimited articles
If you are a member. Log in here