Skip to main content
Therapeutic Advances in Hematology logoLink to Therapeutic Advances in Hematology
. 2019 Dec 5;10:2040620719891356. doi: 10.1177/2040620719891356

Emerging treatment options for patients with p53-pathway-deficient CLL

Marisa J L Aitken 1,2, Hun J Lee 3, Sean M Post 4,
PMCID: PMC6896129  PMID: 31839919

Abstract

Over the past 40 years, p53 has been the most widely studied protein in cancer biology. Originally thought to be an oncogene due to its stabilization in many cancers, it is now considered to be one of the most critical tumor suppressors in a cell’s ability to combat neoplastic transformation. Due to its critical roles in apoptosis, cell-cycle arrest, and senescence, TP53 deletions and mutations are commonly observed and are often a portent of treatment failures and poor clinical outcomes. This is particularly true in chronic lymphocytic leukemia (CLL), as patients with p53 alterations have historically had dismal outcomes. As such, the tremendous efforts made to better understand the functions of p53 in CLL have contributed substantially to recent advances in treating patients with p53-pathway-deficient CLL.

Keywords: 17p deletion, CLL, ibrutinib, idelalisib, TP53 mutation, venetoclax

Introduction

Chronic lymphocytic leukemia (CLL) is the most common leukemia in developed countries.1 Incidence in men exists at nearly a 2:1 ratio compared with women, and the median age at diagnosis is 70 years.2 The clinical course of CLL is highly variable, as some patients live for decades without complications, while others succumb to the disease within months of diagnosis.

A diagnosis of CLL is made by the presence of ⩾5 × 109/L clonal B cells in the peripheral blood for a minimum of 3 months.3 Commonly, lymphocytosis is the only manifestation of CLL at diagnosis; patients are frequently otherwise asymptomatic. The consensus standard of care for patients in early stages of the disease is to offer close follow up without initiating anti-leukemia therapy.3 However, continued proliferation of CD5+ mature-appearing neoplastic cells can result in further leukocytosis, lymphadenopathy, splenomegaly, and autoimmune cytopenias.4 Patients progressing to these symptomatic stages are then eligible for various treatments.

Over the past 2 decades, our understanding of the pathogenesis of CLL has increased dramatically, leading to the development of new therapeutic strategies for such patients requiring treatment. Treatments have evolved from simple alkylating agents to immune-checkpoint inhibitors, genetically engineered cellular therapies, and targeted molecular agents. From 2010 to 2019, eight new agents have been approved by the US Food and Drug Administration (FDA), marking the most prolific period of drug development for CLL on record. All age groups and risk subtypes of CLL patients have benefited from this therapeutic renaissance. However, patients with CLL harboring TP53 abnormalities, usually in the form of del(17p) or TP53 mutations, continue to be a challenging population for durable disease control.

TP53 evaluation

Cytogenetics

Cytogenetic evaluation is a routine and relatively straightforward means to risk-stratify patients with CLL, in which few recurrent cytogenetic abnormalities are seen. Approximately 80% of cases will harbor at least one chromosomal aberration at diagnosis.5 Deletion of the long arm of chromosome 13 [del(13q)] is the most common alteration (~55% of cases), and is associated with favorable prognosis.5 This is followed in frequency by the intermediate-risk category of cytogenetic abnormalities, which encompasses trisomy 12 (10–20% of cases) and deletions in the long arm of chromosome 11 [del(11q); ~15% of cases]. Deletions in the short arm of chromosome 17 [del(17p)] are found in 5–8% of cases at diagnosis, and have been associated with high-risk disease.5 Indeed, numerous prospective clinical trials have confirmed a poor prognosis for patients with del(17p).68 The incidence of del(17p) is even higher in relapsed or refractory CLL (~30% of cases) and confers a similarly dismal prognosis.9 However, notable therapeutic advances discussed in this review have mitigated the poor outcomes associated with del(17p).

A critical gene within the deleted portion of chromosome 17p is TP53, which encodes for the tumor suppressor p53. p53 is often referred to as the ‘guardian of the genome’ in reference to its critical role in maintaining genomic integrity.10 Abrogation of wild-type p53 functions allows cells to accumulate mutations, due to loss of p53-dependent apoptotic and cell-cycle arrest functions, and often results in malignancy. Indeed, germline TP53 mutations are a hallmark of Li-Fraumeni syndrome, wherein patients develop malignancies of various anatomic sites (including hematologic), often before age 30.11

TP53 mutations

Somatic mutations in TP53 are observed in CLL in ~10% of cases at diagnosis and are often associated with del(17p). Indeed, almost 80% of cases with del(17p) will harbor a TP53 mutation in the remaining allele, a phenomenon referred to as loss of heterozygosity (LOH).12 Importantly, mutations in TP53 have also been observed in the absence of 17p deletions and have been associated with poor prognosis, even in this setting.13,14 Such prognoses are evolving in the current era of targeted therapeutics as discussed in this review.

Function

At its simplest, p53 is a transcription factor. In normal cells under basal conditions, the p53 protein exists at levels often below the threshold of antibody-based detection methods. This is largely due to the presence of negative regulators, including mouse double-minute 2 homolog (MDM2). MDM2 binds p53 and inhibits its transcriptional activity by interfering with its interaction with the transcriptional machinery.15 However, most importantly, MDM2 also ubiquitinates p53, thereby targeting p53 for proteasomal degradation.1618 In response to cellular stresses, such as deoxyribonucleic acid (DNA) damage or deregulated oncogene activation, this MDM2-mediated negative regulation of p53 is suppressed, allowing for p53 stabilization and transcription of p53 target genes that drive apoptotic and cell-cycle arrest programs.

Classic chemotherapeutic agents exert cytotoxic effects by directly or indirectly inducing DNA damage. Such genomic injury activates a group of protein kinases that include ataxia–telangiectasia mutated (ATM; in the case of double-strand DNA breaks) and ATM and Rad3 related (ATR; in the case of single-strand DNA breaks or replication stress).19 These kinases begin a cascade of events that include the phosphorylation of both MDM2 and p53. The result is decreased interactions between MDM2 and p53, thus preventing ubiquitination of p53 and thereby facilitating p53 stabilization.2022 As p53 accumulates in the stressed cell and is further phosphorylated, it exerts transcriptional activity, inducing expression of genes that either promote cell-cycle arrest, senescence, or apoptosis. For example, p53-mediated induction of the CDKN1A gene, which encodes the p21 protein, arrests the cell cycle and prevents proliferation of cells that have damaged DNA that could promote tumor formation or further tumor evolution.23

Among p53-responsive genes that induce apoptosis are pro-apoptotic members of the B-call lymphoma 2 (BCL2) family, such as BAX, PUMA, and NOXA.2426 p53 has also been shown to directly decrease levels of the antiapoptotic protein BCL2.27,28 Together, the BCL2 family controls the release of cytochrome c from the outer membrane of mitochondria, resulting in an irreversible apoptotic cascade.29 Thus, upon administration of many chemotherapeutic agents, the desirable result of cell death results in eradication of some or all of the tumor.

In scenarios where p53 cannot bind DNA or cannot be stabilized in order to act as a transcription factor, the desirable cytotoxic effects of chemotherapy are not evident. Importantly, the damage induced by the chemotherapy may also go unrepaired in the absence of wild-type p53 function. Thus, administration of such drugs may accelerate the accumulation of genotoxic insults while failing to induce cell death. This sets the stage for a challenging therapeutic scenario.

Consequences of TP53 aberrations

In CLL (as well as many other malignancies), these normal wild-type functions of p53 are interrupted. As previously described, in addition to 17p deletions, TP53 can also be mutated in CLL. These are often missense mutations that predominantly occur in the DNA-binding domain of TP53 and result in the loss of wild-type p53 functions.30 Indeed, mice that lack Trp53 (the murine homolog of TP53) or express a mutant form (e.g. R172H, R172P, or R270H, corresponding to the R175 and R273 ‘hotspot mutations’ in humans) are tumor prone, often developing tumors of T- or B-cell lineage.3134 Interestingly, some of these mutations have been shown to gain oncogenic activity as they alter the cellular transcriptome. This has been reviewed elsewhere.35 This review will remain focused on the deficiencies of wild-type p53, which confer a fitness advantage to malignant cells.

Even when TP53 or chromosome 17p are not directly influenced, alterations in p53 regulators may likewise render the p53 pathway deficient. Overexpression of MDM2 has been observed in B-cell malignancies, including CLL.36,37 A single-nucleotide polymorphism in MDM2 has also been linked to poor outcomes in CLL.38,39 Likewise, overexpression of MDM4, another negative p53 regulator, has been observed in a subset of p53-wild-type CLL that responds poorly to MDM2 inhibition.40 These observations have been validated in murine models, where increased levels of the p53’s negative regulators MDM2 or MDM4 result in hematologic tumors.41,42

Conversely, decreased expression of MDM2 inhibitors can also abrogate p53 functions. For example, p14ARF, which is encoded from an alternative reading frame in CDKN2A, binds to MDM2 and inhibits MDM2-dependent ubiquitination of p53.4345 As such, mutations, deletions, and hypermethylation of the CDKN2A locus have been associated with aggressive disease courses in CLL.46

Together, these data underscore the importance of p53 deregulation in hematopoietic oncogenesis and response to conventional chemotherapies. These observations also provide substantial rationale for utilizing therapeutic modalities that function without a requirement for wild-type p53.

Treating CLL

Initiation of therapy for CLL is not required for all patients at the time of diagnosis. While cytogenetic [del(17p)] or molecular (TP53 mutation) aberrations may confer a worse prognosis, the presence of these events alone is not an indication for treatment initiation. Patients may start treatment based on disease-related symptoms or evidence of disease progression, such as physical findings of bulky lymph nodes or splenomegaly, or progressive lymphocytosis or cytopenias. Guidelines for initiation of therapy have been reviewed by the International Workshop on CLL.3 Outside of clinical trials, early therapy is generally not recommended before these criteria are met, as there is currently no evidence that starting chemotherapy earlier in asymptomatic patients improves survival in CLL.47 However, early use of targeted molecular agents is an active field of study.48,49

Historic treatments-genotoxic chemotherapies

Alkylating agents

Chlorambucil is an alkylating agent that was approved for medical use in 1957. It exerts its anticancer effects via interference with DNA replication, leading to cellular apoptosis in a p53-dependent manner.50 The original studies using chlorambucil plus prednisone for treatment of CLL had modest overall response rates (ORRs) and complete response (CR) rates of 47% and 7%, respectively.51 Subsequently, patients would almost invariably progress with splenomegaly and bulky lymphadenopathy, the majority of whom succumbed to their disease or due to treatment complications.

Chlorambucil is still widely used in Europe and in elderly patients for palliative disease control.52 In a meta-analysis of seven clinical trials of the German CLL study group, of 3552 patients, 77% received chlorambucil plus anti-CD20 antibody therapy.53 Notably, 152 patients were more than 80-years old at treatment initiation.53 Unfortunately, chlorambucil was especially poor in controlling CLL with TP53 abnormalities, as these patients had significantly inferior ORR (27% versus 87%), shorter progression-free survival (PFS; 5-year PFS: 5% versus 17%), and overall survival (OS; 5-year OS 20% versus 59%) compared with patients without TP53 abnormalities.12,54 Therefore, chlorambucil is currently not recommended for cases of CLL with TP53 abnormalities, except for palliative purposes. These observations align with our biologic understanding of the consequences of administering such therapies to malignancies deficient in wild-type p53.

Purine analogs

Fludarabine is a purine analog discovered in the early 1970s as having cytotoxic activity in vitro.55 In a seminal paper by Rai and colleagues, fludarabine showed increased ORR (63% versus 37%), CR (20% versus 4%), and PFS (25 months versus 14 months) compared with chlorambucil.56 However, further analysis of patients with TP53 abnormalities demonstrated continued poor outcomes compared with standard-risk patients.56 Approximately 9% of CLL patients had TP53 abnormalities at diagnosis, but by the time of relapse, 44% of patients had TP53 abnormalities, suggesting that fludarabine exerts a selection advantage for TP53 aberrant clones.12,57 Nonetheless, fludarabine has been a backbone of frontline CLL chemotherapy since Rai and colleagues’ publication in 2000, and its combination with the cytotoxic agent cyclophosphamide, and the monoclonal antibody rituximab have been the genesis of immunochemotherapy (e.g. fludarabine–cyclophosphamide–rituximab, FCR) in hematologic malignancies.

Bendamustine

Bendamustine is a multifunctional alkylating agent containing a purine-like ring that has demonstrated clinical activity in multiple hematologic malignancies. In addition to causing DNA damage responses, this drug has been observed to trigger intrinsic apoptotic pathways, including upregulation of p53 upregulated modulator of apoptosis (PUMA) and phorbol-12-myristate-13-acetate-induced protein 1 (NOXA) and increasing expression of mitochondrial apoptogenic proteins.58 In addition, bendamustine can cause necrotic cell death, and thus shows some efficacy in cells lacking functional p53 or apoptotic pathways.59

Bendamustine has been studied in the frontline setting for patients with newly diagnosed CLL. In one study, del(17p) was noted in 7% of patients and the median PFS was less than 12 months, consistent with previous reports using genotoxic chemotherapy approaches.60 Bendamustine showed a promising response rate and was better tolerated than FCR, which led to community practices, particularly in the US, to adapt to using bendamustine, particularly for elderly patients who were not eligible for clinical trials. These practices led to a head-to-head study in which it was determined that, in combination with rituximab, bendamustine has a role in the frontline setting for patients lacking del(17p) who are not fit to receive FCR.61

Antibody therapy

Rituximab

Proteins expressed on the surface of leukemia cells represent a bevy of biologic targets. CD20 is a cell-surface protein expressed on pre-B cells and on differentiated B cells of both normal and malignant origin.62 The anti-CD20 antibody rituximab was the first of the targeted antibody therapies to change treatment paradigms for all B-cell malignancies. A combination of signaling-induced cell death, complement-dependent cytotoxicity, and antibody-dependent cellular cytotoxicity is responsible for rituximab-induced cell death.63 These distinct mechanisms of action and manageable side-effect profile allowed for straightforward combinations with already existing chemotherapeutic agents.64 FCR was effective in relapsed CLL and set the standard for frontline therapy for newly diagnosed, fit CLL patients requiring treatment.65,66 With this combination, ORR improved to 92% with a CR of 70%.66 However, even though this treatment modality has proven effective in many patients with CLL, those with TP53 abnormalities continued to have poorer response rates and shorter remission durations.

Ofatumumab

Ofatumumab is also an anti-CD20 antibody that targets a distinct epitope on CD20 that is not targeted by rituximab.67 In fludarabine-refractory CLL patients, ofatumumab was safe and effective as a single agent, with ORR of 58%, PFS of 5.9 months, and OS of 15.4 months.68 While adverse events were minimal, routine use of ofatumumab has not been adapted since it has not demonstrated superiority over rituximab.

Obinutuzumab

Obinutuzumab is another CD20 antibody that was first studied in combination with chlorambucil for elderly patients who were not candidates for standard chemoimmunotherapy.69 Patients receiving both drugs had significantly higher CR rates compared with chlorambucil alone (20.7% versus 7%).69 However, patients with del(17p) did not see such benefit. Importantly, the addition of obinutuzumab did not elicit substantial toxicity, a crucial consideration in these patient populations.69 The results of this study led to trials combining obinutuzumab with fludarabine–cyclophosphamide (FC) or bendamustine, where the combinations demonstrated robust activity, though only one patient harbored del(17p).70 For obinutuzumab–FC, the ORR was 62% with 24% CR/CRi with incomplete hematologic recovery (CRi). Obinutuzumab–bendamustine had an ORR of 90% with 45% CR/CRi.70 Further studies are ongoing to evaluate which patient populations are best to incorporate this novel CD20 antibody and to investigate its role in p53-pathway-deficient CLL.

Alemtuzumab

Alemtuzumab is an anti-CD52 antibody that was designed for CLL patients refractory to both alkylating and purine analog agents; a population usually harboring TP53 abnormalities.71 In the early 2000s, fludarabine-refractory patients had an anticipated survival of only 10 months. This led to the design of a trial treating high-risk CLL patient populations (fludarabine-refractory or harboring TP53 abnormalities) with alemtuzumab. This trial demonstrated an ORR of 33%, but came with significant infectious complications, including cytomegalovirus reactivation and fungal infections.72,73 Alemtuzumab is still available for CLL with abnormal TP53; however, its use has been limited due to this significant toxicity.

Targeted agents

To better treat CLL with deficiencies in the p53 pathway, using agents that kill cells independently of p53 is imperative. Understanding the mechanism of action of currently available targeted agents provides rationale for their utilization and combinations.

B-cell expansion in CLL is largely driven by constitutive B-cell receptor (BCR) signaling.74 The BCR signals through a series of tyrosine kinases, including phosphatidylinositol 3-kinase delta (PI3Kδ) and Bruton’s tyrosine kinase (BTK).75 Understanding the BCR signaling axis has been pivotal in developing effective drugs for CLL. To this end, PI3Kδ inhibitors (idelalisib, duvelisib, and umbralisib) and BTK inhibitors (ibrutinib, acalabrutinib, and zanubrutinib) are in various stages of clinical use. These drugs directly inhibit the pathway that produces and maintains B cells of both normal and malignant origin.

In addition to constitutive BCR signaling, CLL cells often harbor high expression of BCL2, an antiapoptotic protein that sustains cell survival.76,77 As such, methods that inhibit or decrease levels of BCL2 in CLL cells have been shown to activate apoptosis.78 This understanding led to development of venetoclax, a BCL2 inhibitor, in CLL.

Together, these drugs have been revolutionary, as they inhibit critical mediators of CLL emergence and survival. They are discussed individually in the following section. The major clinical trials utilizing these drugs in patients with p53-pathway-deficient CLL are summarized in Table 1.

Table 1.

Summary of major trials including patients with p53-pathway-deficient CLL.

PMID Treatment setting Treatment arm Median age del(17p) TP53 mutation CR/CRi Median PFS
Median OS MRD negativity MRD assessment
All patients p53 deficient
24401022 Frontline Obinutuzumab +chlorambucil 74 22/333 ND 20.7% 26.7 months ND NR 19.5% (BM); 37.7% (PB) ASO PCR
Rituximab + chlorambucil 73 20/330 7.0% 15.2 months ND NR 2.6% (BM); 3.3% (PB)
Chlorambucil 72 10/118 0% 11.1 months ND NR ND
24450857 Relapsed/refractory Idelalisib + rituximab 71 42/110 0% NR; 93% at 6 months NR NR; 92% at 12 months ND
Rituximab + placebo 71 45/110 0% 5.5 months; 46% at 6 months 4 months NR; 80% at 12 months ND
30287523 Relapsed/refractory Duvelisib 69 21/160 20/160 0.6% 13.3 months 12.7 months NR ND
Ofatumumab 69 28/159 18/159 0.6% 9.9 months 9 months NR ND
EHA LB2602 Frontline Ibrutinib 64 6/182 14/182 ND NR ND ND ND
Placebo 64 7/181 13/181 ND 14.8 months ND ND ND
23782158 Relapsed/refractory Ibrutinib 66 28/85 ND 2.3% NR; 75% at 26 months 57% at 26 months NR; 83% at 26 months ND
27637985 Relapsed/refractory Ibrutinib 64 144/144 107/116 2% NR; 63% at 24 months NR; 75% at 24 months ND
24881631 Relapsed/refractory Ibrutinib 67 63/195 ND 2% NR; 88% at 6 months NR; 83% at 6 months NR; 90% at 12 months ND
Ofatumumab 67 64/196 1% 8.1 months; 65% at 6 months 5.8 months NR; 81% at 12 months ND
30501481 Frontline Rituximab + bendamustine 70 14/481 16/174 26% 43 months; 74% at 2 years 7 months NR; 95% at 2 years 8% Flow cytometry
Ibrutinib 71 9/181 15/168 7% NR; 87% at 2 years NR NR; 90% at 2 years 1%
Ibrutinib + rituximab 71 11/180 20/168 12% NR; 88% at 2 years NR NR;94% at 2 years 4%
30522969 Frontline Ibrutinib + obinutuzumab 70 14/113 13/112 19% NR; 79% at 30 months NR NR; 86% at 30 months 35% Flow cytometry
Chlorambucil + obinutuzumab 72 18/116 16/110 8% 19 months; 31% at 30 months 11.3 months NR; 85% at 30 months 25%
26641137 Relapsed/refractory Acalabrutinib 62 18/59 ND 0% NR NR NR ND
EHA LB2606 Relapsed/refractory Acalabrutinib 68 28/155 ND 0% NR; 88% at 1 year NR; 88% at 1 year* NR ND
Rituximab + idelalisib or rituximab + bendamustine 67 21/154 ND 1% 16.5 months; 68% at 1 year 16.2 months* NR ND
31340982 Mixed Zanubrutinib 69 18/94 2.6% NR; 100% at 12 months ND ND ND
26639348 Relapsed/refractory Venetoclax 66 31/102 ND 20% 25 months; 49% at 2 years 16 months NR; 84% at 2 years 5% Flow cytometry
29562156 Relapsed/refractory Venetoclax + Rituximab 64.5 46/173 48/192 26.8% NR; 84.9% at 24 months 81.5% at 24 months 91.9% at 24 months 62.4% Flow cytometry
Rituximab + bendamustine 66 46/169 51/184 8.2% 17 months; 36.3% at 24 months 27.8% at 24 months 86.6% at 24 months 13.3%
31166681 Frontline Venetoclax + obinutuzumab 72 17/200 19/171 49.5% NR; 88.6% at 24 months 73.9% at 24 months NR; 91.8% at 24 months 75.5% (PB); 56.9% (BM) ASO PCR
Chlorambucil + obinutuzumab 71 14/193 13/157 23.1% NR; 63.7% at 24 months 32.7% at 24 months NR; 93.3% at 24 months 35.2% (PB); 17.1% (BM)
28715249 Relapsed/refractory CD19 CAR-T cells 61 14/24 ND 17% 8.5 months ND NR 88% Flow cytometry
30642819 Relapsed/refractory Nivolumab + ibrutinib 65 19/36 ND 0% NR NR NR ND
28424162 Relapsed/refractory Pembrolizumab 72 6/16 1/16 0% 2.4 months ND 11.2 months ND

ASO PCR, allele-specific oligonucleotide polymerase chain reaction; BM, bone marrow; CLL, chronic lymphocytic leukemia; CR, complete response; CRi, CR with incomplete hematologic recovery; MRD, minimal residual disease; ND, no data; NR, not reached; OS, overall survival; PFS, progression-free survival; PB, peripheral blood; PMID, unique identifier number on PubMed.

Note: * study reported “high risk cytogenetic features”, which included p53 abnormalities in addition to other features.

Idelalisib

Idelalisib is an orally available small molecule that specifically inhibits the catalytic subunit of PI3Kδ.79 Mice lacking this isoform have severe B-cell defects, highlighting the importance of this specific protein in the development and function of B cells.8082 In CLL, PI3Kδ signals downstream of the BCR, leading to constitutive activation of the PI3K pathway and upregulation of antiapoptotic proteins.83,84 Chronic BCR signaling subsequently leads to chronic activation of PI3Kδ in B-cell malignancies, including CLL.85 Downstream of PI3Kδ lies a number of effector molecules, including AKT (protein kinase B). This is particularly relevant in the context of p53, as AKT phosphorylates MDM2, enhancing its ability to ubiquitinate p53 and target it for proteasomal degradation.86 Thus, in addition to blocking the upregulation of antiapoptotic proteins, inhibition of PI3Kδ may partly restore p53 activity by allowing p53 stabilization. Indeed, in other cancer models, idelalisib has been shown to induce PUMA.87 In the absence of wild-type p53 function, upregulation of PUMA can push cells toward apoptosis. Thus, idelalisib can be useful in treating malignancies with chronic BCR activation, even in the setting of p53 pathway deficiency.

Idelalisib is currently FDA approved for relapsed CLL when used in combination with rituximab. This approval was based on the double-blind, placebo-controlled phase III study using idelalisib plus rituximab versus placebo plus rituximab. The median PFS in the idelalisib plus rituximab group was 20.3 months, whereas the placebo plus rituximab group had a median PFS of 5.5 months.88,89 OS showed similar trends of 40.6 months versus 34.6 months.89 This study enrolled a challenging patient population, as more than 40% of patients had del(17p), and the majority had received FCR and had a median of three prior therapies. In long-term follow up, where all patients received idelalisib after initial treatment, patients with del(17p) or TP53 mutations had an OS of 28.5 months in the idelalisib–rituximab arm compared with only 14.8 months in the placebo–rituximab arm.89

While clinically effective, many patients do not tolerate idelalisib, and this agent has a more adverse safety profile than other targeted agents, with relatively high rates of diarrhea and infectious pneumonia.90 The FDA issued a black-box warning after frequent immune-mediated occurrences of colitis, pneumonitis, or hepatotoxicity were reported.91 Cessation of the drug, in addition to use of corticosteroids, appear to alleviate the immune reaction.92 Combinations with idelalisib have also been associated with higher rates of infection, including Pneumocystic jirovecii pneumonia.88,93

Duvelisib

Duvelisib is an orally available PI3Kδ inhibitor that demonstrated superiority to ofatumumab in a phase III clinical trial of relapsed refractory CLL patients.94 In this study, 160 patients were randomized to receive single-agent duvelisib or single-agent ofatumumab. Approximately 30 patients in each group had p53 deficiencies. Duvelisib was able to obtain an overall response rate of 74% versus 45% with ofatumumab alone and a median PFS of 13.3 months versus 9.9 months. The efficacy of duvelisib was maintained in p53-deficient patients, as their median PFS was 12.7 months compared with 9 months for patients receiving ofatumumab. Based on these data, the FDA approved the use of duvelisib in the treatment of relapsed refractory CLL.94

Umbralisib

Umbralisib is a newer inhibitor of PI3Kδ that is structurally distinct from idelalisib and duvelisib.95 In a phase I study, 17/20 patients with relapsed refractory CLL had an objective response.95 Of eight patients with high-risk cytogenetic features, six had a response and none had disease progression. Importantly, umbralisib appeared to have a more favorable safety profile than idelalisib and duvelisib, with fewer incidences of autoimmune-like toxicities.95 A study evaluating the use of umbralisib in patients who were intolerant to idelalisib or BTK inhibitors suggests that these patients may benefit from switching to umbralisib.96 Median PFS in this cohort, which comprised 24% p53-deficient patients, was 23.5 months. Importantly, patients with prior PI3Kδ-targeted therapy did not require umbralisib discontinuation, due to recurrence of adverse events that led to discontinuation of the original drug.96

Ibrutinib

Ibrutinib is a tyrosine kinase inhibitor that specifically inhibits Bruton’s tyrosine kinase (BTK), a critical molecule mediating BCR signaling. In a phase I/II study of ibrutinib in relapsed refractory patients with high-risk disease, ORR was 71%, and an additional 20% of patients had a partial response (PR) with lymphocytosis. The latter was subsequently described as an ibrutinib-related phenomenon that does not have negative clinical impact. At 26 months of median follow up, PFS was 75% and OS was 83%.97

Ibrutinib was the first novel agent to obtain full FDA approval for frontline treatment (March 2016) and relapsed refractory settings in patients with del(17p) CLL. In the frontline setting, ibrutinib was compared with its combination with rituximab versus bendamustine–rituximab in older patients.98 Critically, for patients in this study with TP53 abnormalities, the median PFS for the bendamustine plus rituximab group was 6 months compared with the ibrutinib plus rituximab group, where median PFS was not reached at 3 years of follow up.98

These encouraging results with ibrutinib in the p53-deficient patients were further validated in the RESONATE-17 trial. Ibrutinib in del(17p) populations showed 2-year PFS of 63% and OS of 75% in this trial, which enrolled 144 patients, all with centrally confirmed del(17p).99

Ibrutinib was also evaluated in the frontline setting in combination with obinutuzumab in a phase III trial.100 Ibrutinib plus obinutuzumab demonstrated superiority compared with chlorambucil–obinutuzumab, as median PFS was not reached in the ibrutinib group and was 19 months in the chlorambucil–obinutuzumab group. In p53-deficient patients, median PFS was 11.3 months in the chlorambucil–obinutuzumab group but was not reached in the ibrutinib–obinutuzumab group.100

In the longest follow-up study of single-agent ibrutinib, 5-year follow up still showed an ORR of 89% and CR in 29% in treatment-naïve patients, and CR of 10% in the relapsed refractory population. However, the del(17p) population had a median PFS of 26 months compared with 51 months for the whole relapsed refractory population.101

Ibrutinib has also been shown effective in an in vivo model of de novo p53-mutated CLL.102 Importantly, this study also demonstrated that frontline ibrutinib treatment in this model did not place undue pressure on the remaining wild-type Trp53 alleles to undergo LOH. While these findings must be fully substantiated clinically, they are of great interest, since most single trials do not have substantial numbers of patients harboring only heterozygous p53 mutations.

Ibrutinib has been a clear breakthrough in treating CLL patients with TP53 abnormalities and has been incorporated into all guidelines for the frontline and relapse settings. While outcomes with this drug are substantially better than those seen with chemoimmunotherapy in similar populations, ibrutinib alone may not be optimal for overcoming TP53 abnormalities.

Acalabrutinib

Acalabrutinib is a second-generation, more selective, irreversible inhibitor of BTK. This was tested in a phase I/II study of relapsed refractory CLL patients with a median of three prior lines of therapy in which 31% had TP53 abnormalities. The acalabrutinib arm had an ORR of 95%, partial response rate of 85%, and partial response with lymphocytosis in 10%. Interestingly, del(17p) patients had a 100% ORR, and no observations of Richter’s transformation were noted in that trial.103

In a phase III study, single-agent acalabrutinib was compared with rituximab–idelalisib (IR) or rituximab–bendamustine (BR) in a relapsed refractory population.104 PFS was not reached in the acalabrutinib arm versus 16.5 months in the IR or BR groups. This improvement in PFS was maintained in the p53-deficient subgroups.104

Acalabrutinib has also demonstrated efficacy in a small study of patients deemed intolerant to ibrutinib.105 Patients with del(17p) had an ORR of 67%, and median PFS was not reached in this subgroup at 13.6 months of follow up.105 Of 33 patients in this study who were previously deemed intolerant to ibrutinib, only 3 discontinued acalabrutinib due to adverse events.105 Another phase II study has generated similar results, demonstrating that acalabrutinib is tolerable and effective in ibrutinib-intolerant patients.106 Indeed, a head-to-head comparison of ibrutinib versus acalabrutinib is currently underway for relapsed refractory patients with high-risk CLL [ClinicalTrials.gov identifier: NCT02477696].

Zanubrutinib

Zanubrutinib is an orally available kinase inhibitor with greater selectivity for BTK than ibrutinib.107 This selectivity is desirable, since the off-target effects of ibrutinib are thought to largely mediate the adverse events observed clinically.107 In a phase I study, ORR was 100% in patients with p53 abnormalities.107 This high rate of activity is likely due to the fact that zanubrutinib has a longer half-life compared with acalabrutinib or ibrutinib, thus prolonging exposure of CLL cells to specific BTK inhibition.107 Zanubrutinib is now being compared with ibrutinib in an ongoing phase III trial of relapsed refractory CLL [ClinicalTrials.gov identifier: NCT03734016].108

The evidence for the use of BTK inhibitors in TP53-aberrant CLL is thus promising, and highlights how targeted agents not requiring the p53 pathway for their efficacy are critical for patients with TP53 alterations.

Venetoclax

Venetoclax is an orally available inhibitor of BCL2, an antiapoptotic protein that sustains cell survival and is highly expressed in CLL.76 BCL2 binds to pro-apoptotic proteins BIM, BAD, BID, and NOXA, thereby inhibiting initiation of the apoptotic cascade.109 Inhibition of BCL2 by venetoclax thus allows pro-apoptotic proteins to promote cell death. Intact p53 regulates the expression of several pro-apoptotic proteins such as PUMA and BAX, which promote apoptosis. In the absence of wild-type p53 function, venetoclax still holds promise, as it directly antagonizes BCL2 and promotes cell death in a p53-independent manner.110

This lack of p53-dependency is highlighted by studies showing that in relapsed refractory CLL patients with TP53 mutations or chromosome 17p deletions, single-agent venetoclax led to a 20% CR rate, with 5% of patients having no minimal residual disease (MRD) via flow cytometry.111 In this difficult-to-treat population, single-agent venetoclax led to 69% PFS at 16 months.111

Venetoclax has been studied in combination with obinutuzumab (VO) versus chlorambucil–obinutuzumab (CO) in previously untreated patients.112 The use of venetoclax was associated with a longer PFS, including in the p53-aberrant group, where 24 month PFS estimates were 73.9% in the VO arm versus 32.7 in the CO arm.112 Further, 70.8% of patients with aberrant p53 treated with VO achieved MRD negativity in peripheral blood compared with only 9.1% in CO.112

In relapsed refractory CLL, venetoclax plus rituximab (VR) was compared with bendamustine plus rituximab (BR).113 In this study, venetoclax was stopped after 2 years of treatment. The 2-year rate of PFS in del(17p) patients was 81.5% in the VR group versus 27.8% in the BR group.113 Post-treatment follow up of this study confirmed these results, showing that median PFS in the VR group for patients with TP53 mutations was 36 months, versus 12.9 months in the BR group.114 Likewise, del(17p) patients did not reach median PFS, versus 15.4 months in the BR group.114 Notably, aberrant p53 was associated with an increased risk of disease progression after venetoclax cessation.114

Ibrutinib plus venetoclax

Since chronic BCR signaling is exacerbated by overexpression of BCL2, dual inhibition of these molecules is biologically justified. In the frontline setting, the combination of ibrutinib and venetoclax has been studied to combine mechanistically distinct drugs to minimize the risk of disease resistance developing. In patients with high-risk CLL with TP53 aberrations, 80% achieved CR with 61% having no MRD.115 While longer follow up is needed to determine the durability of the disease response, this treatment combination represents a completely chemotherapy-free regimen that is highly effective in treating patients with TP53 alterations.

Currently, a phase III study is investigating the role of venetoclax in combination with ibrutinib and obinutuzumab versus ibrutinib plus obinutuzumab in treatment-naïve young patients with CLL. This phase III trial builds from the efficacy of an earlier trial utilizing this combination,116 and hopes to answer the question of whether this time-limited three-drug combination is effective versus continuous ibrutinib dosing in CLL patients aged younger than 70 years [ClinicalTrials.gov identifier: NCT03701282]. Yet another study evaluating this combination is also underway, wherein each drug is given as a sequential therapy, beginning with ibrutinib as induction therapy, obinutuzumab as consolidation, and venetoclax as a 1-year maintenance [ClinicalTrials.gov identifier: NCT03755947].

Immunological therapies

Checkpoint inhibitors

Nivolumab is an antibody against programmed-cell death protein 1 (PD-1), the blockade of which releases the ‘checkpoint inhibition’ of antitumor T-cell responses, allowing for T-cell-induced tumor killing.117 Nivolumab in combination with ibrutinib in a high-risk [del(17p) and del(11q)] relapsed refractory CLL population led to an ORR of 61%.118 Grade 3–4 immune-related toxicities included rash in 8% of patients and elevated alanine transaminase in 2%.118 While this combination was well tolerated, the response rate was similar to single-agent ibrutinib in CLL. Therefore, it is unclear if the addition of nivolumab potentiates the clinical efficacy of ibrutinib. However, it has been noted that 65% of cases with Richter’s transformation responded to the combination of ibrutinib and nivolumab, demonstrating that this combination may be useful in this setting.118,119

Likewise, pembrolizumab, another anti-PD-1-antibody has shown efficacy as a single agent in patients with Richter’s transformation.120 However, no patients with CLL responded to pembrolizumab in this study.120 A study investigating the combination of pembrolizumab with either idelalisib or ibrutinib is currently underway [ClinicalTrials.gov identifier: NCT02332980].

Chimeric antigen receptor T cells

Modifying one’s own T cells to attack cancer is no longer science fiction. June and colleagues were the first to report the use of autologous chimeric antigen receptor (CAR)-modified T cells in a multirefractory CLL patient, who then achieved a CR.121 This technology programs T cells to recognize CD19 on the surface of normal and malignant B cells and then selectively kill CD19-expressing cells.121

Currently, similar CAR-T cells are being evaluated in multiple prospective trials. A phase I CD19 CAR-T trial enrolled very high risk and ibrutinib-refractory CLL patients. Most patients had either complex karyotype or del(17p) (23/24).122 ORR was 71% (17/24), and for patients who were alive at the restaging time point, CR was 21% (4/19) and partial response was 53% (10/19).122 Toxicities were similar to previous CAR-T studies, with cytokine release syndrome and neurotoxicity observed in 83% and 33%, respectively. In those who had a clinical response and underwent bone marrow biopsy, 88% (15/17) had no MRD by flow cytometry, and 58% (7/12) had no detectable disease by deep IGH sequencing.122 This demonstrates that patients who respond to CAR-T therapy may be able to achieve a deep remission without MRD.

Other CAR therapies are also in development, such as ‘off the shelf’ allogeneic CAR-T-cell therapy,123 and natural killer (NK) CAR-cell therapy, the latter of which is being actively developed for refractory CLL [ClinicalTrials.gov identifier: NCT02727803].

Allogeneic transplantation

At this time, the only truly ‘curative’ treatment option for hematologic malignancy is allogeneic stem-cell transplantation, which potentially offers a graft versus leukemia effect, leading to a cure. This possible benefit is substantial, but the risk is also high. Given the option, many patients with TP53 abnormalities seek allogeneic transplantation. At 6 years post-allogeneic transplant, data from four large centers showed a PFS of 40–45% and OS of 50–60% in this patient population.124126 These were patients who had matching donors and were selected to be ‘fit’. Treatment-related mortality and severe graft versus host disease was reported in 16–25% and 50–55%, respectively. These results make it daunting to recommend allogeneic transplantation in many elderly patients, as risk will undoubtedly be worse for elderly patients who are under-represented in these clinical trials. However, for a young, fit, high-risk CLL patient with a perfectly matched donor, the opportunity of a potentially curative therapy may be worth the risks associated with stem-cell transplantation. Indeed, the evolving reality of targeted, orally available, and well-tolerated therapies is reason to pause prior to recommending transplant. However, this higher-risk option still represents a viable option while awaiting longer-term outcomes for patients treated with such targeted agents.

p53-targeted therapy

Indirect activation of p53 function by inhibition of MDM2 has been achieved preclinically with a small-molecule antagonist, Nutlin-3a.127 However, the development of this agent has not been fully deployed in the clinical setting due to other exciting therapeutic options, deflating clinical interest in MDM2 as a pharmacologic target. Given that p53 mutations represent a significant mechanism driving poor outcomes in patients with CLL, therapies that restore wild-type p53 function may still be useful additions to the armamentarium of antileukemic agents. In fact, a recently opened phase III clinical trial [ClinicalTrials.gov identifier: NCT03745716] is attempting to reactivate mutant p53 functions using APR-246 (formally known as PRIMA-1MET) in patients with refractory myelodysplastic syndrome. Successful results from this trial could provide impetus to explore the activity of APR-246 in patients with CLL harboring p53 mutations.

Resistance

In cases of CLL that develop resistance to BTK inhibitors ibrutinib or acalabrutinib, a substantially higher percentage appear to harbor del(17p).128,129 Such an association suggests that being aware of resistance mechanisms in this population may be particularly prudent.

Restoration of BCR signaling and resistance to ibrutinib have been observed via mutations in BTK at Cys481, thus preventing ibrutinib from binding.128131 Activating mutations in the downstream substrate PLCγ2 leading to autonomous BCR signaling are also commonly observed.128131 In such cases, transition of therapy to venetoclax or a PI3Kδ inhibitor can suppresses these resistant clones.128,129,132 Development of small molecules to specifically address these resistance mutations may also hold promise. LOXO305 is a reversible BTK inhibitor that retains activity against the most common BTK C481S alteration in vitro.133 A phase I trial is underway with this agent [ClinicalTrials.gov identifier: NCT03740529]. Likewise, ARQ 531 is a reversible BTK inhibitor with other kinase inhibitory activity that has been shown in preclinical studies to be effective against both BTK and PLCγ2 mutant clones.134

Resistance to venetoclax can arise from a mutation in BCL2 at Gly101 or Asp103. Mutations at these residue can reduce the affinity of venetoclax for BCL2 substantially. These mutations have been associated with venetoclax resistance and impending relapse in patients with CLL.135,136 Other antiapoptotic proteins besides BCL2, such as MCL1 or BCL-XL, may also be upregulated in CLL cells, leading to disease progression despite venetoclax administration. Importantly, inhibitors of BCR signaling (ibrutinib, idelalisib) and blockade of CD20 (obinutuzumab, rituximab) have demonstrated downregulation of MCL1, which sensitizes leukemic cells to BCL2 inhibition by venetoclax, lending further credence to the notion of combination therapy.137

Pre-emptively understanding these mechanisms of resistance and their impact on the p53 pathway will be key to the sustained success of these agents in the clinic. Understanding the known mechanisms of resistance or anticipating the development of resistance may be useful for keeping patients in deep, durable remissions.

Summary

Patients with CLL have benefited greatly from the explosion of new treatment options. We are rapidly moving away from genotoxic drugs in the frontline setting, and the new reality is that patients with CLL are being treated with targeted small-molecule inhibitors, achieving greater responses and deeper remissions in the context of improved quality of life. Likewise, TP53-abnormal populations have also seen vast improvements in their survival with these new combinations. Likewise, novel CAR-T, NK CARs, and allogeneic CAR therapies have the potential to provide the next steps forward in improving the quality and quantity of life for all patients with CLL.

Acknowledgments

We thank Mrs. Xiaorui Zhang for critically reviewing the manuscript.

Footnotes

Funding: The author(s) received no financial support for the research, authorship, and/or publication of this article.

Conflict of interest statement: The author(s) declare that there is no conflict of interest.

Contributor Information

Marisa J. L. Aitken, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA The University of Texas MD Anderson Cancer Center UT Health Graduate School of Biomedical Sciences, Houston, TX, USA.

Hun J. Lee, Department of Lymphoma and Multiple Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA

Sean M. Post, Department of Leukemia, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Houston, TX 77030-4000, USA.

References

  • 1. Jemal A, Siegel R, Ward E, et al. Cancer statistics, 2007. CA Cancer J Clin 2007; 57: 43–66. [DOI] [PubMed] [Google Scholar]
  • 2. National Cancer Institute Surveillance, Epidemiology, and the End Results Program. Cancer fact sheets: chronic lymphocytic leukemia (CLL), https://seer.cancer.gov/statfacts/html/clyl.html (accessed 1 July 2019).
  • 3. Hallek M, Cheson BD, Catovsky D, et al. iwCLL guidelines for diagnosis, indications for treatment, response assessment, and supportive management of CLL. Blood 2018; 131: 2745–2760. [DOI] [PubMed] [Google Scholar]
  • 4. Rozman C, Montserrat E. Chronic lymphocytic leukemia. N Engl J Med 1995; 333: 1052–1057. [DOI] [PubMed] [Google Scholar]
  • 5. Dohner H, Stilgenbauer S, Benner A, et al. Genomic aberrations and survival in chronic lymphocytic leukemia. N Engl J Med 2000; 343: 1910–1916. [DOI] [PubMed] [Google Scholar]
  • 6. Grever MR, Lucas DM, Dewald GW, et al. Comprehensive assessment of genetic and molecular features predicting outcome in patients with chronic lymphocytic leukemia: results from the US intergroup phase III trial E2997. J Clin Oncol 2007; 25: 799–804. [DOI] [PubMed] [Google Scholar]
  • 7. Catovsky D, Richards S, Matutes E, et al. Assessment of fludarabine plus cyclophosphamide for patients with chronic lymphocytic leukaemia (the LRF CLL4 trial): a randomised controlled trial. Lancet 2007; 370: 230–239. [DOI] [PubMed] [Google Scholar]
  • 8. Hallek M, Fischer K, Fingerle-Rowson G, et al. Addition of rituximab to fludarabine and cyclophosphamide in patients with chronic lymphocytic leukaemia: a randomised, open-label, phase 3 trial. Lancet 2010; 376: 1164–1174. [DOI] [PubMed] [Google Scholar]
  • 9. Stilgenbauer S, Zenz T, Winkler D, et al. Subcutaneous alemtuzumab in fludarabine-refractory chronic lymphocytic leukemia: clinical results and prognostic marker analyses from the CLL2H study of the German chronic lymphocytic leukemia study group. J Clin Oncol 2009; 27: 3994–4001. [DOI] [PubMed] [Google Scholar]
  • 10. Lane DP. Cancer. p53, guardian of the genome. Nature 1992; 358: 15–16. [DOI] [PubMed] [Google Scholar]
  • 11. Malkin D, Li FP, Strong LC, et al. Germ line p53 mutations in a familial syndrome of breast cancer, sarcomas, and other neoplasms. Science 1990; 250: 1233–1238. [DOI] [PubMed] [Google Scholar]
  • 12. Zenz T, Eichhorst B, Busch R, et al. TP53 mutation and survival in chronic lymphocytic leukemia. J Clin Oncol 2010; 28: 4473–4479. [DOI] [PubMed] [Google Scholar]
  • 13. Zenz T, Krober A, Scherer K, et al. Monoallelic TP53 inactivation is associated with poor prognosis in chronic lymphocytic leukemia: results from a detailed genetic characterization with long-term follow-up. Blood 2008; 112: 3322–3329. [DOI] [PubMed] [Google Scholar]
  • 14. Rossi D, Cerri M, Deambrogi C, et al. The prognostic value of TP53 mutations in chronic lymphocytic leukemia is independent of Del17p13: implications for overall survival and chemorefractoriness. Clin Cancer Res 2009; 15: 995–1004. [DOI] [PubMed] [Google Scholar]
  • 15. Momand J, Zambetti GP, Olson DC, et al. The mdm-2 oncogene product forms a complex with the p53 protein and inhibits p53-mediated transactivation. Cell 1992; 69: 1237–1245. [DOI] [PubMed] [Google Scholar]
  • 16. Haupt Y, Maya R, Kazaz A, et al. Mdm2 promotes the rapid degradation of p53. Nature 1997; 387: 296–299. [DOI] [PubMed] [Google Scholar]
  • 17. Honda R, Tanaka H, Yasuda H. Oncoprotein MDM2 is a ubiquitin ligase E3 for tumor suppressor p53. FEBS Lett 1997; 420: 25–27. [DOI] [PubMed] [Google Scholar]
  • 18. Kubbutat MH, Jones SN, Vousden KH. Regulation of p53 stability by Mdm2. Nature 1997; 387: 299–303. [DOI] [PubMed] [Google Scholar]
  • 19. Abraham RT. PI 3-kinase related kinases: ‘big’ players in stress-induced signaling pathways. DNA Repair (Amst) 2004; 3: 883–887. [DOI] [PubMed] [Google Scholar]
  • 20. Shieh SY, Ikeda M, Taya Y, et al. DNA damage-induced phosphorylation of p53 alleviates inhibition by MDM2. Cell 1997; 91: 325–334. [DOI] [PubMed] [Google Scholar]
  • 21. Maya R, Balass M, Kim ST, et al. ATM-dependent phosphorylation of Mdm2 on serine 395: role in p53 activation by DNA damage. Genes Dev 2001; 15: 1067–1077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Khosravi R, Maya R, Gottlieb T, et al. Rapid ATM-dependent phosphorylation of MDM2 precedes p53 accumulation in response to DNA damage. Proc Natl Acad Sci U S A 1999; 96: 14973–14977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Waldman T, Kinzler KW, Vogelstein B. p21 is necessary for the p53-mediated G1 arrest in human cancer cells. Cancer Res 1995; 55: 5187–5190. [PubMed] [Google Scholar]
  • 24. Miyashita T, Reed JC. Tumor suppressor p53 is a direct transcriptional activator of the human bax gene. Cell 1995; 80: 293–299. [DOI] [PubMed] [Google Scholar]
  • 25. Nakano K, Vousden KH. PUMA, a novel proapoptotic gene, is induced by p53. Mol Cell 2001; 7: 683–694. [DOI] [PubMed] [Google Scholar]
  • 26. Oda E, Ohki R, Murasawa H, et al. Noxa, a BH3-only member of the Bcl-2 family and candidate mediator of p53-induced apoptosis. Science 2000; 288: 1053–1058. [DOI] [PubMed] [Google Scholar]
  • 27. Miyashita T, Harigai M, Hanada M, et al. Identification of a p53-dependent negative response element in the bcl-2 gene. Cancer Res 1994; 54: 3131–3135. [PubMed] [Google Scholar]
  • 28. Miyashita T, Krajewski S, Krajewska M, et al. Tumor suppressor p53 is a regulator of Bcl-2 and Bax gene expression in vitro and in vivo. Oncogene 1994; 9: 1799–1805. [PubMed] [Google Scholar]
  • 29. Schuler M, Bossy-Wetzel E, Goldstein JC, et al. p53 induces apoptosis by caspase activation through mitochondrial cytochrome c release. J Biol Chem 2000; 275: 7337–7342. [DOI] [PubMed] [Google Scholar]
  • 30. Bouaoun L, Sonkin D, Ardin M, et al. TP53 Variations in human cancers: new lessons from the IARC TP53 database and genomics data. Hum Mutat 2016; 37: 865–876. [DOI] [PubMed] [Google Scholar]
  • 31. Donehower LA, Harvey M, Slagle BL, et al. Mice deficient for p53 are developmentally normal but susceptible to spontaneous tumours. Nature 1992; 356: 215–221. [DOI] [PubMed] [Google Scholar]
  • 32. Jacks T, Remington L, Williams BO, et al. Tumor spectrum analysis in p53-mutant mice. Curr Biol 1994; 4: 1–7. [DOI] [PubMed] [Google Scholar]
  • 33. Lang GA, Iwakuma T, Suh YA, et al. Gain of function of a p53 hot spot mutation in a mouse model of Li-Fraumeni syndrome. Cell 2004; 119: 861–872. [DOI] [PubMed] [Google Scholar]
  • 34. Olive KP, Tuveson DA, Ruhe ZC, et al. Mutant p53 gain of function in two mouse models of Li-Fraumeni syndrome. Cell 2004; 119: 847–860. [DOI] [PubMed] [Google Scholar]
  • 35. Mantovani F, Collavin L, Del Sal G. Mutant p53 as a guardian of the cancer cell. Cell Death Differ 2019; 26: 199–212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Xu-Monette ZY, Medeiros LJ, Li Y, et al. Dysfunction of the TP53 tumor suppressor gene in lymphoid malignancies. Blood 2012; 119: 3668–3683. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Watanabe T, Hotta T, Ichikawa A, et al. The MDM2 oncogene overexpression in chronic lymphocytic leukemia and low-grade lymphoma of B-cell origin. Blood 1994; 84: 3158–3165. [PubMed] [Google Scholar]
  • 38. Post SM, Pant V, Abbas H, et al. Prognostic impact of the MDM2SNP309 allele in leukemia and lymphoma. Oncotarget 2010; 1: 168–174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Gryshchenko I, Hofbauer S, Stoecher M, et al. MDM2 SNP309 is associated with poor outcome in B-cell chronic lymphocytic leukemia. J Clin Oncol 2008; 26: 2252–2257. [DOI] [PubMed] [Google Scholar]
  • 40. Bo MD, Secchiero P, Degan M, et al. MDM4 (MDMX) is overexpressed in chronic lymphocytic leukaemia (CLL) and marks a subset of p53wild-type CLL with a poor cytotoxic response to Nutlin-3. Br J Haematol 2010; 150: 237–239. [DOI] [PubMed] [Google Scholar]
  • 41. Jones SN, Hancock AR, Vogel H, et al. Overexpression of Mdm2 in mice reveals a p53-independent role for Mdm2 in tumorigenesis. Proc Natl Acad Sci U S A 1998; 95: 15608–15612. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Xiong S, Pant V, Suh YA, et al. Spontaneous tumorigenesis in mice overexpressing the p53-negative regulator Mdm4. Cancer Res 2010; 70: 7148–7154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Kamijo T, Weber JD, Zambetti G, et al. Functional and physical interactions of the ARF tumor suppressor with p53 and Mdm2. Proc Natl Acad Sci U S A 1998; 95: 8292–8297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Zhang Y, Xiong Y, Yarbrough WG. ARF promotes MDM2 degradation and stabilizes p53: ARF-INK4a locus deletion impairs both the Rb and p53 tumor suppression pathways. Cell 1998; 92: 725–734. [DOI] [PubMed] [Google Scholar]
  • 45. Zhang Y, Xiong Y. Control of p53 ubiquitination and nuclear export by MDM2 and ARF. Cell Growth Differ 2001; 12: 175–186. [PubMed] [Google Scholar]
  • 46. Pinyol M, Cobo F, Bea S, et al. p16(INK4a) gene inactivation by deletions, mutations, and hypermethylation is associated with transformed and aggressive variants of non-Hodgkin’s lymphomas. Blood 1998; 91: 2977–2984. [PubMed] [Google Scholar]
  • 47. CLL Trialists’ Collaborative Group. Chemotherapeutic options in chronic lymphocytic leukemia: a meta-analysis of the randomized trials. J Natl Cancer Inst 1999; 91: 861–868. [DOI] [PubMed] [Google Scholar]
  • 48. Langerbeins P, Bahlo J, Rhein C, et al. The CLL12 trial protocol: a placebo-controlled double-blind phase III study of ibrutinib in the treatment of early-stage chronic lymphocytic leukemia patients with risk of early disease progression. Future Oncol 2015; 11: 1895–1903. [DOI] [PubMed] [Google Scholar]
  • 49. Langerbeins P, Bahlo J, Rhein C, et al. Ibrutinib versus placebo in patients with asymptomatic, treatment-naïve early stage CLL: primary endpoint results of the phase 3 double-blind randomized CLL12 TRIAL. Hematol Oncol 2019; 37: 38–40.31187520 [Google Scholar]
  • 50. Di Antonio M, McLuckie KI, Balasubramanian S. Reprogramming the mechanism of action of chlorambucil by coupling to a G-quadruplex ligand. J Am Chem Soc 2014; 136: 5860–5863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Sawitsky A, Rai KR, Glidewell O, et al. Comparison of daily versus intermittent chlorambucil and prednisone therapy in the treatment of patients with chronic lymphocytic leukemia. Blood 1977; 50: 1049–1059. [PubMed] [Google Scholar]
  • 52. Sylvan SE, Asklid A, Johansson H, et al. First-line therapy in chronic lymphocytic leukemia: a Swedish nation-wide real-world study on 1053 consecutive patients treated between 2007 and 2013. Haematologica 2019; 104: 797–804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Al-Sawaf O, Bahlo J, Robrecht S, et al. Outcome of patients aged 80 years or older treated for chronic lymphocytic leukaemia. Br J Haematol 2018; 183: 727–735. [DOI] [PubMed] [Google Scholar]
  • 54. Gonzalez D, Martinez P, Wade R, et al. Mutational status of the TP53 gene as a predictor of response and survival in patients with chronic lymphocytic leukemia: results from the LRF CLL4 trial. J Clin Oncol 2011; 29: 2223–2229. [DOI] [PubMed] [Google Scholar]
  • 55. Chun HG, Leyland-Jones B, Cheson BD. Fludarabine phosphate: a synthetic purine antimetabolite with significant activity against lymphoid malignancies. J Clin Oncol 1991; 9: 175–188. [DOI] [PubMed] [Google Scholar]
  • 56. Rai KR, Peterson BL, Appelbaum FR, et al. Fludarabine compared with chlorambucil as primary therapy for chronic lymphocytic leukemia. N Engl J Med 2000; 343: 1750–1757. [DOI] [PubMed] [Google Scholar]
  • 57. Schnaiter A, Paschka P, Rossi M, et al. NOTCH1, SF3B1, and TP53 mutations in fludarabine-refractory CLL patients treated with alemtuzumab: results from the CLL2H trial of the GCLLSG. Blood 2013; 122: 1266–1270. [DOI] [PubMed] [Google Scholar]
  • 58. Roue G, Lopez-Guerra M, Milpied P, et al. Bendamustine is effective in p53-deficient B-cell neoplasms and requires oxidative stress and caspase-independent signaling. Clin Cancer Res 2008; 14: 6907–6915. [DOI] [PubMed] [Google Scholar]
  • 59. Leoni LM, Bailey B, Reifert J, et al. Bendamustine (Treanda) displays a distinct pattern of cytotoxicity and unique mechanistic features compared with other alkylating agents. Clin Cancer Res 2008; 14: 309–317. [DOI] [PubMed] [Google Scholar]
  • 60. Fischer K, Cramer P, Busch R, et al. Bendamustine in combination with rituximab for previously untreated patients with chronic lymphocytic leukemia: a multicenter phase II trial of the German chronic lymphocytic leukemia study group. J Clin Oncol 2012; 30: 3209–3216. [DOI] [PubMed] [Google Scholar]
  • 61. Eichhorst B, Fink AM, Bahlo J, et al. First-line chemoimmunotherapy with bendamustine and rituximab versus fludarabine, cyclophosphamide, and rituximab in patients with advanced chronic lymphocytic leukaemia (CLL10): an international, open-label, randomised, phase 3, non-inferiority trial. Lancet Oncol 2016; 17: 928–942. [DOI] [PubMed] [Google Scholar]
  • 62. Riley JK, Sliwkowski MX. CD20: a gene in search of a function. Semin Oncol 2000; 27: 17–24. [PubMed] [Google Scholar]
  • 63. Weiner GJ. Rituximab: mechanism of action. Semin Hematol 2010; 47: 115–123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Mohammed R, Milne A, Kayani K, et al. How the discovery of rituximab impacted the treatment of B-cell non-Hodgkin’s lymphomas. J Blood Med 2019; 10: 71–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Badoux XC, Keating MJ, Wang X, et al. Fludarabine, cyclophosphamide, and rituximab chemoimmunotherapy is highly effective treatment for relapsed patients with CLL. Blood 2011; 117: 3016–3024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Parikh SA, Keating MJ, O’Brien S, et al. Frontline chemoimmunotherapy with fludarabine, cyclophosphamide, alemtuzumab, and rituximab for high-risk chronic lymphocytic leukemia. Blood 2011; 118: 2062–2068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Teeling JL, French RR, Cragg MS, et al. Characterization of new human CD20 monoclonal antibodies with potent cytolytic activity against non-Hodgkin lymphomas. Blood 2004; 104: 1793–1800. [DOI] [PubMed] [Google Scholar]
  • 68. Wierda WG, Kipps TJ, Mayer J, et al. Ofatumumab as single-agent CD20 immunotherapy in fludarabine-refractory chronic lymphocytic leukemia. J Clin Oncol 2010; 28: 1749–1755. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Goede V, Fischer K, Busch R, et al. Obinutuzumab plus chlorambucil in patients with CLL and coexisting conditions. N Engl J Med 2014; 370: 1101–1110. [DOI] [PubMed] [Google Scholar]
  • 70. Brown JR, O’Brien S, Kingsley CD, et al. Obinutuzumab plus fludarabine/cyclophosphamide or bendamustine in the initial therapy of CLL patients: the phase 1b GALTON trial. Blood 2015; 125: 2779–2785. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Ferrajoli A, O’Brien S, Keating MJ. Alemtuzumab: a novel monoclonal antibody. Expert Opin Biol Ther 2001; 1: 1059–1065. [DOI] [PubMed] [Google Scholar]
  • 72. Keating MJ, O’Brien S, Kontoyiannis D, et al. Results of first salvage therapy for patients refractory to a fludarabine regimen in chronic lymphocytic leukemia. Leuk Lymphoma 2002; 43: 1755–1762. [DOI] [PubMed] [Google Scholar]
  • 73. Keating MJ, Flinn I, Jain V, et al. Therapeutic role of alemtuzumab (Campath-1H) in patients who have failed fludarabine: results of a large international study. Blood 2002; 99: 3554–3561. [DOI] [PubMed] [Google Scholar]
  • 74. Petlickovski A, Laurenti L, Li X, et al. Sustained signaling through the B-cell receptor induces Mcl-1 and promotes survival of chronic lymphocytic leukemia B cells. Blood 2005; 105: 4820–4827. [DOI] [PubMed] [Google Scholar]
  • 75. Stevenson FK, Krysov S, Davies AJ, et al. B-cell receptor signaling in chronic lymphocytic leukemia. Blood 2011; 118: 4313–4320. [DOI] [PubMed] [Google Scholar]
  • 76. Kitada S, Andersen J, Akar S, et al. Expression of apoptosis-regulating proteins in chronic lymphocytic leukemia: correlations with in vitro and in vivo chemoresponses. Blood 1998; 91: 3379–3389. [PubMed] [Google Scholar]
  • 77. Hanada M, Delia D, Aiello A, et al. bcl-2 gene hypomethylation and high-level expression in B-cell chronic lymphocytic leukemia. Blood 1993; 82: 1820–1828. [PubMed] [Google Scholar]
  • 78. Kitada S, Takayama S, De Riel K, et al. Reversal of chemoresistance of lymphoma cells by antisense-mediated reduction of bcl-2 gene expression. Antisense Res Dev 1994; 4: 71–79. [DOI] [PubMed] [Google Scholar]
  • 79. Yang Q, Modi P, Newcomb T, et al. Idelalisib: first-in-class PI3K delta inhibitor for the treatment of chronic lymphocytic leukemia, small lymphocytic leukemia, and follicular lymphoma. Clin Cancer Res 2015; 21: 1537–1542. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Okkenhaug K, Bilancio A, Farjot G, et al. Impaired B and T cell antigen receptor signaling in p110δ PI 3-kinase mutant mice. Science 2002; 297: 1031–1034. [DOI] [PubMed] [Google Scholar]
  • 81. Clayton E, Bardi G, Bell SE, et al. A crucial role for the p110δ subunit of phosphatidylinositol 3-kinase in B cell development and activation. J Exp Med 2002; 196: 753–763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Jou ST, Carpino N, Takahashi Y, et al. Essential, nonredundant role for the phosphoinositide 3-kinase p110δ in signaling by the B-cell receptor complex. Mol Cell Biol 2002; 22: 8580–8591. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Lannutti BJ, Meadows SA, Herman SE, et al. CAL-101, a p110δ selective phosphatidylinositol-3-kinase inhibitor for the treatment of B-cell malignancies, inhibits PI3K signaling and cellular viability. Blood 2011; 117: 591–594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Longo PG, Laurenti L, Gobessi S, et al. The Akt/Mcl-1 pathway plays a prominent role in mediating antiapoptotic signals downstream of the B-cell receptor in chronic lymphocytic leukemia B cells. Blood 2008; 111: 846–855. [DOI] [PubMed] [Google Scholar]
  • 85. Rodriguez A, Villuendas R, Yanez L, et al. Molecular heterogeneity in chronic lymphocytic leukemia is dependent on BCR signaling: clinical correlation. Leukemia 2007; 21: 1984–1991. [DOI] [PubMed] [Google Scholar]
  • 86. Ogawara Y, Kishishita S, Obata T, et al. Akt enhances Mdm2-mediated ubiquitination and degradation of p53. J Biol Chem 2002; 277: 21843–21850. [DOI] [PubMed] [Google Scholar]
  • 87. Yang S, Zhu Z, Zhang X, et al. Idelalisib induces PUMA-dependent apoptosis in colon cancer cells. Oncotarget 2017; 8: 6102–6113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Furman RR, Sharman JP, Coutre SE, et al. Idelalisib and rituximab in relapsed chronic lymphocytic leukemia. N Engl J Med 2014; 370: 997–1007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. 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: 1391–1402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. De Weerdt I, Koopmans SM, Kater AP, et al. Incidence and management of toxicity associated with ibrutinib and idelalisib: a practical approach. Haematologica 2017; 102: 1629–1639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. 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: 195–203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Coutre SE, Barrientos JC, Brown JR, et al. Management of adverse events associated with idelalisib treatment: expert panel opinion. Leuk Lymphoma 2015; 56: 2779–2786. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. 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: 297–311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. 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: 2446–2455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Burris HA, III, Flinn IW, Patel MR, et al. Umbralisib, a novel PI3Kδ and casein kinase-1epsilon inhibitor, in relapsed or refractory chronic lymphocytic leukaemia and lymphoma: an open-label, phase 1, dose-escalation, first-in-human study. Lancet Oncol 2018; 19: 486–496. [DOI] [PubMed] [Google Scholar]
  • 96. Mato AR, Schuster SJ, Lamanna N, et al. A phase 2 study to assess the safety and efficacy of umbralisib in patients with chronic lymphocytic leukemia (CLL) who are intolerant to prior BTK or PI3K delta inhibitor therapy. Hematol Oncol 2019; 37: 88–89. [Google Scholar]
  • 97. Byrd JC, Furman RR, Coutre SE, et al. Targeting BTK with ibrutinib in relapsed chronic lymphocytic leukemia. N Engl J Med 2013; 369: 32–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Woyach JA, Ruppert AS, Heerema NA, et al. Ibrutinib regimens versus chemoimmunotherapy in older patients with untreated CLL. N Engl J Med 2018; 379: 2517–2528. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. 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: 1409–1418. [DOI] [PubMed] [Google Scholar]
  • 100. Moreno C, Greil R, Demirkan F, et al. Ibrutinib plus obinutuzumab versus chlorambucil plus obinutuzumab in first-line treatment of chronic lymphocytic leukaemia (iLLUMINATE): a multicentre, randomised, open-label, phase 3 trial. Lancet Oncol 2019; 20: 43–56. [DOI] [PubMed] [Google Scholar]
  • 101. O’Brien S, Furman RR, Coutre S, et al. Single-agent ibrutinib in treatment-naive and relapsed/refractory chronic lymphocytic leukemia: a 5-year experience. Blood 2018; 131: 1910–1919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Lee HJ, Gallardo M, Ma H, et al. p53-independent ibrutinib responses in an Emu-TCL1 mouse model demonstrates efficacy in high-risk CLL. Blood Cancer J 2016; 6: e434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Byrd JC, Harrington B, O’Brien S, et al. Acalabrutinib (ACP-196) in relapsed chronic lymphocytic leukemia. N Engl J Med 2016; 374: 323–332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Ghia P, Pluta A, Wach M, et al. Acalabrutinib vs rituximab plus idelalisib (IdR) or bendamustine (BR) by investigator choice in relapsed/refractory (RR) chronic lymphocytic leukemia: phase 3 ASCEND study. Hematol Oncol 2019; 37: 86–87. [Google Scholar]
  • 105. Awan FT, Schuh A, Brown JR, et al. Acalabrutinib monotherapy in patients with chronic lymphocytic leukemia who are intolerant to ibrutinib. Blood Adv 2019; 3: 1553–1562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Rogers KA, Thompson PA, Allan JN, et al. Phase 2 study of acalabrutinib in ibrutinib (IBR)-intolerant patients (pts) with relapsed/refractory (R/R) chronic lymphocytic leukemia (CLL). J Clin Oncol 2019; 37: 7530–7530. [Google Scholar]
  • 107. Tam CS, Trotman J, Opat S, et al. Phase 1 study of the selective BTK inhibitor zanubrutinib in B-cell malignancies and safety and efficacy evaluation in CLL. Blood 2019; 134: 851–859. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Hillmen P, Brown JR, Byrd JC, et al. ALPINE: phase III zanubrutinib (BGB-3111) versus ibrutinib in patients with relapsed/refractory (R/R) chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL). J Clin Oncol 2019; 37: TPS7572–TPS7572. [Google Scholar]
  • 109. Hardwick JM, Soane L. Multiple functions of BCL-2 family proteins. Cold Spring Harb Perspect Biol 2013; 5. pii: a008722. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Anderson MA, Deng J, Seymour JF, et al. The BCL2 selective inhibitor venetoclax induces rapid onset apoptosis of CLL cells in patients via a TP53-independent mechanism. Blood 2016; 127: 3215–3224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Roberts AW, Davids MS, Pagel JM, et al. Targeting BCL2 with venetoclax in relapsed chronic lymphocytic leukemia. N Engl J Med 2016; 374: 311–322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. 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: 2225–2236. [DOI] [PubMed] [Google Scholar]
  • 113. Seymour JF, Kipps TJ, Eichhorst B, et al. Venetoclax-rituximab in relapsed or refractory chronic lymphocytic leukemia. N Engl J Med 2018; 378: 1107–1120. [DOI] [PubMed] [Google Scholar]
  • 114. 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: 269–277. [DOI] [PubMed] [Google Scholar]
  • 115. Jain N, Keating M, Thompson P, et al. Ibrutinib and venetoclax for first-line treatment of CLL. N Engl J Med 2019; 380: 2095–2103. [DOI] [PubMed] [Google Scholar]
  • 116. Rogers KA, Huang Y, Ruppert AS, et al. Phase 2 study of combination obinutuzumab, ibrutinib, and venetoclax in treatment-naive and relapsed/refractory chronic lymphocytic leukemia. Blood 2018; 132: 693–693. [Google Scholar]
  • 117. Iwai Y, Ishida M, Tanaka Y, et al. Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade. Proc Natl Acad Sci U S A 2002; 99: 12293–12297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Younes A, Brody J, Carpio C, et al. Safety and activity of ibrutinib in combination with nivolumab in patients with relapsed non-Hodgkin lymphoma or chronic lymphocytic leukaemia: a phase 1/2a study. Lancet Haematol 2019; 6: e67–e78. [DOI] [PubMed] [Google Scholar]
  • 119. Jain N, Basu S, Thompson PA, et al. Nivolumab combined with ibrutinib for CLL and richter transformation: a phase II trial. Blood 2016; 128: 59. [Google Scholar]
  • 120. Ding W, LaPlant BR, Call TG, et al. Pembrolizumab in patients with CLL and Richter transformation or with relapsed CLL. Blood 2017; 129: 3419–3427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Porter DL, Levine BL, Kalos M, et al. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N Engl J Med 2011; 365: 725–733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. 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: 3010–3020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Sheridan C. Allogene and celularity move CAR-T therapy off the shelf. Nat Biotechnol 2018; 36: 375–377. [DOI] [PubMed] [Google Scholar]
  • 124. Sorror ML, Storer BE, Sandmaier BM, et al. Five-year follow-up of patients with advanced chronic lymphocytic leukemia treated with allogeneic hematopoietic cell transplantation after nonmyeloablative conditioning. J Clinical Oncol 2008; 26: 4912–4920. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125. 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: 3284–3288. [DOI] [PubMed] [Google Scholar]
  • 126. Khouri IF, Bassett R, Poindexter N, et al. Nonmyeloablative allogeneic stem cell transplantation in relapsed/refractory chronic lymphocytic leukemia: long-term follow-up, prognostic factors, and effect of human leukocyte histocompatibility antigen subtype on outcome. Cancer 2011; 117: 4679–4688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Kojima K, Konopleva M, McQueen T, et al. Mdm2 inhibitor Nutlin-3a induces p53-mediated apoptosis by transcription-dependent and transcription-independent mechanisms and may overcome Atm-mediated resistance to fludarabine in chronic lymphocytic leukemia. Blood 2006; 108: 993–1000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Kanagal-Shamanna R, Jain P, Patel KP, et al. Targeted multigene deep sequencing of Bruton tyrosine kinase inhibitor-resistant chronic lymphocytic leukemia with disease progression and Richter transformation. Cancer 2019; 125: 559–574. [DOI] [PubMed] [Google Scholar]
  • 129. Ahn IE, Underbayev C, Albitar A, et al. Clonal evolution leading to ibrutinib resistance in chronic lymphocytic leukemia. Blood 2017; 129: 1469–1479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. Woyach JA, Furman RR, Liu TM, et al. Resistance mechanisms for the Bruton’s tyrosine kinase inhibitor ibrutinib. N Engl J Med 2014; 370: 2286–2294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Burger JA, Landau DA, Taylor-Weiner A, et al. Clonal evolution in patients with chronic lymphocytic leukaemia developing resistance to BTK inhibition. Nat Commun 2016; 7: 11589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. 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: 65–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Brandhuber B, Gomez E, Smith S, et al. LOXO-305, a next generation reversible BTK inhibitor, for overcoming acquired resistance to irreversible BTK inhibitors. Clin Lymphoma Myeloma Leuk 2018; 18: S216. [Google Scholar]
  • 134. Reiff SD, Mantel R, Smith LL, et al. The BTK inhibitor ARQ 531 targets ibrutinib-resistant CLL and richter transformation. Cancer Discov 2018; 8: 1300–1315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. 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: 342–353. [DOI] [PubMed] [Google Scholar]
  • 136. Tausch E, Close W, Dolnik A, et al. Venetoclax resistance and acquired BCL2 mutations in chronic lymphocytic leukemia. Haematologica 2019; 104: e434–e437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137. Bagacean C, Tomuleasa C, Tempescul A, et al. Apoptotic resistance in chronic lymphocytic leukemia and therapeutic perspectives. Crit Rev Clin Lab Sci 2019; 56: 321–332. [DOI] [PubMed] [Google Scholar]

Articles from Therapeutic Advances in Hematology are provided here courtesy of SAGE Publications

RESOURCES