Article synopsis
Allogeneic stem cell transplantation (HSCT) offers the only potentially curative approach to the treatment of chronic lymphocytic leukemia (CLL). However, this is applicable only to a minority of CLL patients in view of the advanced age at presentation. Moreover, HSCT is associated with significant treatment related mortality and morbidity, largely due to chronic graft versus host disease (GVHD). The judicious choice of which patients merit this approach therefore remains important. Internationally accepted guidelines suggest that HSCT is indicated in patients who are fit enough, have a suitable matched donor, have 17p deletion or TP53 mutations or have relapsed relatively quickly after chemo-immunotherapy. There are several new agents that are in clinical trials or recently approved in CLL that demonstrate impressive responses and durable durations of response in high risk patients who might be candidates for transplant. HSCT must always be considered in view of other, potentially less toxic therapies which could be offered. Therefore the choice of HSCT versus a novel agent is one that must be gauged on a patient by patient basis, and this will change as data mature on the use of these novel agents in CLL.
Keywords: Chronic lymphocytic leukemia, allogeneic hematopoietic stem cell transplantation, novel substances, high risk patients, GvL, GvHD
Introduction: how the availability of immunochemotherapy and novel substances are changing CLL treatment
Chronic lymphocytic leukemia (CLL) is the most common leukemia in adults in the Western world, and is characterized by the progressive accumulation of mature typically CD5-positive B lymphocytes within the blood, bone marrow, and secondary lymphoid organs1–3. Although CLL is mostly an indolent disease, there are subgroups of patients which die within a few years from diagnosis despite intensive therapy. Over the past decade, significant advances in the understanding of the pathogenesis of CLL have lead to the development of a range of novel treatment options for patients requiring therapy. In young patients without significant comorbidities, immunochemotherapy with fludarabine, cyclophosphamide and the anti-CD20 monoclonal antibody (mAb) rituximab (FCR) has been established as the first-line standard of care treatment4,5. While this regimen leads to high overall response rates (ORR) and a long progression-free survival (PFS), it is unsuitable for certain subgroups of patients: these include patients with p53 abnormalities who respond poorly to purine-analogue-based immunochemotherapy and relapse often and early6–8, and elderly patients with comorbidities unable to tolerate FCR-associated toxicities9.
In the latter, chlorambucil is a widely accepted therapeutic option, and the combination with rituximab is generally well tolerated and improves PFS10,11. A recently published pivotal phase 3 trial by the German CLL Study Group (GCLLSG) showed that the type 2 anti-CD20 antibody obinutuzumab was superior to rituximab when each was combined with chlorambucil12. Ofatumumab is another fully humanized anti-CD20 mAb that has revealed high efficacy in untreated and relapsed/ refractory patients, and even in patients pre-treated with rituximab13–15. Several recent clinical studies indicate that novel agents interfering with B-cell receptor (BCR) signaling, such as the BTK inhibitor ibrutinib, the PI3kp110δ inhibitor idelalisib, or the BCL2 inhibitor navitoclax, are well tolerated and very active, even for the treatment of relapsed and fludarabine-refractory CLL, and various combinations with immunochemotherapy are currently being tested in registration studies or are under clinical development16–22. While these early results appear very encouraging, it is yet to be seen how they will translate into long-lasting remissions and disease control. In addition, a recent report indicates that patients can become resistant to ibrutinib therapy because of mutations of drug binding sites within the BCR pathway, and similar resistance mechanisms to other substances are likely23.
The only curative treatment option in CLL so far is allogeneic hematopoietic stem cell transplantation (HSCT)24. HSCT takes advantage of the graft-versus-leukemia (GvL) effect mediated by differentiated transplanted effector cells, which are capable to mount an antitumor immune response and induce long-lasting clinical remission25. However, HSCT is only suitable for a selected group of patients, and the challenges that HSCT has to face in 2014 are the following:
To identify and predict which patients and specific subgroups of patients benefit most from HSCT, and in which novel substances are unlikely to alter the biological course of their disease,
to recognize the appropriate time point when HSCT should be offered,
to determine if and how HSCT should be best combined with novel therapeutic options.
This review summarizes the current knowledge on HSCT in CLL and critically discusses its role in the era of novel treatment strategies.
The unmet need of poor risk CLL patients before the availability of novel substances
Although immunochemotherapy has significantly improved the outcome for the majority of CLL patients, there are subgroups of patients who have repeatedly been identified as having a poor response to therapy. The pivotal report by Döhner and colleagues predicted that patients with del17p- typically require therapy within one year of diagnosis and have a median overall survival (OS) of just 32 months6. This lack of chemosensitivity is biologically explained by the malfunction of the tumor suppressor protein p53, whose gene locus is located on the short arm of chromosome 1726. In CLL, deletion of 17p- leads to the inactivation of the TP53 gene. This is often accompanied by inactivating mutations of the second locus of TP53, leading to a complete loss of function27,28. Within the pivotal CLL8 trial, which demonstrated the superiority of frontline FCR over fludarabine and cyclophosphamide alone, del17p- was the strongest negative predictive factor for response to therapy and survival, and the clinical responses that were achieved were not durable. Even though there is retrospective data indicating that some patients with del17p- might experience an indolent course despite the mutation29, similar unfavorable outcomes have been observed in other prospective trials using combinations of rituximab with bendamustine30 or fludarabine alone31.
Until recently, the only therapy that appeared to be able to overcome the negative impact of p53 abnormalities in the first-line treatment setting, both in terms of its predictive value and its effect on the response to treatment, is the anti-CD52 monoclonal antibody alemtuzumab and combinations with chlorambucil, high-dose corticosteroids, rituximab, and FCR32–37. These approaches however are associated with high hematological and non-hematological toxicities and severe infectious complications, and are therefore unsuitable for the majority of elderly CLL patients. In the relapsed setting, the management of patients with TP53 abnormalities is even more challenging. Several clinical studies have demonstrated that FCR and combinations with high-dose corticosteroids, alemtuzumab or alternative regimens consisting of rituximab, oxaliplatin, cytarabine and fludarabine (OFAR) have only limited and short-term efficacy and are associated with high toxicity rates37–43. However, depending on doses and application routes, it also seems feasible that alemtuzumab-based regimens can serve as a means to “bridge” the time to HSCT.
Summary
del17p- and/ or p53 abnormalities are negative predictive factors for response to therapy and survival. This can be partly overcome by treatment with the anti-CD52 antibody alemtuzumab, alone and in combination, but comes at the cost of high non-hematological and hematological toxicities.
Indication of transplantation: the 2007 EBMT consensus criteria and beyond
In line with the experiences from these (immuno)chemotherapy-based clinical trials, the EBMT transplant consensus from 2007 states that HSCT is a reasonable treatment option in relapsed/ fludarabine-refractory patients and patients with p53 abnormalities with indication for treatment44. This is also reflected in the 2008 iwCLL guidelines, which recommend that patients with resistant disease, a short time to progression, and del(17p) should be offered investigative clinical protocols including HSCT45. In a more recently published Perspective, three risk categories were suggested based on the predicted effectiveness of FCR-like treatment, and the “highest-risk” category included patients in which treatment with FCR is unlikely to yield acceptable response or remission rates or prolong survival46. Features of “highest-risk” include TP53 loss/mutation, purine analog-refractoriness, a very short response to prior FCR and failure to achieve CR after FCR, and these patients were considered to be prime candidates for investigational agents in clinical trials and HSCT. These definitions are summarized in table 1.
Table 1.
EBMT criteria Dreger et al. 200744 |
|
iwCLL criteria Hallek et al. 200845 |
|
Highest risk in risk category model Zenz et al. 201246 |
|
Summary
Internationally accepted guidelines suggest that HSCT is indicated in patients who are fit enough for this approach, have a suitable matched donor, have 17p deletion or TP53 mutations or have relapsed relatively quickly after chemo-immunotherapy.
Evidence for the efficacy of HSCT in CLL
The first myeloablative treatment based transplantation strategies were developed more than 20 years ago, but were unsuitable for the majority of patients due to their high morbidity and mortality47,48. After it was recognized that engraftment and GvL activity can be achieved without preceding myeloablative treatment49–51, non-myeloablative reduced intensity conditioning (RIC) strategies have made HSCT accessible to a larger cohort of CLL patients, including the elderly and those with comorbidities. This is reflected in the number of RIC-HSCTs in the European Society of Blood and Marrow Transplantation (EBMT) registry: according to the 2012 annual activity survey, 3% of all HSCT indications were performed in CLL (n=475), mostly from unrelated donors, making CLL the most frequent indication for HSCT among lymphomas52.
Several large studies have demonstrated that in contrast to other intensive therapies, the relapse incidence after HSCT seems to decrease over time, indicating that RIC HSCT provides long-term disease control in about 40% of patients and also overcomes the negative prognostic effect of p53 abnormalities and fludarabine-refractoriness53–61. The results from the largest reported prospective studies are summarized in table 2. The curative potential of HCST was also confirmed in patients with SF3B1 and NOTCH1 gene mutations62, which have been identified as novel recurrent genetic mutations in CLL and are mostly associated with resistance or poor response to conventional treatment63–66. A smaller recently published prospective trial of 40 patients using RIC with fludarabine, total body irradiation and rituximab demonstrated a positive effect of rituximab on OS and EFS in multivariate analyses67.
Table 2.
Fred Hutchinson Cancer Center | German CLL Study Group | MD Anderson Cancer Center | Dana Farber Cancer Intitute | |
---|---|---|---|---|
| ||||
Sorror et al. 200854 | Dreger et al. 201053,62 | Khouri et al. 201156 | Brown et al. 201357 | |
Number of patients | 82 | 90 | 86 | 76 |
Conditioning regimen | Flu/low-dose TBI | Flu/Cy ± ATG | Flu/Cy ± R | Flu/Bu |
Donors, % (sibling/MUR) | 63/37 | 41/59 | 50/50 | 37/63 |
Median follow-up, months | 60 | 72 | 37 | 61 |
Early mortality, % (<100d) | <10 | <3 | <3 | <3 |
NRM, % | 23 | 23 | 17 | 16 |
Acute grade 3-4 GvHD, % | 20 | 14 | 7 | 17 |
Severe chronic GvHD, % | 53 | 55 | 56 | 48 |
Median PFS, % | 39 (5yrs) | 38 (6yrs) | 36 (6yrs) | 43 (6yrs) |
Median OS, % | 50 (5yrs) | 58 (6yrs) | 51 (6yrs) | 63 (6yrs) |
Abbreviations: Flu – fludarabine; TBI – total body irradiation; Cy – cyclophosphamide; ATG - antithymocyte globulin; R – rituximab; Bu – busulfan; MUR – matched unrelated donor; NRM – non relapse mortality; GvHD – graft versus host disease; PFS – progression free survival; OS – overall survival.
HSCT seems particularly active in patients with complete or partial disease remission at the time of transplantation: in patients with chemo-sensitive disease, the five year OS could be increased to up to 80%53,57,68. To achieve a good remission state is however challenging, and as discussed before, some regimens that appear suitable come to the cost of high toxicities. Recent studies indicate that modified OFAR and alemtuzumab based regimens are the most favorable strategies to help prepare patients for successful HSCT by achieving good remissions43,69–71. In general, pre-transplant characteristics as assessed by the EBMT risk score seem to be of predictive value for OS: this score uses five patient-specific pre-transplant variables (age, disease status, time from diagnosis to transplant, donor type, and donor–recipient sex combination)72. A retrospective EBMT analysis demonstrated that there was a significant difference in OS at 5 years between patients with score 1–3 and patients having a higher score, and also supported the use of matched unrelated donors as equivalent alternative to HLA-matched sibling donors in CLL68.
Summary
RIC HSCT provides long-term disease control in about 40% of patients, including patients with adverse prognostic markers, but remission status at the time of transplantation and pre-transplant characteristics are predictive of HSCT outcome.
Post-transplantation monitoring by MRD kinetics and GvL activity
The curative effect of HSCT in some patients supports out current understanding of the importance of minimal residual disease (MRD) as a quantification of treatment response. MRD denotes a subclinical disease burden remaining after specific therapy. For CLL, this is defined as a contamination of five CLL cells or less per nl peripheral blood in the absence of clinical signs or symptoms of the disease73. Patients showing less than one CLL cell in 10.000 benign leukocytes in peripheral blood or bone marrow are considered as being MRD negative45. MRD levels have been demonstrated to be an independent predictor of PFS and OS after immunochemotherapy, and add significantly to the prognostic power of known pre-treatment parameters74–76. Recent data indicate that they can potentially also be used to de-escalate treatment based on the MRD depth of response77. After HSCT, MRD kinetics rather than levels seems to identify patients that are at risk of clinical relapse, long before clinical signs become apparent53,78,79. This is most likely mediated by ongoing GvL activity of donor T lymphocytes and their continuous immunotherapeutic activity, which is highly sensitive to immunomodulation by immune suppression or donor lymphocyte infusions (DLI)80,81.
Summary
after HSCT, MRD kinetics aid in the assessment of response and indicate the level of ongoing GvL-mediated immunotherapeutic activity.
Adverse events and risk of GvHD in CLL
GvL activity in CLL seems to be closely correlated to graft-versus-host disease (GvHD), as patients with chronic GvHD (cGvHD) have a reduced risk of relapse57,82. Accordingly, an increased relapse rate was observed when donor T cells were depleted55,60,68. However, cGvHD remains a significant problem, and is largely responsible for nonrelapse mortality (NRM) rates of up to 23% and affects up to 60% of patients in the large clinical trials summarized in table 2. Apart from its impact on NRM, cGVHD is the major determinant of quality of life after HSCT83,84. The clinical symptoms of cGVHD however decrease over time in many affected patients, and therapeutic immunosuppression could be terminated after one to two years in many patients in the trials summarized in table 2.
Other acute side effects of RIC HSCT in the early transplant phase include nausea, mucositis, and infections. Due to substantial improvements of supportive and anti-infective treatments and the availability of dedicated transplant units, these are considerably easier to manage than in the era of myeloablative HSCT, which is reflected in very low early mortality rates of less than 10% in the first 100 days after HSCT (see table 2).
Summary
HSCT is associated with significant treatment related mortality and morbidity, largely due to chronic GvHD.
Management of relapse after HSCT
Even though HSCT can be curative in up to 40% of patients, a significant proportion still relapses after HSCT. To date, there is no standard treatment or guidelines available for patients who failed HSCT and are unresponsive to post-HSCT immunomodulation based interventions. In a retrospective analysis of 40 patients from the MDACC, median time to HSCT failure was 7 months, and the most common salvage treatment regimens were re-treatment with rituximab- and alemtuzumab-based immunochemotherapy and treatment with thalidomide or lenalidomide and ibrutinib85. This led to a median OS from time of progression of 53 months, indicating that post-HSCT relapses are sensitive to salvage therapy. Interestingly, there were no differences between FCR, alemtuzumab or combination chemotherapy in OS, while the majority of patients that had received ibrutinib were still alive at the time of last follow-up.
Summary
the clinical management of relapse after HSCT is challenging but seems to be sensitive to immunochemotherapy treatment.
Autologous hematopoietic stem cell transplantation
Long before the advent of fludarabine or antibody-based strategies, there was realistic hope that myeloablative therapy followed by autologous stem cell transplantation (autoSCT) might be an effective and potentially curative front-line treatment option for suitable patients with CLL. Since then, several prospective trials have demonstrated that autoSCT can prolong EFS and PFS if used as part of early front-line treatment, but fails to improve OS and lacks the potential to overcome the negative impact of biomarkers that confer resistance to chemotherapy or early relapse86–89. In addition, it is associated with increased risk of late adverse events such as secondary malignancies60,89,90. Therefore, autoSCT does currently not play a role in the treatment of CLL, and patients that have benefited from this approach in the past are also most likely to respond to conventional immunochemotherapy.
Summary
autologous SCT does no longer play a role in the treatment of CLL.
Outcome of poor risk CLL patients in the era of novel substances and treatments
Due to the availability of novel substances and treatment strategies, the standard of care in CLL is changed dramatically. These new approaches include new mAbs, immune modulatory agents, substances interfering with the BCR signaling pathway, and novel cellular therapies. As extensive reviews have been published elsewhere, we will focus on the impact on patients with high risk CLL91–93.
Although representing a ‘passive’ immunotherapy, mAbs display enhanced antitumoural activity by engaging the immune system through increased complement-dependent cytotoxicity and antibody-dependent cellular cytotoxicity. Anti-CD20 mAbs are now integral components of CLL therapy5,12–15, but the new generation antibody ofatumumab seems to be more effective in refractory patients and in patients pretreated with rituximab, yielding a response rate of almost 60%33. Lenalidomide has been demonstrated to have pleiotropic effects on immune cells in both preclinical and clinical studies, primarily by enhancing antitumoural immunity in effector cells94–99. Several clinical trials have demonstrated that lenalidomide as a single agent and in combination with rituximab has activity both in untreated and relapsed/ refractory CLL and in patients with del17p-, but is associated with nonnegligible toxicities such as tumour flare reaction (TFR) and increased risk of opportunistic infections100–107. The combination of lenalidomide with ofatumumab is currently being investigated in relapsed/ refractory CLL.
The most striking results have been observed in recently published prospective trials using the BCR signalling inhibitors ibrutinib and idelalisib: ibrutinib is a BTK inhibitor that primarily blocks BCR associated anti-apoptosis pathways. In addition, it affects BCR- and chemokine-controlled retention and homing of CLL cells in their growth- and survival-supporting lymph node and bone marrow microenvironment108–110. In a phase I/II study in 85 heavily pre-treated patients with relapsed or refractory CLL, the ORR was 71%, with a PFS of 75% and an OS of 83% at 26 months, and response was independent of del17p-111. Toxicities were mild and only a few serious adverse events were observed. Idelalisib is an inhibitor of PI3K110δ, which is also a component of CLL signalling pathways involved in cell survival, clonal expansion and malignant cells retention in lymphoid tissues112,113. In a phase I study conducted on 54 heavily pretreated CLL patients with relapsed/ refractory disease, including patients with del17p- (24%), 81% of patients achieved nodal responses with an overall response rate of 72% and a very acceptable safety profile20. A phase III trial was then initiated on 220 patients with relapsed CLL receiving idelalisib in combination with rituximab versus rituximab plus placebo21. Due to overwhelming efficacy, the study was interrupted after the first interim analysis: the ORR was 81% for the combination therapy versus 13% for rituximab monotherapy while OS at 12 months was 92% versus 80%, and PFS 93% versus 46%. Multiple ongoing randomized and non-randomized trials are now investigating combinations of these substances with ofatumumab, rituximab, bendamustine and other novel agents.
BCL-2 antagonists such as navitoclax and ABT-199 mainly work by triggering apoptosis via targeting the BCL2 family. In a phase I study with 29 patients with relapsed/ refractory CLL, lymphocytosis was reduced by more than 50% in 19 of 21 patients with baseline lymphocytosis, and PR or stabilization of disease was achieved in almost half of the patients, again including patients with del17p- CLL22. Another promising group of agents that have shown efficacy in high risk CLL patients are cyclin dependent kinase (CDK) inhibitors such as flavopiridol. In a recently published phase I study, the combination with cyclophosphamide and rituximab was tolerable and active in high-risk CLL patients without TLS toxicity, confirming previous findings that flavopiridol can overcome the negative impact of del17p-114,115.
However, a recent report indicates that patients can become resistant to ibrutinib therapy because of mutations of drug binding sites within the BCR pathway, and similar resistance mechanisms to other substances are likely are currently under investigation23.
A very exciting new active immunotherapy strategy is chimeric antigen receptor (CAR) T-cell therapy. CAR technology has recently emerged as a novel and promising perspective to specifically target malignant cells with precisely engineered T-cells. It uses the single chain variable fragment from an antibody molecule fused with an internal T-cell signaling domain to form a chimeric antigen receptor, which is then transduced into T cells116. A major advantage of this approach is that it eliminates MHC restriction, enabling the same CAR to be used for several different patients. In a pivotal report, a heavily pre-treated high-risk patient with refractory CLL received autologous T cells that had been modified with CARs directed at CD19, a B-cell surface antigen, resulting in remission induction and lasting tumour control117. Since then, several clinical trials have reported impressive results with anti CD19 CARs, both in CLL and acute lymphoblastic leukaemia117–121. However, it has also become clear that the success of CAR therapy is depending on the inclusion of lympho-reducing conditioning chemotherapy and the choice of CAR design.117,119,120,122. In addition, CAR T-cell therapy can be associated with severe complications such as cytokine release syndrome, a potentially lethal complication, and lasting normal B-cell depletion119,123.
Summary
there are several new agents and immunotherapy approaches that are in clinical trials or recently approved in CLL that demonstrate impressive responses and durable durations of response in high risk patients who might be candidates for transplant.
Discussion: potential for the combination of HSCT and novel therapies
Recent clinical trials have taught us that novel agents are efficacious, very tolerable, and have the ability to abrogate the negative predictive effect of fludarabine-resistance and del17p-. However, similar to targeted treatment of chronic myeloid leukemia with BCR-ABL antagonists, resistance mechanisms are emerging and give reasons for concern about the long-term curative effect of those substances. In addition, one has to keep in mind that the majority of those agents are only available in clinical trials, or once they are approved come at a very high treatment cost and only in selected countries, which makes them inaccessible for the majority of patients in need. Patient numbers with specific mutations are also still small, and it is therefore uncertain if the observations relating to del17p- patients can be extrapolated to p53 abnormalities (i.e. isolated mutations, with/without del17p-), and to other mutations known to confer an adverse prognosis or poor response to treatment. Similarly, the outcome of patients that are relapsing following novel treatments is not known, and one can only speculate how such treatment will influence the biology and chemosensitivity of the relapsed disease.
In the context of CAR therapy, although this is a very promising treatment approach and seems to be highly efficacious in patients that would have otherwise had no further therapeutic option, further studies are needed to fully investigate the clinical use of CAR T-cell therapy and treatment-related toxicities, and its optimal combination with existing treatment approaches. Similarly to novel agents, this is only available in few selected sites and within clinical trials; due to the complexity involved in cell collection, genetic manipulation, application and clinical management this is very likely (and desired) to stay in the hand of dedicated specialized CAR manufacturing and treatment centers.
On the other hand, long-term follow up data from large prospective trials of HSCT reaching almost a decade in a few centers, have lead us to believe that despite being suitable for only selected subgroups of patients and coming at the cost of rather extensive GvHD and reduction of quality of life, HSCT has curative potential in about half of the patients undergoing this procedure. Although there is no prospective data on whether HSCT can change the natural biological course of high-risk CLL, there is some retrospective data using a donor versus no-donor comparison approach which indicates that OS was significantly improved in patients with donor124. This leads us to believe that HSCT does indeed have the potential to correct the natural dismal course of disease of high risk CLL. However, HSCT should always be restricted to patients meeting the EBMT transplant/ highest risk patient criteria (see table 1) and should never be part of first-line treatment in the general patient population.
As there are no direct comparisons between HSCT and novel agents, general evidence-based recommendations are very difficult to make at this point. Instead, we need to understand the limitations of each approach, and carefully weigh the chances and risks of each procedure on a case-to-case basis. In general, the availability of treatments, their expected benefit and side effects, and individual treatment-histories and pre-transplant characteristics as determined by the EBMT risk score need to be taken into consideration. With the data that is available up to now, it seems feasible to consider HSCT in highest risk patients, i.e. patients who are relapsed/ refractory and exhibit p53 abnormalities or del11q-, are suitable for transplant in terms of age, concomitant diseases, have a well-matched donor, and are willing to undergo this procedure. As the success of HSCT is however highly dependent on the remission state of the time of HSCT, it seems very desirable to focus on achieving disease control first. This can be facilitated by novel substances. As they are also well tolerable and show only moderate toxicities, they seem a good option to bridge the time until HSCT, and maybe even to postpone HSCT to a later point in the disease. How these substances should be best combined, if there is the option to completely eliminate the chemotherapy backbone from induction or second-line treatment, and whether they will have an affect on GvL and immunmodulation, is the major focus of ongoing preclinical and clinical studies.
Summary
the judicious choice of which patients merit this approach remains important. HSCT must always be considered in view of other, potentially less toxic therapies which could be offered. The choice of HSCT versus a novel agent is one that must be gauged on a patient by patient basis, and this will change as data mature on the use of novel agents in CLL.
Key points.
HSCT offers the only potentially curative approach to the treatment of CLL but is suitable only for a minority of patients and is associated with significant treatment-related mortality and morbidity
Guidelines suggest that HSCT is indicated in fit CLL patients with a suitable matched donor, del17p-/ TP53 mutations or who have relapsed shortly after chemo-immunotherapy (high-risk patients).
HSCT must always be considered in view of other, potentially less toxic therapies.
Several new agents demonstrate impressive and durable responses in high risk patients who might be candidates for transplant.
The choice of HSCT versus a novel agent is one that must be gauged on a patient by patient basis.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Chiorazzi N, Rai KR, Ferrarini M. Chronic lymphocytic leukemia. The New England journal of medicine. 2005;352:804–15. doi: 10.1056/NEJMra041720. [DOI] [PubMed] [Google Scholar]
- 2.Siegel R, Ma J, Zou Z, Jemal A. Cancer statistics, 2014. CA: a cancer journal for clinicians. 2014;64:9–29. doi: 10.3322/caac.21208. [DOI] [PubMed] [Google Scholar]
- 3.Howlader N, Noone AM, Krapcho M, Neyman N, Aminou R, Altekruse SF, Kosary CL, Ruhl J, Tatalovich Z, Cho H, Mariotto A, Eisner MP, Lewis DR, Chen HS, Feuer EJ, Cronin KA, editors. SEER Cancer Statistics Review, 1975–2009 (Vintage 2009 Populations) National Cancer Institute; Bethesda, MD: Apr, 2012. http://seer.cancer.gov/csr/1975_2009_pops09/, based on November 2011 SEER data submission, posted to the SEER web site. [Google Scholar]
- 4.Tam CS, O’Brien S, Wierda W, et al. Long-term results of the fludarabine, cyclophosphamide, and rituximab regimen as initial therapy of chronic lymphocytic leukemia. Blood. 2008;112:975–80. doi: 10.1182/blood-2008-02-140582. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.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;376:1164–74. doi: 10.1016/S0140-6736(10)61381-5. [DOI] [PubMed] [Google Scholar]
- 6.Döhner H, Stilgenbauer S, Benner A, et al. Genomic Aberrations and Survival in Chronic Lymphocytic Leukemia. New England Journal of Medicine. 2000;343:1910–6. doi: 10.1056/NEJM200012283432602. [DOI] [PubMed] [Google Scholar]
- 7.Zenz T, Eichhorst B, Busch R, et al. TP53 Mutation and Survival in Chronic Lymphocytic Leukemia. Journal of Clinical Oncology. 2010;28:4473–9. doi: 10.1200/JCO.2009.27.8762. [DOI] [PubMed] [Google Scholar]
- 8.Fink AM, Bottcher S, Ritgen M, et al. Prediction of poor outcome in CLL patients following first-line treatment with fludarabine, cyclophosphamide and rituximab. Leukemia. 2013;27:1949–52. doi: 10.1038/leu.2013.190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Shanafelt T. Treatment of older patients with chronic lymphocytic leukemia: key questions and current answers. ASH Education Program Book. 2014;2013:158–67. doi: 10.1182/asheducation-2013.1.158. [DOI] [PubMed] [Google Scholar]
- 10.Hillmen P, Gribben JG, Follows GA, et al. Rituximab Plus Chlorambucil As First-Line Treatment for Chronic Lymphocytic Leukemia: Final Analysis of an Open-Label Phase II Study. Journal of Clinical Oncology. 2014 doi: 10.1200/JCO.2013.49.6547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Foà R, Del Giudice I, Cuneo A, et al. Chlorambucil plus rituximab with or without maintenance rituximab as first-line treatment for elderly chronic lymphocytic leukemia patients. American Journal of Hematology. 2014;89:480–6. doi: 10.1002/ajh.23668. [DOI] [PubMed] [Google Scholar]
- 12.Goede V, Fischer K, Busch R, et al. Obinutuzumab plus chlorambucil in patients with CLL and coexisting conditions. N Engl J Med. 2014;370:1101–10. doi: 10.1056/NEJMoa1313984. [DOI] [PubMed] [Google Scholar]
- 13.Wierda WG, Kipps TJ, Mayer J, et al. Ofatumumab as single-agent CD20 immunotherapy in fludarabine-refractory chronic lymphocytic leukemia. J Clin Oncol. 2010;28:1749–55. doi: 10.1200/JCO.2009.25.3187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Wierda WG, Kipps TJ, Durig J, et al. Chemoimmunotherapy with O-FC in previously untreated patients with chronic lymphocytic leukemia. Blood. 2011;117:6450–8. doi: 10.1182/blood-2010-12-323980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.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;118:5126–9. doi: 10.1182/blood-2011-04-348656. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Byrd JC, Furman RR, Coutre SE, et al. Targeting BTK with Ibrutinib in Relapsed Chronic Lymphocytic Leukemia. New England Journal of Medicine. 2013;369:32–42. doi: 10.1056/NEJMoa1215637. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Jaglowski SM, Jones JA, Flynn JM, et al. A phase 1b/2 study evaluating activity and tolerability of the BTK inhibitor ibrutinib in combination with ofatumumab in patients with chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL) and related diseases. ASCO Meeting Abstracts. 2014;32:7009. [Google Scholar]
- 18.Byrd JC, Brown JR, O’Brien S, et al. Ibrutinib versus Ofatumumab in Previously Treated Chronic Lymphoid Leukemia. New England Journal of Medicine. 2014 doi: 10.1056/NEJMoa1400376. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Burger JA, Keating MJ, Wierda WG, et al. Ibrutinib In Combination With Rituximab (iR) Is Well Tolerated and Induces a High Rate Of Durable Remissions In Patients With High-Risk Chronic Lymphocytic Leukemia (CLL): New, Updated Results Of a Phase II Trial In 40 Patients. ASH Annual Meeting Abstracts. 2013;122:675. [Google Scholar]
- 20.Brown JR, Byrd JC, Coutre SE, et al. Idelalisib, an inhibitor of phosphatidylinositol 3-kinase p110delta, for relapsed/refractory chronic lymphocytic leukemia. Blood. 2014;123:3390–7. doi: 10.1182/blood-2013-11-535047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.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: 10.1056/NEJMoa1315226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Roberts AW, Seymour JF, Brown JR, et al. Substantial Susceptibility of Chronic Lymphocytic Leukemia to BCL2 Inhibition: Results of a Phase I Study of Navitoclax in Patients With Relapsed or Refractory Disease. Journal of Clinical Oncology. 2012;30:488–96. doi: 10.1200/JCO.2011.34.7898. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Woyach JA, Furman RR, Liu T-M, et al. Resistance Mechanisms for the Bruton’s Tyrosine Kinase Inhibitor Ibrutinib. New England Journal of Medicine. 2014;370:2286–94. doi: 10.1056/NEJMoa1400029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Gribben JG, Riches JC. Immunotherapeutic strategies including transplantation: eradication of disease. ASH Education Program Book. 2013;2013:151–7. doi: 10.1182/asheducation-2013.1.151. [DOI] [PubMed] [Google Scholar]
- 25.Kolb H-J. Graft-versus-leukemia effects of transplantation and donor lymphocytes. Blood. 2008;112:4371–83. doi: 10.1182/blood-2008-03-077974. [DOI] [PubMed] [Google Scholar]
- 26.Isobe M, Emanuel BS, Givol D, Oren M, Croce CM. Localization of gene for human p53 tumour antigen to band 17p13. Nature. 1986;320:84–5. doi: 10.1038/320084a0. [DOI] [PubMed] [Google Scholar]
- 27.Zenz T, Kröber 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; 2008 doi: 10.1182/blood-2008-04-154070. [DOI] [PubMed] [Google Scholar]
- 28.Dicker F, Herholz H, Schnittger S, et al. The detection of TP53 mutations in chronic lymphocytic leukemia independently predicts rapid disease progression and is highly correlated with a complex aberrant karyotype. Leukemia. 2008;23:117–24. doi: 10.1038/leu.2008.274. [DOI] [PubMed] [Google Scholar]
- 29.Tam CS, Shanafelt TD, Wierda WG, et al. De novo deletion 17p13.1 chronic lymphocytic leukemia shows significant clinical heterogeneity: the M. D. Anderson and Mayo Clinic experience. Blood. 2009;114:957–64. doi: 10.1182/blood-2009-03-210591. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.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. Journal of Clinical Oncology. 2012;30:3209–16. doi: 10.1200/JCO.2011.39.2688. [DOI] [PubMed] [Google Scholar]
- 31.Woyach JA, Ruppert AS, Heerema NA, et al. Chemoimmunotherapy With Fludarabine and Rituximab Produces Extended Overall Survival and Progression-Free Survival in Chronic Lymphocytic Leukemia: Long-Term Follow-Up of CALGB Study 9712. Journal of Clinical Oncology. 2011;29:1349–55. doi: 10.1200/JCO.2010.31.1811. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Hillmen P, Skotnicki AB, Robak T, et al. Alemtuzumab Compared With Chlorambucil As First-Line Therapy for Chronic Lymphocytic Leukemia. Journal of Clinical Oncology. 2007;25:5616–23. doi: 10.1200/JCO.2007.12.9098. [DOI] [PubMed] [Google Scholar]
- 33.Zent CS, Call TG, Shanafelt TD, et al. Early treatment of high-risk chronic lymphocytic leukemia with alemtuzumab and rituximab. Cancer. 2008;113:2110–8. doi: 10.1002/cncr.23824. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Pettitt AR, Jackson R, Carruthers S, et al. Alemtuzumab in Combination With Methylprednisolone Is a Highly Effective Induction Regimen for Patients With Chronic Lymphocytic Leukemia and Deletion of TP53: Final Results of the National Cancer Research Institute CLL206 Trial. Journal of Clinical Oncology. 2012;30:1647–55. doi: 10.1200/JCO.2011.35.9695. [DOI] [PubMed] [Google Scholar]
- 35.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;118:2062–8. doi: 10.1182/blood-2011-01-329177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.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. Journal of Clinical Oncology. 2009;27:3994–4001. doi: 10.1200/JCO.2008.21.1128. [DOI] [PubMed] [Google Scholar]
- 37.Zent CS, Taylor RP, Lindorfer MA, et al. Chemoimmunotherapy for relapsed/refractory and progressive 17p13-deleted chronic lymphocytic leukemia (CLL) combining pentostatin, alemtuzumab, and low-dose rituximab is effective and tolerable and limits loss of CD20 expression by circulating CLL cells. American Journal of Hematology. 2014;89:757–65. doi: 10.1002/ajh.23737. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Bowen DA, Call TG, Jenkins GD, et al. Methylprednisolone-rituximab is an effective salvage therapy for patients with relapsed chronic lymphocytic leukemia including those with unfavorable cytogenetic features. Leukemia & Lymphoma. 2007;48:2412–7. doi: 10.1080/10428190701724801. [DOI] [PubMed] [Google Scholar]
- 39.Badoux XC, Keating MJ, Wang X, et al. Cyclophosphamide, fludarabine, alemtuzumab, and rituximab as salvage therapy for heavily pretreated patients with chronic lymphocytic leukemia. Blood. 2011;118:2085–93. doi: 10.1182/blood-2011-03-341032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Badoux XC, Keating MJ, Wang X, et al. Fludarabine, cyclophosphamide, and rituximab chemoimmunotherapy is highly effective treatment for relapsed patients with CLL. Blood. 2011;117:3016–24. doi: 10.1182/blood-2010-08-304683. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.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;99:3554–61. doi: 10.1182/blood.v99.10.3554. [DOI] [PubMed] [Google Scholar]
- 42.Tsimberidou AM, Wierda WG, Plunkett W, et al. Phase I-II Study of Oxaliplatin, Fludarabine, Cytarabine, and Rituximab Combination Therapy in Patients With Richter’s Syndrome or Fludarabine-Refractory Chronic Lymphocytic Leukemia. Journal of Clinical Oncology. 2008;26:196–203. doi: 10.1200/JCO.2007.11.8513. [DOI] [PubMed] [Google Scholar]
- 43.Brown JR, Messmer B, Werner L, et al. A phase I study of escalated dose subcutaneous alemtuzumab given weekly with rituximab in relapsed chronic lymphocytic leukemia/small lymphocytic lymphoma. Haematologica. 2013;98:964–70. doi: 10.3324/haematol.2013.086207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Dreger P, Corradini P, Kimby E, et al. Indications for allogeneic stem cell transplantation in chronic lymphocytic leukemia: the EBMT transplant consensus. Leukemia. 2007;21:12–7. doi: 10.1038/sj.leu.2404441. [DOI] [PubMed] [Google Scholar]
- 45.Hallek M, Cheson BD, Catovsky D, et al. Guidelines for the diagnosis and treatment of chronic lymphocytic leukemia: a report from the International Workshop on Chronic Lymphocytic Leukemia updating the National Cancer Institute Working Group 1996 guidelines. Blood. 2008;111:5446–56. doi: 10.1182/blood-2007-06-093906. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Zenz T, Gribben JG, Hallek M, Doehner H, Keating MJ, Stilgenbauer S. Risk categories and refractory CLL in the era of chemoimmunotherapy. Blood. 2012;119:4101–7. doi: 10.1182/blood-2011-11-312421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Rabinowe S, Soiffer R, Gribben J, et al. Autologous and allogeneic bone marrow transplantation for poor prognosis patients with B-cell chronic lymphocytic leukemia. Blood. 1993;82:1366–76. [PubMed] [Google Scholar]
- 48.Khouri IF, Keating MJ, Vriesendorp HM, et al. Autologous and allogeneic bone marrow transplantation for chronic lymphocytic leukemia: preliminary results. Journal of Clinical Oncology. 1994;12:748–58. doi: 10.1200/JCO.1994.12.4.748. [DOI] [PubMed] [Google Scholar]
- 49.Khouri IF, Keating M, Körbling M, et al. Transplant-lite: induction of graft-versus-malignancy using fludarabine-based nonablative chemotherapy and allogeneic blood progenitor-cell transplantation as treatment for lymphoid malignancies. Journal of Clinical Oncology. 1998;16:2817–24. doi: 10.1200/JCO.1998.16.8.2817. [DOI] [PubMed] [Google Scholar]
- 50.Slavin S, Nagler A, Naparstek E, et al. Nonmyeloablative Stem Cell Transplantation and Cell Therapy as an Alternative to Conventional Bone Marrow Transplantation With Lethal Cytoreduction for the Treatment of Malignant and Nonmalignant Hematologic Diseases. Blood. 1998;91:756–63. [PubMed] [Google Scholar]
- 51.McSweeney PA, Niederwieser D, Shizuru JA, et al. Hematopoietic cell transplantation in older patients with hematologic malignancies: replacing high-dose cytotoxic therapy with graft-versus-tumor effects. Blood. 2001;97:3390–400. doi: 10.1182/blood.v97.11.3390. [DOI] [PubMed] [Google Scholar]
- 52.Passweg JR, Baldomero H, Peters C, et al. Hematopoietic SCT in Europe: data and trends in 2012 with special consideration of pediatric transplantation. Bone Marrow Transplant. 2014;49:744–50. doi: 10.1038/bmt.2014.55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Dreger P, Doehner 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;116:2438–47. doi: 10.1182/blood-2010-03-275420. [DOI] [PubMed] [Google Scholar]
- 54.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. Journal of Clinical Oncology. 2008;26:4912–20. doi: 10.1200/JCO.2007.15.4757. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Schetelig J, van Biezen A, Brand R, et al. Allogeneic hematopoietic cell transplantation for chronic lymphocytic leukemia with 17p- deletion: A retrospective EBMT analysis. Journal of Clinical Oncology. 2008;26:5094–100. doi: 10.1200/JCO.2008.16.2982. [DOI] [PubMed] [Google Scholar]
- 56.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;117:4679–88. doi: 10.1002/cncr.26091. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Brown JR, Kim HT, Armand P, et al. Long-term follow-up of reduced-intensity allogeneic stem cell transplantation for chronic lymphocytic leukemia: prognostic model to predict outcome. Leukemia. 2013;27:362–9. doi: 10.1038/leu.2012.228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Moreno C, Villamor N, Colomer D, et al. Allogeneic Stem-Cell Transplantation May Overcome the Adverse Prognosis of Unmutated VH Gene in Patients With Chronic Lymphocytic Leukemia. Journal of Clinical Oncology. 2005;23:3433–8. doi: 10.1200/JCO.2005.04.531. [DOI] [PubMed] [Google Scholar]
- 59.Dreger P, Brand R, Hansz J, et al. Treatment-related mortality and graft-versus-leukemia activity after allogeneic stem cell transplantation for chronic lymphocytic leukemia using intensity-reduced conditioning. Leukemia. 2003;17:841–8. doi: 10.1038/sj.leu.2402905. [DOI] [PubMed] [Google Scholar]
- 60.Gribben JG, Zahrieh D, Stephans K, et al. Autologous and allogeneic stem cell transplantations for poor-risk chronic lymphocytic leukemia. Blood. 2005;106:4389–96. doi: 10.1182/blood-2005-05-1778. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Schetelig J, Thiede C, Bornhäuser M, et al. Evidence of a Graft-Versus-Leukemia Effect in Chronic Lymphocytic Leukemia After Reduced-Intensity Conditioning and Allogeneic Stem-Cell Transplantation: The Cooperative German Transplant Study Group. Journal of Clinical Oncology. 2003;21:2747–53. doi: 10.1200/JCO.2003.12.011. [DOI] [PubMed] [Google Scholar]
- 62.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;121:3284–8. doi: 10.1182/blood-2012-11-469627. [DOI] [PubMed] [Google Scholar]
- 63.Wang L, Lawrence MS, Wan Y, et al. SF3B1 and Other Novel Cancer Genes in Chronic Lymphocytic Leukemia. New England Journal of Medicine. 2011;365:2497–506. doi: 10.1056/NEJMoa1109016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Quesada V, Conde L, Villamor N, et al. Exome sequencing identifies recurrent mutations of the splicing factor SF3B1 gene in chronic lymphocytic leukemia. Nat Genet. 2012;44:47–52. doi: 10.1038/ng.1032. [DOI] [PubMed] [Google Scholar]
- 65.Stilgenbauer S, Schnaiter A, Paschka P, et al. Gene mutations and treatment outcome in chronic lymphocytic leukemia: results from the CLL8 trial. Blood. 2014;123:3247–54. doi: 10.1182/blood-2014-01-546150. [DOI] [PubMed] [Google Scholar]
- 66.Baliakas P, Hadzidimitriou A, Sutton LA, et al. Recurrent mutations refine prognosis in chronic lymphocytic leukemia. Leukemia. 2014 doi: 10.1038/leu.2014.196. [DOI] [PubMed] [Google Scholar]
- 67.Michallet M, Socié G, Mohty M, et al. Rituximab, fludarabine, and total body irradiation as conditioning regimen before allogeneic hematopoietic stem cell transplantation for advanced chronic lymphocytic leukemia: Long-term prospective multicenter study. Experimental Hematology. 2013;41:127–33. doi: 10.1016/j.exphem.2012.10.008. [DOI] [PubMed] [Google Scholar]
- 68.Michallet M, Sobh M, Milligan D, et al. The impact of HLA matching on long-term transplant outcome after allogeneic hematopoietic stem cell transplantation for CLL: a retrospective study from the EBMT registry. Leukemia. 2010;24:1725–31. doi: 10.1038/leu.2010.165. [DOI] [PubMed] [Google Scholar]
- 69.Tsimberidou AM, Wierda WG, Wen S, et al. Phase I-II Clinical Trial of Oxaliplatin, Fludarabine, Cytarabine, and Rituximab Therapy in Aggressive Relapsed/Refractory Chronic Lymphocytic Leukemia or Richter Syndrome. Clinical Lymphoma Myeloma and Leukemia. 2013;13:568–74. doi: 10.1016/j.clml.2013.03.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Stilgenbauer S, Cymbalista F, Leblond V, et al. Subcutaneous Alemtuzumab Combined with Oral Dexamethasone, Followed by Alemtuzumab Maintenance or Allo-SCT In CLL with 17p- or Refractory to Fludarabine - Interim Analysis of the CLL2O Trial of the GCLLSG and FCGCLL/MW. ASH Annual Meeting Abstracts. 2010;116:920. [Google Scholar]
- 71.Krejci M, Doubek M, Brychtova Y, et al. Fludarabine with cytarabine followed by reduced-intensity conditioning and allogeneic hematopoietic stem cell transplantation in patients with poor-risk chronic lymphocytic leukemia. Annals of Hematology. 2013;92:249–54. doi: 10.1007/s00277-012-1579-y. [DOI] [PubMed] [Google Scholar]
- 72.Gratwohl A, Stern M, Brand R, et al. Risk score for outcome after allogeneic hematopoietic stem cell transplantation. Cancer. 2009;115:4715–26. doi: 10.1002/cncr.24531. [DOI] [PubMed] [Google Scholar]
- 73.Boettcher S, Ritgen M, Dreger P. Allogeneic stem cell transplantation for chronic lymphocytic leukemia: Lessons to be learned from minimal residual disease studies. Blood Reviews. 2011;25:91–6. doi: 10.1016/j.blre.2011.01.001. [DOI] [PubMed] [Google Scholar]
- 74.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. Journal of Clinical Oncology. 2005;23:2971–9. doi: 10.1200/JCO.2005.04.021. [DOI] [PubMed] [Google Scholar]
- 75.Boettcher S, Ritgen M, Fischer K, et al. Minimal Residual Disease Quantification Is an Independent Predictor of Progression-Free and Overall Survival in Chronic Lymphocytic Leukemia: A Multivariate Analysis From the Randomized GCLLSG CLL8 Trial. Journal of Clinical Oncology. 2012;30:980–8. doi: 10.1200/JCO.2011.36.9348. [DOI] [PubMed] [Google Scholar]
- 76.Santacruz R, Villamor N, Aymerich M, et al. The prognostic impact of minimal residual disease in patients with chronic lymphocytic leukemia requiring first-line therapy. Haematologica. 2014;99:873–80. doi: 10.3324/haematol.2013.099796. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Strati P, Keating MJ, O’Brien SM, et al. Eradication of bone marrow minimal residual disease may prompt early treatment discontinuation in CLL. Blood. 2014;123:3727–32. doi: 10.1182/blood-2013-11-538116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Farina L, Carniti C, Dodero A, et al. Qualitative and quantitative polymerase chain reaction monitoring of minimal residual disease in relapsed chronic lymphocytic leukemia: early assessment can predict long-term outcome after reduced intensity allogeneic transplantation. Haematologica. 2009;94:654–62. doi: 10.3324/haematol.2008.000273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Logan AC, Zhang B, Narasimhan B, et al. Minimal residual disease quantification using consensus primers and high-throughput IGH sequencing predicts post-transplant relapse in chronic lymphocytic leukemia. Leukemia. 2013;27:1659–65. doi: 10.1038/leu.2013.52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Richardson SE, Khan I, Rawstron A, et al. Risk-stratified adoptive cellular therapy following allogeneic hematopoietic stem cell transplantation for advanced chronic lymphocytic leukaemia. British Journal of Haematology. 2013;160:640–8. doi: 10.1111/bjh.12197. [DOI] [PubMed] [Google Scholar]
- 81.Machaczka M, Johansson J-E, Remberger M, et al. Allogeneic hematopoietic stem cell transplant with reduced-intensity conditioning for chronic lymphocytic leukemia in Sweden: does donor T-cell engraftment 3 months after transplant predict survival? Leukemia & Lymphoma. 2012;53:1699–705. doi: 10.3109/10428194.2012.666661. [DOI] [PubMed] [Google Scholar]
- 82.Toze CL, Galal A, Barnett MJ, et al. Myeloablative allografting for chronic lymphocytic leukemia: evidence for a potent graft-versus-leukemia effect associated with graft-versus-host disease. Bone Marrow Transplant. 2005;36:825–30. doi: 10.1038/sj.bmt.1705130. [DOI] [PubMed] [Google Scholar]
- 83.Dreger P. The Evolving Role of Stem Cell Transplantation in Chronic Lymphocytic Leukemia. Hematology/Oncology Clinics of North America. 2013;27:355–69. doi: 10.1016/j.hoc.2013.01.007. [DOI] [PubMed] [Google Scholar]
- 84.Pidala J, Kurland B, Chai X, et al. Patient-reported quality of life is associated with severity of chronic graft-versus-host disease as measured by NIH criteria: report on baseline data from the Chronic GVHD Consortium. Blood. 2009;117:4651–7. doi: 10.1182/blood-2010-11-319509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Benjamini O, Rozovski U, Jain P, et al. Outcome Of Chronic Lymphocytic Leukemia (CLL) Patients That Failed Allogeneic Stem Cell Transplantation. Blood - ASH Annual Meeting Abstracts. 2013;122:2880. [Google Scholar]
- 86.Michallet M, Dreger P, Sutton L, et al. Autologous hematopoietic stem cell transplantation in chronic lymphocytic leukemia: results of European intergroup randomized trial comparing autografting versus observation. Blood. 2010;117:1516–21. doi: 10.1182/blood-2010-09-308775. [DOI] [PubMed] [Google Scholar]
- 87.Sutton L, Chevret S, Tournilhac O, et al. Autologous stem cell transplantation as a first-line treatment strategy for chronic lymphocytic leukemia: a multicenter, randomized, controlled trial from the SFGM-TC and GFLLC. Blood. 2011;117:6109–19. doi: 10.1182/blood-2010-11-317073. [DOI] [PubMed] [Google Scholar]
- 88.Brion A, Mahe B, Kolb B, et al. Autologous transplantation in CLL patients with B and C Binet stages: final results of the prospective randomized GOELAMS LLC 98 trial. Bone Marrow Transplant. 2012;47:542–8. doi: 10.1038/bmt.2011.117. [DOI] [PubMed] [Google Scholar]
- 89.Dreger P, Doehner H, McClanahan F, et al. Early autologous stem cell transplantation for chronic lymphocytic leukemia: long-term follow-up of the German CLL Study Group CLL3 trial. Blood. 2012;119:4851–9. doi: 10.1182/blood-2011-09-378505. [DOI] [PubMed] [Google Scholar]
- 90.Milligan DW, Kochethu G, Dearden C, et al. High incidence of myelodysplasia and secondary leukaemia in the UK Medical Research Council Pilot of autografting in chronic lymphocytic leukaemia. British Journal of Haematology. 2006;133:173–5. doi: 10.1111/j.1365-2141.2006.05982.x. [DOI] [PubMed] [Google Scholar]
- 91.Hallek M. Signaling the end of chronic lymphocytic leukemia: new frontline treatment strategies. Blood. 2013;122:3723–34. doi: 10.1182/blood-2013-05-498287. [DOI] [PubMed] [Google Scholar]
- 92.Jones JA, Byrd JC. How will B-cell-receptor-targeted therapies change future CLL therapy? Blood. 2014;123:1455–60. doi: 10.1182/blood-2013-09-453092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Davila M, Bouhassira DG, Park J, et al. Chimeric antigen receptors for the adoptive T cell therapy of hematologic malignancies. Int J Hematol. 2014;99:361–71. doi: 10.1007/s12185-013-1479-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Ramsay AG, Johnson AJ, Lee AM, et al. Chronic lymphocytic leukemia T cells show impaired immunological synapse formation that can be reversed with an immunomodulating drug. The Journal of clinical investigation. 2008;118:2427–37. doi: 10.1172/JCI35017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Shanafelt TD, Ramsay AG, Zent CS, et al. Long-term repair of T-cell synapse activity in a phase II trial of chemoimmunotherapy followed by lenalidomide consolidation in previously untreated chronic lymphocytic leukemia (CLL) Blood. 2013;121:4137–41. doi: 10.1182/blood-2012-12-470005. [DOI] [PubMed] [Google Scholar]
- 96.Ramsay AG, Clear AJ, Fatah R, Gribben JG. Multiple inhibitory ligands induce impaired T-cell immunologic synapse function in chronic lymphocytic leukemia that can be blocked with lenalidomide: establishing a reversible immune evasion mechanism in human cancer. Blood. 2012;120:1412–21. doi: 10.1182/blood-2012-02-411678. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Ramsay AG, Evans R, Kiaii S, Svensson L, Hogg N, Gribben JG. Chronic lymphocytic leukemia cells induce defective LFA-1-directed T-cell motility by altering Rho GTPase signaling that is reversible with lenalidomide. Blood. 2013;121:2704–14. doi: 10.1182/blood-2012-08-448332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Aue G, Njuguna N, Tian X, et al. Lenalidomide-induced upregulation of CD80 on tumor cells correlates with T-cell activation, the rapid onset of a cytokine release syndrome and leukemic cell clearance in chronic lymphocytic leukemia. Haematologica. 2009;94:1266–73. doi: 10.3324/haematol.2009.005835. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Lapalombella R, Andritsos L, Liu Q, et al. Lenalidomide treatment promotes CD154 expression on CLL cells and enhances production of antibodies by normal B cells through a PI3-kinase-dependent pathway. Blood. 2010;115:2619–29. doi: 10.1182/blood-2009-09-242438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.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;24:5343–9. doi: 10.1200/JCO.2005.05.0401. [DOI] [PubMed] [Google Scholar]
- 101.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;27:5404–9. doi: 10.1200/JCO.2008.21.1169. [DOI] [PubMed] [Google Scholar]
- 102.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;31:584–91. doi: 10.1200/JCO.2012.42.8623. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Andritsos LA, Johnson AJ, Lozanski G, et al. Higher doses of lenalidomide are associated with unacceptable toxicity including life-threatening tumor flare in patients with chronic lymphocytic leukemia. J Clin Oncol. 2008;26:2519–25. doi: 10.1200/JCO.2007.13.9709. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.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;111:5291–7. doi: 10.1182/blood-2007-12-130120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Chen CI, Bergsagel PL, Paul H, et al. Single-agent lenalidomide in the treatment of previously untreated chronic lymphocytic leukemia. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2011;29:1175–81. doi: 10.1200/JCO.2010.29.8133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Badoux XC, Keating MJ, Wen S, et al. Lenalidomide as initial therapy of elderly patients with chronic lymphocytic leukemia. Blood. 2011;118:3489–98. doi: 10.1182/blood-2011-03-339077. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Strati P, Keating MJ, Wierda WG, et al. Lenalidomide induces long-lasting responses in elderly patients with chronic lymphocytic leukemia. Blood. 2013;122:734–7. doi: 10.1182/blood-2013-04-495341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Ponader S, Chen SS, Buggy JJ, et al. The Bruton tyrosine kinase inhibitor PCI-32765 thwarts chronic lymphocytic leukemia cell survival and tissue homing in vitro and in vivo. Blood. 2012;119:1182–9. doi: 10.1182/blood-2011-10-386417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Herman SE, Gordon AL, Hertlein E, et al. Bruton tyrosine kinase represents a promising therapeutic target for treatment of chronic lymphocytic leukemia and is effectively targeted by PCI-32765. Blood. 2011;117:6287–96. doi: 10.1182/blood-2011-01-328484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.de Rooij MF, Kuil A, Geest CR, et al. The clinically active BTK inhibitor PCI-32765 targets B-cell receptor- and chemokine-controlled adhesion and migration in chronic lymphocytic leukemia. Blood. 2012;119:2590–4. doi: 10.1182/blood-2011-11-390989. [DOI] [PubMed] [Google Scholar]
- 111.Byrd JC, Furman RR, Coutre SE, et al. Targeting BTK with ibrutinib in relapsed chronic lymphocytic leukemia. The New England journal of medicine. 2013;369:32–42. doi: 10.1056/NEJMoa1215637. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.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;118:3603–12. doi: 10.1182/blood-2011-05-352492. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Herman SE, Lapalombella R, Gordon AL, et al. The role of phosphatidylinositol 3-kinase-delta in the immunomodulatory effects of lenalidomide in chronic lymphocytic leukemia. Blood. 2011;117:4323–7. doi: 10.1182/blood-2010-11-315705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Stephens DM, Ruppert AS, Maddocks K, et al. Cyclophosphamide, alvocidib (flavopiridol), and rituximab, a novel feasible chemoimmunotherapy regimen for patients with high-risk chronic lymphocytic leukemia. Leukemia Research. 2013;37:1195–9. doi: 10.1016/j.leukres.2013.06.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Woyach JA, Lozanski G, Ruppert AS, et al. Outcome of patients with relapsed or refractory chronic lymphocytic leukemia treated with flavopiridol: impact of genetic features. Leukemia. 2012;26:1442–4. doi: 10.1038/leu.2011.375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.June CH, Blazar BR, Riley JL. Engineering lymphocyte subsets: tools, trials and tribulations. Nat Rev Immunol. 2009;9:704–16. doi: 10.1038/nri2635. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Porter DL, Levine BL, Kalos M, Bagg A, June CH. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N Engl J Med. 2011;365:725–33. doi: 10.1056/NEJMoa1103849. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.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;119:2709–20. doi: 10.1182/blood-2011-10-384388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Brentjens RJ, Riviere 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;118:4817–28. doi: 10.1182/blood-2011-04-348540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Kalos M, Levine BL, Porter DL, et al. T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Sci Transl Med. 2011;3:95ra73. doi: 10.1126/scitranslmed.3002842. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Grupp SA, Kalos M, Barrett D, et al. Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N Engl J Med. 2013;368:1509–18. doi: 10.1056/NEJMoa1215134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Kochenderfer JN, Dudley ME, Carpenter RO, et al. Donor-derived CD19-targeted T cells cause regression of malignancy persisting after allogeneic hematopoietic stem cell transplantation. Blood. 2013;122:4129–39. doi: 10.1182/blood-2013-08-519413. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Xu XJ, Tang YM. Cytokine release syndrome in cancer immunotherapy with chimeric antigen receptor engineered T cells. Cancer letters. 2014;343:172–8. doi: 10.1016/j.canlet.2013.10.004. [DOI] [PubMed] [Google Scholar]
- 124.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. Annals of Oncology. 2014;25:200–6. doi: 10.1093/annonc/mdt511. [DOI] [PubMed] [Google Scholar]