Chronic lymphocytic leukemia (CLL) represents the most prevalent adult leukemia. Treatment with chemotherapy over the past 3 decades has been palliative. The introduction of therapeutic antibodies has increased the number of treatment options for this disease. Despite this increase, our true understanding of the mechanism of action of antibody therapy in CLL remains limited. Rituximab, a CD20 antibody, is currently widely used in combination-based strategies for both previously untreated symptomatic CLL and as salvage therapy. Recent data suggest that the addition of rituximab to fludarabine with or without cyclophosphamide prolongs survival in younger patients with CLL. Other improved CD20 antibodies with promising clinical activity, including ofatumumab and GA-101, are coming forward. Alemtuzumab, a CD52 antibody, likewise has demonstrated benefit in both symptomatic, previously untreated CLL and in patients with relapsed disease but has less selectivity. Development of other therapeutic antibodies targeting alternative B-cell–specific antigens in CLL has been less successful, although many promising candidate antibodies and/or small modular immune pharmaceuticals (SMIPs) are coming forward. In addition, recent efforts to combine currently applied therapeutic antibodies with other biologic and targeted therapies with efficacy in CLL offers the potential to move toward alternative non–chemotherapy-based treatment approaches.

Chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL) is the most common leukemia, with an incidence rate of 2 to 6 cases per 100 000 people per year.1  The median survival is highly variable with some patients exhibiting an indolent natural history, whereas others develop aggressive disease with a survival of less than 2 to 3 years. Genomic features such as immunoglobulin herpesvirus (IGHV) mutational status, β2-microglobulin, ZAP70 expression, interphase cytogenetics, and complex karyotype on metaphase cytogenetics, recently reviewed by Zenz et al,2  provide further differentiation of disease prognosis. Because there is no proven survival benefit with early treatment, CLL patients are observed until symptoms appear.

Therapy for CLL has evolved significantly from 1970 when alkylator-based therapy such as chlorambucil or cyclophosphamide was used. Late 1990 saw multiple randomized phase 3 studies comparing fludarabine to alkylator-based therapy and demonstrating improved response and progression-free survival (PFS), as recently reviewed in Ricci et al.3  Success prompted phase 3 studies combining fludarabine with cyclophosphamide, in which further improved response and PFS were noted.3  Treatment intensification in CLL from alkylator to fludarabine and cyclophosphamide combinations resulted in increased cellular immune suppression and myelosuppression. In addition, chemotherapy intensification did not greatly improve treatment outcomes in patients with high-risk genomic features, such as those with IGHV unmutated disease, del(17p13.1), and p53 mutations.2  These patients all display poorer outcomes, with markedly reduced survivals compared with patients with normal genomic features or good-risk features, as recently reviewed by Zenz et al.4  Of all prognostic factors examined in CLL, patients with mutated or deleted p53 respond very poorly to standard therapies that mainly act through mechanisms relying on an intact p53 pathway. Identifying therapies that circumvent p53 is therefore a priority for the treatment of this high-risk population. Attempts to intensify chemotherapy beyond fludarabine/alkylator-based combinations have been pursued with enhanced toxicity but little evidence of clinical benefit. As with many other types of cancers, treatment outcomes of CLL patients with chemotherapy-based approaches reached a plateau with no improvements in survival or hints of cure in even a subset of patients. This review will focus on how the clinical application of therapeutic monoclonal antibodies more than the past decade has impacted the therapeutic approach to CLL and point to potential opportunities in the future with other targeted therapies currently being explored.

History of monoclonal antibodies in B-cell malignancies

Monoclonal antibodies have a fixed effector cell binding region (Fc) and a variable region with affinity toward a specific antigen. Antibodies can mediate cytotoxicity toward tumor cells via both direct and indirect mechanisms based upon the target. Direct cytotoxicity of tumor cells can occur though transmembrane signaling, and recruitment of effector cells (natural killer [NK] cells, macrophages, neutrophils) that mediate antibody-dependent cell cytotoxicity (ADCC) and complement that mediates complement-dependent cytotoxicity (CDC). Indirect cytotoxicity can occur by interfering with both the interaction of a tumor cell with the microenvironment-generated survival signal and with its binding to soluble factors that enhance tumor cell survival. Given the specificity of antibodies for a single antigen and the multiple mechanisms by which they can mediate cytotoxicity, antibody-based cancer therapy was seen as a potential “silver bullet” therapy for patients with CLL, particularly if the antigen is selectively expressed on B cells. Numerous target antigens offered the opportunity to selectively target B cells, including CD19, CD37, CD20, and idiotype. Murine antibodies derived from mouse plasma cell hybridoma cells directed toward these targets were the first-generation agents evaluated in multiple clinical studies from 1980. These studies were impaired by production issues that limited antibody supply, diminished antibody activity toward the tumor cell, and development of human antibody–mouse antibody reactions with repeated administration. As a consequence, very modest activity with essentially all murine antibody-based treatments was observed, limiting the development of this modality. Technologic advances allowing engineering of mouse-derived antibodies including a minimal mouse component of the variable complementarity-determining region in the final product (chimeric or humanized) represented a major advance for this modality. In general, chimeric and humanized therapeutic antibodies directed toward human B-cell antigens mediate improved ADCC and CDC compared with their murine counterparts. In addition, chimeric and humanized antibodies generally lack human anti–mouse antibody even on repeated administration. Concurrent with advances in chimeric and humanization technologies were improvements in the ability to produce larger amounts of antibodies. These advances fostered the rebirth of antibody-based therapeutics, impacting treatment of many diseases including CLL. This review will summarize evaluations of antibody and peptide therapies that directly target CLL cells that are either approved or under clinical investigation at this time (Table 1).

Table 1

Newer monoclonal antibodies in clinical development

AgentTargetHumanized/chimericDirect cell deathADCCCDCDevelopment status
MDX-1342 CD19 Humanized Yes Yes No Phase 1 
XmAb5574 CD19 Humanized Modest Yes No Phase 1 
Ofatumumab CD20 Humanized Yes Yes Yes Phase 3 
GA-101 CD20 Humanized Yes Yes Modest Phase 2 
PRO131921 CD20 Humanized Yes Yes Yes Phase 1/2 
Veltuzumab CD20 Humanized Yes Yes Yes Phase 1/2 
LFB-R603 CD20 Chimeric Yes Yes Yes Phase 1 
Lumiliximab CD23 Primatized Yes Yes Yes Phase 3 
TRU-016 CD37 Humanized Yes Yes No Phase 1 
SGN40 CD40 Humanized Yes Yes No Phase 1/2 
HCD122 CD40 Humanized No Yes No Phase 1 
MDX-1411 CD70 Humanized No Yes No Phase 1 
Milatuzumab CD74 Humanized Yes No No Phase 1/2 
Ipilimumab CTLA-4 Humanized Yes No No Phase 2 
Bevacizumab VEGF Humanized No No No Phase 2 
AgentTargetHumanized/chimericDirect cell deathADCCCDCDevelopment status
MDX-1342 CD19 Humanized Yes Yes No Phase 1 
XmAb5574 CD19 Humanized Modest Yes No Phase 1 
Ofatumumab CD20 Humanized Yes Yes Yes Phase 3 
GA-101 CD20 Humanized Yes Yes Modest Phase 2 
PRO131921 CD20 Humanized Yes Yes Yes Phase 1/2 
Veltuzumab CD20 Humanized Yes Yes Yes Phase 1/2 
LFB-R603 CD20 Chimeric Yes Yes Yes Phase 1 
Lumiliximab CD23 Primatized Yes Yes Yes Phase 3 
TRU-016 CD37 Humanized Yes Yes No Phase 1 
SGN40 CD40 Humanized Yes Yes No Phase 1/2 
HCD122 CD40 Humanized No Yes No Phase 1 
MDX-1411 CD70 Humanized No Yes No Phase 1 
Milatuzumab CD74 Humanized Yes No No Phase 1/2 
Ipilimumab CTLA-4 Humanized Yes No No Phase 2 
Bevacizumab VEGF Humanized No No No Phase 2 

ADCC indicates antibody-dependent cell-mediated cytotoxicity; and CDC, complement-dependent cytotoxicity.

Rituximab target and mechanism of action

Rituximab is a chimeric murine/human antibody directed against CD20. CD20 is expressed relatively selectively on B cells from the pre–B-cell stage until postgerminal cells differentiate to become plasma cells. CD20 knockout mice demonstrate normal B-cell development and function, but CD19-induced calcium responses and B-cell receptor signaling are significantly altered.5  Unlike other antigens, CD20 is neither shed nor internalized in resting normal B cells.6  These data support CD20 as an ideal target for antibody-based therapy in mature B-cell malignancies.

Rituximab was the first approved therapeutic antibody for the treatment of cancer. Not surprisingly, the majority of mechanism of action studies of therapeutically used antibodies come from preclinical studies with rituximab. As with most immunoglobulin G1 (IgG1) therapeutic antibodies, rituximab can mediate CDC, ADCC, and direct apoptosis with a cross-linking antibody (Figure 1).7  Extensive investigation of each mechanism has been pursued in lymphoma and CLL. Although CDC is relevant to rituximab-mediated cytotoxicity in some B-cell lines, CLL cells express dim CD20 and only a small subset of cells are susceptible to CDC by rituximab.8,9  Very elegant preclinical in vitro and in vivo studies have demonstrated that CLL cells are prone to CD20-shaving after treatment, and this diminishes the ability of CDC to occur.10-12  Attempts to abrogate the phenomenon of shaving have been undertaken by administering very low doses of rituximab on a thrice weekly schedule with very modest clinical activity.11,13  Thus, although a very strong hypothesis with supportive preclinical data support shaving as a reason for modest rituximab clinical activity when administered at higher doses, it is not clear what CDC contributes to tumor elimination. Monocytes mediate antibody-dependent cellular phagocytosis (ADCP),14  and NK cells mediate ADCC14  against rituximab-labeled CLL cells in vitro. Despite this, the function of both monocytes15,16  and NK cells17,18  to mediate ADCP and ADCC, respectively, are certainly compromised in vitro and are likely compromised in vivo in CLL patients. The mechanism of innate immune ADCP and ADCC is not known but may be from the increased T regulatory cells documented in CLL patients with active disease.19  A recent study demonstrated that T regulatory cells can dramatically dampen ADCC mediated toward rituximab-labeled tumor cells.20  With the increased T regulatory cells in CLL patients,19  diminished monocyte15,16  and NK cell17,18  function, the contribution of these cells to tumor elimination is not clear. In addition, single nucleotide polymorphisms of FcγRIIIa and FcγRIIa that enhance ADCC and are associated with improved response to rituximab,21,22  in lymphoma have no impact on rituximab treatment response in CLL.23,24  Strategies directed at either reversing the innate immune dysfunction with immune modulating agents such as interleukin-21,14  TLR agonists,25  or agents that deplete T regulatory cells26  offer the best opportunity to optimize immune cell participation in tumor clearance by rituximab. Finally, several groups have demonstrated that rituximab can mediate both caspase-dependent and -independent apoptosis in vitro (reviewed in Jaglowski and Byrd27 ) and in vivo.28  Apoptosis appears to be the most important mechanism of action in CLL and involves the activation of the p38 mitogen-activated protein kinase, a pathway that requires an intact p53 gene, and caspase 9 cleavage.7-9  Ofatumumab, a second-generation fully humanized anti-CD20 that recognizes a different CD20 epitope than rituximab, has similar ADCC, stronger CDC, and requires cross-linking to induce direct apoptosis similar to riutximab.29,30  GA101 binds with high affinity to the CD20 epitope and, as a result, induction of ADCC is 5 to 100 times greater than with rituximab.31-34  Type II anti-CD20 antibodies such as B1 and GA101 promote direct apoptosis without a cross-linking antibody.35  Other differences between type I (rituximab, ofatumumab) and type II (GA101, B1) anti-CD20 antibodies lie predominately in their ability to redistribute CD20 into plasma membrane lipid rafts.36  Type II anti-CD20 antibodies do not segregate CD20 into lipid rafts and are very effective at activating a caspase-independent, lysosomal-dependent mechanism of death that is dependent upon homotypic adhesion.35  The relevance of this observation in vivo among CLL patients receiving type II CD20 antibody therapy remains unexplored.

Figure 1

Mechanisms of rituximab-mediated cell death. Rituximab coated B cells are killed by at least 4 different mechanisms. (A) Binding of rituximab to CD20 on B cell surface causes activation of the complement cascade, which generates the membrane attack complex (MAC) that can directly induce B-cell lysis by complement-mediated cytotoxicity (CDC). (B) Binding of rituximab allows interaction with NK cells via Fc receptors III (FcRIII), which leads to antibody-dependent cell-mediated cytotoxicity (ADCC). (C) The Fc portion of rituximab and the deposited complement fragments allow for recognition by both FcR and complement receptors on macrophages, which lead to phagocytosis and ADCC. (D) The crosslinking of several molecules of rituximab and CD20 in the lipid raft determine the interaction of these complexes with elements of a signaling pathway involving Src kinases that mediate direct apoptosis.

Figure 1

Mechanisms of rituximab-mediated cell death. Rituximab coated B cells are killed by at least 4 different mechanisms. (A) Binding of rituximab to CD20 on B cell surface causes activation of the complement cascade, which generates the membrane attack complex (MAC) that can directly induce B-cell lysis by complement-mediated cytotoxicity (CDC). (B) Binding of rituximab allows interaction with NK cells via Fc receptors III (FcRIII), which leads to antibody-dependent cell-mediated cytotoxicity (ADCC). (C) The Fc portion of rituximab and the deposited complement fragments allow for recognition by both FcR and complement receptors on macrophages, which lead to phagocytosis and ADCC. (D) The crosslinking of several molecules of rituximab and CD20 in the lipid raft determine the interaction of these complexes with elements of a signaling pathway involving Src kinases that mediate direct apoptosis.

Close modal

Early rituximab studies and single-agent CLL trials

Although the phase 3 pivotal approval study of rituximab in non-Hodgkin lymphomas (NHL) demonstrated promising clinical activity, the response among the 33 patients with SLL was modest, with only 12% of patients achieving a partial response (PR).37  Similarly disappointing results were obtained in several other small studies (reviewed in Jaglowski and Byrd27 ). The very modest response to rituximab compared with follicular lymphoma provided some pause for developing this agent in CLL/SLL. Potential reasons for lower response in CLL included diminished CD20 expression,38  altered innate immune function,17,18  and different pharmacokinetic features37  compared with lymphoma. Two trials performed by our group39  and the M. D. Anderson Cancer Center (MDACC)40  administered either thrice weekly doses or higher doses of rituximab weekly (up to 2250 mg/m2 per dose) to relapsed CLL patients with improved response. Benefit was predominately in the blood and nodal compartment with response duration approaching that achieved in follicular B-NHL trials. These 2 studies established a role for single-agent rituximab in relapsed CLL and encouraged subsequent trials of rituximab as a single agent in previously untreated patients where improved efficacy was observed41  and in combination strategies with chemotherapy.

Rituximab chemoimmunotherapy studies in CLL

Numerous phase 2 studies combining rituximab with other therapies used in CLL have been pursued. Due to reference limitations, those impacting current CLL therapy approaches are outlined below.

A phase 2 study performed by the German CLL Study Group (GCLLSG) of fludarabine and rituximab (FR) in both refractory and previously untreated patients resulted in an overall response rate (ORR) of 87% with a subset achieving complete response (CR).42  CALGB 9712 evaluated fludarabine in combination with rituximab given either concurrently or sequentially. Patients in the concurrent arm experienced more severe hematologic and infusion-related toxicity, but the ORR was 90% with a CR of 47% compared with an ORR of 78% and CR of 28% in the sequential arm.43  An evaluation of outcome based on genetic features demonstrated worse 3 year survival among those patients with del(17p13.1) or del(11q22.3) compared with those with normal cytogenetics or other abnormalities, with 33% and 53% of patients with del(17p13.1) and del(11q22.3) respectively surviving at 3 years compared with 86% of patients with normal cytogenetics.44  A retrospective comparison of outcome of patients on this trial to a similarly designed CALGB study evaluating, in part, fludarabine alone demonstrated chemoimmunotherapy improved PFS and overall survival (OS).45  Long-term follow-up data recently presented shows no increased risk of treatment-related acute myeloid leukemia (AML).24  An Italian phase 2 study of sequential FR confirmed good response rates, with 78% of patients achieving a CR, but only patients who had stable disease (SD) or better with fludarabine remained on the study to receive rituximab.46 

The combination of fludarabine, cyclophosphamide, and rituximab (FCR) has been extensively explored. A single-arm study of 300 previously untreated patients with progressive CLL from the MDACC reported an ORR of 95% with 72% of patients attaining a CR, 10% nodular partial remission (nPR), and 13% a PR.47  The 6-year OS and PFS were 77% and 51%, respectively.47  Toxicity in this study included predominately cytopenias and associated infection. Eight patients developed treatment-related myelodysplasia. A second study of 177 previously treated patients at this same institution was pursued using the same schedule of FCR.48  The results of this study demonstrated a CRR in 25% of patients with an ORR of 73%.48  The median time to progression was 28 months, which varied significantly based upon response. The median time to progression for patients achieving CR, nPR, and PR was 39, 33, and 15 months, respectively. Toxicities observed were predominately grade 3 or 4 neutropenia (81%) and grade 3 or greater infection (16%). Whereas patients with del(11q22.3) appeared to benefit from FCR with loss of the adverse PFS observed in fludarabine-monotherapy studies, based on a retrospective evaluation of patients treated at the MDACC,49  those with IGHV unmutated disease and del(17p13.1) continued to have an inferior outcome with FCR.47 50 

Pentostatin is a nucleoside analog that has been suggested to be less myelotoxic than fludarabine while still active in CLL. This prompted a study in patients with previously treated CLL51  substituting pentostatin for fludarabine. This pentostatin, cyclophosphamide, and rituximab (PCR) regimen had an ORR of 75% with a CR rate of 25%. The major toxicities were infections and myelosuppression. When evaluated in previously untreated patients, 91% had responses with a 41% CR rate. Similar to the study with FCR, patients with del(11q22.3) had similar PFS as those without this aberration,52  again suggesting that cyclophosphamide may be an important addition for del(11q22.3) patients. Infections and myelosuppression were again the predominant toxicities observed.

With the approval of bendamustine for clinical use in newly diagnosed CLL, pilot studies combining this agent with rituximab have recently been reported in previously untreated patients where a 90% ORR and 33% CRR was observed.53  A parallel study in relapsed CLL demonstrated a 76% ORR and 13% CRR.54  Toxicity in both of these studies included myelosuppression and infection. A randomized phase 3 study comparing bendamustine and rituximab to FCR is currently ongoing.

Phase 3 studies with rituximab in CLL

Two phase 3 studies, CLL8 from the GCLLSG and the REACH trial, have been presented and confirm improved response rates and overall survivals with the addition of rituximab. In CLL8, at a median observation time of 37.7 months, the ORR for FCR among the 761 previously untreated patients evaluable for response was 95.1% versus 88.4% for FC, and the CRR was 44.1% compared with 21.8% for FC.55,56  The median PFS was 32.8 months for FC and 51.8 months for FCR (P < .001, hazard ratio [HR] .56), and the OS was longer for those patients who received FCR at 84.1% versus 79.0% in the FC arm (P = .01), with statistically significant differences seen in patients with Binet stages A (P = .09, HR .19) and B (P < .001, HR .45) but not in patients with Binet stage C disease (P = .168, HR 1.4). Patients with del(17p13.1) had particularly poor outcome, and shorter overall survival was seen in the FC arm; likewise, there was no significant improvement in CR, PFS, and OS among the subgroup of patients with del(17p13.1) treated with FCR. A trend toward shorter overall survival in the FCR arm in patients with unmutated IGHV status was observed. Patients with del(11q22.3) again appeared to benefit from the addition of cyclophosphamide, with response rates approaching that of patients without this mutation.57  There was not an increased infection rate observed with the addition of rituximab, and more deaths occurred in the FC arm.55  The REACH trial compared FC to FCR in 552 patients with relapsed/refractory CLL. These patients had received a median one line of previous treatment with the majority consisting of single-agent alkylator therapy; patients who had received combination FC or rituximab were not eligible. Treatment with FCR resulted in an ORR of 70%, versus 58% for FC alone, and the CRRs were 24% versus 13% for FCR and FC, respectively. Observed PFS in the FCR arm was 30.6 months compared with 20.6 months in the FC arm.58  Hematologic toxicities remained the most significant adverse events. Collectively, these 2 phase 3 studies provide justification for the use of rituximab as part of combination chemoimmunotherapy in both newly diagnosed and relapsed CLL. The use of cyclophosphamide in this regimen seems to be important for patients with del(11q22.3) as both the MDA and German CLL study group showed no adverse outcome in this group compared with low risk karyotype whereas CALGB 9712 did. For other genetic groups the benefit of cyclophosphamide is uncertain as there has not been a comparison of FCR to FR. A randomized study (CALGB 10404) is ongoing and seeks to answer whether the combination of FCR has an advantage over FR and, if so, whether the increased risk of secondary AML associated with cyclophosphamide justifies that advantage.

Maintenance rituximab in CLL

The use of maintenance rituximab is common in NHL. Contrasting with this approach, no randomized trials have been performed to determine whether benefit is derived in CLL/SLL from extended maintenance therapy using rituximab. A phase 2 study of 75 previously untreated patients with CLL evaluated the efficacy of rituximab maintenance after treatment with fludarabine for 6 cycles.59  All patients received 4 weekly doses of 375 mg/m2 rituximab after therapy, and then those who were minimal residual disease (MRD)–positive went on to consolidation with 4 monthly cycles of 375 mg/m2 rituximab followed by 12 monthly cycles of 150 mg/m2. MRD-positive patients in CR or PR receiving consolidation had a longer PFS than the patients not receiving consolidation (87% vs 32% at 5 years). A randomized study comparing maintenance rituximab is now underway by the Polish CLL group and until results from this trial are available, this approach should only be applied as part of clinical trials.

Rituximab in the treatment of autoimmune complications

Autoimmune complications of CLL occur in 10% to 25% of patients during their disease course. Autoimmune hemolytic anemia (AIHA) is the most common, followed by immune thrombocytopenia (ITP). These occur both as an intrinsic process associated with the pathogenesis of CLL and as a result of treatment with purine analogues such as fludarabine.60  Rituximab was initially described for the treatment of steroid-refractory pure red cell aplasia or AIHA61 ; subsequently, the successful treatment of 2 patients with CLL who developed red cell aplasia with 375 mg/m2 rituximab weekly for 2 weeks was described.62  A series of 8 patients with CLL and steroid-refractory AIHA was treated with a combination of rituximab and dexamethasone, and all patients achieved a remission of their AIHA, with 5 patients achieving a Coombs-negative status. Retreatment was also found to be successful.63  In a series of 14 patients with CLL and AIHA treated with rituximab monotherapy, all but 2 patients had an increase in their hemoglobin levels after treatment.64  Rituximab is effective in patients with chronic refractory ITP as well and is of interest in CLL specifically as fludarabine-related ITP is not responsive to steroids. Three patients who developed ITP while receiving fludarabine and who did not respond to treatment with steroids or IVIG were treated with weekly rituximab for 4 doses. All patients had rapid and dramatic improvements in their platelet counts, and the response durations were 6 months or greater for all 3 patients.65  While randomized data demonstrating the exact benefit of rituximab in autoimmune complications is lacking, these data provide evidence for its effectiveness. In the author's opinion, rituximab represents one of the more active therapies for the treatment of autoimmune complications of CLL not responding to initial steroid treatment.

Ofatumumab

Ofatumumab is a newly approved, human type I CD20 monoclonal antibody. In vitro, it has been demonstrated to mediate CDC against rituximab-resistant Raji cells and CLL cells with low expression of CD20. It appears to have greater potency in CDC than rituximab, as well as a slower off-rate and more stable CD20 binding.29  In addition, it appears to bind a different epitope of CD20 than rituximab.30  A phase 1/2 study of ofatumumab in relapsed/refractory patients demonstrated that it is generally well tolerated, even at high doses and is active, with an ORR of 50%. Infusion-related adverse events are similar to those reported with rituximab and decrease after the first infusion. Infections were fairly common, occurring in 51% of patients, including one fatal infection.66  A planned interim analysis of the seminal study of 138 patients treated with 8 weekly infusions of ofatumumab followed by 4 monthly infusions over a 24-week period has been reported. The patients included in this study were required to be refractory to at least one fludarabine-containing regimen and either refractory to at least one alemtuzumab-containing regimen (FA-refractory group) or to have bulky lymphadenopathy (> 5 cm) rendering them less suitable for alemtuzumab treatment (BF-refractory group). Response criteria were based on physical examination or hematologic criteria alone, limiting assessment of internal lymphadenopathy. The ORR was 58% for the FA-refractory group and 47% for the BR-refractory group with one CR observed in the BF-refractory group. All other responses were partial. The median duration of response was 7.1 months in the FA-refractory group and 5.6 months in the BF-refractory group with most patients progressing during treatment. The presence of del(17p13.1) in the BF-refractory group was associated with lower response, which was most apparent among patients with bulky lymph nodes. The median PFS was 5.7 months in the FA-refractory group and 5.9 months in the BF-refractory group with median OS of 13.7 months and 15.4 months, respectively. A landmark analysis done at week 12 demonstrated that median OS was significantly longer among responding patients compared with those who did not respond although the meaning of this is uncertain. Infusion-related reactions were seen in 64% of patients in the FA-refractory group and 61% in the BF-refractory group, almost all of which were grade 1 or 2. One hundred eighty-nine infectious events were reported, 74% of which were grade 1 or 2. Thirteen infections with onset during treatment resulted in death, including one reported case of progressive multifocal leukoencephalopathy (PML).67  Although the lack of CT scan monitoring of nodal disease in this study has been criticized, it is unclear that such monitoring adds benefit in terms of predicting PFS to therapy in CLL.68,69  From this study and other data presented, it is unlear that ofatumumab offers relative benefit over the use of high dose rituximab previously reported.39,40  Only a randomized comparative study will be able to discern such a difference.

Combination studies are being done to enhance the therapeutic efficacy of ofatumumab. A study evaluating the combination of ofatumumab with fludarabine and cyclophosphamide (O-FC) was recently presented. Sixty-one previously untreated patients received either 500 mg or 1000 mg ofatumumab combined with fludarabine and cyclophosphamide every 4 weeks for a total of 6 courses. The CRR was 32% for patients who received 500 mg ofatumumab and 50% for those who received 1000 mg. The OR rates were 77% and 73% respectively. The most common grade 3-4 toxicities were infections, reported in 11 patients, and hematologic adverse events, including neutropenia in 29 patients, anemia in 8 patients, and thrombocytopenia in 9 patients. Grade 3-4 hemolytic anemia occurred in 3 patients.70 

Currently, NCCN guidelines provide the recommendation for use of ofatumumab in previously treated CLL patients.71  Clinical trials to define the activity of ofatumuamb in previously untreated CLL and in combination with most forms of chemoimmunotherapy explored with rituximab are ongoing. Determining the actual scientific advance of ofatumumab over rituximab will require randomized phase 3 trials.

GA101

GA101 is a type II glycoengineered humanized CD20 monoclonal antibody that binds CD20 in a completely different orientation than rituximab and over a larger surface area.31  It initiates nonapoptotic cell death via an actin-dependent lysosome-mediated mechanism that is reliant on cell-to-cell contact.32  Depletion of CLL cells in whole blood samples has been demonstrated, and it may be more potent than rituximab at similar concentrations.33,34  GA-101 is internalized less by CLL cells compared with type I CD20 antibodies, making it an ideal agent for investigation. A recently reported phase 1 study in 13 relapsed/refractory CLL patients demonstrated that GA101 is relatively well tolerated, with the most common grade 3-4 toxicity being transient neutropenia in 9 patients. One CRi, 7 PRs, and 3 patients with SD were observed. No clear dose-effect relationship was established.72  GA101 is being evaluated in a phase 2 study as a single agent in relapsed/refractory CLL and in combination with chlorambucil in previously untreated elderly patients. A phase 3 study is currently underway in patients with comorbidity comparing monotherapy with chlorambucil, chlorambucil plus rituximab, and chlorambucil plus GA-101.

Target and mechanism of action

Alemtuzumab (Campath-1H, Genzyme) is a recombinant DNA-derived humanized IgG1 kappa monoclonal antibody that recognizes the cell-surface antigen CD52. CD52 is a 21- to 28-kDa, heavily glycosylated, membrane-anchored glycoprotein, highly expressed on all B and T lymphocytes at most stages of differentiation (except plasma cells), as well as on granulocytes, monocytes, macrophages, eosinophils, NK cells, and dendritic cells.73  The CD52 antigen is also expressed on tumor cells, particularly T-cell prolymphocytic leukemia (T-PLL) as well as CLL, hairy cell leukemia, NHL, and acute lymphoblastic leukemia.74  Despite the frequent use of alemtuzumab in clinical trials, detailed mechanistic studies to elucidate specific pathways of cell killing have been hampered by the lack of cell lines expressing CD52. Thus, the mechanism of action of alemtuzumab remains to be completely clarified. Alemtuzumab can act through immunologic mechanisms, such as CDC75,76  and/or ADCC by virtue of its IgG Fc region.77,78  It has also been shown that alemtuzumab can induce direct CLL cell death through a membrane raft-dependent mechanism.79  Unlike rituximab, alemtuzumab has been shown to induce cell death in vitro in CLL cells through a mechanism that is independent of p53 status and caspase activation.79  Moreover several in vivo clinical studies80-82  report that alemtuzumab therapy is effective in subgroup of patients with high-risk cytogenetic markers (such as del(17p13.1), which points to its unique mechanism of action.

Alemtuzumab single-agent activity

The dosing schedule of alemtuzumab was developed empirically using primarily clinical response as a surrogate end point in initial phase 1 studies. The intravenous (IV) dosing schedule currently used as the standard regimen for alemtuzumab therapy comprises a 2-hour IV infusion at a starting dose of 3 mg on day 1, 10 mg on day 2, and 30 mg 3 times weekly for a total of 8 to 12 weeks.

The effectiveness of single-agent alemtuzumab in refractory/relapsed CLL patients has been demonstrated (reviewed in Alinari et al83 ). In this setting, alemtuzumab produced an ORR of 33% to 54%. In the majority of these studies, antitumor effects of alemtuzumab were more significant in blood and bone marrow than in lymph nodes (especially if larger than 5 cm). The reason for this differential response is not clear, although it has been postulated to be related to poor bioavailability of the drug in bulky sites causing a low saturation of the binding sites on the neoplastic cells' surface. Moreover, there may be variability in the immune effector mechanisms in lymph nodes compared with other sites.

Alemtuzumab was initially approved in 2001 as a consequence of the pivotal CAM 211 phase 3 study, in which 93 patients with relapsed or refractory CLL who had failed prior therapy with fludarabine and an alkylating agent were treated with stepped-up dosing followed by 30 mg 3 times weekly for a total of 12 weeks.84  The ORR was 33% (2% CR, 31% PR) and the median duration of response was 8.7 months. Given that the majority of patients treated with IV alemtuzumab experience infusional toxicity combined with the observation that subcutaneous (SC) alemtuzumab had comparable biologic activity with diminished infusion-related events, interest in SC administration has progressively increased. In a phase 2 study by the GCLLSG,80  103 patients with fludarabine-refractory CLL received at least one dose of alemtuzumab, administered subcutaneously at 30 mg 3 times weekly for up to 12 weeks. The ORR was 34% (4% CR, 30% PR). The median PFS was 7.7 months and the median OS was 19.1 months. This trial confirmed earlier studies that alemtuzumab was effective for del(17p13.1) CLL.81,82 

In studies involving patients with relapsed/refractory CLL treated with alemtuzumab, the most common adverse events were cytopenia and infection as a consequence of profound cellular immune suppression. Reactivation of herpesvirus including cytomegalovirus (CMV) were the most common opportunistic infections observed. Prophylaxis against opportunistic infections together with monitoring for CMV reactivation is highly recommended in these patients. Recently, O'Brien et al85  demonstrated that the addition of valganciclovir 450 mg orally twice daily was highly effective for prophylaxis of CMV reactivation in patients receiving alemtuzumab. Our own approach is to administer bactrim (or equivalent) and anti-herpes antiviral (acyclovir, valacylovir) and to monitor for reactivation of CMV with subsequent preemptive early treatment due to the prohibitive cost of valgancyclovir.

Several pilot studies in previously untreated CLL patients86,87  have shown benefit with alemtuzumab. Lundin and colleagues87  treated 41 CLL patients with SC alemtuzumab as first-line therapy for up to 18 weeks, with an observed ORR of 87%, including a 19% CR rate with a median TTF of more than 18 months. The treatment was generally well tolerated, with adverse events mainly comprising local injection site reactions, and with no episodes of febrile neutropenia or major bacterial infections. CMV reactivation occurred in 10% of patients. These promising results promoted a recent phase 3 trial of 297 CLL patients who were prospectively randomized to receive either IV alemtuzumab 30 mg 3 times weekly for up to 12 weeks or oral chlorambucil 40 mg/m2 every 4 weeks for up to 12 cycles.88  Alemtuzumab-treated patients obtained a significantly superior response rate with significantly improved PFS compared with chlorambucil (ORR 83% vs 56% and CRR 24% vs 2%). In addition, this trial prospectively demonstrated the superiority of alemtuzumab versus chlorambucil in patients with del(17p13). No differences in terms of grade 3-4 hematologic toxicities were noticed between the 2 arms; however, 52% of alemtuzumab-treated patients developed CMV reactivation, in contrast to only 2% of the patients treated with chlorambucil. Despite approval for this indication, the use of alemtuzumab as monotherapy for primary therapy of CLL is not generally used, given both the improved results with chemoimmunotherapy and the immune suppression associated with this treatment.

Alemtuzumab as consolidation therapy for CLL

Given that alemtuzumab works best against blood and bone marrow disease, efforts to apply alemtuzumab as a consolidation approach occurred early in its development (reviewed in Alinari et al83 ). O'Brien and colleagues89  administered alemtuzumab 10 or 30 mg IV 3 times weekly to 41 CLL patients with residual disease after their most recent therapy. The ORR was 46% including 56% among the 29 patients treated with 30 mg. Eleven of these 29 patients (38%) achieved a MRD-negative marrow, assessed by polymerase chain reaction. Infectious occurred in 15 patients (37%) with 9 being CMV reactivation. The authors reported that 3 patients developed Epstein-Barr virus (EBV)–positive large B-cell lymphoma (LCL) within 6 weeks after finishing therapy. It is known that T and B lymphocytes subpopulations are heavily depleted by alemtuzumab and remain subnormal for more than a year.83  Therefore the development of these LCLs is probably due to proliferation of EBV-positive B cells in severely immunocompromised patients. Development of LCL represents a major concern in alemtuzumab treated patients, although fortunately it is not that common. The GCLLSG90  reported the results of a phase 3 trial where patients responding to fludarabine-based induction therapy were randomized to receive IV alemtuzumab 30 mg 3 times weekly for a maximum of 12 weeks or observation. Of 21 evaluable patients, 11 were randomized to receive alemtuzumab. This study was prematurely closed because of severe infections in 7 of 11 patients in the alemtuzumab arm. The PFS was significantly improved for patients receiving alemtuzumab at a median follow-up of 21.4 months. The CALGB performed 2 studies administering alemtuzumab after fludarabine91  or fludarabine and rituximab.92  Both of these studies demonstrated the ability of alemtuzumab to improve response to treatment.92  However, reactivation of CMV was observed in both studies and unacceptable infectious toxicity was observed in patients when alemtuzumab was administered after fludarabine and rituximab. A community-based clinical trial administering alemtuzumab after fludarabine and rituximab also noted problematic toxicity with combined chemoimmunotherapy.93  Future attempts to explore this consolidation approach after chemoimmunotherapy regimens should allow an extended recovery time before administration of alemtuzumab. Consolidation with alemtuzumab should only be considered in the context of a clinical trial.

Alemtuzumab combination strategies in CLL

In an attempt to enhance the therapeutic activity observed with fludarabine-based regimens, alemtuzumab has been added to this modality by several investigators (reviewed in Alinari et al83 ). These studies have, in general, demonstrated feasibility of administration with an acceptable toxicity profile. A phase 3 study comparing fludarabine combined with alemtuzumab (FluCAM) versus fludarabine alone was recently reported.94  This study of 335 previously treated CLL patients receiving second line therapy demonstrated a higher ORR, CRR and PFS with the combination therapy. Adverse events included cytopenias and infections and were similar between both treatment arms. The only toxicity uniformly associated with FluCAM was CMV reactivation in 8% of patients.94  Attempts to further intensify this regimen with the addition of cyclophosphamide to FluCAM resulted in increased infectious morbidity.95  Concomitantly, efforts to combine alemtuzumab with the FCR regimen were undertaken in both relapsed96  and previously untreated high risk CLL.97  Although these studies demonstrated feasibility, it is unclear that any benefit was offered over the FCR-based treatment. Combination of alemtuzumab with rituximab98-100  has also been evaluated by several groups with improved response. The FCGCLL/MW and GOELAMS recently presented results in a phase 3 study comparing FCR to the combination of fludarabine, cyclophosphamide, and alemtuzumab (FCCam). Response rates of the first 100 patients were reported in a preliminary analysis with safety data presented for the entire cohort of 178 patients. The ORR in the first 100 patients was 96% for FCR compared with 85% in the FCCam arm (P = .086) with a CRR of 78% in the FCR arm versus 58% in the FCC am arm (P = .072). The percentage of grade 4 neutropenia increased during FCCam treatment (28.4% for cycle 1 and 45.5% for cycle 6), and 44 of the 63 reported serious adverse events (SAE) declared occurred in the FCCam arm. Seven patients died, all of whom were in the FCCam arm.101  Thus, this particular combination appears to be associated with unacceptable infectious toxicities for no obvious clinical benefit.

Efforts to target other B cell–specific antigens are underway as part of clinical trials at this time. Of antibody and peptide therapeutics currently in clinical development, those targeting CD37 appear the most promising. CD37 is expressed on B cells and transformed mature B cell leukemias and lymphomas but not on T cells. TRU-016 is a CD37 small modular immunopharmaceutical (SMIP) that represents a structural modification of a CD37 antibody that lacks the CH1 domain. In vitro studies with the chimeric version of TRU-016 demonstrate that this is a potent inducer of apoptosis and ADCC-dependent cytotoxicity against CLL cells.102  Interim results of a phase 1 study of TRU-016 reported a favorable toxicity profile and partial responses at higher doses.103  Whereas the CD23 antibody lumiliximab is most mature with respect to clinical investigation in CLL, with a completed phase 1 single-agent study showing minimal activity but favorable toxicity profile104  and a phase 1/2 combination study with FCR showing potential benefit,105  results from a phase 3 study comparing FCR plus lumiliximab versus FCR did not confirm benefit in terms of improved response or PFS. Therapeutic antibodies directed at CD40 with partial agonist (SGN40106 ) and blocking properties (HCD122; personal communication, J.C.B., September 5, 2010) have completed phase 1 testing with minimal clinical activity in CLL and likely will be applied as combination therapies. Other new antibodies targeting CD19 (MDX-1342 and XmAb5574) and CD74 (milatuzamab) are either entering into or currently being tested in early phase 1 clinical trials for CLL. Extending beyond new targets for therapeutic antibodies and SMIPs is the opportunity to combine these therapies with new immune-modulating agents (interleukin-21, lenalidomide, CpG oligonucleotides) and targeted therapies (CAL-101, fostamatinib, PCI-32 765, SCH727965, ABT-263, and flavopiridol) with the potential to avoid the immunosuppressive effects of chemotherapy treatments.

Monoclonal antibodies represent an exciting addition to the growing armamentarium of agents used to treat CLL. To date, those agents targeting CD20 and CD52 have shown greatest promise. While both CD20- and CD52-directed antibodies have demonstrated activity as single agents, their greatest contribution (as has been demonstrated with rituximab) may lie in their combination with more traditional chemotherapies. When added to chemotherapy, the use of rituximab has resulted in improved ORR and CR rate and longer PFS and OS compared with chemotherapy alone, and this has been borne out in phase 3 studies. Monoclonal antibodies promise to be a fertile source of preclinical and clinical investigation and will likely continue to revolutionize the way we care for patients with CLL.

This work was supported by grants from Leukemia & Lymphoma Society (P50-CA140158, PO1-CA95426, PO1 CA81534) and by the D. Warren Brown Foundation.

National Institutes of Health

Contribution: S.J. and J.C.B. wrote the entire manuscript; and L.A., R.L., and R.M. wrote components of the manuscript and reviewed and approved the final version.

Conflict-of-interest disclosure: J.C.B. is a consultant for Calistoga Pharmaceuticals and has a financial interest in the development of this compound. The remaining authors declare no competing financial interests.

Correspondence: John C. Byrd, MD, 455B, OSUCCC, 410 W. 12th Ave, Columbus, OH 43210; e-mail: John.byrd@osumc.edu.

1
Rozman
 
C
Montserrat
 
E
Chronic lymphocytic leukemia.
N Engl J Med
1995
, vol. 
333
 
16
(pg. 
1052
-
1057
)
2
Zenz
 
T
Mertens
 
D
Kuppers
 
R
Dohner
 
H
Stilgenbauer
 
S
From pathogenesis to treatment of chronic lymphocytic leukaemia.
Nat Rev Cancer
2010
, vol. 
10
 
1
(pg. 
37
-
50
)
3
Ricci
 
F
Tedeschi
 
A
Morra
 
E
Montillo
 
M
Fludarabine in the treatment of chronic lymphocytic leukemia: a review.
Ther Clin Risk Manag
2009
, vol. 
5
 
1
(pg. 
187
-
207
)
4
Zenz
 
T
Dohner
 
H
Stilgenbauer
 
S
Genetics and risk-stratified approach to therapy in chronic lymphocytic leukemia.
Best Pract Res Clin Haematol
2007
, vol. 
20
 
3
(pg. 
439
-
453
)
5
Uchida
 
J
Lee
 
Y
Hasegawa
 
M
, et al. 
Mouse CD20 expression and function.
Int Immunol
2004
, vol. 
16
 
1
(pg. 
119
-
129
)
6
Press
 
OW
Howell-Clark
 
J
Anderson
 
S
Bernstein
 
I
Retention of B-cell-specific monoclonal antibodies by human lymphoma cells.
Blood
1994
, vol. 
83
 
5
(pg. 
1390
-
1397
)
7
Maloney
 
DG
Mechanism of action of rituximab.
Anticancer Drugs
2001
12 Suppl 2
(pg. 
S1
-
S4
)
8
Bellosillo
 
B
Villamor
 
N
Lopez-Guillermo
 
A
, et al. 
Complement-mediated cell death induced by rituximab in B-cell lymphoproliferative disorders is mediated in vitro by a caspase-independent mechanism involving the generation of reactive oxygen species.
Blood
2001
, vol. 
98
 
9
(pg. 
2771
-
2777
)
9
Golay
 
J
Lazzari
 
M
Facchinetti
 
V
, et al. 
CD20 levels determine the in vitro susceptibility to rituximab and complement of B-cell chronic lymphocytic leukemia: further regulation by CD55 and CD59.
Blood
2001
, vol. 
98
 
12
(pg. 
3383
-
3389
)
10
Li
 
Y
Williams
 
ME
Cousar
 
JB
Pawluczkowycz
 
AW
Lindorfer
 
MA
Taylor
 
RP
Rituximab-CD20 complexes are shaved from Z138 mantle cell lymphoma cells in intravenous and subcutaneous SCID mouse models.
J Immunol
2007
, vol. 
179
 
6
(pg. 
4263
-
4271
)
11
Williams
 
ME
Densmore
 
JJ
Pawluczkowycz
 
AW
, et al. 
Thrice-weekly low-dose rituximab decreases CD20 loss via shaving and promotes enhanced targeting in chronic lymphocytic leukemia.
J Immunol
2006
, vol. 
177
 
10
(pg. 
7435
-
7443
)
12
Beum
 
PV
Kennedy
 
AD
Williams
 
ME
Lindorfer
 
MA
Taylor
 
RP
The shaving reaction: rituximab/CD20 complexes are removed from mantle cell lymphoma and chronic lymphocytic leukemia cells by THP-1 monocytes.
J Immunol
2006
, vol. 
176
 
4
(pg. 
2600
-
2609
)
13
Aue
 
G
Lindorfer
 
MA
Beum
 
PV
, et al. 
Fractionated subcutaneous rituximab is well-tolerated and preserves CD20 expression on tumor cells in patients with chronic lymphocytic leukemia.
Haematologica
2009
, vol. 
95
 
2
(pg. 
329
-
332
)
14
Gowda
 
A
Roda
 
J
Hussain
 
SR
, et al. 
IL-21 mediates apoptosis through up-regulation of the BH3 family member BIM and enhances both direct and antibody-dependent cellular cytotoxicity in primary chronic lymphocytic leukemia cells in vitro.
Blood
2008
, vol. 
111
 
9
(pg. 
4723
-
4730
)
15
Flieger
 
D
Emmerich
 
B
Meyer
 
N
Riethmuller
 
G
Ziegler-Heitbrock
 
HW
Deficient production of tumor necrosis factor by peripheral-blood monocytes in chronic lymphocytic leukemia.
Int J Cancer
1990
, vol. 
45
 
2
(pg. 
280
-
286
)
16
Yang
 
L
Zhao
 
T
Shi
 
X
, et al. 
Functional analysis of a dominant negative mutation of interferon regulatory factor 5.
PLoS One
2009
, vol. 
4
 
5
pg. 
e5500
 
17
Ziegler
 
HW
Kay
 
NE
Zarling
 
JM
Deficiency of natural killer cell activity in patients with chronic lymphocytic leukemia.
Int J Cancer
1981
, vol. 
27
 
3
(pg. 
321
-
327
)
18
Kay
 
NE
Zarling
 
JM
Impaired natural killer activity in patients with chronic lymphocytic leukemia is associated with a deficiency of azurophilic cytoplasmic granules in putative NK cells.
Blood
1984
, vol. 
63
 
2
(pg. 
305
-
309
)
19
Beyer
 
M
Kochanek
 
M
Darabi
 
K
, et al. 
Reduced frequencies and suppressive function of CD4+CD25hi regulatory T cells in patients with chronic lymphocytic leukemia after therapy with fludarabine.
Blood
2005
, vol. 
106
 
6
(pg. 
2018
-
2025
)
20
Gowda
 
A
Ramanunni
 
A
Cheney
 
C
, et al. 
Differential effects of IL-2 and IL-21 on expansion of the CD4+ CD25+ Foxp3+ T regulatory cells with redundant roles in natural killer cell mediated antibody dependent cellular cytotoxicity in chronic lymphocytic leukemia.
mAbs
2010
, vol. 
2
 
1
(pg. 
35
-
41
)
21
Cartron
 
G
Dacheux
 
L
Salles
 
G
, et al. 
Therapeutic activity of humanized anti-CD20 monoclonal antibody and polymorphism in IgG Fc receptor FcgammaRIIIa gene.
Blood
2002
, vol. 
99
 
3
(pg. 
754
-
758
)
22
Weng
 
WK
Levy
 
R
Two immunoglobulin G fragment C receptor polymorphisms independently predict response to rituximab in patients with follicular lymphoma.
J Clin Oncol
2003
, vol. 
21
 
21
(pg. 
3940
-
3947
)
23
Farag
 
SS
Flinn
 
IW
Modali
 
R
Lehman
 
TA
Young
 
D
Byrd
 
JC
Fc gamma RIIIa and Fc gamma RIIa polymorphisms do not predict response to rituximab in B-cell chronic lymphocytic leukemia.
Blood
2004
, vol. 
103
 
4
(pg. 
1472
-
1474
)
24
Woyach
 
JA
Lin
 
TS
Lucas
 
MS
, et al. 
A phase I/II study of rituximab and etanercept in patients with chronic lymphocytic leukemia and small lymphocytic lymphoma.
Leukemia
2009
, vol. 
23
 
5
(pg. 
912
-
918
)
25
Friedberg
 
JW
Kelly
 
JL
Neuberg
 
D
, et al. 
Phase II study of a TLR-9 agonist (1018 ISS) with rituximab in patients with relapsed or refractory follicular lymphoma.
Br J Haematol
2009
, vol. 
146
 
3
(pg. 
282
-
291
)
26
Gowda
 
A
Ramanunni
 
A
Cheney
 
C
, et al. 
Differential effects of IL-2 and IL-21 on expansion of the CD4(+)CD25(+)Foxp3(+) T regulatory cells with redundant roles in natural killer cell mediated antibody dependent cellular cytotoxicity in chronic lymphocytic leukemia.
mAbs
2010
, vol. 
2
 
1
(pg. 
35
-
41
)
27
Jaglowski
 
SM
Byrd
 
JC
Rituximab in chronic lymphocytic leukemia.
Semin Hematol
4
, vol. 
47
 
2
(pg. 
156
-
169
)
28
Byrd
 
JC
Kitada
 
S
Flinn
 
IW
, et al. 
The mechanism of tumor cell clearance by rituximab in vivo in patients with B-cell chronic lymphocytic leukemia: evidence of caspase activation and apoptosis induction.
Blood
2002
, vol. 
99
 
3
(pg. 
1038
-
1043
)
29
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
, vol. 
104
 
6
(pg. 
1793
-
1800
)
30
Teeling
 
JL
Mackus
 
WJ
Wiegman
 
LJ
, et al. 
The biological activity of human CD20 monoclonal antibodies is linked to unique epitopes on CD20.
J Immunol
2006
, vol. 
177
 
1
(pg. 
362
-
371
)
31
Niederfellner
 
GJ
Lammens
 
A
Schwaiger
 
M
, et al. 
Crystal structure analysis reveals that the novel type II anti-CD20 antibody GA101 interacts with a similar epitope as rituximab and ocrelizumab but in a fundamentally different way [abstract].
Blood (ASH Annual Meeting Abstracts)
2009
, vol. 
114
 
22
pg. 
3726
 
32
Alduaij
 
W
Potluri
 
S
Ivanov
 
A
, et al. 
New-generation anti-CD20 monoclonal antibody (GA101) evokes homotypic adhesion and actin-dependent, lysosome-mediated cell death in B-cell lymphoma [abstract].
Blood (ASH Annual Meeting Abstracts)
2009
, vol. 
114
 
22
pg. 
725
 
33
Zenz
 
T
Volden
 
M
Mast
 
T
, et al. 
In vitro activity of the type II anti-CD20 antibody GA101 in refractory, genetic high-risk CLL [abstract].
Blood (ASH Annual Meeting Abstracts)
2009
, vol. 
114
 
22
pg. 
2379
 
34
Patz
 
M
Forcob
 
N
Muller
 
B
, et al. 
Depletion of chronic lymphocytic leukemia cells from whole blood samples mediated by the anti-CD20 antibodies rituximab and GA101 [abstract].
Blood (ASH Annual Meeting Abstracts)
2009
, vol. 
114
 
22
pg. 
2365
 
35
Ivanov
 
A
Beers
 
SA
Walshe
 
CA
, et al. 
Monoclonal antibodies directed to CD20 and HLA-DR can elicit homotypic adhesion followed by lysosome-mediated cell death in human lymphoma and leukemia cells.
J Clin Invest
2009
, vol. 
119
 
8
(pg. 
2143
-
2159
)
36
Cragg
 
MS
Glennie
 
MJ
Antibody specificity controls in vivo effector mechanisms of anti-CD20 reagents.
Blood
2004
, vol. 
103
 
7
(pg. 
2738
-
2743
)
37
Berinstein
 
NL
Grillo-Lopez
 
AJ
White
 
CA
, et al. 
Association of serum Rituximab (IDEC-C2B8) concentration and anti-tumor response in the treatment of recurrent low-grade or follicular non-Hodgkin's lymphoma.
Ann Oncol
1998
, vol. 
9
 
9
(pg. 
995
-
1001
)
38
Ginaldi
 
L
De Martinis
 
M
Matutes
 
E
Farahat
 
N
Morilla
 
R
Catovsky
 
D
Levels of expression of CD19 and CD20 in chronic B cell leukaemias.
J Clin Pathol
1998
, vol. 
51
 
5
(pg. 
364
-
369
)
39
Byrd
 
JC
Murphy
 
T
Howard
 
RS
, et al. 
Rituximab using a thrice weekly dosing schedule in B-cell chronic lymphocytic leukemia and small lymphocytic lymphoma demonstrates clinical activity and acceptable toxicity.
J Clin Oncol
2001
, vol. 
19
 
8
(pg. 
2153
-
2164
)
40
O'Brien
 
SM
Kantarjian
 
H
Thomas
 
DA
, et al. 
Rituximab dose-escalation trial in chronic lymphocytic leukemia.
J Clin Oncol
2001
, vol. 
19
 
8
(pg. 
2165
-
2170
)
41
Hainsworth
 
JD
Litchy
 
S
Barton
 
JH
, et al. 
Single-agent rituximab as first-line and maintenance treatment for patients with chronic lymphocytic leukemia or small lymphocytic lymphoma: a phase II trial of the Minnie Pearl Cancer Research Network.
J Clin Oncol
2003
, vol. 
21
 (pg. 
1746
-
1751
)
42
Schulz
 
H
Klein
 
SK
Rehwald
 
U
, et al. 
Phase 2 study of a combined immunochemotherapy using rituximab and fludarabine in patients with chronic lymphocytic leukemia.
Blood
2002
, vol. 
100
 
9
(pg. 
3115
-
3120
)
43
Byrd
 
JC
Peterson
 
BL
Morrison
 
VA
, et al. 
Randomized phase 2 study of fludarabine with concurrent versus sequential treatment with rituximab in symptomatic, untreated patients with B-cell chronic lymphocytic leukemia: results from Cancer and Leukemia Group B 9712 (CALGB 9712).
Blood
2003
, vol. 
101
 
1
(pg. 
6
-
14
)
44
Byrd
 
JC
Gribben
 
JG
Peterson
 
BL
, et al. 
Select high-risk genetic features predict earlier progression following chemoimmunotherapy with fludarabine and rituximab in chronic lymphocytic leukemia: justification for risk-adapted therapy.
J Clin Oncol
2006
, vol. 
24
 
3
(pg. 
437
-
443
)
45
Byrd
 
JC
Rai
 
K
Peterson
 
BL
, et al. 
Addition of rituximab to fludarabine may prolong progression-free survival and overall survival in patients with previously untreated chronic lymphocytic leukemia: an updated retrospective comparative analysis of CALGB 9712 and CALGB 9011.
Blood
2005
, vol. 
105
 
1
(pg. 
49
-
53
)
46
Del Poeta
 
G
Del Principe
 
MI
Consalvo
 
MA
, et al. 
The addition of rituximab to fludarabine improves clinical outcome in untreated patients with ZAP-70-negative chronic lymphocytic leukemia.
Cancer
2005
, vol. 
104
 
12
(pg. 
2743
-
2752
)
47
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
, vol. 
112
 
4
(pg. 
975
-
980
)
48
Wierda
 
W
O'Brien
 
S
Wen
 
S
, et al. 
Chemoimmunotherapy with fludarabine, cyclophosphamide, and rituximab for relapsed and refractory chronic lymphocytic leukemia.
J Clin Oncol
2005
, vol. 
23
 
18
(pg. 
4070
-
4078
)
49
Tsimberidou
 
AM
Tam
 
C
Abruzzo
 
LV
, et al. 
Chemoimmunotherapy may overcome the adverse prognostic significance of 11q deletion in previously untreated patients with chronic lymphocytic leukemia.
Cancer
2009
, vol. 
115
 
2
(pg. 
373
-
380
)
50
Lin
 
KI
Tam
 
CS
Keating
 
MJ
, et al. 
Relevance of the immunoglobulin VH somatic mutation status in patients with chronic lymphocytic leukemia treated with fludarabine, cyclophosphamide, and rituximab (FCR) or related chemoimmunotherapy regimens.
Blood
2009
, vol. 
113
 
14
(pg. 
3168
-
3171
)
51
Lamanna
 
N
Kalaycio
 
M
Maslak
 
P
, et al. 
Pentostatin, cyclophosphamide, and rituximab is an active, well-tolerated regimen for patients with previously treated chronic lymphocytic leukemia.
J Clin Oncol
2006
, vol. 
24
 
10
(pg. 
1575
-
1581
)
52
Kay
 
NE
Geyer
 
SM
Call
 
TG
, et al. 
Combination chemoimmunotherapy with pentostatin, cyclophosphamide, and rituximab shows significant clinical activity with low accompanying toxicity in previously untreated B chronic lymphocytic leukemia.
Blood
2007
, vol. 
109
 
2
(pg. 
405
-
411
)
53
Fischer
 
K
Cramer
 
P
Stilgenbauer
 
S
, et al. 
Bendamustine Combined with Rituximab (BR) in First-Line Therapy of Advanced CLL: A Multicenter Phase II Trial of the German CLL Study Group (GCLLSG).
2009
, vol. 
Vol 114
 pg. 
205
 
54
Fischer
 
K
Stilgenbauer
 
S
Schweighofer
 
CD
, et al. 
Bendamustine in Combination with Rituximab (BR) for Patients with Relapsed Chronic Lymphocytic Leukemia (CLL): A Multicentre Phase II Trial of the German CLL Study Group (GCLLSG).
2008
, vol. 
Vol 112
 pg. 
330
 
55
Hallek
 
M
Fingerle-Rowson
 
G
Fink
 
A-M
, et al. 
First-Line Treatment with Fludarabine (F), Cyclophosphamide (C), and Rituximab (R) (FCR) Improves Overall Survival (OS) in Previously Untreated Patients (pts) with advanced chronic lymphocytic leukemia (CLL): results of a randomized phase III trial on behalf of an International Group of Investigators and the German CLL Study Group.
Blood. ASH Annual Meeting Abstracts
2009
, vol. 
114
 
22
pg. 
535
 
56
Hallek
 
M
Fingerle-Rowson
 
G
Fink
 
A-M
, et al. 
Immunochemotherapy with fludarabine (F), cyclophosphamide (C), and rituximab (R) (FCR) versus fludarabine and cyclophosphamide (FC) improves response rates and progression-free survival (PFS) of previously untreated patients (pts) with advanced chronic lymphocytic leukemia (CLL).
Blood. ASH Annual Meeting Abstracts
2008
, vol. 
112
 
11
pg. 
325
 
57
Stilgenbauer
 
S
Zenz
 
T
Winkler
 
D
, et al. 
Genomic Aberrations, VH Mutation status and outcome after fludarabine and cyclophosphamide (FC) or FC plus rituximab (FCR) in the CLL8 trial.
Blood. ASH Annual Meeting Abstracts
2008
, vol. 
112
 
11
pg. 
781
 
58
Robak
 
T
Moiseev
 
SI
Dmoszynska
 
A
, et al. 
Rituximab, Fludarabine, and cyclophosphamide (R-FC) prolongs progression free survival in relapsed or refractory chronic lymphocytic leukemia (CLL) compared with FC alone: final results from the International Randomized Phase III REACH Trial.
Blood. ASH Annual Meeting Abstracts
2008
, vol. 
112
 
11
(pg. 
lba
-
1
)
59
Del Poeta
 
G
Del Principe
 
MI
Buccisano
 
F
, et al. 
Consolidation and maintenance immunotherapy with rituximab improve clinical outcome in patients with B-cell chronic lymphocytic leukemia.
Cancer
2008
, vol. 
112
 
1
(pg. 
119
-
128
)
60
Dearden
 
C
Disease-Specific Complications of Chronic Lymphocytic Leukemia.
Hematology
2008
, vol. 
2008
 
1
(pg. 
450
-
456
)
61
Zecca
 
M
De Stefano
 
P
Nobili
 
B
Locatelli
 
F
Anti-CD20 monoclonal antibody for the treatment of severe, immune-mediated, pure red cell aplasia and hemolytic anemia.
Blood
2001
, vol. 
97
 
12
(pg. 
3995
-
3997
)
62
Ghazal
 
H
Successful treatment of pure red cell aplasia with rituximab in patients with chronic lymphocytic leukemia.
Blood
2002
, vol. 
99
 
3
(pg. 
1092
-
1094
)
63
Gupta
 
N
Kavuru
 
S
Patel
 
D
, et al. 
Rituximab-based chemotherapy for steroid-refractory autoimmune hemolytic anemia of chronic lymphocytic leukemia.
Leukemia
2002
, vol. 
16
 
10
(pg. 
2092
-
2095
)
64
D'Arena
 
G
Laurenti
 
L
Capalbo
 
S
, et al. 
Rituximab therapy for chronic lymphocytic leukemia-associated autoimmune hemolytic anemia.
Am J Hematol
2006
, vol. 
81
 
8
(pg. 
598
-
602
)
65
Hegde
 
UP
Wilson
 
WH
White
 
T
Cheson
 
BD
Rituximab treatment of refractory fludarabine-associated immune thrombocytopenia in chronic lymphocytic leukemia.
Blood
2002
, vol. 
100
 
6
(pg. 
2260
-
2262
)
66
Coiffier
 
B
Lepretre
 
S
Pedersen
 
LM
, et al. 
Safety and efficacy of ofatumumab, a fully human monoclonal anti-CD20 antibody, in patients with relapsed or refractory B-cell chronic lymphocytic leukemia: a phase 1-2 study.
Blood
2008
, vol. 
111
 
3
(pg. 
1094
-
1100
)
67
Wierda
 
WG
Kipps
 
TJ
Mayer
 
J
, et al. 
Ofatumumab as single-agent CD20 immunotherapy in fludarabine-refractory chronic lymphocytic leukemia.
J Clin Oncol
4
1
, vol. 
28
 
10
(pg. 
1749
-
1755
)
68
Eichhorst
 
BF
Fischer
 
K
Fink
 
A-M
, et al. 
The Performance of Computed Tomography (CT) Scans or Ultrasound Has No Clinical Relevance in Treatment of Patients with Chronic Lymphocytic Leukemia (CLL): Results of a Metaanalysis from Three Phase III Studies of the German CLL Study Group (GCLLSG).
2008
, vol. 
Vol 112
 pg. 
1053
 
69
Blum
 
KA
Young
 
D
Broering
 
S
, et al. 
Computed tomography scans do not improve the predictive power of 1996 national cancer institute sponsored working group chronic lymphocytic leukemia response criteria.
J Clin Oncol
2007
, vol. 
25
 
35
(pg. 
5624
-
5629
)
70
Wierda
 
WG
Kipps
 
TJ
Durig
 
J
, et al. 
Ofatumumab Combined with Fludarabine and Cyclophosphamide (O-FC) Shows High Activity in Patients with Previously Untreated Chronic Lymphocytic Leukemia (CLL): Results From a Randomized, Multicenter, International, Two-Dose, Parallel Group, Phase II Trial.
Blood (ASH Annual Meeting Abstracts)
2009
, vol. 
114
 
22
pg. 
207
 
71
Zelenetz
 
AD
Abramson
 
JS
Advani
 
RH
, et al. 
NCCN Clinical Practice Guidelines in Oncology: Non-Hodgkin's Lymphomas.
J Natl Compr Canc Netw
3
, vol. 
8
 
3
(pg. 
288
-
334
)
72
Morschhauser
 
F
Cartron
 
G
Lamy
 
T
, et al. 
Phase I Study of RO5072759 (GA101) in Relapsed/Refractory Chronic Lymphocytic Leukemia.
Blood (ASH Annual Meeting Abstracts)
2009
, vol. 
114
 
22
pg. 
884
 
73
Rossmann
 
ED
Lundin
 
J
Lenkei
 
R
Mellstedt
 
H
Osterborg
 
A
Variability in B-cell antigen expression: implications for the treatment of B-cell lymphomas and leukemias with monoclonal antibodies.
Hematol J
2001
, vol. 
2
 
5
(pg. 
300
-
306
)
74
Ginaldi
 
L
De Martinis
 
M
Matutes
 
E
, et al. 
Levels of expression of CD52 in normal and leukemic B and T cells: correlation with in vivo therapeutic responses to Campath-1H.
Leuk Res
1998
, vol. 
22
 
2
(pg. 
185
-
191
)
75
Golay
 
J
Manganini
 
M
Rambaldi
 
A
Introna
 
M
Effect of alemtuzumab on neoplastic B cells.
Haematologica
2004
, vol. 
89
 
12
(pg. 
1476
-
1483
)
76
Zent
 
CS
Chen
 
JB
Kurten
 
RC
Kaushal
 
GP
Marie Lacy
 
H
Schichman
 
SA
Alemtuzumab (CAMPATH 1H) does not kill chronic lymphocytic leukemia cells in serum free medium.
Leuk Res
2004
, vol. 
28
 
5
(pg. 
495
-
507
)
77
Crowe
 
JS
Hall
 
VS
Smith
 
MA
Cooper
 
HJ
Tite
 
JP
Humanized monoclonal antibody CAMPATH-1H: myeloma cell expression of genomic constructs, nucleotide sequence of cDNA constructs and comparison of effector mechanisms of myeloma and Chinese hamster ovary cell-derived material.
Clin Exp Immunol
1992
, vol. 
87
 
1
(pg. 
105
-
110
)
78
Hale
 
G
Clark
 
M
Waldmann
 
H
Therapeutic potential of rat monoclonal antibodies: isotype specificity of antibody-dependent cell-mediated cytotoxicity with human lymphocytes.
J Immunol
1985
, vol. 
134
 
5
(pg. 
3056
-
3061
)
79
Mone
 
AP
Cheney
 
C
Banks
 
AL
, et al. 
Alemtuzumab induces caspase-independent cell death in human chronic lymphocytic leukemia cells through a lipid raft-dependent mechanism.
Leukemia
2006
, vol. 
20
 
2
(pg. 
272
-
279
)
80
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
, vol. 
27
 
24
(pg. 
3994
-
4001
)
81
Lozanski
 
G
Heerema
 
NA
Flinn
 
IW
, et al. 
Alemtuzumab is an effective therapy for chronic lymphocytic leukemia with p53 mutations and deletions.
Blood
2004
, vol. 
103
 
9
(pg. 
3278
-
3281
)
82
Stilgenbauer
 
S
Dohner
 
H
Campath-1H-induced complete remission of chronic lymphocytic leukemia despite p53 gene mutation and resistance to chemotherapy.
N Engl J Med
2002
, vol. 
347
 
6
(pg. 
452
-
453
)
83
Alinari
 
L
Lapalombella
 
R
Andritsos
 
L
Baiocchi
 
RA
Lin
 
TS
Byrd
 
JC
Alemtuzumab (Campath-1H) in the treatment of chronic lymphocytic leukemia.
Oncogene
2007
, vol. 
26
 
25
(pg. 
3644
-
3653
)
84
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
, vol. 
99
 
10
(pg. 
3554
-
3561
)
85
O'Brien
 
S
Ravandi-Kashani
 
F
Wierda
 
WG
, et al. 
A Randomized Trial of Valacyclovir Versus Valganciclovir To Prevent CMV Reactivation in Patients with CLL Receiving Alemtuzumab.
ASH Annual Meeting Abstracts
2005
, vol. 
106
 
11
pg. 
2960
 
86
Osterborg
 
A
Fassas
 
AS
Anagnostopoulos
 
A
Dyer
 
MJ
Catovsky
 
D
Mellstedt
 
H
Humanized CD52 monoclonal antibody Campath-1H as first-line treatment in chronic lymphocytic leukaemia.
Br J Haematol
1996
, vol. 
93
 
1
(pg. 
151
-
153
)
87
Lundin
 
J
Kimby
 
E
Bjorkholm
 
M
, et al. 
Phase II trial of subcutaneous anti-CD52 monoclonal antibody alemtuzumab (Campath-1H) as first-line treatment for patients with B-cell chronic lymphocytic leukemia (B-CLL).
Blood
2002
, vol. 
100
 
3
(pg. 
768
-
773
)
88
Hillmen
 
P
Skotnicki
 
AB
Robak
 
T
, et al. 
Alemtuzumab compared with chlorambucil as first-line therapy for chronic lymphocytic leukemia.
J Clin Oncol
2007
, vol. 
25
 
35
(pg. 
5616
-
5623
)
89
O'Brien
 
SM
Kantarjian
 
HM
Thomas
 
DA
, et al. 
Alemtuzumab as treatment for residual disease after chemotherapy in patients with chronic lymphocytic leukemia.
Cancer
2003
, vol. 
98
 
12
(pg. 
2657
-
2663
)
90
Wendtner
 
CM
Ritgen
 
M
Schweighofer
 
CD
, et al. 
Consolidation with alemtuzumab in patients with chronic lymphocytic leukemia (CLL) in first remission–experience on safety and efficacy within a randomized multicenter phase III trial of the German CLL Study Group (GCLLSG).
Leukemia
2004
, vol. 
18
 
6
(pg. 
1093
-
1101
)
91
Byrd
 
JC
Peterson
 
BL
Rai
 
KR
, et al. 
Fludarabine followed by alemtuzumab consolidation for previously untreated chronic lymphocytic leukemia: final report of Cancer and Leukemia Group B study 19901.
Leuk Lymphoma
2009
, vol. 
50
 
10
(pg. 
1589
-
1596
)
92
Lin
 
TS
Donohue
 
KA
Byrd
 
JC
, et al. 
Consolidation Therapy with Subcutaneous (SC) Alemtuzumab After Fludarabine and Rituximab (FR) Induction Therapy Improves the Complete Response (CR) Rate in Chronic Lymphocytic Leukemia (CLL) and Eradicates Minimal Residual Disease (MRD) but Is Associated with Severe Infectious Toxicity: Final Analysis of CALGB Study 10101.
2009
, vol. 
Vol 114
 pg. 
210
 
93
Hainsworth
 
JD
Vazquez
 
ER
Spigel
 
DR
, et al. 
Combination therapy with fludarabine and rituximab followed by alemtuzumab in the first-line treatment of patients with chronic lymphocytic leukemia or small lymphocytic lymphoma: a phase 2 trial of the Minnie Pearl Cancer Research Network.
Cancer
2008
, vol. 
112
 
6
(pg. 
1288
-
1295
)
94
Engert
 
A
Gercheva
 
L
Robak
 
T
, et al. 
Improved Progression-Free Survival (PFS) of Alemtuzumab (Campath(R), MabCampath(R)) Plus Fludarabine (Fludara(R)) Versus Fludarabine Alone as Second-Line Treatment of Patients with B-Cell Chronic Lymphocytic Leukemia: Preliminary Results From a Phase III Randomized Trial [abstract].
Blood (Am Soc Hematol Ann Meeting)
2009
, vol. 
114
  
Abstract 537
95
Elter
 
T
James
 
R
Stilgenbauer
 
S
, et al. 
Chemoimmuno-Therapy with Fludarabine, Cyclophosphamide and Alemtuzumab (FC-Cam) in Patients with Relapsed or Genetic High-Risk CLL: Final Analysis of the CLL2L Trial of the German CLL Study Group [abstract].
Blood (Am Soc Hematol Ann Meeting)
2009
, vol. 
114
  
Abstract 209
96
Badoux
 
XC
Keating
 
M
O'Brien
 
S
, et al. 
Chemoimmunotherapy with Cyclophosphomide, Fludarabine, Alemtuzumab and Rituximab (CFAR) Is Effective in Relapsed Patients with Chronic Lymphocytic Leukemia (CLL) [abstract].
Blood (Am Soc Hematol Ann Meeting)
2009
, vol. 
114
  
Abstract 3431
97
Parikh
 
SA
Keating
 
M
O'Brien
 
S
, et al. 
Frontline combined chemoimmunotherapy with fludarabine, cyclophosphamide, alemtuzumab and rituximab (CFAR) in high-risk chronic lymphocytic leukemia [abstract].
Blood (Am Soc Hematol Ann Meeting)
2009
, vol. 
114
  
Abstract 208
98
Zent
 
CS
Call
 
TG
Shanafelt
 
TD
, et al. 
Early treatment of high-risk chronic lymphocytic leukemia with alemtuzumab and rituximab.
Cancer
2008
, vol. 
113
 
8
(pg. 
2110
-
2118
)
99
Faderl
 
S
Thomas
 
DA
O'Brien
 
S
, et al. 
Experience with alemtuzumab plus rituximab in patients with relapsed and refractory lymphoid malignancies.
Blood
2003
, vol. 
101
 
9
(pg. 
3413
-
3415
)
100
Faderl
 
S
Ferrajoli
 
A
Wierda
 
W
O'Brien
 
S
Lerner
 
S
Keating
 
MJ
Alemtuzumab by continuous intravenous infusion followed by subcutaneous injection plus rituximab in the treatment of patients with chronic lymphocytic leukemia recurrence.
Cancer
2010
, vol. 
116
 
10
(pg. 
2360
-
2365
)
101
Lepretre
 
S
Aurran
 
T
Mahe
 
B
, et al. 
Immunochemotherapy with fludarabine (F), cyclophosphamide (C), and rituximab (R) (FCR) versus fludarabine (F), cyclophosphamide (C) and mabCampath (Cam) (FCCam) in previously untreated patients (pts) with advanced B-chronic lymphocytic leukemia (B-CLL): experience on safety and efficacy within a randomised multicenter phase III trial of the French Cooperative Group On CLL and WM (FCGCLL/MW) and the “Groupe Ouest-Est d'Etudes Des Leucemies Aigues Et Autres Maladies Du sang” (GOELAMS): CLL2007FMP (for fit medically patients).
Blood. ASH Annual Meeting Abstracts
2009
, vol. 
114
 
22
pg. 
538
 
102
Zhao
 
X
Lapalombella
 
R
Joshi
 
T
, et al. 
Targeting CD37-positive lymphoid malignancies with a novel engineered small modular immunopharmaceutical.
Blood
2007
, vol. 
110
 
7
(pg. 
2569
-
2577
)
103
Andritsos
 
L
Furman
 
RR
Flinn
 
IW
, et al. 
A phase 1 trial of TRU-016, an anti-CD37 small modular immunopharmaceutical (SMIPTM) protein in relapsed and refractory CLL: early promising clinical activity.
Blood. ASH Annual Meeting Abstracts
2009
, vol. 
114
 pg. 
3424
 
104
Byrd
 
JC
O'Brien
 
S
Flinn
 
IW
, et al. 
Phase 1 study of lumiliximab with detailed pharmacokinetic and pharmacodynamic measurements in patients with relapsed or refractory chronic lymphocytic leukemia.
Clin Cancer Res
2007
, vol. 
13
 
15 Pt 1
(pg. 
4448
-
4455
)
105
Byrd
 
JC
Kipps
 
TJ
Flinn
 
IW
, et al. 
Phase 1/2 study of lumiliximab combined with fludarabine, cyclophosphamide, and rituximab in patients with relapsed or refractory chronic lymphocytic leukemia.
Blood
1
21
, vol. 
115
 
3
(pg. 
489
-
495
)
106
Furman
 
RR
Forero-Torres
 
A
Shustov
 
A
Drachman
 
JG
A phase I study of dacetuzumab (SGN-40, a humanized anti-CD40 monoclonal antibody) in patients with chronic lymphocytic leukemia.
Leuk Lymphoma
2
, vol. 
51
 
2
(pg. 
228
-
235
)
Sign in via your Institution