Unlabeled and radiolabeled anti-CD20 monoclonal antibodies have had a significant impact in the care of patients with follicular lymphoma (FL) over the past decade. More recently, bendamustine has demonstrated activity in refractory FL, and has been explored as initial therapy and in novel combinations. Whereas outcomes for this patient population have significantly improved, there remains substantial unmet need for patients who require more effective and better-tolerated therapies. Novel anti-CD20 antibodies and other immunotherapies against different B-cell antigens are under active investigation. The proteosome inhibitor bortezomib and the immunomodulatory agent lenalidomide have demonstrated single-agent activity and are currently in randomized trials. Other novel compounds have demonstrated activity in broad-based clinical studies in B-cell malignancies. However, considerable challenges remain in efficiently demonstrating which patient subsets can benefit from these novel compounds and which combinations may have the greatest clinical benefit in further improving outcomes for patients with FL.

There are three great ironies in the current development of novel therapies for patients with follicular lymphoma (FL). First, more successful frontline approaches have made the assessment of new agents as part of initial therapy a more difficult and long-term endeavor. Second, patients with resistant disease need new agents. However, the demonstration of efficacy in this patient population can be difficult. Third, in some cases, the more targeted a drug—and the more rational the design—the more challenging it may be to establish single-agent activity. Indeed, to a large extent, this is the situation we currently face. The anti-CD20 monoclonal antibody rituximab has become part of the standard of care for nearly all patients with FL and has been associated with significant improvements in overall survival. The radioimmunotherapy agents yttrium-90 ibritumomab tiuxetan and iodine-131 tositumomab have substantial activity, and are well tolerated. The chemotherapeutic agent bendamustine was recently approved by the US Food and Drug Administration (FDA) for treatment of patients with rituximab refractory indolent lymphoma and is positioned to become a component of frontline therapy. In the current environment, it is remarkable that, through clinical trials, FL patients have access to more novel drugs than ever.

Targeted agents can be broadly classified into three groups: (1) those that target the surface of the cell (eg, monoclonal antibodies), (2) those that are directed against intracellular processes (eg, proteasome inhibitors), and (3) those that target the cancer microenvironment (eg, immunomodulatory drugs).

Most therapeutic monoclonal antibodies are designed to target glycoproteins on the surface of cancer cells. From the cell surface, the antibody is able to interact with other components of the immune system, resulting in antibody-dependent cell-mediated cytotoxicity (ADCC) or complement-dependent cytotoxicity (CDC), the putative dominant mechanisms of action of rituximab. Despite taking place at the cell surface, the effects of antibody binding may extend to the intracellular domain, where alterations in cell signaling can induce cell apoptosis. Monoclonal antibodies may also have effects beyond the cancer cell itself, resulting in a “vaccinal effect.” The choice of antigen, the properties of the antibody (ie, the degree and site of binding, the ability to interact with the immune system or alter signal transduction), and the decision to coadminister additional drugs all may enhance efficacy in certain settings.

Several novel antibodies under preclinical and clinical assessments incorporate structural modifications that are hoped to deliver clinical advantages, in comparison with rituximab. Antibody binding to effector cells through activating Fc receptors (FcγRIII) is requisite for ADCC and has therefore been central to new antibody development. The importance of CDC and direct cytotoxicity in elimination of cancer cells have been debated, but have also been the focus of novel antibody design. Finally, engineering away from murine or chimeric protein structures toward humanized or fully human antibodies has resulted in decreased immunogenicity, changes in pharmacokinetics, and, most importantly, potential enhancement of therapeutic effects.

The fully human, anti-CD20 ofatumumab was engineered to target an epitope more proximal to the surface of B cells and to have a slower off-rate than rituximab.1,2  Classified as a type I antibody, ofatumumab is able to induce translocation of CD20 into detergent-insoluble lipid rafts.1  As a result, ofatumumab is able to induce significantly greater CDC than rituximab at lower target-antigen concentrations.1  Additionally, the Fc portion of ofatumumab binds more strongly with FcγRIII and appears to produce greater ADCC than rituximab in vitro.3  Ofatumumab was recently approved by the FDA for treatment of patients with chronic lymphocytic leukemia (CLL) that is refractory to fludarabine and alemtuzumab, a patient population that is poorly responsive to rituximab. A phase I/II study in patients with previously treated FL evaluated four weekly infusions at doses from 300 to 1000 mg.4  Toxicity was similar to that of rituximab, and clinical response rates ranged from 20% to 62%, depending on the cohort. The median time to progression was 8.8 months. To assess whether a novel anti-CD20 might overcome resistance to rituximab, ofatumumab was assessed with eight weekly infusions at one of two dose levels (500 or 1000 mg) in 116 rituximab refractory patients with FL.5  Those subjects who received the higher dose demonstrated an overall response rate (ORR) of 10%, including one complete response (CR). Given the refractory nature of this population (median four prior regimens) one could argue that comparisons of ofatumumab to rituximab might be better suited for a less resistant patient population. Ongoing studies include those employing ofatumumab in combination with chemotherapy (NCT00494780).

Veltuzumab (hA20) is a humanized IgG1 with identical anti-CD20 complementarity-determining regions such as rituximab.6  Preclinical studies demonstrated activity similar to that of rituximab, although with a slower off-rate and enhanced CDC in several B-cell lymphoma cell lines.6  In a multicenter phase I/II study, no grade 3 or grade 4 toxicity was observed, and 44% of patients with previously treated FL achieved an objective response (27% CR rate) with intravenous doses of 80 to 750 mg/m2 weekly for 4 weeks.7  Preliminary results of a phase I/II study of subcutaneously injected veltuzumab have been reported, with doses ranging from 80 to 320 mg/m2 every 2 weeks for four doses.8  Tolerability and bioavailability were good. Of 15 patients, including 12 with FL, there were eight responders, including three CRs. It is noteworthy that veltuzumab has been associated with responses in patients treated with very low doses administered either intravenously or subcutaneously.

GA101 is a humanized antibody generated from Chinese hamster ovary cells that have been engineered to produce antibodies with afucosylated Fc region carbohydrates. The result is an antibody that binds more strongly to FcγRIII and has improved ADCC.9  As a type II antibody, GA101 lacks the ability to induce CDC, but appears to be better at direct induction of cell death than type I antibodies.9  Data from two phase I clinical trials have been reported. In the first trial, 21 subjects, including 13 with FL, were treated with nine doses of GA101 ranging from 50 to 2000 mg.10  No dose-limiting toxicity was observed, and side effects consisted primarily of grade 1/2 infusion reactions. Five CRs/unconfirmed CRs (CRu) and four partial responses (PRs) were demonstrated. In the second trial, 22 patients (10 with FL) received four weekly doses (100 to 2000 mg), followed by maintenance therapy of one dose every 3 months for up to 2 years.11  Half of all patients were refractory to prior rituximab. Five PRs were observed, and toxicity was similar to the first trial.

AME-133 is a human IgG1 antibody with high affinity for CD20 and an Fc region capable of binding to FcγRIII with significantly greater affinity than rituximab.12  Preclinical studies suggested that AME-133 was more effective than rituximab at natural killer cell activation and could overcome the effects of less favorable FcγRIII polymorphisms.13  A phase I/II trial in patients with relapsed FL (nonrituximab refractory) and at least one allele corresponding to the low affinity FcγRIII is ongoing.

A target antigen should be preferentially expressed on malignant cells, stably expressed on the cell surface, and mediate an important cellular function. The fact that CD20 may not completely fulfill these criteria, yet is remarkably effective as an antibody target, demonstrates that a nonideal antigen can still be valuable.

CD22 is widely expressed on normal and malignant B cells, and it plays a role in the mediation of B-cell activation and adhesion. Unlike CD20, CD22 constitutively internalizes and is degraded without recycling. Epratuzumab is a humanized IgG1 anti-CD22 antibody associated with both ADCC and direct cytotoxicity in preclinical studies.14  Phase I/II studies demonstrated significant objective response rates across various dose levels in both FL and diffuse, large B-cell lymphoma (DLBCL).15,16  Toxicity consisted primarily of infusion-related reactions. Concurrent administration of epratuzumab and rituximab was evaluated in two studies.17,18  Objective responses were noted in about two-thirds of patients with FL and about half of a smaller number of DLBCL subjects, including some CRs. An ongoing study of the Cancer and Leukemia Group B (CALGB) is evaluating the combination of epratuzumab + rituximab in the initial treatment for FL (NCT00553501).

CD37 is expressed at high concentrations on the surface of B cells and mature B-cell lymphomas. TRU-016 is a small, modular immunopharmaceutical comprised of a IgG1 variable regions (VL and VH) and a small, engineered constant region.19  The small size of the SMIP theoretically allows for improved tumor penetration relative to larger monoclonal antibodies. Preclinical testing demonstrated the activity of TRU-016 alone and in combination with other cytotoxic agents in B-cell lymphoma cell lines.20  A phase I trial of TRU-016 in patients with CLL is underway with trials in FL likely to follow (NCT00614042).21 

Milatuzumab is a humanized antibody against CD74. Although the precise role of CD74 is unclear, it appears to function both in antigen presentation, as the invariant chain of class II MHC, and as an accessory-signaling molecule in B-cell differentiation.22–24  Unlike CD20, CD74 is present on the cell surface for a very brief period of time (∼ 10 minutes) before it is endocytosed. Preclinical data demonstrated that milatuzumab was active in vitro and in vivo against B-cell lymphomas and that the mechanism of action was distinct from that of rituximab.25  We are conducting a phase I trial in patients with previously treated B-cell lymphomas (NCT00504972). CD74 may serve as an ideal target for immunoconjugates or combinations with other antibodies that rely on ADCC or CDC.

Galiximab is a chimeric human primate anti-CD80 antibody with single-agent activity and excellent tolerability in previously treated FL.26  Based on promising preclinical data, a phase I/II trial was conducted with the combination of galiximab + rituximab in patients with previously treated FL.27  An ORR of 66% and a 12.1-month median progression-free survival (PFS) were observed. The CALGB subsequently evaluated the combination as initial treatment in FL using four weekly doses of both agents, followed by an “extended induction” of both agents administered once every 2 months for an additional four doses each.28  The ORR was 70%, including a 44% CR/CRu rate. The response rate correlated with the Follicular Lymphoma International Prognostic Index (FLIPI) score, with 75% of patients with a low-risk FLIPI obtaining a CR. This regimen is an example of the potential for combination biologic therapy as an initial approach to FL, which may allow for a less toxic regimen that is sufficiently effective to delay the need for a chemotherapy-containing regimen. Despite the apparent activity of the combination in patients with low-risk FLIPI scores, development of galiximab appears to be on hold, a result of the challenges of drug development in the new era.

Antibodies that target two antigens are termed “bispecific.” Blinatumomab is an anti-CD3/anti-CD19 bispecific antibody that engages T cells and malignant B cells. Phase I studies have demonstrated tolerability and clinical activity in B-cell lymphoma.29  Antibody 22–20 is a hexavalent bispecific antibody composed of epratuzumab conjugated with four Fab regions from veltuzumab.30  The agent is a potent inducer of ADCC and direct cytotoxicity, but not CDC, despite an ability to translocate the target antigens into lipid rafts. Further clinical evaluation will be required to determine if this new class of antibodies is as successful as preclinical testing might suggest.

The use of a radionuclide or a toxin conjugated to a B-cell specific monoclonal antibody allows for another mechanism to induce cell killing beyond the direct antibody and immune effects. Unlike naked antibodies, which appear to exert their effects largely from the cell surface, conjugated antibodies often benefit from internalization. Inotuzumab ozogamicin (CMC-544) is a humanized anti-CD22 antibody conjugated to calicheamicin. In a phase I study, toxicity included thrombocytopenia, asthenia, nausea, and neutropenia. The ORR for all patients treated at all doses was 39%; in FL, 68% of patients responded at the maximum tolerated dose. A follow-up study enrolled 110 patients treated with the combination of CMC-544 and rituximab.31  For 38 patients with recurrent FL, the ORR was 87%, with a median PFS of 23.6 months. Further studies of this drug as a single agent or in combination are ongoing (NCT00868608, NCT0155496).

SAR3419 is an antibody-drug conjugate comprised of a humanized anti-CD19 IgG1 antibody to the tubulin inhibitor DM4, a maytansinoid derivative. A phase I study was conducted in 29 subjects (12 with FL), and used an infusion every 3 weeks, with doses ranging from 10 to 270 mg/m2/dose.32  Dose-limiting toxicity consisted of blurred vision due to corneal changes. Notably, hematologic toxicity was not a significant issue with this regimen. Five objective responses were observed. An additional study evaluating other dosing schedules is currently ongoing (NCT00796731).

The anti-CD22 antibody epratuzumab conjugated with 90Y-DOTA was studied in a phase I trial of fractionated radioimmunotherapy.33  Patients received two or three weekly doses of study drug, with increasing doses of 90Y. The highest total dose received was 45 mCi/m2 (15 mCi/m2 × 3), significantly higher than the maximum dose of 90Y-ibritumomab tiuxetan (a flat dose of 32 mCi). Of the 34 patients with previously treated FL, 74% responded with 62% obtaining a CR/CRu. There was a significant relationship between radiation dose and response rate. We are conducting a phase I/II study of fractionated 90Y-epratuzumab in combination with veltuzumab in patients with untreated follicular lymphoma (NCT01147393).

Intracellular processes relevant to FL include signaling through the B-cell receptor and the PI3K-AKT-mTOR pathway. The development of agents that target these pathways is covered in another article in this section by Drs. Witzig and Gupta. Other relevant targets include bcl-6, the mdm2-p53 axis, the Bcl-2 family, and the ubiquitin-proteasome pathway.

The proteasome plays a vital role in the regulation of several cell mechanisms important to lymphoma, including the cell cycle (via the cyclins, the cyclin-depending kinases (cdk), and the cdk inhibitors), tumor suppression (via p53), and gene transcription (via NF-κB [nuclear factor-κ light-chain enhancer of activated B cells]).34  The precise mechanism by which proteasome inhibition induces cell death is unclear and may be related to a variety of these pathways, depending on the setting, highlighting the fact that more than drugs with one mechanism of action may have advantages over those that target a single tyrosine kinase.

Bortezomib is a small molecule inhibitor of the proteosome approved in the United States for the treatment of multiple myeloma and mantle cell lymphoma. Bortezomib has been evaluated in a number of phase II studies in patients with FL.35–37  Interestingly, the median time to response to bortezomib in FL appears longer than in other B-cell malignancies, the significance of which is unclear.38  DeVos and colleagues39  evaluated the combination of bortezomib with rituximab in a phase II study that enrolled subjects with previously treated FL or marginal zone lymphoma not refractory to prior rituximab. In arm A, patients received bortezomib 1.3 mg/m2 twice weekly for 2weeks in each 3-week cycle + rituximab 375 mg/m2 weekly for the first 4 weeks only (N = 41), whereas in arm B, patients received bortezomib 1.6 mg/m2 weekly for 4 weeks in each 5-week cycle + rituximab 375 mg/m2 weekly for the first 4 weeks only (N = 40). The response rates and time to progression were similar (49% vs 43% and 7 months vs 10 months in arm A vs arm B, respectively), whereas the weekly regimen appeared slightly better tolerated. A phase II randomized trial of rituximab alone or in combination with bortezomib is ongoing. The National Cancer Institute of Canada recently reported preliminary results from a phase II trial in which bortezomib 1.3 mg/m2 on days 1 and 8 was added to standard rituximab, cyclophosphamide, vincristine, and prednisone (R-CVP) in patients with advanced-stage FL.40  Six of 95 patients reported grade 3 neurotoxicity, with no grade 4 events. The overall response and CR rates were 84.6% and 47%, respectively. In a similar phase I trial, two different schedules of bortezomib were substituted for vincristine in R-CVP in patients with previously treated indolent or mantle cell lymphoma.41  Two of four patients treated with twice weekly bortezomib experienced grade 3/4 neurotoxicity. The ORR was 64% in the twice-weekly group, suggesting that targeted agents have the potential to replace traditional cytotoxic chemotherapeutic agents as components of some regimens.

The novel proteasome inhibitor MLN9708 has demonstrated activity in preclinical lymphoma models and is currently undergoing evaluation in a phase I trial in patients with relapsed lymphoma.42  MLN9708 also exists in oral form and is undergoing evaluation in multiple myeloma. Carfilzomib is a novel proteasome inhibitor with significant activity in multiple myeloma and no reported evidence of neurotoxicity. Two phase I studies have been conducted in patients with relapsed hematologic malignancies. Further experience in FL will be required to determine its activity and safety profile.43,44 

Bcl-2 Inhibitors

The Bcl-2 family is a group of proteins that share at least one of four Bcl homology domains (BH1-BH4). Family members may be either proapoptotic (eg, Bax, Bak) or antiapoptotic (eg, Bcl- XL, Bcl-2), depending on the domains expressed. The 14;18 translocation, which occurs in the vast majority of FL, results in juxtaposition of the BCL2 gene next to the immunoglobulin heavy chain gene, resulting in constitutive expression. At least three Bcl-2 inhibitors are currently in development in B-cell malignancies. Apogossypol is an orally active analog of gossypol. The negative enantiomer of gossypol, AT-101, demonstrated some activity in patients with CLL, but was associated with dose-limiting hepatotoxicity and gastrointestinal toxicity.45  A subsequent two-stage phase II trial of AT-101 + rituximab in patients with FL did not meet the threshold to proceed to stage II, perhaps related to limitations in the dose.46  In preclinical testing, apogossypol was less toxic than gossypol and had greater activity.47  Clinical testing will be required to validate the preclinical results. ABT-263 is an orally available, small molecule BH3 mimetic that primarily inhibits Bcl-XL. In a phase I/II study, patients with previously treated lymphoid malignancies were treated with cycles of either 14 days of ABT-263 followed by 1 week off or 21 days of continuous ABT-263.48  Thrombocytopenia was common due to the expression of Bcl-XL in megakaryocytes, but appeared to be better with continuous dosing over intermittent dosing. Activity was seen primarily in CLL, suggesting that combination with other agents may be required for maximal effect in other lymphomas. Obatoclax is novel BH3 mimetic that appears to sensitize rituximab-resistant cells to treatment with bortezomib.49  The combination has undergone phase I testing in patients with mantle cell lymphoma and is currently undergoing additional clinical evaluation in combination with multiple other agents.50 

mdm2 Inhibitors

Dysfunction of the p53 tumor suppressor has been implicated in multiple cancers. The human homolog of murine double-minute protein 2 (mdm2) is a natural inhibitor of p53 and is frequently upregulated in FL. Nutlin-3 is a novel small molecule antagonist of mdm2 with preclinical activity in mantle cell lymphoma.51  This drug and other small molecule mdm2 inhibitors are most likely to demonstrate activity in those patients with unmutated TP53 genes, offering a potential pretreatment predictor of response.

Bcl-6 Inhibitors

The bcl-6 protein is a transcriptional repressor expressed in germinal center B cells. Translocations involving the BCL6 gene are common in FL and appear to be a harbinger of high-grade transformation.52  Hsp90 stabilizes bcl-6, allowing it to have a greater effect on target genes. The hsp90 inhibitor PU-H71 has demonstrated preclinical activity in bcl-6–dependent large-cell lymphomas, making it potentially relevant for clinical development in B-cell malignancies derived from the germinal center, including FL.53 

Lenalidomide

The immunomodulatory agent lenalidomide is approved for the treatment of patients with myeloma and myelodysplastic syndromes. The precise mechanism of action of lenalidomide remains unclear, but likely involves multiple effects on the tumor microenvironment, including downregulation of tumor necrosis factor-α, interleukin-6, interleukin-8, vascular endothelial growth factor, and activation of T and NK cells.54,55  Several studies have demonstrated the utility of this novel agent in non-Hodgkin lymphoma, including FL. In an international phase II study, patients with previously treated indolent lymphoma received lenalidomide 25 mg once daily on days 1 to 21 of every 28-day cycle.56  The ORR was 23% (27% in FL), with a median PFS of 4.4 months. The most common grade 3/4 events were neutropenia and thrombocytopenia. Based on preclinical data that the activation of NK cells by lenalidomide may improve rituximab-induced ADCC, several groups have undertaken studies of the combination. Sixteen patients with relapsed indolent lymphoma received lenalidomide, initially as described previously, but subsequently lowered to 20 mg due to tumor lysis, plus four weekly doses of rituximab beginning on day 15 of cycle 1 and repeated for four additional doses the patient had less than a CR after cycle 2.57  In addition to two patients with grade 3 tumor lysis, fatigue (12%), neutropenia (18%), and hyponatremia (18%) were the most common grade 3/4 adverse events. Twelve patients responded, including five patients with a CR/CRu. An ongoing phase II study, under the auspices of the CALGB, randomizes patients to lenalidomide alone or in combination with rituximab (NCT00238238). Patients are required to have relapsed disease following rituximab therapy, but must not be rituximab resistant. The primary objectives are overall response and time-to-disease progression, with one aim to determine whether rituximab augments the effects of lenalidomide. In an approach that could change the management of patients with newly diagnosed, low FLIPI-risk FL, Fowler and colleagues58  tested lenalidomide 20 mg on days 1 to 21 + rituximab on day 1 for up to six cycles in patients with untreated indolent lymphoma. Preliminary results in 19 patients demonstrated an ORR of 86%, with a 75% CR/CRu rate. Longer follow-up with a larger number of patients will be required to determine whether this approach is worth carrying forward to a phase III study.

Targeting T-Regulatory Cells

Another approach to targeting the tumor microenvironment in FL involves interference with T-regulatory (T-regs) cells in tumor masses that may interfere with the host immune response against the tumor. Denileukin diftitox is an immunotoxin directed against the CD25 antigen expressed on T-regs. Ongoing studies are assessing the combination of denileukin diftitox in combination with rituximab, with the hope that depletion fo T-regs will enhance the antilymphoma effects of the anti-CD20 antibody (NCT00460109).

Therapeutic monoclonal antibodies have provided significant benefit for patients with FL. Essentially all patients with B-cell lymphoma receive rituximab at multiple times over their treatment. Novel anti-CD20 agents offer the potential for enhanced activity relative to that of rituximab, although clinical data have not been compelling to date. Agents directed against different targets offer the possibility of combination with rituximab or significant activity on their own. Many challenges exist in determining the optimal use of novel agents. New anti-CD20 antibodies will require randomized comparative trials or demonstration of effectiveness in rituximab refractory patients (elusive thus far). Novel combinations also require vetting through comparative studies with rituximab alone. Whether or not such combinations are effective, and warrant the associated costs, remains to be seen.

The broad array of novel agents, heterogeneous clinical behavior, and range of standard therapies presents difficulties in development of novel agents for FL. One way to overcome these challenges may be to explore the use of new agents as part of initial therapy, where the disease may be somewhat more uniform in its characteristics and where the variables of prior therapy are eliminated. The CALGB has been exploring novel combinations of biologic agents (without chemotherapy) as initial treatment for FL.

Despite the challenges, the promise of novel therapeutic agents in FL suggests the possibility that better tolerated and more effective treatments are on the horizon to help clinicians and researchers continue to improve outcomes for patients.

Conflict-of-interest disclosure: J.P.L. has either consulted for and/or received honoraria from Hospira, Cell Therapeutics, Pfizer, Celgene, GlaxoSmithKline, Biogen IDEC, Celgene, Calistoga, Johnson & Johnson, EMD Serono, sanofi-aventis, Millenium, Biotest, Cephalon, Pharmion, Eisai, Cougar Biotechnology, Genentech, Novartis, and Immunomedics. P.M. has either consulted for and/or received honoraria from Facet and Genentech.

Off-label drug use: Off-label and investigational lymphoma therapies.

John P. Leonard, MD, Clinical Director, Center for Lymphoma and Myeloma, and Professor, Division of Hematology and Medical Oncology, Weill Cornell Medical College and New York Presbyterian Hospital, 525 East 68th St., Box 403, New York, NY 10065; Phone: (646) 962-2068; Fax: (646) 962-1605; e-mail: jpleonar@med.cornell.edu

1
Teeling
JL
French
RR
Cragg
MS
et al
Characterization of new human CD20 monoclonal antibodies with potent cytolytic activity against non-Hodgkin lymphomas
Blood
2004
104
1793
1800
2
Teeling
JL
Mackus
WJM
Wiegman
LJJM
et al
The biological activity of human CD20 monoclonal antibodies is linked to unique epitopes on CD20
J Immunol
2006
177
362
371
3
Craigen
JL
Mackus
WJM
Engleberts
P
et al
Ofatumumab, a human Mab targeting a membrane-proximal small-loop epitope on CD20, induces potent NK cell-mediated ADCC [abstract 1725]
Blood
2009
114
4
Hagenbeek
A
Gadeberg
O
Johnson
P
et al
First clinical use of ofatumumab, a novel fully human anti-CD20 monoclonal antibody in relapsed or refractory follicular lymphoma: results of a phase 1/2 trial
Blood
2008
111
5486
5495
5
Hagenbeek
A
Fayad
L
Delwail
V
et al
Evaluation of ofatumumab, a vovel human CD20 monoclonal antibody, as single agent therapy in rituximab-refractory follicular lymphoma [abstract 935]
Blood
2009
114
6
Stein
R
Qu
Z
Chen
S
et al
Characterization of a new humanized anti-CD20 monoclonal antibody, IMMU-106, and its use in combination with the humanized anti-CD22 antibody, epratuzumab, for the therapy of non-Hodgkin's lymphoma
Clin Cancer Res
2004
10
2868
2878
7
Morschhauser
F
Leonard
JP
Fayad
L
et al
Humanized anti-CD20 antibody, veltuzumab, in refractory/recurrent non-Hodgkin's lymphoma: phase I/II results
J Clin Oncol
2009
27
3346
3353
8
Negrea
OG
Allen
SL
Rai
KR
et al
Subcutaneous injections of low doses of humanized anti-CD20 veltuzumab for treatment of indolent B-cell malignancies [abstract 3757]
Blood
2009
114
9
Mössner
E
Brünker
P
Moser
S
et al
Increasing the efficacy of CD20 antibody therapy through the engineering of a new type II anti-CD20 antibody with enhanced direct and immune effector cell-mediated B-cell cytotoxicity
Blood
2010
115
4393
4402
10
Salles
G
Morschhauser
F
Lamy
T
et al
Phase I study of RO5072759 (GA101) in patients with relapsed/refractory CD20+ non-Hodgkin lymphoma (NHL) [abstract 1704]
Blood
2009
114
11
Sehn
LH
Assouline
SE
Stewart
DA
et al
A phase I study of GA101 (RO5072759) monotherapy followed by maintenance in patients with multiply relapsed/refractory CD20+ malignant disease [abstract 934]
Blood
2009
114
12
Bowles
JA
Wang
S-Y
Link
BK
et al
Anti-CD20 monoclonal antibody with enhanced affinity for CD16 activates NK cells at lower concentrations and more effectively than rituximab
Blood
2006
108
2648
2654
13
Weiner
GJ
Bowles
JA
Link
BK
Campbell
MA
Wooldridge
JE
Breitmeyer
JB
Anti-CD20 monoclonal sntibody (mAb) with enhanced affinity for CD16 activates NK cells at lower concentrations and more effectively than rituximab® [abstract 348]
Blood
2005
106
14
Carnahan
J
Stein
R
Qu
Z
et al
Epratuzumab, a CD22-targeting recombinant humanized antibody with a different mode of action from rituximab
Mol Immunol
2007
44
1331
1341
15
Leonard
JP
Coleman
M
Ketas
JC
et al
Phase I/II trial of epratuzumab (humanized anti-CD22 antibody) in indolent non-Hodgkin's lymphoma
J Clin Oncol
2003
21
3051
3059
16
Leonard
JP
Coleman
M
Ketas
JC
et al
Epratuzumab, a humanized anti-CD22 antibody, in aggressive non-Hodgkin's lymphoma: phase I/II clinical trial results
Clin Cancer Res
2004
10
5327
5334
17
Leonard
JP
Coleman
M
Ketas
J
et al
Combination antibody therapy with epratuzumab and rituximab in relapsed or refractory non-Hodgkin's lymphoma
J Clin Oncol
2005
23
5044
5051
18
Strauss
SJ
Morschhauser
F
Rech
J
et al
Multicenter phase II trial of immunotherapy qith the humanized anti-CD22 antibody, epratuzumab, in combination with rituximab, in refractory or recurrent non-Hodgkin's lymphoma
J Clin Oncol
2006
24
3880
3886
19
Zhao
X
Lapalombella
R
Joshi
T
et al
Targeting CD37-positive lymphoid malignancies with a novel engineered small modular immunopharmaceutical
Blood
2007
110
2569
2577
20
Baum
PR
Cerveny
C
Gordon
B
et al
Evaluation of the effect of TRU-016, an anti-CD37 directed SMIP in combination with other therapeutic drugs in models of non-Hodgkin's lymphoma [abstract 8571]
J Clin Oncol
2009
27
21
Andritsos
L
Furman
R
Flinn
IW
et al
A phase I trial of TRU-016, an anti-CD37 small modular immunopharmaceutical (SMIP) in relapsed and refractory CLL [abstract 3017]
J Clin Oncol
2009
27
22
Cresswell
P
Assembly, transport, and function of MHC class II molecules
Ann Rev Immunol
1994
12
259
291
23
Leng
L
Metz
CN
Fang
Y
et al
MIF signal transduction initiated by binding to CD74
J Exp Med
2003
197
1467
1476
24
Matza
D
Lantner
F
Bogoch
Y
Flaishon
L
Hershkoviz
R
Shachar
I
Invariant chain induces B cell maturation in a process that is independent of its chaperonic activity
Proc Natl Acad Sci U S A
2002
99
3018
3023
25
Stein
R
Qu
Z
Cardillo
TM
et al
Antiproliferative activity of a humanized anti-CD74 monoclonal antibody, hLL1, on B-cell malignancies
Blood
2004
104
3705
3711
26
Czuczman
MS
Thall
A
Witzig
TE
et al
Phase I/II study of galiximab, an anti-CD80 antibody, for relapsed or refractory follicular lymphoma
J Clin Oncol
2005
23
4390
4398
27
Friedberg
JW
Leonard
JP
Younes
A
et al
Updated results from a phase II study of galiximab (anti-CD80) in combination with rituximab for relapsed or refractory, follicular NHL [abstract 2435]
Blood
2005
106
28
Czuczman
MS
Leonard
JP
Johnson
JL
et al
FLIPI score is applicable and predictive of response to upfront immunotherapy in CALGB 50402: phase II trial of extended induction galiximab ([G] anti-CD80 monoclonal antibody) plus rituximab [R] [abstract 1003]
Blood
2008
112
29
Bargou
R
Leo
E
Zugmaier
G
et al
Tumor regression in cancer patients by very low doses of a T cell engaging antibody
Science
2008
321
5891
974
977
30
Rossi
EA
Goldenberg
DM
Cardillo
TM
Stein
R
Chang
C-H
Hexavalent bispecific antibodies represent a new class of anticancer therapeutics: 1. Properties of anti-CD20/CD22 antibodies in lymphoma
Blood
2009
113
6161
6171
31
Dang
NH
Smith
MR
Offner
F
et al
Anti-CD22 immunoconjugate inotuzumab ozogamicin (CMC-544) + rituximab: clinical activity including survival in patients with recurrent/refractory follicular or ‘aggressive’ lymphoma [abstract 584]
Blood
2009
114
32
Younes
A
Gordon
L
Kim
S
et al
Phase I multi-dose escalation study of the anti-CD19 maytansinoid immunoconjugate SAR3419 administered by intravenous (IV) infusion every 3 weeks to patients with relapsed/refractory B-cell non-Hodgkin's lymphoma (NHL) [abstract 585]
Blood
2009
114
33
Kraeber-Bodere
F
Morschhauser
F
Huglo
D
et al
Fractionated radioimmunotherapy in NHL with DOTA-conjugated, humanized anti-CD22 IgG, epratuzumab: results at high cumulative doses of 90Y [abstract 8502]
J Clin Oncol
2008
26
34
Palombella
VJ
Rando
OJ
Goldberg
AL
Maniatis
T
The ubiquitin proteasome pathway is required for processing the NF-κB1 precursor protein and the activation of NF-κB
Cell
1994
78
773
785
35
O'Connor
OA
Wright
J
Moskowitz
C
et al
Phase II clinical experience with the novel proteasome inhibitor bortezomib in patients with indolent non-Hodgkin's lymphoma and mantle cell lymphoma
J Clin Oncol
2005
23
676
684
36
Strauss
SJ
Maharaj
L
Hoare
S
et al
Bortezomib therapy in patients with relapsed or refractory lymphoma: potential correlation of in vitro sensitivity and tumor necrosis factor alpha response with clinical activity
J Clin Oncol
2006
24
2105
2112
37
Goy
A
Younes
A
McLaughlin
P
et al
Phase II study of proteasome inhibitor bortezomib in relapsed or refractory B-cell non-Hodgkin's lymphoma
J Clin Oncol
2005
23
667
675
38
O'Connor
OA
Portlock
C
Moskowitz
C
et al
Time to treatment response in patients with follicular lymphoma treated with bortezomib is longer compared with other histologic subtypes
Clin Cancer Res
2010
16
719
726
39
de Vos
S
Goy
A
Dakhil
SR
et al
Multicenter randomized phase II study of weekly or twice-weekly bortezomib plus rituximab in patients with relapsed or refractory follicular or marginal-zone B-cell lymphoma
J Clin Oncol
2009
27
5023
5030
40
Sehn
LH
Macdonald
DA
Rubin
SH
et al
Bortezomib added to CVP-R is safe and effective for previously untreated advanced stage follicular lymphoma: a phase II study by the NCIC Clinical Trials Group [abstract 407]
Blood
2009
114
41
Gerecitano
J
Portlock
CS
Hamlin
PA
Jr
et al
Rituximab, cyclophosphamide, bortezomib and prednisone (R-CBorP): final results of a phase I trial evaluating two dosing schedules and the safety of overlapping pegfilgrastim p with relapsed/refractory indolent and mantle cell lymphomas [abstract 3708]
Blood
2009
114
42
Donelan
J
Bannerman
B
Bano
K
et al
Antitumor activity of MLN9708, a second-generation proteasome inhibitor, in preclinical models of lymphoma [abstract 2724]
Blood
2009
114
43
Alsina
M
Trudel
S
Vallone
M
Molineaux
C
Kunkel
L
Goy
A
Phase 1 single aagent antitumor activity of twice weekly consecutive day dosing of the proteasome inhibitor carfilzomib (PR-171) in hematologic malignancies [abstract 411]
Blood
2007
110
44
O'Connor
OA
Stewart
AK
Vallone
M
et al
A phase 1 dose escalation study of the safety and pharmacokinetics of the novel proteasome inhibitor carfilzomib (PR-171) in patients with hematologic malignancies
Clin Cancer Res
2009
15
7085
7091
45
James
DF
Castro
JE
Loria
O
Prada
CE
Aguillon
RA
Kipps
TJ
AT-101, a small molecule Bcl-2 antagonist, in treatment naive CLL patients (pts) with high risk features; preliminary results from an ongoing phase I trial [abstract 6605]
J Clin Oncol
2006
24
46
Kingsley
E
Richards
D
Garbo
L
et al
An open-label, multicenter, phase II study of AT-101 in combination with rituximab® in patients with untreated, grade 1–2, follicular non-Hodgkin's lymphoma (FL) [abstract 8582]
J Clin Oncol
2009
27
47
Kitada
S
Kress
CL
Krajewska
M
Jia
L
Pellecchia
M
Reed
JC
Bcl-2 antagonist apogossypol (NSC736630) displays single-agent activity in Bcl-2-transgenic mice and has superior efficacy with less toxicity compared with gossypol (NSC19048)
Blood
2008
111
3211
3219
48
Wilson
WH
O'Connor
OA
Czuczman
MS
et al
Phase 1/2a study of ABT-263 in relapsed or refractory lymphoid malignancies [abstract 1711]
Blood
2009
114
49
Hernandez-Ilizaliturri
F
Tsio
P-C
Campagna
R
et al
Obatoclax and bortezomib therapy results in disruption of the p53-mediated apoptosis/autophagy pathway and is associated with potent synergistic anti-tumor activity against rituximab-chemotherapy-sensitive and -resistant B-cell lymphoma [abstract 288]
Blood
2009
114
50
Goy
A
Ford
P
Feldman
T
et al
A phase 1 trial of the pan Bcl-2 family inhibitor obatoclax mesylate (GX15–070) in combination with bortezomib in patients with relapsed/refractory mantle cell lymphoma [abstract 2569]
Blood
2007
110
51
Tabe
Y
Sebasigari
D
Jin
L
et al
MDM2 antagonist nutlin-3 displays antiproliferative and proapoptotic activity in mantle cell lymphoma
Clin Cancer Res
2009
15
933
942
52
Akasaka
T
Lossos
IS
Levy
R
BCL6 gene translocation in follicular lymphoma: a harbinger of eventual transformation to diffuse aggressive lymphoma
Blood
2003
102
1443
1448
53
Cerchietti
LC
Lopes
EC
Yang
SN
et al
A purine scaffold Hsp90 inhibitor destabilizes BCL-6 and has specific antitumor activity in BCL-6-dependent B cell lymphomas
Nat Med
2009
15
1369
1376
54
Anderson
KC
Lenalidomide and thalidomide: mechanisms of action—similarities and differences
Semin Hematol
2005
42
S3
S8
55
Chang
DH
Liu
N
Klimek
V
et al
Enhancement of ligand-dependent activation of human natural killer T cells by lenalidomide: therapeutic implications
Blood
2006
108
618
621
56
Witzig
TE
Wiernik
PH
Moore
T
et al
Lenalidomide oral monotherapy produces durable responses in relapsed or refractory indolent non-Hodgkin's lymphoma [abstract 1169]
J Clin Oncol
2009
27
712
719
57
Dutia
M
DeRoock
I
Chee
K
et al
R2: preliminary results of a phase II study of lenalidomide and rituximab in relapsed/refractory indolent non-Hodgkin's lymphoma (NHL) [abstract 1679]
Blood
2009
114
670
58
Fowler
N
McLaughlin
P
Hagemeister
FB
et al
A biologic combination of lenalidomide and rituximab for front-line therapy of indolent B-cell non-Hodgkin's lymphoma [abstract 1714]
Blood
2009
114