Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2011 Apr 1.
Published in final edited form as: Semin Hematol. 2010 Apr;47(2):124–132. doi: 10.1053/j.seminhematol.2010.01.006

Antigenic Modulation and Rituximab Resistance

Ronald P Taylor 1, Margaret A Lindorfer 1
PMCID: PMC2848181  NIHMSID: NIHMS170991  PMID: 20350659

Abstract

Several types of B cell lymphoma have been successfully treated with rituximab (RTX), and approval by the FDA for use of RTX in the treatment of rheumatoid arthritis has increased interest in targeting CD20 on B cells for other indications. Although large amounts of RTX can be infused into humans with no apparent dose-limiting toxicity, recent evidence suggests that the body’s effector mechanisms, including complement-mediated cytotoxicity and NK cell-mediated killing, can be saturated or exhausted at high burdens of RTX-opsonized B cells. One of the consequences of this saturation phenomenon is that the opsonized B cells are instead processed by a different pathway mediated by FcγR on effector cells. In this alternative pathway both RTX and CD20 are removed (“shaved”) from the B cells and are taken up by monocytes/macrophages. This process, formerly called antigenic modulation, appears to occur in several compartments in the body and may play a key role in the development of resistance to rituximab therapy.

INTRODUCTION

The mechanisms of B cell resistance to rituximab (RTX)-mediated cytotoxicity, can, like much of immunology, be divided into innate and adaptive components. Innate components include expression of CD20 at levels below the thresholds required to trigger RTX-mediated cytotoxicity, and the activity and level of complement control proteins on malignant cells.14 A patient’s innate cytotoxic effector mechanisms will play critical roles, as demonstrated by differences in efficacy which correlate with polymorphisms in C1q and polymorphisms in and effector cell densities of FcγR.59 In addition, as noted by Smith,10 differences in resistance of cells in different compartments “could reflect differential access to antibody or to effector mechanisms,” emphasizing the importance of tumor microenvironments.

Treatment with RTX does not destroy all malignant B cells, and over the long term (periods in excess of one year) surviving B cells appear to acquire an adaptive resistance to later RTX therapies,11,12 although in many cases the malignant cells still express CD20. The surviving cells may take up residence in compartments in which they will be resistant to later attack by RTX, but there is no evidence for or against this possibility. Over the short term, that is, at the time of RTX treatment, there are several adaptive mechanisms by which targeted cells can escape RTX-mediated cytotoxicity. As noted below, deposition of C3 activation fragments on RTX-opsonized cells can block binding to FcγRIIIa on NK cells, inhibiting antibody-dependent cellular cytotoxicity (ADCC).13 Although not yet formally proven for RTX, there is evidence that sublytic complement attack on nucleated cells induces Ca2+ fluxes, leading to protein synthesis and at least temporary upregulation of defenses against complement-dependent cytotoxicity (CDC).14 RTX treatment may acutely exhaust effector mechanisms responsible for killing IgG-opsonized substrates. Under these conditions an alternative processing pathway which removes RTX/CD20 complexes from B cells predominates, thus insuring the resistance of CD20-negative cells to RTX therapy. Table I summarizes some of the mechanisms by which resistance to RTX develops and also presents potential therapeutic strategies to overcome this resistance. In this review we will focus our discussion on the loss of efficacy of RTX treatment due to blockade or saturation of host effector mechanisms.

Table I.

Mechanisms of Resistance to Rituximab Therapy and Potential Therapeutic Strategies to Overcome Resistance

Mechanism Potential Therapeutic Strategies Ref
Loss of epitope
Permanent loss of CD20 (i.e. outgrowth of a CD20-negative clone) Target alternative epitope (e.g. CD52) 11,64
Temporary mAb-mediated loss of CD20 (i.e. Shaving) Lower, more frequent doses of mAb; IVIG or other agents to block FcRγI 15,17,62,72
Reduced ADCC
Exhaustion of ADCC Lower, more frequent doses of mAb; increase activity or number of NK cells 3941,46,47
Blockade of ADCC by deposited C3 activation fragments Engineer mAbs, or adjust the dose in order to maximize ADCC while minimizing C3 fragment deposition 13,32
Polymorphisms in effector molecules (i.e. FcγR) Screen polymorphisms prior to therapy; engineer Fc region of mAbs to maximize ADCC for all patients 57,4345
Reduced CDC
Increased surface expression of complement control proteins Block the action of CD55 or CD59 73
Exhaustion of CDC (i.e. temporary depletion of complement components) Infusion of fresh frozen plasma; lower, more frequent doses of mAb. 15,17,52
Sublytic C attack leading to increased resistance to CDC mAb designed to maximize C activation and thereby minimize sublytic C attack 14,33,35
Tumor microenvironment
Protective factors in tumor microenvironment Mobilize malignant cells to periphery; attack stromal factors 7476

CYTOTOXIC MECHANISMS: VARIATIONS ON IMMUNE COMPLEX PROCESSING

Seven years ago our laboratory initiated a clinical study in chronic lymphocytic leukemia (CLL), to determine whether infusion of RTX promotes complement activation and deposition of C3 activation fragments on circulating CLL B cells.15 We had demonstrated that in vitro binding of RTX to B cells in the presence of complement leads to deposition of C3 fragments on the cells; our findings in primates indicated that within minutes of RTX infusion, C3 fragments were deposited on RTX-opsonized cells.3,16 In both cases, C3b/iC3b was co-localized with cell-bound RTX. In the clinical study, we found that after infusion of only 30 mg of RTX, C3 fragments were demonstrable on circulating B cells. However, an unexpected observation set the stage for future investigations: The number of circulating CLL B cells decreased considerably after infusion of 30 mg but then substantially increased after infusion of the remaining ~600 mg RTX (~ 5 hours later). Although the final concentration of RTX in the bloodstream (>100 ug/ml) was considerably higher than the concentration after infusion of the first 30 mg (<5 ug/ml), it appeared that the cytotoxic action of RTX had been saturated or exhausted. That is, B cells were not cleared from the bloodstream with a high level of efficacy. This finding has been quite durable (Table II); we observed this pattern in all of our studies of CLL patients receiving single agent RTX therapy in doses of ≥100 mg.17 These observations dramatically emphasize the need for detailed analyses of the cytotoxic mechanisms of RTX in different compartments in the body and in the bloodstream in particular.

Table II.

Key Measurements on CLL Blood Samples, Before, During and After RTX Infusion

Before treatment After 30 mg After 375 mg/m2
Normalized lymphocyte count Higha 100% Low 20% Intermediate 60%
Plasma RTX concentration 0 1–5 ug/ml >100 ug/ml
CH50 Normal Normal Depleted (10–90%)
CD20/cell, normalized 100 2–10 2–10
C3 fragments deposited No Yes Yes
a

Pretreatments levels varied from 5,000–100,000 lymphocytes/uL.15,17

An overwhelming body of evidence indicates that RTX-mediated cytotoxicity must rely on the body’s immune mechanisms.13,9,15,1823 These cytotoxic reactions are initiated by recognition of the RTX-CD20-B cell immune complex, either by FcγR on effector cells (NK cells and monocyte/macrophages) or by C1q, the first component of the classical pathway of complement. Recognition of IgG–opsonized substrate cells by FcγRIIIa on NK cells can promote ADCC.18,24,25 IgG-opsonized cells can also bind to macrophages via several different FcγR, setting in motion cytotoxic pathways that lead to ADCC and phagocytosis.19,26 The classic studies of Frank and Schreiber, which examined bloodstream clearance mechanisms mediated by the mononuclear phagocytic system (MPS), revealed that binding of a few thousand IgG molecules to a circulating cell such as an erythrocyte (E) leads to its rapid clearance due to recognition by FcγR on tissue macrophages in the liver (Kupffer cells) and spleen.26,27 Studies of polymorphisms in CD16 (FcγRIIIa), first reported by Cartron et al. and confirmed by others57, indicate that individuals homozygous for the higher affinity binding FcγRIIIa allotype (158 Valine) have considerably better responses to RTX therapy. This result underlines the importance of ADCC and phagocytosis in the RTX mechanism.

IgG-opsonized cells can bind C1q, promoting several consecutive steps in the classical complement activation pathway, including covalent deposition of C3 activation fragments on the cells, followed by CDC due to assembly and penetration of the cell membrane by numerous copies of the cytolytic membrane attack complex (MAC).23 RTX can mediate CDC of B cells that express high levels of CD20. The results in certain mouse models,28,29 as well as the report of Racila et al.,8 which correlated the response of patients with follicular lymphoma to polymorphisms in C1q, all suggest that CDC may play a role in the therapeutic action of RTX. However, primary B cells from most CLL patients are refractory to RTX-mediated CDC due to relatively low levels of expression of CD20. Upon infusion of RTX, circulating CLL cells that are not lysed become heavily opsonized with C3 fragments;3,17 these C3 fragments may promote clearance of the opsonized cells by the MPS, due to uptake mediated by complement receptors on fixed cells.30,31 Conversely, several lines of evidence developed by Weiner and colleagues suggest that C3 fragments bound to RTX-opsonized B cells may actually interfere with NK cell-promoted ADCC due to C3b/iC3b-mediated blockade of access of NK cell CD16 to the Fc region of cell-bound RTX.13,32 Thus, it will be important to determine the relative impact of C3 fragment deposition in targeting RTX-opsonized circulating cells to the MPS compared to the effect of complement activation on sessile, non-circulating cells in tumors.

These potential problems may be overcome with second generation anti-CD20 mAbs such as ofatumumab. Ofatumumab mediates CDC of B cells more effectively than RTX, most likely due to increased binding of C1q to opsonized cells.33 Indeed, ofatumumab makes much more efficient use of complement than does RTX and can, in the presence of limited amounts of complement, mediate much higher levels of C3b deposition and CDC than does RTX in the presence of much larger amounts of complement.3335 Based on these observations, treatment of patients with high B cell burdens with rituximab is more likely to consume complement due to inefficient cell killing compared to treatment with comparable amounts of ofatumumab, and future clinical studies should allow for evaluation of this prediction.

SATURATION and/or EXHAUSTION

A key question is whether the body’s effector mechanisms associated with immune complex processing, complement activation, phagocytosis and/or ADCC, can be saturated or exhausted, leading to persistence and proliferation of malignant B cells that would otherwise be eliminated. That is, the B cell burden may be so high that although large doses of RTX saturate CD20 on targeted B cells in different body compartments, the necessary effector mechanisms within these compartments are not adequate to effectively kill targeted cells.

Insight into this question may be drawn from studies in systemic lupus erythematosus (SLE). SLE is an autoimmune disease characterized by chronic proliferation of large quantities of immune complexes in the circulation and in tissues.3638 The kinetics of clearance from the circulation of a model particulate immune complex, 51Cr-labeled IgG-opsonized E, are reduced substantially in SLE, likely due to competition by circulating immune complexes.36 These IgG-containing complexes can bind to FcγR on monocytes/macrophages, and then the immune complexes, along with FcγR, are internalized. This process appears to down-modulate expression of FcγRII and FcγRIII, compromising the activity of monocyte/macrophages in clearing IgG-opsonized particles.38

In another example of saturation and exhaustion, Bowles and Weiner reported that NK cell-mediated ADCC of RTX-opsonized cells comes at a price, in that CD16 (FcγRIIIa) on the NK cells is severely down-regulated.39 The consequence of down-regulation is inhibition of killing of additional RTX-opsonized B cells until CD16 can be re-expressed. In vitro experiments reported by Bhat and Watzl suggest that one NK cell can kill at most 4 IgG-opsonized cells.40 In vivo findings of Beredeja et al. indicate that after NK cell-mediated ADCC, re-expression of CD16 on the NK cell and restoration of full cytotoxic activity may require 24 hours or more.41 Varchetta et al. reported that as a consequence of NK cell-mediated ADCC of trastuzumab-opsonized cells, CD16 on the NK cells is down-modulated, suggesting that NK cell-mediated ADCC of other targets may also be subject to saturation.42 In the future, anti-CD20 mAbs with Fc regions engineered to maximize ADCC may provide increased efficacy and methods to increase NK cell cytotoxicity and/or numbers are under development.4347

Treatment of immune thrombocytopenia (ITP) with IgG anti-E antibodies (anti-D) makes explicit use of saturation of effector mechanisms.48 This treatment leads to modest decreases in the hematocrit due to clearance of IgG-opsonized E; however, the decoy IgG-E immune complexes saturate the clearance capacity of the MPS, sparing IgG-opsonized platelets and providing effective therapy for certain patients.49 Part of this saturation phenomenon is likely due to down-modulation of FcγRIII. Song et al. examined a mouse model for this system and reported that as a result of targeting E by infusion of specific IgG antibodies, splenic FcγRIII was down-modulated.50 It is likely that splenic FcγRIII was internalized coincident with phagocytosis of IgG-opsonized E.

The complement system can be exhausted as a consequence of RTX treatment. RTX-opsonized B cells at densities seen in CLL (100,000 B cells per ul) substantially depleted complement activity in serum.15 Although the cells were saturated with bound RTX, CDC was reduced considerably compared to CDC seen for lower cell burdens, where complement activity was adequate. The first CLL patient we studied had a moderate burden of circulating B cells (< 20,000 per ul), yet RTX treatment led to massive depletion of complement immediately after the first infusion.15 His complement titer was partially restored one week later, but after the second weekly RTX treatment (in a 4-week cycle) complement remained low for more than a month, suggesting that the RTX-B cell immune complexes, in several compartments in addition to the bloodstream, were continuing to activate and deplete complement. Complement titers were decreased in other CLL patients who received RTX therapy; the time for restoration of full complement activity after RTX treatment was variable, from days to weeks.15 Therefore we suggested that fresh frozen plasma as a complement source may enhance RTX therapeutic activity,15,51 and recent clinical studies, although not well controlled have provided some evidence in favor of this paradigm.52 However, infusion of two units of fresh frozen plasma may not be adequate to provide substantial long-term restoration of complement titers, and additional studies will be required to determine if this approach has therapeutic utility.52

EXHAUSTION CAN RESULT IN INCREASED RESISTANCE: SHAVING

High levels of RTX-opsonized B cells can saturate effector mechanisms that would otherwise promote RTX-mediated cytotoxicity. Under these conditions the RTX/CD20 complexes on B cells will be subject to an alternative processing pathway, the “shaving reaction.”25,53 In this process RTX and CD20 are removed from opsonized cells in a reaction mediated by FcγR on acceptor cells, but the cells are not killed! This phenomenon was originally described in other systems as antigenic modulation: loss of expression of surface antigens after binding of antibody.54,55 Loss of the target of infused mAb T101 (CD5) on circulating malignant T cells was reported by Bertram et al. more than 20 years ago.54 The shaving reaction resembles trogocytosis (from the Greek, to gnaw or nibble), in which formation of an immunological synapse leads to removal and internalization of ligands associated with donor cells by cognate receptors on acceptor cells. Trogocytosis has been documented for B cells, T cells, neutrophils and NK cells.56

By definition, macrophages are “big eaters” and might not be expected to engage in trogocytosis. However, macrophages can make use of FcγR, especially FcγRI, to endocytose small IgG-opsonized substrates by taking them into the cell without surrounding them.5759 Therefore macrophages and other cells that express FcγR may be capable of either wholesale phagocytosis and/or ADCC (most likely mediated by FcγRII and FcγRIII38), or trogocytosis, in which the target cell-bound RTX (the ligand for FcγR) and CD20 are removed and internalized by the acceptor monocyte/macrophage but the target cell is left alive and intact.56 Our investigations of low-dose RTX for treatment of CLL (see below) suggest that phagocytosis and shaving of RTX-opsonized B cells occur simultaneously. That is, some RTX-opsonized cells that enter the liver are effectively bound to Kupffer cells and phagocytosed. However, other RTX-opsonized cells may pass through the gauntlet of Kupffer cells and escape with some loss of bound RTX and CD20. We have recently obtained additional evidence that macrophages can mediate either shaving or phagocytosis. After RTX-opsonized CD20-positive mouse 38C13 B cells29 were incubated with adherent mouse peritoneal macrophages, the cells were separated and examined. Macrophages had either phagocytosed entire cells, or had trogocytosed small amounts of RTX and plasma membrane from the B cells. Moreover, the recovered B cells had lost CD20 (Lindorfer and Taylor, unpublished observations).

The effector cell phenotype may influence the outcome of an encounter with a target cell. THP-1 cells and freshly isolated monocytes promote shaving of RTX-opsonized B cells, but do not mediate ADCC or phagocytosis.53 On the other hand, NK cells promote ADCC of RTX-opsonized cells, and also take up RTX and CD20 from the cells.25 This process will lead to down-regulation of NK cell CD16, thus suppressing/exhausting its ability to execute ADCC.39 Neutrophils, which express several FcγR, also mediate shaving of RTX-opsonized cells, but in the absence of complement activation and/or other mediators, they do not kill the cells.25

It is an oversimplification to assign all phagocytosis/ADCC to FcγRII and FcγRIII, and all trogocytosis to FcγRI. However, we suggest this provides a reasonable paradigm to explain the transition from clearance and destruction of RTX-opsonized cells to shaving that occurs when CLL patients with high burdens of circulating cells are treated with standard doses of RTX (Table II). The large burden of B cells cleared from the circulation (~2 X 1011) after infusion of only 30 mg of RTX will likely saturate the phagocytic capacity of liver Kupffer cells (~2 X 1010) as well as the ADCC capacity of circulating NK cells (~1 X 109) and in many cases complement activity will be reduced.9,15,60 After RTX-opsonized B cells are cleared, CLL B cells will re-equilibrate into the circulation from other compartments,61 and be opsonized by RTX. However, based on the SLE immune complex model38 as well as the ITP model,48,50 FcγRII and FcγRIII on tissue macrophages and FcγRIII on NK cells will be substantially reduced, but FcγRI on macrophages and monocytes will be available to promote shaving. The result is that cells which enter the circulation after the first round of clearance will not only escape ADCC and phagocytosis by the MPS, but these cells will also have CD20 removed by the shaving reaction. This adaptive resistance may allow them to escape targeting by RTX and proliferate.

Therefore, we initiated a pilot clinical trial in CLL to evaluate the effects of much lower, more frequent (thrice-weekly) doses of RTX on the biology of circulating malignant B cells.17 The testable hypothesis was that at RTX doses of ~35 mg (20 mg/m2), the plasma concentration of RTX would be quite low (< 5 ug/ml) after the first round of clearance of opsonized cells. As additional CLL cells entered the circulation from other compartments, these B cells would not be opsonized by RTX, and cell-associated CD20 would be preserved. It would then be possible to target circulating cells with an additional low RTX dose two days later, when the body’s effector systems, previously saturated after the first RTX dose, would have recovered and could then mediate additional clearance of opsonized B cells. In five out of six patients who received the 20 mg/m2 dose, CD20 was largely preserved and in 4 of 6 patients additional RTX infusions continued to promote clearance of malignant cells. This trial tested mechanisms leading to loss of CD20 on CLL B cells, and the low dose paradigm may not be adequate for targeting B cells that do not re-equilibrate into the bloodstream.17 However, these results, taken in context with our findings in CLL patients who received RTX doses >100 mg (60 mg/m2), provide compelling evidence that the body’s effector mechanisms responsible for clearing RTX-opsonized cells can be saturated, thus sparing a population of RTX-opsonized cells. This phenomenon evolves into a perfect storm: CD20 will be removed from RTX-opsonized cells, allowing them to proliferate under conditions which support continued shaving of re-expressed CD20. On the basis of these considerations it is not surprising that single agent RTX therapy at standard doses has modest efficacy in CLL.1 Finally, in collaboration with A. Wiestner and his colleagues, we have found that subcutaneous delivery of lower, more frequent doses of RTX has the potential to be an effective and convenient paradigm for CLL treatment.62

SHAVING OUTSIDE THE BLOODSTREAM

The clinical success of RTX in non-Hodgkins lymphoma is well-documented, suggesting that RTX therapy is more effective at killing B cells in tissues than in the bloodstream.9,10 However, approximately 50% of patients treated with single agent RTX do not respond, and responding patients often suffer relapses one to two years later. It is therefore important to ask how effective are the body’s effector mechanisms in tissue compartments, and, can these mechanisms also be saturated or exhausted?

It is certainly reasonable that CD20-negative B cell lymphomas can develop long after RTX therapy, likely due to outgrowth/mutation of a population of CD20-negative cells.63 However, several immunohistochemical investigations have described CD20-negative B cells (positive for CD79a, CD19, and other B cell markers) that appeared within a few months or even days after completion of RTX infusions.6371 Under these conditions, RTX was likely present in the circulation and tissues. These studies made use of reagents that would reveal CD20 even in the presence of bound RTX, thus precluding simple steric blockade as an explanation of the findings. In many of these reports, B cells lacking CD20 were demonstrable in tumors of patients with B cell lymphomas after RTX treatment.

RTX therapy is also used for other indications including rheumatoid arthritis (RA) or to eliminate B cells before kidney transplants.70,71 In these investigations B cells were cleared from the bloodstream for long periods. However, in the RA study, B cells lacking CD20 were demonstrable in the bone marrow and synovium. In the transplant study, splenectomies were performed 3–13 days after RTX infusion. B cells that were CD79a-positive but CD20-negative were found in the spleens of patients who received RTX infusions ≥35 mg/m2. The results of these two investigations suggest that although bloodstream clearance was effective due to the large capacity of the MPS to clear “normal” levels of opsonized B cells, cytotoxic mechanisms within tissue compartments responsible for eliminating normal B cells may have been exhausted. We suggest that a substantial fraction of the B cells in tissues had lost CD20 due to the shaving reaction. In principle it should be possible to test this hypothesis by sterile culture of isolated B cells for a few days in the absence of RTX, to allow for CD20 to be re-expressed. Examination of the levels of CD16 on monocyte/macrophages and NK cells within tissue compartments in which shaving is suspected could be quite informative. If CD16 were down-regulated, this would be consistent with the overall working hypothesis we have presented.

CONCLUSIONS and FUTURE DIRECTIONS

As we have noted in the introduction, there is increasing evidence that long after patients with B cell lymphomas are successfully treated with RTX, they suffer relapses and in many cases their disease is refractory to additional RTX treatments that include chemotherapy, although their B cells do express CD20.11,12 The reason(s) for this apparent adaptive RTX resistance remain unknown, and it is not unreasonable to speculate that one or more of the short-term mechanisms that allow cells to chronically escape RTX-mediated cytotoxicity may play key roles in this phenomenon. The elucidation of these long-term resistance “pathways” constitutes a significant challenge not only for anti-CD20 therapies, but also likely with respect to other mAb-based therapies in the treatment of cancer. Many different approaches for abrogating resistance to immunotherapy are currently under investigation (Table I).

Our discovery of the shaving reaction (or perhaps rediscovery of antigenic modulation) was based on clinical observations in CLL. Additional clinical studies as well as in vitro experiments have provided important new insights into how RTX-opsonized B cells are either killed by or escape the body’s immune effector mechanisms. RTX is now used in combination with a variety of chemotherapeutic regimens, with a higher level of clinical efficacy, and therefore mechanisms of resistance will change. However, it is likely that many of the lessons learned in analyzing innate and adaptive mechanisms of resistance of targeted cells to RTX will be applicable toward understanding the action of other immunotherapeutic mAbs.9

Acknowledgments

Work from our laboratory cited in this review was supported in part by the University of Virginia NIH Cancer Center Support Grant, an NIH Bench to Bedside Award, The Commonwealth Foundation for Cancer Research, The James and Rebecca Craig Foundation, The Lymphoma Research Foundation, GenMab, and CLL Topics. We especially thank C. Venkat and the late P.C. Venkat of CLL Topics for their support.

Financial disclosure: RPT has received research support from GenMab.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Reference List

  • 1.Golay J, Zaffaroni L, Vaccari T, Lazari M, Borleri G, Bernasconi S, et al. Biologic response of B lymphoma cells to anti-CD20 monoclonal antibody rituximab in vitro: CD55 and CD59 regulate complement mediated cell lysis. Blood. 2000;95:3900–3908. [PubMed] [Google Scholar]
  • 2.Manches O, Lui G, Chaperot L, Gressin R, Molens JP, Jacob MC, et al. In vitro mechanisms of action of rituximab on primary non-Hodgkin’s lymphomas. Blood. 2003;101:949–954. doi: 10.1182/blood-2002-02-0469. [DOI] [PubMed] [Google Scholar]
  • 3.Kennedy AD, Solga MD, Schuman TA, Chi AW, Lindorfer MA, Sutherland WM, et al. An anti-C3b(i) mAb enhances complement activation, C3b(i) deposition, and killing of CD20+ cells by Rituximab. Blood. 2003;101:1071–1079. doi: 10.1182/blood-2002-03-0876. [DOI] [PubMed] [Google Scholar]
  • 4.van Meerten T, van Rijn RS, Hol S, Hagenbeek A, Ebeling SB. Complement-induced cell death by rituximab depends on CD20 expression level and acts complementary to antibody-dependent cellular cytotoxicity. Clin Cancer Res. 2006;12:4027–4035. doi: 10.1158/1078-0432.CCR-06-0066. [DOI] [PubMed] [Google Scholar]
  • 5.Cartron G, Dacheux L, Salles G, Solal-Celigny P, Bardos P, Colombat P, et al. Therapeutic activity of humanized anti-CD20 monoclonal antibody and polymorphism in IgG Fc receptor FcγRIIIa gene. Blood. 2002;99:754–758. doi: 10.1182/blood.v99.3.754. [DOI] [PubMed] [Google Scholar]
  • 6.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;21:3940–3947. doi: 10.1200/JCO.2003.05.013. [DOI] [PubMed] [Google Scholar]
  • 7.Hatjiharissi E, Xu L, Santos DD, Hunter ZR, Ciccarelli BT, Verselis S, et al. Increased natural killer cell expression of CD16, augmented binding and ADCC activity to rituximab among individuals expressing the FcγRIIIa-158 V/V and V/F polymorphism. Blood. 2007;110:2561–2564. doi: 10.1182/blood-2007-01-070656. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Racila E, Link BK, Weng WK, Witzig TE, Ansell S, Maurer MJ, et al. A polymorphism in the complement component C1qA correlates with prolonged response following rituximab therapy of follicular lymphoma. Clin Canc Res. 2008;14:6697–6703. doi: 10.1158/1078-0432.CCR-08-0745. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Glennie MJ, French R, Cragg MS, Taylor RP. Mechanisms of killing by anti-CD20 monoclonal antibodies. Mol Immunol. 2007;44:3823–3837. doi: 10.1016/j.molimm.2007.06.151. [DOI] [PubMed] [Google Scholar]
  • 10.Smith MR. Rituximab (monoclonal anti-CD20 antibody): mechanisms of action and resistance. Oncogene. 2003;22:7359–7368. doi: 10.1038/sj.onc.1206939. [DOI] [PubMed] [Google Scholar]
  • 11.Davis TA, Grillo-Lopez AJ, White CA, McLaughlin P, Czuczman MS, Link BK, et al. Rituximab anti-CD20 monoclonal antibody therapy in non-Hodgkin’s lymphoma: safety and efficacy of re-treatment. J Clin Oncol. 2000;18:3135–3143. doi: 10.1200/JCO.2000.18.17.3135. [DOI] [PubMed] [Google Scholar]
  • 12.Martin A, Conde E, Arnan M, Canales MA, Deben G, Sancho JM, et al. R-ESHAP as salvage therapy for patients with relapsed or refractory diffuse large B-cell lymphoma: the influence of prior exposure to rituximab on outcome. A GEL/TAMO study. Haematologica. 2008;93:1829–1836. doi: 10.3324/haematol.13440. [DOI] [PubMed] [Google Scholar]
  • 13.Wang SY, Racila E, Taylor RP, Weiner GJ. NK cell activation and antibody dependent cellular cytotoxicity induced by Rituximab-coated target cells is inhibited by the C3b component of complement. Blood. 2008;111:1456–1463. doi: 10.1182/blood-2007-02-074716. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Reiter Y, Ciobotariu A, Jones J, Morgan BP, Fishelson Z. Complement membrane attack complex, perforin, and bacterial exotoxins induce in K562 cells calcium-dependent cross-protection from lysis. J Immunol. 1995;155:2203–2210. [PubMed] [Google Scholar]
  • 15.Kennedy AD, Beum PV, Solga MD, DiLillo DJ, Lindorfer MA, Hess CE, et al. Rituximab infusion promotes rapid complement depletion and acute CD20 loss in chronic lymphocytic leukemia. J Immunol. 2004;172:3280–3288. doi: 10.4049/jimmunol.172.5.3280. [DOI] [PubMed] [Google Scholar]
  • 16.Beum PV, Lindorfer MA, Hall BE, George TC, Frost K, Morrissey PJ, et al. Quantitative analysis of protein co-localization on B cells opsonized with rituximab and complement using the ImageStream multispectral imaging flow cytometer. J Immunol Meth. 2006;317:90–99. doi: 10.1016/j.jim.2006.09.012. [DOI] [PubMed] [Google Scholar]
  • 17.Williams ME, Densmore JJ, Pawluczkowycz AW, Beum PV, Kennedy AD, Lindorfer MA, et al. Thrice-weekly low-dose rituximab decreases CD20 loss via shaving and promotes enhanced targeting in chronic lymphocytic leukemia. J Immunol. 2006;177:7435–7443. doi: 10.4049/jimmunol.177.10.7435. [DOI] [PubMed] [Google Scholar]
  • 18.Golay J, Manganini M, Facchinetti V, Gramigna R, Broady R, Borleri G, et al. Rituximab-mediated antibody-dependent cellular cytotoxicity against neoplastic B cells is stimulated strongly by interleukin-2. Haematologica. 2003;88:1002–1012. [PubMed] [Google Scholar]
  • 19.Lefebvre M-L, Krause SW, Salcedo M, Nardin A. Ex vivo-activated human macrophages kill chronic lymphocytic leukemia cells in the presence of Rituximab: mechanism of antibody-dependent cellular cytotoxicity and impact of human serum. J Immunother. 2006;29:388–397. doi: 10.1097/01.cji.0000203081.43235.d7. [DOI] [PubMed] [Google Scholar]
  • 20.Tedder TF, Baras A, Xiu Y. Fcγ receptor-dependent effector mechanisms regulate CD19 and CD20 antibody immunotherapies for B lymphocyte malignancies and autoimmunity. Springer Semin Immun. 2006;28:351–364. doi: 10.1007/s00281-006-0057-9. [DOI] [PubMed] [Google Scholar]
  • 21.Taylor RP, Lindorfer MA. Immunotherapeutic mechanisms of anti-CD20 monoclonal antibodies. Curr Opin Immunol. 2008;20:444–449. doi: 10.1016/j.coi.2008.05.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Gong Q, Ou Q, Ye S, Lee WP, Cornelius J, Diehl L, et al. Importance of cellular microenvironment and circulatory dynamics in B cell immunotherapy. J Immunol. 2005;174:817–826. doi: 10.4049/jimmunol.174.2.817. [DOI] [PubMed] [Google Scholar]
  • 23.Walport MJ. Complement. N Engl J Med. 2001;344:1058–1066. doi: 10.1056/NEJM200104053441406. [DOI] [PubMed] [Google Scholar]
  • 24.Dall’Ozzo S, Tartas S, Paintaud G, Cartron G, Colombat P, Bardos P, et al. Rituximab-dependent cytotoxicity by natural killer cells: influence of FCGR3A polymorphism on the concentration-effect relationship. Cancer Res. 2004;64:4664–4669. doi: 10.1158/0008-5472.CAN-03-2862. [DOI] [PubMed] [Google Scholar]
  • 25.Beum PV, Lindorfer MA, Taylor RP. Within peripheral blood mononuclear cells, antibody-dependent cellular cytotoxicity of rituximab-opsonized Daudi cells is promoted by NK cells and inhibited by monocytes due to shaving. J Immunol. 2008;181:2916–2924. doi: 10.4049/jimmunol.181.4.2916. [DOI] [PubMed] [Google Scholar]
  • 26.Schreiber AD, Frank MM. Role of antibody and complement in the immune clearance and destruction of erythrocytes: in vivo effects of IgG and IgM complement fixing sites. J Clin Invest. 1972;51:575–582. doi: 10.1172/JCI106846. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Kabbash I, Esdaile JM, Henker S, De Cary F, Danoff D, Fuks A, et al. Reticuloendothelial system Fc receptor function in systemic lupus erythematosus: effect of decreased sensitization on clearance of autologous erythocytes. J Rheum. 1987;14:487–489. [PubMed] [Google Scholar]
  • 28.Di Gaetano N, Cittera E, Nota R, Vecchi A, Grieco V, Scanziani E, et al. Complement activation determines the therapeutic activity of Rituximab in vivo. J Immunol. 2003;171:1581–1587. doi: 10.4049/jimmunol.171.3.1581. [DOI] [PubMed] [Google Scholar]
  • 29.Golay J, Cittera E, Di Gaetano N, Manganini M, Mosca M, Nebuloni M, et al. The role of complement in the therapeutic activity of rituximab in a murine B lymphoma model homing in lymph nodes. Haematologica. 2006;91:176–183. [PubMed] [Google Scholar]
  • 30.Helmy KY, Katschke KJ, Gorgani NN, Kjavin NM, Elliott JM, Diehl L, et al. CRIg: A macrophage complement receptor required for phagocytosis of circulating pathogens. Cell. 2006;124:915–927. doi: 10.1016/j.cell.2005.12.039. [DOI] [PubMed] [Google Scholar]
  • 31.Hinglais N, Kazatchkine MD, Mandet C, Appay M, Bariety J. Human liver Kupffer cells express CR1, CR3, and CR4 complement receptor antigens. Lab Invest. 1989;61:509–514. [PubMed] [Google Scholar]
  • 32.Wang SY, Veeramani S, Racila E, Cagley J, Frtizinger D, Vogel CW, et al. Depletion of the C3 component of complement enhances the ability of rituximab-coated target cells to activate human NK cells and improves the efficacy of monoclonal antibody therapy in an in vivo model. Blood. 2009 doi: 10.1182/blood-2009-01-200469. 10.1182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Pawluczkowycz AW, Beurskens FJ, Beum PV, Lindorfer MA, van de Winkel JGJ, Parren PWHI, et al. Binding of submaximal C1q promotes complement-dependent cytotoxicity (CDC) of B cells opsonized with anti-CD20 mAbs ofatumumab (OFA) or rituximab (RTX): Considerably higher levels of CDC are induced by OFA than by RTX. J Immunol. 2009;183:749–758. doi: 10.4049/jimmunol.0900632. [DOI] [PubMed] [Google Scholar]
  • 34.Taylor RP, Pawluczkowycz AW, Beum PV, Lindorfer MA, Beurskens FB, van de Winkel J, et al. Complement activation and complemented-mediated killing of B cells promoted by anti-CD20 monoclonal antibodies (mAb) rituximab and ofatumumab are rapid, and ofatumumab kills cells more rapidly and with greater efficacy. Blood. 2007;118:695a. [Google Scholar]
  • 35.Teeling JL, French RR, Cragg MS, van den Brakel J, Pluyter M, Huang H, et al. Characterization of new human CD20 monoclonal antibodies with potent cytolytic activity against non-Hodgkin’s lymphomas. Blood. 2004;104:1793–1800. doi: 10.1182/blood-2004-01-0039. [DOI] [PubMed] [Google Scholar]
  • 36.Frank MM, Hamburger MI, Lawley TJ, Kimberly RP, Plotz PH. Defective reticuloendothelial system Fc-receptor function in systemic lupus erythematosus. N Eng J Med. 1979;300:518–523. doi: 10.1056/NEJM197903083001002. [DOI] [PubMed] [Google Scholar]
  • 37.Manderson AP, Botto M, Walport MJ. The role of complement in the development of systemic lupus erythematosus. Annu Rev Immunol. 2004;22:431–456. doi: 10.1146/annurev.immunol.22.012703.104549. [DOI] [PubMed] [Google Scholar]
  • 38.Kavai M, Szegedi G. Immune complex clearance by monocytes and macrophages in systemic lupus erythematosus. Autoimmun Rev. 2007;6:497–502. doi: 10.1016/j.autrev.2007.01.017. [DOI] [PubMed] [Google Scholar]
  • 39.Bowles JA, Weiner GJ. CD16 polymorphisms and NK activation induced by monoclonal antibody-coated target cells. J Immunol Methods. 2005;304:88–99. doi: 10.1016/j.jim.2005.06.018. [DOI] [PubMed] [Google Scholar]
  • 40.Bhat R, Watzl C. Serial killing of tumor cells by human natural killer cells: enhancement by therapeutic antibodies. PLoS ONE. 2007;2:1–7. doi: 10.1371/journal.pone.0000326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Berdeja JG, Hess A, Lucas DM, O’Donnell P, Ambinder RF, Diehl LF, et al. Systemic interleukin-2 and adoptive transfer of lymphokine-activated killer cells improves antibody-dependent cellular cytotoxicity in patients with relapsed B-cell lymphoma treated with rituximab. Clin Cancer Res. 2007;13:2392–2399. doi: 10.1158/1078-0432.CCR-06-1860. [DOI] [PubMed] [Google Scholar]
  • 42.Varchetta S, Gibelli N, Oliviero B, Nardini E, Gennari R, Gatti G, et al. Elements related to heterogeneity of antibody-dependent cell cytotoxicity in patients under trastuzumab therapy for primary operable breast cancer overexpressing Her2. Cancer Res. 2007;67:11991–11999. doi: 10.1158/0008-5472.CAN-07-2068. [DOI] [PubMed] [Google Scholar]
  • 43.Bowles JA, Wang S-Y, Link BK, Beuerlein G, Campbell M-A, Marquis D, 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. doi: 10.1182/blood-2006-04-020057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.de Romeuf C, Dutertre CA, Le Garff-Tavernier M, Fournier N, Gaucher C, Glacet A, et al. Chronic lymphocytic leukaemia cells are efficiently killed by an anti-CD20 monoclonal antibody selected for improved engagement of FcγRIIIA/CD16. Br J Haematol. 2008;140:635–643. doi: 10.1111/j.1365-2141.2007.06974.x. [DOI] [PubMed] [Google Scholar]
  • 45.Stavenhagen JB, Gorlatov S, Tuaillon N, Rankin CT, Li H, Burke S, et al. Fc opitimization of therapeutic antibodies enhances their ability to kill tumor cells in vitro and controls tumor expansion in vivo via low-affinity activating Fcγ receptors. Canc Res. 2007;67:8882–8990. doi: 10.1158/0008-5472.CAN-07-0696. [DOI] [PubMed] [Google Scholar]
  • 46.Klingemann HG. Natural killer cell-based immunotherapeutic strategies. Cytotherapy. 2005;7:16–22. doi: 10.1080/14653240510018000. [DOI] [PubMed] [Google Scholar]
  • 47.Lundqvist A, Yokoyama H, Smith A, Berg M, Childs R. Bortexomib treatment and regulatory T-cell depletion enchance the antitumor effects of adoptively infused NK cells. Blood. 2009;113:6120–6127. doi: 10.1182/blood-2008-11-190421. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Scaradavou A, Woo B, Woloski BMR, Cunningham-Rundles S, Ettinger LJ, Aledort LM, et al. Intravenous anti-D treatment of immune thrombocytopenic purpura: experience in 272 patients. Blood. 1997;89:2689–2700. [PubMed] [Google Scholar]
  • 49.Taylor RP, Lindorfer MA. Drug Insight: the mechanism of action of rituximab in autoimmune disease--the immune complex decoy hypothesis. Nat Clin Pract Rheum. 2007;3:86–95. doi: 10.1038/ncprheum0424. [DOI] [PubMed] [Google Scholar]
  • 50.Song S, Crow AR, Siragam V, Freedman J, Lazarus AH. Monoclonal antibodies that mimic the action of anti-D in the amelioration of murine ITP act by a mechanism distinct from that of IVIg. Blood. 2005;105:1546–1548. doi: 10.1182/blood-2004-05-1886. [DOI] [PubMed] [Google Scholar]
  • 51.Taylor R. Fresh frozen plasma as a complement source. Lancet Oncol. 2007;8:370–371. doi: 10.1016/S1470-2045(07)70114-6. [DOI] [PubMed] [Google Scholar]
  • 52.Klepfish A, Rachmilewitz EA, Kotsianidis I, Patchenko P, Schattner A. Adding fresh frozen plasma to rituximab for the treatment of patients with refractory advanced CLL. Quart J Med. 2008;101:737–740. doi: 10.1093/qjmed/hcn085. [DOI] [PubMed] [Google Scholar]
  • 53.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;176:2600–2609. doi: 10.4049/jimmunol.176.4.2600. [DOI] [PubMed] [Google Scholar]
  • 54.Bertram JH, Gill PS, Levine AM, Boquiren D, Hoffman FM, Meyer P, et al. Monoclonal antibody T101 in T cell malignancies: A clinical, pharmacokinetic, and immunologic correlation. Blood. 1986;68:752–761. [PubMed] [Google Scholar]
  • 55.Schroff RW, Klein RA, Farrell MM, Stevenson HC. Enhancing effects of monocytes on modulation of a lymphocyte membrane antigen. J Immunol. 1984;133:2270–2277. [PubMed] [Google Scholar]
  • 56.Hudrisier D, Aucher A, Puaux AL, Bordier C, Joly E. Capture of target cell membrane components via trogocytosis is triggered by a selected set of surface molecules on T or B cells. J Immunol. 2007;178:3637–3647. doi: 10.4049/jimmunol.178.6.3637. [DOI] [PubMed] [Google Scholar]
  • 57.Guyre C, Keler T, Swink S, Vitale L, Graziano R, Fanger M. Receptor modulation by FcγRI-specific fusion proteins is dependent on receptor number and modified by IgG. J Immunol. 2001;167:6303–6311. doi: 10.4049/jimmunol.167.11.6303. [DOI] [PubMed] [Google Scholar]
  • 58.Davis W, Harrison PT, Hutchinson MJ, Allen JM. Two distinct regions of FcγRI initiate separate signalling pathways involved in endocytosis and phagocytosis. EMBO J. 1995;14:432–441. doi: 10.1002/j.1460-2075.1995.tb07019.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Lovdal T, Andersen E, Brech ABT. Fc receptor mediated endocytosis of small soluble immunoglobulin G immune complexes in Kupffer and endothelial cells from rat liver. J Cell Sci. 2000;113:3255–3266. doi: 10.1242/jcs.113.18.3255. [DOI] [PubMed] [Google Scholar]
  • 60.Lundin J, Porwit-MacDonald A, Rossmann ED, Karlsson C, Edman P, Rezvany MR, et al. Cellular immune reconstitution after subcutaneous alemtuzumab (anti-CD52 monoclonal antibody, CAMPATH-1H) treatment as first-line therapy for B-cell chronic lymphocytic leukaemia. Leukemia. 2004;18:484–490. doi: 10.1038/sj.leu.2403258. [DOI] [PubMed] [Google Scholar]
  • 61.Cooper IA, Ding JC, Adams PB, Quinn MA, Brettell M. Intensive leukapheresis in the management of cytopeias in patients with chronic lymphocytic leukaemia (CLL) and lymphocytic lymphoma. Amer J Hematol. 1979;6:387–398. doi: 10.1002/ajh.2830060411. [DOI] [PubMed] [Google Scholar]
  • 62.Aue G, Lindorfer MA, Beum PV, Pawluczkowycz AW, Vire B, Hughes T, et al. Fractionated subcutaneous rituximab is well tolerated and preserves CD20 expression on tumor cells in patients with chronic lymphocytic leukemia. Haematologica. 2009 doi: 10.3324/haematol.2009.012484. Epub 10.3324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Kennedy GA, Tey SK, Cobcroft R, Marlton P, Cull G, Grimmett K, et al. Incidence and nature of CD20-negative relapses following rituximab therapy in aggressive B-cell non-Hodgkin’s lymphoma: a retrospective review. Br J Haemotol. 2002;119:412–416. doi: 10.1046/j.1365-2141.2002.03843.x. [DOI] [PubMed] [Google Scholar]
  • 64.Davis TA, Czerwinski DK, Levy R. Therapy of B-cell lymphoma with anti-CD20 antibodies can result in the loss of CD20 antigen expression. Clin Canc Res. 1999;5:611–5. [PubMed] [Google Scholar]
  • 65.Foran JM, Norton AJ, Micallef INM, Taussig DC, Amess JAL, Rohatiner AZS, et al. Loss of CD20 expression following treatment with rituximab (chimaeric monoclonal anti-CD20): a retrospective cohort analysis. Br J Haematol. 2001;114:881–883. doi: 10.1046/j.1365-2141.2001.03019.x. [DOI] [PubMed] [Google Scholar]
  • 66.Haidar JH, Shamseddine A, Salem Z, Abou Mrad Y, Nasr MR, Zaatari G, et al. Loss of CD20 expression in relapsed lymphomas after rituximab therapy. Eur J Haematol. 2003;70:330–332. doi: 10.1034/j.1600-0609.2003.00007.x. [DOI] [PubMed] [Google Scholar]
  • 67.Alvaro-Naranjo T, Jaen-Martinez J, Guma-Padro J, Bosch-Princep R, Salvado-Usach MT. CD20-negative DLBCL transformation after rituximab treatment in follicular lymphoma: a new case report and review of the literature. Ann Hematol. 2003;82:585–588. doi: 10.1007/s00277-003-0694-1. [DOI] [PubMed] [Google Scholar]
  • 68.Clarke LE, Bayer MG, Ehmann C, Helm KF. Cutaneous B-cell lymphoma with loss of CD20 immunoreactivity after rituximab therapy. J Cutaneous Path. 2003;30:459–462. doi: 10.1034/j.1600-0560.2003.00078.x. [DOI] [PubMed] [Google Scholar]
  • 69.Goteri G, Olivieri A, Ranaldi R, Lucesole M, Filosa A, Capretti R, et al. Bone marrow histopathological and molecular changes of small B-cell lymphomas after rituximab therapy: comparison with clinical response and patients’ outcome. Intl J Immunopath Pharmacol. 2006;19:421–431. doi: 10.1177/039463200601900218. [DOI] [PubMed] [Google Scholar]
  • 70.Teng YKO, Levarht EWN, Hashemi M, Bajema IM, Toes REM, Huizinga TWJ, et al. Immunohistochemical analysis as a means to predict responsiveness to rituximab treatment. Arth Rheum. 2007;56:3909–3918. doi: 10.1002/art.22967. [DOI] [PubMed] [Google Scholar]
  • 71.Toki D, Ishida H, Horita S, Setoguchi K, Yamaguchi Y, Tanabe K. Impact of low-dose rituximab on splenic B cells in ABO-incompatible renal transplant recipients. Transplant Intl. 2009;22:447–454. doi: 10.1111/j.1432-2277.2008.00821.x. [DOI] [PubMed] [Google Scholar]
  • 72.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;179:4263–4271. doi: 10.4049/jimmunol.179.6.4263. [DOI] [PubMed] [Google Scholar]
  • 73.Macor P, Tripodo C, Zorzet S, Piovan E, Bossi F, Marzari R, et al. In vivo targeting of human neutralizing antibodies against CD55 and CD59 to lymphoma cells increases the antitumor activity of rituximab. Cancer Res. 2007;67:10556–10563. doi: 10.1158/0008-5472.CAN-07-1811. [DOI] [PubMed] [Google Scholar]
  • 74.Burger JA, Ghia P, Rosenwald A, Caligaris-Cappio F. The microenvironment in mature B-cell malignancies: a target for new treatment strategies. Blood. 2009;114:3367–3375. doi: 10.1182/blood-2009-06-225326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Mayo Clinic. Everolimus and alemtuzxumab in treating patients with recurrent chronic lymphocytic leukemia or small lymphocytic lymphoma. clinicaltrials.gov NCT00935792. 2009 [Google Scholar]
  • 76.Ghosh AK, Kay NE, Secreto CR, Shanafelt TD. Curcumin inhibiits prosurvival pathways in chronic lymphocytic leukemia B cells and may overcome their stromal protection in combination with EGCG. Clin Canc Res. 2009;15:1250–1258. doi: 10.1158/1078-0432.CCR-08-1511. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES