A specialized form of trogocytosis occurs when Fcγ receptors on acceptor cells take up and internalize donor cell-associated immune complexes composed of specific monoclonal antibodies (mAbs) bound to target antigens on donor cells. This trogocytosis reaction, an example of antigenic modulation, has been described in recent clinical correlative studies and in vitro investigations for several mAbs used in cancer immunotherapy, including rituximab and ofatumumab. We discuss the impact of Fcγ-receptor–mediated trogocytosis on the efficacy of cancer immunotherapy and other mAb-based therapies.

Trogocytosis, as first described by Joly and Hudrisier,1  is characterized by the transfer of cell surface molecules from a donor cell to an acceptor cell and proceeds in discreet steps2-5 : firstly, the two cells form an immunologic synapse,6,7  due to recognition of cognate ligands on the donor cell by cell-surface receptors on the acceptor cell. Secondly, in an energy-requiring process that includes actin polymerization, membrane remodeling, and signaling, portions of the plasma membranes of the cells merge, and chelated ligands on the donor cell along with sections of its plasma membrane are pinched off and taken up by the acceptor cell. Thirdly, captured material is then either displayed on the surface of the acceptor cell, or internalized, processed, and degraded. This reaction has been demonstrated for a variety of acceptor cells, including T and B lymphocytes, monocyte/macrophages, dendritic cells, neutrophils, and natural killer (NK) cells, and plays an important role in antigen presentation and processing. In all cases, only small portions of donor cells are taken up by acceptor cells, and because donor cells are only marginally perturbed, trogocytosis (gnawing or nibbling) is an apt description of the process.

We will focus our discussion on a specialized form of trogocytosis, mediated by Fcγ receptors (FcγR) on effector cells, including monocytes, macrophages, neutrophils, and NK cells, which manifests as an alternative reaction during monoclonal antibody (mAb)-based cancer therapy.8  Reaction of a therapeutic IgG mAb with its target on a cancer cell would normally be expected to mediate cell killing by well-defined mechanisms: antibody-dependent cellular cytotoxicity, antibody-dependent cellular phagocytosis, complement-dependent cytotoxicity, and/or direct killing (eg, programmed cell death).9-11  The first two reactions require participation of effector cells expressing FcγR. However, evidence based on clinical correlative studies and in vitro investigations indicates that many immunotherapeutic mAbs can mediate either cell killing or trogocytosis of their targeted antigens in the presence of effector cells (Table 1).8,12-16  In the later reaction, once the mAbs bind to target antigens on the donor cells, an immunologic synapse is formed. The acceptor cell then removes the mAb-antigen immune complexes from the donor cell and subsequently internalizes them.13,17-19  Therefore, targeted donor cells can escape relatively unharmed from the intended cytotoxic immunotherapy.8,12,20 Table 2 summarizes laboratory and clinical parameters used to define FcγR-mediated trogocytosis. Although our first studies with THP-1 acceptor cells indicated that FcγRI plays a major role in trogocytosis, it is now clear that depending upon the cell type, virtually all FcγR can promote trogocytosis.21,22 

Table 1

Important examples of trogocytosis mediated by mAbs

Target EpitopemAbKey observations
CD3 Muromonab-CD3 (OKT3) Early clinical report of antigenic modulation. In vitro experiments demonstrated requirements for monocytes and intact IgG.36,37  
CD5 T101 Loss of CD5 was induced by treatment of patients with CLL or cutaneous T-cell lymphoma with mAb T101. In vitro studies indicated FcγR on monocytes and intact IgG were required.38-44,75,76  
CD8 Hit8a When whole blood samples are probed with CD8 mAbs for standard flow cytometry analyses, CD8 is transferred to and internalized by monocytes and neutrophils.19,21,30  
CD20 RTX (Rituxan) Both mAbs promote in vivo and in vitro trogocytosis.8,12-14,17,18,20,22,52,55-58,61,64,67  
 OFA (Arzerra) 
 CD20-6 This type II CD20 mAb promotes trogocytosis.57  
CD22 Epratuzumab Treatment of SLE patients with epratuzumab resulted in trogocytosis of CD22 and other cell surface proteins from circulating B cells.15,66  
CD25 Daclizumab (Zenapax) Treatment of patients with multiple sclerosis with daclizumab resulted in trogocytosis of CD25 from T cells.15  
CD33 Gemtuzumab ozogamicin (Mylotarg) Trogocytosis, mediated by FcγRIIb on LSECs, may explain why some patients who receive Mylotarg therapy experience severe liver damage.22,68-70,72  
EGF-R Cetuximab (Erbitux) Cetuximab mediates trogocytosis by monocytes of the EGF receptor on 3 different cell lines in vitro.18,77  
Her2/Neu Trastuzumab (Herceptin) Trastuzumab mediates trogocytosis by monocytes of the Her2/Neu antigen on a breast cancer cell line in vitro.18  
Target EpitopemAbKey observations
CD3 Muromonab-CD3 (OKT3) Early clinical report of antigenic modulation. In vitro experiments demonstrated requirements for monocytes and intact IgG.36,37  
CD5 T101 Loss of CD5 was induced by treatment of patients with CLL or cutaneous T-cell lymphoma with mAb T101. In vitro studies indicated FcγR on monocytes and intact IgG were required.38-44,75,76  
CD8 Hit8a When whole blood samples are probed with CD8 mAbs for standard flow cytometry analyses, CD8 is transferred to and internalized by monocytes and neutrophils.19,21,30  
CD20 RTX (Rituxan) Both mAbs promote in vivo and in vitro trogocytosis.8,12-14,17,18,20,22,52,55-58,61,64,67  
 OFA (Arzerra) 
 CD20-6 This type II CD20 mAb promotes trogocytosis.57  
CD22 Epratuzumab Treatment of SLE patients with epratuzumab resulted in trogocytosis of CD22 and other cell surface proteins from circulating B cells.15,66  
CD25 Daclizumab (Zenapax) Treatment of patients with multiple sclerosis with daclizumab resulted in trogocytosis of CD25 from T cells.15  
CD33 Gemtuzumab ozogamicin (Mylotarg) Trogocytosis, mediated by FcγRIIb on LSECs, may explain why some patients who receive Mylotarg therapy experience severe liver damage.22,68-70,72  
EGF-R Cetuximab (Erbitux) Cetuximab mediates trogocytosis by monocytes of the EGF receptor on 3 different cell lines in vitro.18,77  
Her2/Neu Trastuzumab (Herceptin) Trastuzumab mediates trogocytosis by monocytes of the Her2/Neu antigen on a breast cancer cell line in vitro.18  

CLL, chronic lymphocytic leukemia; EGF-R, epidermal growth factor receptor; LSEC, liver sinusoidal endothelial cells; OFA, ofatumumab; RTX, rituximab; SLE, systemic lupus erythematosus.

Table 2

Criteria for identifying FcγR-mediated trogocytosis

Criteria are as follows:
In vitro experiments with cell lines or with primary cells
• In the presence of acceptor cells that express FcγR, the mAb promotes transfer of its targeted antigen from the donor cell to the acceptor cell 
• Both the mAb, its target antigen, small portions of the plasma membrane of the donor cell, and the operant FcγR of the acceptor cell are internalized by
the acceptor cell 
• The reaction is fast, going to completion in less than 1 hour at 37°C 
• The F(ab′)2 fragments of the mAb have little or no activity in mediating trogocytosis, and inhibitors of FcγR, such as specific mAbs or high concentrations
of human IgG, inhibit the reaction 
• In vitro culture of trogocytosed cells in the absence of mAb and acceptor cells leads to re-expression of the trogocytosed antigen 
Clinical correlative measurements specific for CLL 
• Infusion of the mAb first promotes rapid clearance of a substantial fraction of circulating CLL cells 
• Within 1 to 24 hours, additional cells appear in the circulation, but they express much lower levels of the mAb-targeted antigen. 
• Trogocytosis of the targeted antigen continues for days to weeks until the plasma concentration of the mAb falls to a very low level (eg, <1 ug/ml).
At this point, the level of the targeted antigen returns to close to pre-infusion levels on circulating cells. 
Criteria are as follows:
In vitro experiments with cell lines or with primary cells
• In the presence of acceptor cells that express FcγR, the mAb promotes transfer of its targeted antigen from the donor cell to the acceptor cell 
• Both the mAb, its target antigen, small portions of the plasma membrane of the donor cell, and the operant FcγR of the acceptor cell are internalized by
the acceptor cell 
• The reaction is fast, going to completion in less than 1 hour at 37°C 
• The F(ab′)2 fragments of the mAb have little or no activity in mediating trogocytosis, and inhibitors of FcγR, such as specific mAbs or high concentrations
of human IgG, inhibit the reaction 
• In vitro culture of trogocytosed cells in the absence of mAb and acceptor cells leads to re-expression of the trogocytosed antigen 
Clinical correlative measurements specific for CLL 
• Infusion of the mAb first promotes rapid clearance of a substantial fraction of circulating CLL cells 
• Within 1 to 24 hours, additional cells appear in the circulation, but they express much lower levels of the mAb-targeted antigen. 
• Trogocytosis of the targeted antigen continues for days to weeks until the plasma concentration of the mAb falls to a very low level (eg, <1 ug/ml).
At this point, the level of the targeted antigen returns to close to pre-infusion levels on circulating cells. 

This escape mechanism provides insight into possible biological functions of FcγR-mediated trogocytosis. For example, during IgG immune responses to infectious microorganisms, trogocytosis could allow effector cells to remove IgG-opsonized pathogens from the surface of infected cells without destroying the cells. In the immune adherence reaction, complement (C3b)-opsonized polyclonal antibody/pathogen immune complexes are bound to primate erythrocytes via CR1, the receptor for C3b.23  Both in vitro experiments and preclinical studies in nonhuman primate models have demonstrated that the immune complexes are transferred to and processed by acceptor macrophages, and this reaction represents another example of trogocytosis in immune defense.23-26  A similar reaction has been demonstrated to remove CR2 (the receptor for C3d) and bound C3d-tagged immune complexes from opsonized B cells, and likely plays a role in immune responses mediated by complement.27-29  Trogocytosis could also provide a means of removing small amounts of autoantibodies and self-antigens from weakly opsonized cells, allowing them to escape phagocytosis and destruction in conditions associated with autoimmune diseases.30  Similarly, Griffin et al reported that when IgG-opsonized B cells were allowed to form immune complexed “caps,” then the immune complexes could be removed by macrophages in a reaction that spared the B cells from phagocytosis.31 

Antigenic modulation, first described in the pre-mAb era,32  occurs when treatment of cells with specific antibodies promotes upregulation of resistance of the cells to cytotoxicity mediated by the antibodies. Several mechanisms can mediate this resistance, including endocytosis by the cell of both the antibody and the targeted cell-surface antigen (including CD22 and CD4033 ), or capping of the complexes on the cell surface.34,35  Therefore, it is not surprising that trogocytosis mediated by therapeutic mAbs was first described as “antigenic modulation” more than 30 years ago, when specific mouse mAbs were under investigation as immunosuppressive agents following renal transplants36,37  and as immunotherapeutic agents for cancer.38  The reaction is well-documented for IgG2a mAb T101 (CD5-specific), which was tested for treatment of chronic lymphocytic leukemia (CLL) and cutaneous T-cell lymphoma.39-44  The mAb infusion initially promoted clearance of substantial numbers of cells. However, within hours, more cells entered the bloodstream, and many of these cells had considerably reduced CD5 levels. In vitro investigations revealed that some direct internalization of mAb T101 by B cells occurred41,42  (as noted above, one of the first mechanisms for antigenic modulation), but the major mechanism of CD5 loss was indeed mediated by FcγR on monocytes, in a reaction identical to FcγR trogocytosis, but not so named, some 30 years ago.

Ten years ago, our laboratory reported that when patients with CLL were treated with the usual 375 mg/m2 doses of rituximab (RTX), the following patterns were observed: firstly, after infusion of only 30 mg of RTX, the circulating cells were well-opsonized, and a large fraction of cells was rapidly cleared from the circulation.12  Then, soon after completion of the infusion, circulating CLL cell counts increased considerably, reaching levels close to or exceeding pre-infusion values. This observation would appear to present a contradiction: if 30 mg of RTX mediates clearance of a large burden of cells, why does the remainder of infused RTX (∼500 to 700 mg) not clear more cells? The key to resolving this question was revealed by measuring CD20 levels on circulating cells before, during, and after mAb infusion. CLL cells in the bloodstream at the end of the infusion largely represent cells that had re-equilibrated from other compartments; initially, these CLL cells had levels of CD20 comparable to that of circulating cells before RTX infusion.45  After RTX infusion, the cells had only 5% to 10% of the levels found on cells before RTX infusion. This rapid reduction in CD20 is most likely a consequence of trogocytosis of re-equilibrated cells in the circulation, due to the action of FcγR-expressing cells, which may include Kupffer cells as well as liver sinusoidal endothelial cells (LSECs).17,46-49 

An important question must focus on the conditions that favor killing of targeted cells (the preferred outcome) vs escape via trogocytosis. In almost all of these treated CLL patients, complement titers in the bloodstream were reduced considerably after CD20 mAb infusions, indicating this effector function was exhausted. Although RTX weakly promotes complement-dependent cytotoxicity on binding to CLL cells, it can activate and consume complement, thus covalently tagging cells with C3b/iC3b,8,12  and this will facilitate clearance based on recognition of C3b/iC3b-opsonized cells by complement receptors on tissue macrophages.8,26  Additional reports indicate that other mAb-promoted cytotoxic effector functions, including NK-cell–mediated antibody-dependent cellular cytotoxicity and phagocytosis by macrophages, can also be exhausted after CD20 mAb infusion under conditions of high CLL cell burdens.50-53  Thus, trogocytosis takes over as an alternative reaction after the body’s normal cytotoxic effector functions are exhausted. We have extended and generalized these studies to include ofatumumab (OFA), a next generation CD20 mAb. We have recorded virtually identical patterns of CD20 down-modulation for more than 65 CLL patients treated with either RTX or OFA, at doses between 100 mg and 1 g.8,12,20,54  Finally, certain mAbs may be designed to simply block an active site on a cell and not promote cell killing by effector functions. Under these conditions, trogocytosis can occur even in the absence of effector function exhaustion.

Trogocytosis is rapid; in vitro investigations in model systems, reported by both our laboratory and several other laboratories reveal that at 37°C, monocytes and macrophages as well as NK cells and neutrophils, remove IgG immune-complexed substrates from mAb-opsonized cells in 20 to 30 minutes. We have found that trogocytosed CLL cells can be identified in blood samples taken from CLL patients within 0.5 to 2 hours of initiating CD20 mAb infusions.8,12,14,18,22,55-58  Finally, in another example of antigenic modulation, CD20 mAbs, along with CD20, can also be internalized directly by B cells, but in vitro, this internalization process is considerably slower than trogocytosis.14,59,60 

Although we have no direct evidence that CD20 mAbs promote trogocytosis of cells in tissues, we demonstrated that RTX mediated rapid loss of CD20 from human Z138 cells growing in the lungs of SCID mice.61  Also, 30 years ago, mAb T101 was found to promote CD5 loss from CLL cells in tissues in treated CLL patients.39,44  By analogy, it is likely that CD20 reductions in the tissues would accompany CD20 mAb therapy, and anecdotal reports suggest that this may occur.62,63  Moreover, CD20 (or other trogocytosed targets) is not restored on targeted cells until the “pressure” mediated by the specific mAbs in the circulation is eliminated due to natural clearance of the mAbs from the bloodstream.40,43,54,56  Indeed, we have found that for CLL patients who receive doses of 300 mg and 1 g of OFA on days 1 and 8, CD20 remains considerably depressed on day 29, as OFA will still be present in the bloodstream.8 

Given that 30 mg doses of RTX (or OFA) are adequate to clear ∼80% of circulating cells, we reasoned that trogocytosis could be substantially reduced, and effector functions allowed to better recover, by infusing considerably less mAb during each infusion (∼20 mg/m2), and thus reducing the number of mAb-opsonized cells that would be circulating (and subject to trogocytosis) after effector functions were exhausted. Thrice-weekly low-dose infusions over an extended time period should allow for multiple opportunities to efficiently target and eliminate malignant cells. Three recent low-dose trials, based on intravenous RTX alone, subcutaneous infusion of RTX alone, or low-dose RTX combined with alemtuzumab and pentostatin, although all limited in scope, provide quite favorable clinical and correlative evidence in favor of this paradigm.52,54,64  We suggest that modified dosing schedules with these, or other mAbs now in development, have the potential to address the issue of trogocytosis, and thus enhance mAb efficacy, and may provide a revolutionary paradigm shift in the treatment of cancer patients with certain immunotherapeutic mAbs.53  In contrast, the results of a dose-escalation trial for CLL indicated a higher level of efficacy for single agent RTX at (higher) weekly doses of 2250 mg/m,2,65  although all remissions were partial. It is possible that the very high RTX doses in part blocked Fcγ receptors13,19,21,22  and thus had the net effect of decreasing trogocytosis.

Epratuzumab, specific for CD22 on B cells, is being investigated for the treatment of B-cell lymphomas, as well as for systemic lupus erythematosus (SLE).15,66  Daclizumab, specific for CD25, a component of the IL2-receptor on T cells, is under investigation for treatment of multiple sclerosis.16  We note that neither of these mAbs is particularly effective at promoting killing of targeted cells. Both mAbs have advanced to clinical trials, and correlative studies in these trials have revealed that these mAbs promote in vivo downregulation of their respective targets on circulating cells. Moreover, in vitro investigations indicated that acceptor monocytes could promote trogocytosis of CD22 and CD25 from B cells and T cells, respectively.15,16 

Trogocytosis of CD22 on B cells mediated by epratuzumab also induces loss of other B-cell markers, including CD19, CD20, CD21, and CD79b. That is, CD22 is not the only protein removed from the B-cell surface, but a microdomain surrounding CD22 is also taken up, thus giving rise to an innocent bystander reaction in which sections of B-cell membranes containing CD22-epratuzumab immune complexes are removed together with nearby cell-surface proteins. This finding recapitulates clinical correlative measurements indicating that these markers, along with CD22, were reduced on B cells of SLE patients treated with epratuzumab, and it was suggested that these changes might therefore reduce autoimmune complications in SLE.15  Likewise, in vitro binding of RTX to B cells promoted the transfer of CD20 and substantial amounts of CD19 to acceptor monocytes and neutrophils.55,67  We reported that B-cell proteins, including CD19 and CD55, are partially removed from Z138 cells, along with CD20, during trogocytosis mediated by RTX.13  We also observed reduced levels of CD19 in our correlative studies, but the CD19 gate was set broadly enough to include CLL B cells with considerably lower CD19 levels.8,12  Thus, as a consequence of trogocytosis, altered levels of critical phenotypic markers could confound enumeration of specific subpopulations during the course of mAb-based therapy. In fact, Masuda et al30  reported that CD3 and its ligand TCRαβ are also transferred to neutrophils during CD8 mAb-mediated in vitro trogocytosis, and found that this could give rise to false-positive flow cytometry results because granulocytes would appear to be CD8+.19 

Mylotarg (gemtuzumab ozogamicin), a humanized IgG4, CD33-specific mAb conjugated to calicheamicin, is an ADC that was used to treat acute myeloid leukemia. When this ADC is internalized by targeted CD33-positive cells, hydrolysis releases calicheamicin from the mAb, thus allowing it to intercalate into cell DNA and induce apoptotic death.68,69  In ∼10% of treated patients, Mylotarg therapy has been associated with severe liver toxicity that can be fatal, and poisoning of the LSECs due to internalization of the ADC is a very likely pathologic mechanism.70-72  The internalization of Mylotarg by CD33+ cells is relatively slow (takes many hours), and this could allow enough time for circulating Mylotarg-opsonized cells to transfer their deadly cargo to LSECs via trogocytosis when they circulate through the liver. FcγRIIb on LSECs can clear circulating immune complexes,46,47,49  and acceptor cell-associated FcγRIIb can promote trogocytosis and remove RTX (human IgG1)-CD20 complexes from opsonized B cells.22  The affinity of FcγRIIb for IgG4 is modestly lower than for human IgG1,73  but it is quite reasonable to expect that aggregated Mylotarg complexes bound to circulating CD33+ cells could reach the multivalent avidity threshold necessary for chelation by a cluster of FcγRIIb on the LSEC, thereby allowing trogocytosis. Because only ∼10% of patients treated with Mylotarg experience severe hepatic injury, it would be important to determine if subtle differences in FcγR activity on their LSECs, or other factors related to immune complex processing, or the unusual properties of human IgG4 antibodies in the circulation,74  could identify those individuals most susceptible to adverse reactions. Also, because Mylotarg was removed from the market,72  it may not be possible to unambiguously determine how it induces liver pathology. At the least, we strongly suggest that future ADC should be examined in appropriate in vitro model systems to determine if they are susceptible to potentially pathologic trogocytosis. This test would especially apply to ADC designed to target circulating cells.

In conclusion, FcγR-mediated trogocytosis is associated with and complicates a variety of mAb-based therapies, likely substantially reducing efficacy in cancer immunotherapy. In view of the continuing interest in the use of mAbs in virtually all aspects of biomedicine, it is very likely that other examples of trogocytosis will be discovered and reported to compromise mAb efficacy in future clinical trials. We recommend that this issue should first be addressed in the laboratory or in preclinical investigations, with a particular emphasis on ADC. In the case of mAb-based immunotherapies for cancer, alternative dosing paradigms may be quite effective in reducing trogocytosis, and thus enhancing the efficacy of antitumor mAbs. Future efforts to specifically suppress trogocytosis without impacting on other FcγR-mediated activities may be most rewarding.

Contribution: R.P.T. and M.A.L. wrote the paper.

Conflict-of-interest disclosure: Our laboratory has received funding from Genmab and GlaxoSmithKline for some of the studies cited in this paper.

Correspondence: Ronald P. Taylor, Department of Biochemistry and Molecular Genetics, Box 800733, University of Virginia Health Sciences Center, Charlottesville, VA 22908-0733; e-mail: rpt@virginia.edu.

1
Joly
 
E
Hudrisier
 
D
What is trogocytosis and what is its purpose?
Nat Immunol
2003
, vol. 
4
 
9
pg. 
815
 
2
Tabiasco
 
J
Espinosa
 
E
Hudrisier
 
D
Joly
 
E
Fournié
 
JJ
Vercellone
 
A
Active trans-synaptic capture of membrane fragments by natural killer cells.
Eur J Immunol
2002
, vol. 
32
 
5
(pg. 
1502
-
1508
)
3
Tabiasco
 
J
Vercellone
 
A
Meggetto
 
F
Hudrisier
 
D
Brousset
 
P
Fournié
 
JJ
Acquisition of viral receptor by NK cells through immunological synapse.
J Immunol
2003
, vol. 
170
 
12
(pg. 
5993
-
5998
)
4
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
, vol. 
178
 
6
(pg. 
3637
-
3647
)
5
Aucher
 
A
Magdeleine
 
E
Joly
 
E
Hudrisier
 
D
Capture of plasma membrane fragments from target cells by trogocytosis requires signaling in T cells but not in B cells.
Blood
2008
, vol. 
111
 
12
(pg. 
5621
-
5628
)
6
Taylor
 
RP
Gnawing at Metchnikoff’s paradigm.
Blood
2013
, vol. 
122
 
17
(pg. 
2922
-
2924
)
7
Dustin
 
ML
What counts in the immunological synapse?
Mol Cell
2014
, vol. 
54
 
2
(pg. 
255
-
262
)
8
Beurskens
 
FJ
Lindorfer
 
MA
Farooqui
 
M
, et al. 
Exhaustion of cytotoxic effector systems may limit monoclonal antibody-based immunotherapy in cancer patients.
J Immunol
2012
, vol. 
188
 
7
(pg. 
3532
-
3541
)
9
Glennie
 
MJ
French
 
RR
Cragg
 
MS
Taylor
 
RP
Mechanisms of killing by anti-CD20 monoclonal antibodies.
Mol Immunol
2007
, vol. 
44
 
16
(pg. 
3823
-
3837
)
10
Taylor
 
RP
Lindorfer
 
MA
Immunotherapeutic mechanisms of anti-CD20 monoclonal antibodies.
Curr Opin Immunol
2008
, vol. 
20
 
4
(pg. 
444
-
449
)
11
Golay
 
J
Introna
 
M
Mechanism of action of therapeutic monoclonal antibodies: promises and pitfalls of in vitro and in vivo assays.
Arch Biochem Biophys
2012
, vol. 
526
 
2
(pg. 
146
-
153
)
12
Kennedy
 
AD
Beum
 
PV
Solga
 
MD
, et al. 
Rituximab infusion promotes rapid complement depletion and acute CD20 loss in chronic lymphocytic leukemia.
J Immunol
2004
, vol. 
172
 
5
(pg. 
3280
-
3288
)
13
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
)
14
Beum
 
PV
Peek
 
EM
Lindorfer
 
MA
, et al. 
Loss of CD20 and bound CD20 antibody from opsonized B cells occurs more rapidly because of trogocytosis mediated by Fc receptor-expressing effector cells than direct internalization by the B cells.
J Immunol
2011
, vol. 
187
 
6
(pg. 
3438
-
3447
)
15
Rossi
 
EA
Goldenberg
 
DM
Michel
 
R
Rossi
 
DL
Wallace
 
DJ
Chang
 
CH
Trogocytosis of multiple B-cell surface markers by CD22 targeting with epratuzumab.
Blood
2013
, vol. 
122
 
17
(pg. 
3020
-
3029
)
16
Zhang
 
Y
McClellan
 
M
Efros
 
L
, et al. 
Daclizumab reduces CD25 levels on T cells through monocyte-mediated trogocytosis.
Mult Scler
2014
, vol. 
20
 
2
(pg. 
156
-
164
)
17
Daubeuf
 
S
Lindorfer
 
MA
Taylor
 
RP
Joly
 
E
Hudrisier
 
D
The direction of plasma membrane exchange between lymphocytes and accessory cells by trogocytosis is influenced by the nature of the accessory cell.
J Immunol
2010
, vol. 
184
 
4
(pg. 
1897
-
1908
)
18
Beum
 
PV
Mack
 
DA
Pawluczkowycz
 
AW
Lindorfer
 
MA
Taylor
 
RP
Binding of rituximab, trastuzumab, cetuximab, or mAb T101 to cancer cells promotes trogocytosis mediated by THP-1 cells and monocytes.
J Immunol
2008
, vol. 
181
 
11
(pg. 
8120
-
8132
)
19
Masuda
 
S
Iwasaki
 
S
Tomaru
 
U
, et al. 
Mechanism of Fcγ receptor-mediated trogocytosis-based false-positive results in flow cytometry.
PLoS ONE
2012
, vol. 
7
 
12
pg. 
e52918
 
20
Baig
 
NA
Taylor
 
RP
Lindorfer
 
MA
, et al. 
Induced resistance to ofatumumab-mediated cell clearance mechanisms, including complement-dependent cytotoxicity, in chronic lymphocytic leukemia.
J Immunol
2014
, vol. 
192
 
4
(pg. 
1620
-
1629
)
21
Iwasaki
 
S
Masuda
 
S
Baba
 
T
, et al. 
Plasma-dependent, antibody- and Fcγ receptor-mediated translocation of CD8 molecules from T cells to monocytes.
Cytometry A
2011
, vol. 
79
 
1
(pg. 
46
-
56
)
22
Boross
 
P
Jansen
 
JH
Pastula
 
A
van der Poel
 
CE
Leusen
 
JHW
Both activating and inhibitory Fc γ receptors mediate rituximab-induced trogocytosis of CD20 in mice.
Immunol Lett
2012
, vol. 
143
 
1
(pg. 
44
-
52
)
23
Nelson
 
RA
The immune-adherence phenomenon; an immunologically specific reaction between microorganisms and erythrocytes leading to enhanced phagocytosis.
Science
1953
, vol. 
118
 
3077
(pg. 
733
-
737
)
24
Taylor
 
RP
Sutherland
 
WM
Martin
 
EN
, et al. 
Bispecific monoclonal antibody complexes bound to primate erythrocyte complement receptor 1 facilitate virus clearance in a monkey model.
J Immunol
1997
, vol. 
158
 
2
(pg. 
842
-
850
)
25
Lindorfer
 
MA
Nardin
 
A
Foley
 
PL
, et al. 
Targeting of Pseudomonas aeruginosa in the bloodstream with bispecific monoclonal antibodies.
J Immunol
2001
, vol. 
167
 
4
(pg. 
2240
-
2249
)
26
Lindorfer
 
MA
Kohl
 
J
Taylor
 
RP
Ackerman
 
ME
Nimmerjahn
 
F
Interactions between the complement system and Fcg receptors.
Antibody Fc: Linking Adaptive and Innate Immunity
2014
Philadelphia, PA
Elsevier
(pg. 
49
-
74
)
27
Lindorfer
 
MA
Jinivizian
 
HB
Foley
 
PL
Kennedy
 
AD
Solga
 
MD
Taylor
 
RP
B cell complement receptor 2 transfer reaction.
J Immunol
2003
, vol. 
170
 
7
(pg. 
3671
-
3678
)
28
Whipple
 
EC
Shanahan
 
RS
Ditto
 
AH
Taylor
 
RP
Lindorfer
 
MA
Analyses of the in vivo trafficking of stoichiometric doses of an anti-complement receptor 1/2 monoclonal antibody infused intravenously in mice.
J Immunol
2004
, vol. 
173
 
4
(pg. 
2297
-
2306
)
29
Whipple
 
EC
Ditto
 
AH
Shanahan
 
RS
, et al. 
Low doses of antigen coupled to anti-CR2 mAbs induce rapid and enduring IgG immune responses in mice and in cynomolgus monkeys.
Mol Immunol
2007
, vol. 
44
 
4
(pg. 
377
-
388
)
30
Masuda
 
S
Iwasaki
 
S
Tomaru
 
U
Baba
 
T
Katsumata
 
K
Ishizu
 
A
Possible implication of Fc γ receptor-mediated trogocytosis in susceptibility to systemic autoimmune disease.
Clin Dev Immunol
2013
, vol. 
2013
 pg. 
345745
 
31
Griffin
 
FM
Griffin
 
JA
Silverstein
 
SC
Studies on the mechanism of phagocytosis. II. The interaction of macrophages with anti-immunoglobulin IgG-coated bone marrow-derived lymphocytes.
J Exp Med
1976
, vol. 
144
 
3
(pg. 
788
-
809
)
32
Old
 
LJ
Boyse
 
EA
Antigenic properties of experimental leukemias. I. Serological studies in vitro with spontaneous and radiation-induced leukemias.
J Natl Cancer Inst
1963
, vol. 
31
 (pg. 
977
-
995
)
33
Press
 
OW
Farr
 
AG
Borroz
 
KI
Anderson
 
SK
Martin
 
PJ
Endocytosis and degradation of monoclonal antibodies targeting human B-cell malignancies.
Cancer Res
1989
, vol. 
49
 
17
(pg. 
4906
-
4912
)
34
Stackpole
 
CW
Jacobson
 
JB
Lardis
 
MP
Antigenic modulation in vitro. I. Fate of thymus-leukemia (TL) antigen-antibody complexes following modulation of TL antigenicity from the surfaces of mouse leukemia cells and thymocytes.
J Exp Med
1974
, vol. 
140
 
4
(pg. 
939
-
953
)
35
Kammer
 
GM
Smith
 
JA
Mitchell
 
R
Capping of human T cell specific determinants: kinetics of capping and receptor re-expression and regulation by the cytoskeleton.
J Immunol
1983
, vol. 
130
 
1
(pg. 
38
-
44
)
36
Chatenoud
 
L
Baudrihaye
 
MF
Kreis
 
H
Goldstein
 
G
Schindler
 
J
Bach
 
JF
Human in vivo antigenic modulation induced by the anti-T cell OKT3 monoclonal antibody.
Eur J Immunol
1982
, vol. 
12
 
11
(pg. 
979
-
982
)
37
Rinnooy Kan
 
EA
Platzer
 
E
Welte
 
K
Wang
 
CY
Modulation induction of the T3 antigen by OKT3 antibody is monocyte dependent.
J Immunol
1984
, vol. 
133
 
6
(pg. 
2979
-
2985
)
38
Dillman
 
RO
Shawler
 
DL
Sobol
 
RE
, et al. 
Murine monoclonal antibody therapy in two patients with chronic lymphocytic leukemia.
Blood
1982
, vol. 
59
 
5
(pg. 
1036
-
1045
)
39
Schroff
 
RW
Farrell
 
MM
Klein
 
RA
Oldham
 
RK
Foon
 
KA
T65 antigen modulation in a phase I monoclonal antibody trial with chronic lymphocytic leukemia patients.
J Immunol
1984
, vol. 
133
 
3
(pg. 
1641
-
1648
)
40
Foon
 
KA
Schroff
 
RW
Bunn
 
PA
, et al. 
Effects of monoclonal antibody therapy in patients with chronic lymphocytic leukemia.
Blood
1984
, vol. 
64
 
5
(pg. 
1085
-
1093
)
41
Schroff
 
RW
Klein
 
RA
Farrell
 
MM
Stevenson
 
HC
Enhancing effects of monocytes on modulation of a lymphocyte membrane antigen.
J Immunol
1984
, vol. 
133
 
4
(pg. 
2270
-
2277
)
42
Schroff
 
RW
Farrell
 
MM
Klein
 
RA
Stevenson
 
HC
Warner
 
NL
Induction and enhancement by monocytes of antibody-induced modulation of a variety of human lymphoid cell surface antigens.
Blood
1985
, vol. 
66
 
3
(pg. 
620
-
626
)
43
Bertram
 
JH
Gill
 
PS
Levine
 
AM
, et al. 
Monoclonal antibody T101 in T cell malignancies: a clinical, pharmacokinetic, and immunologic correlation.
Blood
1986
, vol. 
68
 
3
(pg. 
752
-
761
)
44
Dillman
 
RO
Beauregard
 
J
Shawler
 
DL
, et al. 
Continuous infusion of T101 monoclonal antibody in chronic lymphocytic leukemia and cutaneous T-cell lymphoma.
J Biol Response Mod
1986
, vol. 
5
 
5
(pg. 
394
-
410
)
45
Tam
 
CS
Otero-Palacios
 
J
Abruzzo
 
LV
, et al. 
Chronic lymphocytic leukaemia CD20 expression is dependent on the genetic subtype: a study of quantitative flow cytometry and fluorescent in-situ hybridization in 510 patients.
Br J Haematol
2008
, vol. 
141
 
1
(pg. 
36
-
40
)
46
Løvdal
 
T
Andersen
 
E
Brech
 
A
Berg
 
T
Fc receptor mediated endocytosis of small soluble immunoglobulin G immune complexes in Kupffer and endothelial cells from rat liver.
J Cell Sci
2000
, vol. 
113
 
pt 18
(pg. 
3255
-
3266
)
47
Mousavi
 
SA
Sporstøl
 
M
Fladeby
 
C
Kjeken
 
R
Barois
 
N
Berg
 
T
Receptor-mediated endocytosis of immune complexes in rat liver sinusoidal endothelial cells is mediated by FcgammaRIIb2.
Hepatology
2007
, vol. 
46
 
3
(pg. 
871
-
884
)
48
Taylor
 
RP
Lindorfer
 
MA
Antigenic modulation and rituximab resistance.
Semin Hematol
2010
, vol. 
47
 
2
(pg. 
124
-
132
)
49
Ganesan
 
LP
Kim
 
J
Wu
 
Y
, et al. 
FcγRIIb on liver sinusoidal endothelium clears small immune complexes.
J Immunol
2012
, vol. 
189
 
10
(pg. 
4981
-
4988
)
50
Berdeja
 
JG
Hess
 
A
Lucas
 
DM
, 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
, vol. 
13
 
8
(pg. 
2392
-
2399
)
51
Bhat
 
R
Watzl
 
C
Serial killing of tumor cells by human natural killer cells—enhancement by therapeutic antibodies.
PLoS ONE
2007
, vol. 
2
 
3
pg. 
e326
 
52
Zent
 
CS
Taylor
 
RP
Lindorfer
 
MA
, et al. 
Chemoimmunotherapy for relapsed/refractory and progressive 17p13-deleted chronic lymphocytic leukemia (CLL) combining pentostatin, alemtuzumab, and low-dose rituximab is effective and tolerable and limits loss of CD20 expression by circulating CLL cells.
Am J Hematol
2014
, vol. 
89
 
7
(pg. 
757
-
765
)
53
Taylor
 
RP
Lindorfer
 
MA
Analyses of CD20 monoclonal antibody-mediated tumor cell killing mechanisms: rational design of dosing strategies.
Mol Pharmacol
2014
, vol. 
86
 
5
(pg. 
485
-
491
)
54
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
)
55
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
, vol. 
181
 
4
(pg. 
2916
-
2924
)
56
Nijmeijer
 
BA
van Schie
 
MLJ
Halkes
 
CJM
Griffioen
 
M
Willemze
 
R
Falkenburg
 
JH
A mechanistic rationale for combining alemtuzumab and rituximab in the treatment of ALL.
Blood
2010
, vol. 
116
 
26
(pg. 
5930
-
5940
)
57
Pedersen
 
AE
Jungersen
 
MB
Pedersen
 
CD
Monocytes mediate shaving of B-cell-bound anti-CD20 antibodies.
Immunology
2011
, vol. 
133
 
2
(pg. 
239
-
245
)
58
Pham
 
T
Mero
 
P
Booth
 
JW
Dynamics of macrophage trogocytosis of rituximab-coated B cells.
PLoS ONE
2011
, vol. 
6
 
1
pg. 
e14498
 
59
Beers
 
SA
French
 
RR
Chan
 
HT
, et al. 
Antigenic modulation limits the efficacy of anti-CD20 antibodies: implications for antibody selection.
Blood
2010
, vol. 
115
 
25
(pg. 
5191
-
5201
)
60
Lim
 
SH
Vaughan
 
AT
Ashton-Key
 
M
, et al. 
Fc gamma receptor IIb on target B cells promotes rituximab internalization and reduces clinical efficacy.
Blood
2011
, vol. 
118
 
9
(pg. 
2530
-
2540
)
61
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
)
62
Teng
 
YKO
Levarht
 
EWN
Hashemi
 
M
, et al. 
Immunohistochemical analysis as a means to predict responsiveness to rituximab treatment.
Arthritis Rheum
2007
, vol. 
56
 
12
(pg. 
3909
-
3918
)
63
Laurent
 
C
de Paiva
 
GR
Ysebaert
 
L
, et al. 
Characterization of bone marrow lymphoid infiltrates after immunochemotherapy for follicular lymphoma.
Am J Clin Pathol
2007
, vol. 
128
 
6
(pg. 
974
-
980
)
64
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
2010
, vol. 
95
 
2
(pg. 
329
-
332
)
65
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
)
66
Rossi
 
EA
Chang
 
CH
Goldenberg
 
DM
Anti-CD22/CD20 Bispecific antibody with enhanced trogocytosis for treatment of Lupus.
PLoS ONE
2014
, vol. 
9
 
5
pg. 
e98315
 
67
Jones
 
JD
Hamilton
 
BJ
Rigby
 
WFC
Rituximab mediates loss of CD19 on B cells in the absence of cell death.
Arthritis Rheum
2012
, vol. 
64
 
10
(pg. 
3111
-
3118
)
68
van Der Velden
 
VHJ
te Marvelde
 
JG
Hoogeveen
 
PG
, et al. 
Targeting of the CD33-calicheamicin immunoconjugate Mylotarg (CMA-676) in acute myeloid leukemia: in vivo and in vitro saturation and internalization by leukemic and normal myeloid cells.
Blood
2001
, vol. 
97
 
10
(pg. 
3197
-
3204
)
69
Arceci
 
RJ
Sande
 
J
Lange
 
B
, et al. 
Safety and efficacy of gemtuzumab ozogamicin in pediatric patients with advanced CD33+ acute myeloid leukemia.
Blood
2005
, vol. 
106
 
4
(pg. 
1183
-
1188
)
70
Giles
 
FJ
Kantarjian
 
HM
Kornblau
 
SM
, et al. 
Mylotarg (gemtuzumab ozogamicin) therapy is associated with hepatic venoocclusive disease in patients who have not received stem cell transplantation.
Cancer
2001
, vol. 
92
 
2
(pg. 
406
-
413
)
71
Rajvanshi
 
P
Shulman
 
HM
Sievers
 
EL
McDonald
 
GB
Hepatic sinusoidal obstruction after gemtuzumab ozogamicin (Mylotarg) therapy.
Blood
2002
, vol. 
99
 
7
(pg. 
2310
-
2314
)
72
Tallman
 
MS
McDonald
 
GB
DeLeve
 
LD
, et al. 
Incidence of sinusoidal obstruction syndrome following Mylotarg (gemtuzumab ozogamicin): a prospective observational study of 482 patients in routine clinical practice.
Int J Hematol
2013
, vol. 
97
 
4
(pg. 
456
-
464
)
73
Ravetch
 
JV
Nimmerjahn
 
F
Paul
 
WE
Fc receptors and their role in immune regulation and inflammation.
Fundamental Immunology
2008
Philadelphia, PA
Lippincott Williams & Wilkins
(pg. 
684
-
705
)
74
Labrijn
 
AF
Buijsse
 
AO
van den Bremer
 
ETJ
, et al. 
Therapeutic IgG4 antibodies engage in Fab-arm exchange with endogenous human IgG4 in vivo.
Nat Biotechnol
2009
, vol. 
27
 
8
(pg. 
767
-
771
)
75
Shawler
 
DL
Johnson
 
DE
McCallister
 
TJ
Bartholomew
 
RM
Dillman
 
RO
Mechanisms of human CD5 modulation and capping induced by murine monoclonal antibody T101.
Clin Immunol Immunopathol
1988
, vol. 
47
 
2
(pg. 
219
-
229
)
76
Foss
 
FM
Raubitscheck
 
A
Mulshine
 
JL
, et al. 
Phase I study of the pharmacokinetics of a radioimmunoconjugate, 90Y-T101, in patients with CD5-expressing leukemia and lymphoma.
Clin Cancer Res
1998
, vol. 
4
 
11
(pg. 
2691
-
2700
)
77
Madsen
 
CB
Lavrsen
 
K
Steentoft
 
C
, et al. 
Glycan elongation beyond the mucin associated Tn antigen protects tumor cells from immune-mediated killing.
PLoS ONE
2013
, vol. 
8
 
9
pg. 
e72413
 
Sign in via your Institution