To the editor:

There remains considerable uncertainty regarding the role of complement as an effector system for anti-cancer monoclonal antibodies (mAbs) such as rituximab and alemtuzumab. We were, therefore, interested to read the intriguing findings of Wang et al,1  indicating that complement activation can be detrimental to antibody immunotherapy. They suggest that breakdown products of the complement component C3 block anti-lymphoma mAb activity and render natural killer (NK) cells less active in antibody-dependent cellular cytotoxicity and the treatment of the 38C13 lymphoma with anti-idiotype mAb.1  Clearly, these are important observations, well supported by in vitro experimentation. However, we feel it is important to stress that the beneficial effect of complement inactivation in vivo is not the general experience with therapeutic mAb, where the absence of complement typically either reduces efficacy2,3  or makes no difference4,5  (reviewed in Lim et al6 ).

By way of example, Figure 1 shows that depletion of normal B cells with anti-CD20 mAb is equally effective regardless of whether the mice are deficient in C1q or C3 or the mAb is engineered not to engage C1q.

Figure 1

Complement does not effect the ability of anti-CD20 mAb to deplete B cells in vivo. (A) BALB/c human CD20 transgenic mice7  (WT or C1q−/−) received 250 μg of anti–human CD20 mAb (Rit m2a7) or mAb lacking C1q binding activity (K322A mutation7 ) intravenously on day 0. The number of circulating B cells was then assessed by flow cytometry for CD19 and B220. The results are expressed as percentage of B cells observed at time 0. (B) Experiments show adoptive transfer of hCD20 Tg (target: high carboxyfluorescein succinimidyl ester [Sigma-Aldrich]) and WT (nontarget: low carboxyfluorescein succinimidyl ester) splenic B cells into recipient mice carrying various effector defects (C1q−/−, C3−/− [Jackson Laboratories], Fcγ chain−/−, and clodronate [Sigma-Aldrich], treated). Twenty-four hours later, mice received control or Rit m2a mAb (10 μg, intravenously), and 16 hours later splenic B cells were analyzed by flow cytometry to determine the target:nontarget ratio. Bars represent mean ± SD, n = 3; each condition is representative of at least 2 independent experiments. The data clearly demonstrate that Rit m2a only deletes target cells (low target:nontarget ratio), and that activatory FcR (absent in Fcγ chain−/− mice) and macrophages (deleted in clodronate-treated mice), but not complement components C1q or C3, are important for target-cell deletion.

Figure 1

Complement does not effect the ability of anti-CD20 mAb to deplete B cells in vivo. (A) BALB/c human CD20 transgenic mice7  (WT or C1q−/−) received 250 μg of anti–human CD20 mAb (Rit m2a7) or mAb lacking C1q binding activity (K322A mutation7 ) intravenously on day 0. The number of circulating B cells was then assessed by flow cytometry for CD19 and B220. The results are expressed as percentage of B cells observed at time 0. (B) Experiments show adoptive transfer of hCD20 Tg (target: high carboxyfluorescein succinimidyl ester [Sigma-Aldrich]) and WT (nontarget: low carboxyfluorescein succinimidyl ester) splenic B cells into recipient mice carrying various effector defects (C1q−/−, C3−/− [Jackson Laboratories], Fcγ chain−/−, and clodronate [Sigma-Aldrich], treated). Twenty-four hours later, mice received control or Rit m2a mAb (10 μg, intravenously), and 16 hours later splenic B cells were analyzed by flow cytometry to determine the target:nontarget ratio. Bars represent mean ± SD, n = 3; each condition is representative of at least 2 independent experiments. The data clearly demonstrate that Rit m2a only deletes target cells (low target:nontarget ratio), and that activatory FcR (absent in Fcγ chain−/− mice) and macrophages (deleted in clodronate-treated mice), but not complement components C1q or C3, are important for target-cell deletion.

Close modal

Several possibilities exist to explain these apparently contradictory findings. The first is that the anti-Id/38C13 lymphoma model is unusually dependent on NK cells, which, in turn, are particularly sensitive to blocking by C3b, consistent with the interpretation given by Wang et al. However, this seems unlikely, since previous work from this group has shown that granulocytes also play a role in this model.8  In addition, successful mAb immunotherapy in mice, including those with anti-CD20 mAb, commonly require an intact mononuclear phagocytic system, but not NK cells (reviewed in Lim et al6 ).

This leaves 2 further potential explanations. The first is that idiotype may be an atypical target for mAb. In support of this, Golay et al have demonstrated that therapy of a hCD20+ variant of 38C13 with CD20 mAb was completely dependent upon complement activity, with no role for NK cells or neutrophils.3  These findings are clearly counter to the results described by Wang et al, perhaps indicating that impact of complement on therapy is determined by the degree of complement activation, with the high levels evoked through CD20 mAb usually being beneficial to therapy, and lower levels, as occur with anti-idiotype mAb, being detrimental as observed. Second, it must be considered that cobra venom factor (CVF) treatment has functions other than simple complement depletion. Rather than removing complement passively, CVF acts as an unregulated analog of C3b, resulting in uncontrolled activation of C3 with systemic release of breakdown products that impact on innate and acquired immune activation. Hence, CVF treatment provides a useful model of acute respiratory distress syndrome where it leads to acute organ damage.9  The release of cytokines and chemokines and accumulation of neutrophils after CVF are precisely the inflammatory conditions that might influence the growth of a small number (5000) of passaged tumor cells, particularly with an immunogenic lymphoma such as 38C13.10  In accordance with this supposition, the results from Wang et al show that CVF alone slows the growth of 38C13 to that seen after treatment with anti-Id mAb. Therefore, the synergistic activity of anti-Id mAb and CVF leading to long-term survival is consistent with the development of acquired immunity. Thus, we feel that an adjuvant effect of CVF must be considered as a plausible alternative explanation of the current in vivo data, especially given the weight of prevailing evidence showing that complement is not deleterious to direct targeting antibody immunotherapy.

Acknowledgments: The authors thank the following for the provision of mice: Prof Mark Shlomchik (Yale School of Medicine, New Haven, CT) for hCD20 Tg; Prof Marina Botto (Imperial College, London, United Kingdom) and Dr Aras Kadioglu (University of Leicester, Leicester, United Kingdom) for C1q−/−; and Prof Jan van de Winkel (University Medical, Utrecht, The Netherlands) for FcγR−/−.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Martin J. Glennie, Tenovus Research Laboratory, Cancer Sciences Division (mailpoint 88), University of Southampton School of Medicine, Tremona Rd, Southampton, United Kingdom SO16 6YD; e-mail: mjg@soton.ac.uk.

1
Wang
 
S-Y
Veeramani
 
S
Racila
 
E
, 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
, vol. 
114
 
26
(pg. 
5322
-
5330
)
2
Cragg
 
MS
Bayne
 
MB
Tutt
 
AL
, et al. 
A new anti-idiotype antibody capable of binding rituximab on the surface of lymphoma cells.
Blood
2004
, vol. 
104
 
8
(pg. 
2540
-
2542
)
3
Golay
 
J
Cittera
 
E
Di Gaetano
 
N
, et al. 
The role of complement in the therapeutic activity of rituximab in a murine B lymphoma model homing in lymph nodes.
Haematologica
2006
, vol. 
91
 
2
(pg. 
176
-
183
)
4
Hamaguchi
 
Y
Uchida
 
J
Cain
 
DW
, et al. 
The peritoneal cavity provides a protective niche for B1 and conventional B lymphocytes during anti-CD20 immunotherapy in mice.
J Immunol
2005
, vol. 
174
 
7
(pg. 
4389
-
4399
)
5
Minard-Colin
 
V
Xiu
 
Y
Poe
 
JC
, et al. 
Lymphoma depletion during CD20 immunotherapy in mice is mediated by macrophage FcgammaRI, FcgammaRIII, and FcgammaRIV.
Blood
2008
, vol. 
112
 
4
(pg. 
1205
-
1213
)
6
Lim
 
SH
Beers
 
SA
French
 
RR
Johnson
 
PW
Glennie
 
MJ
Cragg
 
MS
Anti-CD20 monoclonal antibodies: historical and future perspectives.
Haematologica
 
Prepublished on September 22, 2009, as doi 10.3324/haematol.2008.001628
7
Beers
 
SA
Chan
 
CH
James
 
S
, et al. 
Type II (tositumomab) anti-CD20 monoclonal antibody out performs type I (rituximab-like) reagents in B-cell depletion regardless of complement activation.
Blood
2008
, vol. 
112
 
10
(pg. 
4170
-
4177
)
8
van Ojik
 
HH
Bevaart
 
L
Dahle
 
CE
, et al. 
CpG-A and B oligodeoxynucleotides enhance the efficacy of antibody therapy by activating different effector cell populations.
Cancer Res
2003
, vol. 
63
 
17
(pg. 
5595
-
5600
)
9
Younger
 
JG
Sasaki
 
N
Delgado
 
J
, et al. 
Systemic and lung physiological changes in rats after intravascular activation of complement.
J Appl Physiol
2001
, vol. 
90
 
6
(pg. 
2289
-
2295
)
10
Franki
 
SN
Steward
 
KK
Betting
 
DJ
Kafi
 
K
Yamada
 
RE
Timmerman
 
JM
Dendritic cells loaded with apoptotic antibody-coated tumor cells provide protective immunity against B-cell lymphoma in vivo.
Blood
2008
, vol. 
111
 
3
(pg. 
1504
-
1511
)
Sign in via your Institution