Survey
* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project
* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project
Inmunol3/2006 copia 13/12/06 16:38 Página 173 Revisión Inmunología Vol. 25 / Núm 3/ Julio-Septiembre 2006: 173-187 Control of complement activation by cancer cells and its implications in antibody-mediated cancer immunotherapy R. Pio Division of Oncology (Center for Applied Medical Research) and Department of Biochemistry. School of Medicine, University of Navarra, Pamplona, Spain. CONTROL DE LA ACTIVACIÓN DEL COMPLEMENTO EN CÉLULAS MALIGNAS Y SU IMPLICACIÓN EN LA INMUNOTERAPIA ANTITUMORAL CON ANTICUERPOS MONOCLONALES Recibido: 10 Junio 2006 Aceptado: 5 Septiembre 2006 RESUMEN El complemento es una parte esencial del sistema inmune innato que participa en la eliminación de células extrañas al organismo. Debido al gran número de alteraciones genéticas y epigenéticas asociadas a la carcinogénesis, la transformación neoplásica puede incrementar la capacidad de la célula maligna para activar el complemento. Este hecho está sustentado por estudios clínicos que demuestran una activación del complemento en pacientes con cáncer. Sin embargo, las células malignas suelen desarrollar mecanismos de protección que les hacen resistentes al complemento. A la luz de recientes investigaciones sobre los mecanismos de regulación de la actividad del complemento, el papel de las proteínas inhibidoras, tanto de membrana como solubles, en la protección de las células neoplásicas es cada vez más evidente. Esto podría limitar la eficacia de la inmunoterapia antitumoral basada en anticuerpos monoclonales que, entro otros mecanismos, pueden activar el sistema del complemento. Se han sugerido, y testado, distintas estrategias para la supresión de los mecanismos de control de la activación del complemento. En estudios in vitro e in vivo, la protección mediada por proteínas reguladoras ha podido ser bloqueada mediante la inhibición de su actividad o de su expresión por parte de la célula tumoral. También se han evaluado estrategias dirigidas a incrementar la capacidad de los anticuerpos para fijar el complemento o los mecanismos efectores asociados a su activación. Sin duda, un mejor conocimiento del papel del complemento y de sus mecanismos de control en cáncer ayudará al diseño de inmunoterapias antitumorales más eficaces. PALABRAS CLAVE: Neoplasias / Proteínas inhibidoras del complemento / Inmunoterapia / Anticuerpos. ABSTRACT Complement is a central part of the innate immune system, providing a highly effective means for destruction of non-self cells. Given the numerous genetic and epigenetic changes associated with carcinogenesis, neoplastic transformation may be accompanied by an increased capacity of the malignant cells to activate complement. This is supported by clinical data that demonstrate complement activation in cancer patients. However, malignant cells are often resistant to complement activation by the use of various protective mechanisms. In light of recent advances in the knowledge of the mechanisms regulating complement activity, it begins to be clear that membrane-bound and soluble complement inhibitory proteins play a key role in the protection of neoplastic cells from complement attack. This may hamper the clinical efficacy of cancer immunotherapy strategies based on the use of monoclonal antibodies that, among other mechanisms, can activate complement. Some attempts have been already made to modulate antibody-mediated complement activation. In in vitro and in vivo studies, protection by complement regulatory proteins has been overcome by inhibiting their activities or their expression by the target cells. Other strategies have been aimed to increase the complement-fixing capacity of the therapeutic antibodies or to improve complement-mediated effector mechanisms. Undoubtedly, a better understanding of the role of complement and its mechanisms of control in malignant cells would help to design more efficient complement-mediated cancer immunotherapies. KEY WORDS: Neoplasms / Complement inactivator proteins / Immunotherapy /Antibodies. 173 Inmunol3/2006 copia 13/12/06 16:38 Página 174 CONTROL OF COMPLEMENT ACTIVATION BY CANCER CELLS AND ITS IMPLICATIONS IN ANTIBODY-MEDIATED ... THE COMPLEMENT SYSTEM AND ITS REGULATION Complement is a central part of the innate immune system, providing a highly effective means for destruction of invading microorganisms, clearance of immune complexes and elimination of dead and apoptotic cells(1). The complement system is one of the most ancient parts of the immune system, present in evolution long before the development of adaptive immunity, with which it co-operates(2-4). Complement can enhance humoral immunity(5), modifies T cell immunity(6), shapes the development of the natural antibody repertoire(7), and regulates tolerance to nuclear self antigens(8,9). Complement is composed of both circulating and membrane-bound proteins and can be activated via three distinct pathways: the classical, the alternative and the lectin pathway. The contact of the first component with an activator on the target cell (i.e. an immunoglobulin, an activating surface or certain carbohydrate structures) leads to the generation of a cascade of activations in a precise order depending on the pathway that is activated. Complement proteins can be either zymogens that become active enzymes upon activation of complement, effectors, control proteins or receptors. The three complement pathways share the common step of activating the central component C3, and differ in the mechanism of target recognition. The classical pathway of complement is initiated by the binding of C1q to Fc regions of antigen-bound immunoglobulins (IgG or IgM). C1q together with C1r and C1s, two serine protease proenzymes, constitute C1, the first component of the classical pathway(10). Conformational changes in C1q lead to the activation of C1r, which, in turn, activates C1s(11). The activation of the C1q complex subsequently activates complement through the cleavage of C4 and C2 to yield the classical pathway C3 convertase (C4b2a) that is able to cleave C3. The alternative pathway is the phylogenetically oldest one and is triggered by low-level activation of C3 to C3b by spontaneously hydrolyzed C3 and activated factor B(12,13). C3b can attach to the target cell membrane, and bind to factor B that is cleaved by factor D to form the alternative pathway C3 convertase (C3bBb). The lectin pathway is activated following the recognition and binding of mannose-binding lectin (MBL) to repetitive carbohydrate patterns containing mannose and N-acetylglucosamine residues on pathogen surfaces (14,15). MBL forms a C1-like complex with MBLassociated serine proteases (MASP). Conformational changes in MBL lead to the cleavage and activation of complement components C4, C2, which continue activation as in the classical pathway(11). In all three pathways, cleavage and activation of C3 results in the deposition of C3b on the surface of the target cell, leading to the activation of the C5–C9 components and the formation of the cytolytic membrane 174 VOL. 25 NUM. 3/ 2006 attack complex (MAC) that binds to cell membranes, disrupts the membrane’s integrity and facilitates cell lysis in a process known as complement-dependent cytotoxicity (CDC). When C3b is converted to iC3b, complement-dependent cellular cytotoxicity (CDCC) links with antibody-dependent cellular cytotoxicity (ADCC) through the interaction of iC3b with complement receptor 3 (CR3; CD11b/CD18) on mononuclear phagocytes(16), natural killer cells(17), and lymphocytes(18). Eventually, the complement cascade of proteolytic enzymes releases the anaphylatoxins C4a, C3a, and C5a. These small bioactive peptides act as chemotaxins and leukocyte activators, mediating important proinflammatory responses(19). To protect host cells from bystander killing and as a mechanism of regulation, activation of the complement cascade is highly controlled by several regulatory proteins. Control proteins regulate complement at three main levels: they can inhibit the protease activity of the proteins involved in the activation cascade, facilitate the decay and destruction of convertases, and control the formation of the MAC. At least six complement regulators can be found soluble in plasma: C1 inhibitor, factor I, factor H, C4b-binding protein (C4bp), vitronectin (S-protein) and clusterin (SP40,40). C1 inhibitor prevents the cleavage of C4 and C2(20). Factor I cleaves and inactivates C4b and C3b (21). Factor H and C4bp have a decay-accelerating activity for the alternative and the classical C3 convertases, respectively(22,23). These two proteins are also cofactors for factor I. Vitronectin and clusterin inhibit the insertion of the MAC into the membrane(24,25). Complement activation can be also controlled by membranebound complement regulatory proteins (mCRPs) such as complement receptor type 1 (CR1, CD35), membrane cofactor protein (MCP, CD46), decay-accelerating factor (DAF, CD55), and CD59 (protectin). CR1 functions as a cofactor for factor I and dissociates the C3 and C5 convertases(26,27). CR1 has a restricted tissue distribution (erythrocytes, most leucocytes and tissue macrophages). CD46 is ubiquitously expressed except on erythrocytes and works like CR1 in acting as a cofactor protein for factor I-mediated cleavage of C3b(28). CD55 is a glycosyl phosphatidylinositol (GPI)-anchored protein that inhibits the activation of C3 and C5 by preventing the formation of new C3 and C5 convertases and accelerating the decay of preformed convertases(29). Finally, CD59, also a GPI-anchored protein, prevents the formation of the MAC at the terminal step of the complement activation cascade(30,31). Both CD55 and CD59 are expressed on the surface of virtually all cell types. mCRPs are usually species-specific, protecting the cells only from autologous or homologous complement(32). Structurally, CR1, CD46 and CD55 are closely related to the soluble regulatory proteins factor H and C4bp. They belong to the RCA (regulators of complement activation) family of Inmunol3/2006 copia 13/12/06 16:38 Página 175 INMUNOLOGÍA Figure 1. Structure characteristics of human mCRPs CR1, CD46, CD55, CD59 and the soluble regulators factor H and C4bp. These regulators, except for CD59, contain extracellular short consensus repeats (SCR) domains. SCR domains, also known as sushi domains or complement control protein (CCP) domains, are sixty amino acids long and have four invariant cysteine residues forming two disulfide-bridges per domain. CD46 and CD55 contain four SCR domains each, and factor H twenty. C4bp exists in several isoforms. In the figure the major isoform is depicted, which consists of seven α-chains (with eight SCR domains each) and one β-chain (with three SCR domains) linked together in their C-terminal parts. CD59 does not contain SCR domains; its extracellular domain consists of a seventy amino acid chain with five disulfide bonds. Both CD55 and CD59 are glycophosphatidylinositol (GPI)-anchored proteins, while CR1 and CD46 are inserted into the cell membrane through their transmembrane domains. genes and contain a variable number of short consensus repeat (SCR) domains, each 60 amino acids long (Fig. 1). These proteins, together with complement receptor type 2 (CR2) are encoded on the long arm of chromosome 1, in a region referred to as RCA gene cluster(33,34). COMPLEMENT ACTIVATION ON MALIGNANT CELLS Carcinogenesis is a multistep process in which cancer cells evolve from normal cells through the acquisition of several sequential genetic and epigenetic abnormalities. These abnormalities result in essential alterations in cell physiology that dictates malignant growth(35). During the evolution of a cancer, the sequential alterations produce changes in cell morphology and result in the expression of many neoantigens. For example, a change in glycosylation phenotype is considered a hallmark of cancer cells(36). Warren et al. found that cell surface carbohydrates of tumor cells showed an altered structure when parent cells and virustransformed cells were compared(37). In addition, abnormalities can also be found in the expression and structure of membranebound proteins. Hundreds of genes that are either inactive in the normal tissue of origin or expressed at relatively low levels are activated in cancer. These elements that R. PIO distinguish cancer cells from their normal counterparts may well be recognized by the immune system(38). This is the basis of the immune surveillance hypothesis, which proposes that the immune system surveys the body for tumor-associated antigens, eliminating many or most tumors(39). A corollary to this hypothesis is that tumor cells in progressive cancers develop active mechanisms to escape immune recognition or resist immune attack. Although there is not irrefutable evidence for the existence of an effective immune surveillance, a wealth of published data support the role of the immune system as a primary defense against neoplasia and the importance of the protective mechanisms developed by the tumors(39,40). In this sense, clinical studies have suggested that the complement immune system is activated in patients with cancer in response to the expression of tumor associated antigens(41-43). In the sera of patients with neoplastic diseases and in the membrane of the malignant cells an elevation in complement activity or in levels of complement components has been observed in many studies. Patients with ovarian cancer showed elevated levels of C3a and soluble C5b-9 in the intraperitoneal ascitic fluid, suggesting an activation of complement on the ovarian cancer cells(44). Niculescu et al. reported that C5b-9 deposits may be seen on cell membranes of primary breast cancers(45). Yamakawa et al. detected deposition of C3 and C5b-9 in neoplastic thyroid tissues, which was confirmed later by the report of elevated C3d, C4d, and C5 deposits on papillary thyroid carcinomas(46). MBL complement activation pathway was significantly increased in patients with colorectal cancer compared with healthy persons(47), and MASP-2 concentration in serum proved to be an independent prognostic marker with high MASP-2 levels predicting recurrence and poor survival(48). Deposition of C3 and C5b-9 was sporadically found on the tumor cells and the surrounding stroma(49). In lung tumors, immunohistochemical analysis revealed a minimal deposition of C3b with an apparently lack of activation of the lytic MAC(50). Elevated complement levels correlating with tumor size were found in lung cancer patients(51). Complement components C3c and C4 were significantly elevated in almost all 96 patients with lung cancer when compared with the levels in a control group(52). High complement hemolytic activity and C3 levels were observed in serum from children with neuroblastoma(53). Patients with carcinomas of the digestive tract and with gliomas showed also increased serum complement activity(54,55). In vivo alterations in the classical pathway activation were described in patients with chronic lymphatic leukemia (CLL)(56,57). Besides, a strong positive correlation was found between the length of survival of the patients with CLL and the initial activity of the classical pathway of complement(58). In contrast to these results, 175 Inmunol3/2006 copia 13/12/06 16:38 Página 176 CONTROL OF COMPLEMENT ACTIVATION BY CANCER CELLS AND ITS IMPLICATIONS IN ANTIBODY-MEDIATED ... VOL. 25 NUM. 3/ 2006 COMPLEMENT REGULATORY PROTEINS IN CANCER Figure 2. Protecting mechanisms for complement resistance of cancer cells. Extracellular protectors limit the activation of complement proteins on the surface of the target cells, mainly controlling C3 deposition and the assembling of the membrane attack complex (MAC). These regulators include mCRPs, soluble complement regulators such as factor H, ecto-proteases that can cleave complement components deposited on the cell membrane, sialic acid residues that can limit C3 deposition, and ecto-protein kinases capable of phosphorylating several substrates essential for the formation of the MAC. There are also important intracellular mechanisms that can induce the elimination of the MAC by exo- and endocytosis. The present review is focused on mCRPs and soluble regulators. A review of other significant complement resistance mechanisms can be found in Jurianz et al.(60). patients with advanced metastatic brain tumors had reduced complement titers(55). Mangano et al. reported normal total complement and C3 levels in patients with breast, gastric and colorectal carcinomas, whereas levels in those patients who displayed metastases fell below the normal range(59). Although most in vivo observations support that many cancers activate the autologous complement system, it is also well-known that the efficiency of complement-mediated tumor cytotoxicity is hampered by various protective mechanisms(60). Figure 2 summarizes the mechanisms used by tumor cells to resist CDC. Many of these resistance mechanisms are also used by normal cells to avoid accidental activation or bystander effects due to local activation of complement. However, it is also reasonable to assume that to prevent injury by activated complement, cancer cells develop additional mechanisms to inhibit complement activation. In vitro studies have shown that lung cancer cell lines are extremely resistant to CDC, and this resistance is much higher than that observed in normal cells such as human nasal epithelium primary cell cultures(61,62). One of the best understood protective mechanism used by cancer cells is the over-expression of membrane-associated and/or soluble complement regulatory proteins. Many current hypotheses propose that expression of these proteins on the neoplastic cell membrane protects tumors from complement activation. 176 Expression and role of mCRPs in tumors Several studies have analyzed the expression of complement regulators in primary tumors and cancer cell lines. mCRPs, which serve as important regulators for complement-mediated self-injury, are overexpressed on many tumor cell types. This topic has been extensively reviewed(63,64). With the exception of CR1, most cancers express at least two if not three mCRPs. As noted by Fishelson et al., it is surprising the large variation in mCRP expression that can be observed among tumor types and even among different specimens of the same tumor type. In an illustrative study, the authors analyzed sixteen metastatic melanoma lesions and observed that nine expressed both CD46 and CD59, two had CD59 only, one had CD46 only, and four had neither CD46 nor CD59(65). The obvious consequence for a high expression of one or more mCRPs by a tumor cell would be a higher resistance to complement activation. In melanoma cell lines, levels of cell membrane CD59 were found to regulate their sensitivity to homologous CDC(66). An inverse correlation was found between CD59 expression and the extent of CDC when the cells were sensitized with an anti-GD3 ganglioside monoclonal antibody (mAb). Treatment of the cells with a neutralizing antibody against CD59 or a phosphatidylinositol-specific phospholipase C to remove CD59 from the cell membrane enhanced CDC(66). In prostate tissue, no differences in CD46 expression were observed between normal prostate epithelial cells and malignant cells. However, expression of CD59 showed a slight increase in the metastatic cases(67). Xu et al. previously reported high CD59 levels in prostate tumors correlating with disease progression, as measured by tumor stage, Gleason grade or prostate-specific antigen levels(68). Jarvis et al. demonstrated an upregulation of CD59 in metastatic PC-3 and DU145 prostate cancer cell lines, which after inhibition of CD59 with a neutralizing antibody showed higher sensitivity to CDC(69). In a series of colorectal cancer patients, immunohistochemical expression of CD59 was identified as a marker of poor prognosis(70). Blocking of CD59 with anti-CD59 mAbs on colonic, prostate, breast and ovarian carcinoma cells led to a dependent increase in CDC(71,72). On the other hand, an overexpression of rat CD59 in human neuroblastoma cells conferred resistance to killing by rat complement in vitro and increased the capacity of tumor growth in vivo(73). Overexpression of CD59 in melanoma cells also increased the resistance of these cells to CDC(74). With regard to the role of CD55 in complement activation, Loberg et al. reported an increased expression of CD55 and Inmunol3/2006 copia 13/12/06 16:38 Página 177 INMUNOLOGÍA its receptor CD97 in prostatic tumors compared to normal tissue. Additionally, CD97 expression was associated with the transition to the malignant phenotype(67). Inhibition of CD55 in prostate tumor PC-3 cells using siRNA-mediated knockdown expression resulted in a significant decrease in overall tumor burden in a mouse model of metastasis(75). Expression of CD55 and CD97 was also higher in medullary thyroid carcinomas(76). Interestingly, CD97 mRNA expression directly correlated with the histopathological stage of tumors, showing higher levels in advanced stages. In colorectal carcinoma, CD55 was seen as a marker for poor prognosis(77). In contrast, Madjd et al. reported that loss of CD55 was related to poor prognosis in breast cancer(78). High expression of CD55 was significantly associated with low-grade lymph node negativity and with good prognosis. Survival analysis showed that CD55 overexpression was associated with a more favorable outcome. Deficiency of the homologue of human CD55 in mice significantly enhanced T cell responses to active immunization(79), suggesting that over-expression of CD55 in tumors may modulate tumor immunity mediated by the adaptive response. CD46 is found in most tumors and is perhaps the mCRP with the least level of variation between tumors and normal tissue. Nevertheless, it has been reported that breast tumors expressed high levels of CD46 that correlated with tumor grade and recurrence(80). Higher levels of CD46 in breast cancer were associated with estrogen receptor-positive samples, and lower levels with a loss of differentiation and epidermal growth factor receptor positivity(81). Soluble forms of the mCRPs have also been detected in cancer patients; although it has to be pointed out that these forms are not restricted to malignant diseases and can also be found under normal conditions(82,83). Sera from cancer patients contained soluble forms of CD46(84,85). The stroma of breast, colorectal, lung, renal, ovarian, gastric and cervical carcinomas also contained mCRPs(49,50,86-88). Cell lines from different cancers are also known to release soluble forms of the mCRPs(44,72,86,87,89-92). These forms are able to bind to the tumor cell, and should be considered in the resistance of tumor cells to complement activation. In colorectal cancer, soluble CD55 was present in stool specimens and was proposed as a diagnostic marker of poor prognosis(93,94). Soluble forms of C1 inhibitor, CD59 and CD46, and the soluble inhibitor factor H, were present at higher concentrations in intraperitoneal ascitic fluid from ovarian cancer patients than in serum samples. The presence of these regulators, together with the expression of mCRPs may explain why ovarian malignant cells had surface deposits of C1q and C3 activation products, but not of C5b-9(44). R. PIO Expression and role of soluble complement regulators in tumors Less attention has been paid to the expression and contribution of fluid-phase regulators in the control of complement activation on cancer cells. However, it has been suggested that some of these regulators are also important in the resistance of tumor cells to complement activation and CDC. This is the case for the soluble regulator factor H(44,95-98). Factor H is a 150 kDa glycoprotein present in human plasma which inhibits the formation and activity of the C3 convertase(22,23,99). Besides, alternative splicing of factor H mRNA yields a 42 kDa protein, named factor H-like protein 1 (FHL-1), which shares the complement inhibitory activities of factor H (100,101). Expression of factor H and/or FHL-1 has been described in primary tumors and cell lines from different origins (97,102-107). In fact, a clinically approved immunoassay for the detection of bladder cancer in urine is based in the quantification of factor H(106,108). Obviously, factor H expression may have consequences in the resistance of cancer cells against complement. H2 glioblastoma cells were exceptionally resistant to CDC but, although these cells strongly expressed CD59, CD46 and CD55, a combined neutralization of these molecules did not increase their sensitivity to complement killing(97,109). It was later demonstrated that H2 cells produced factor H and FHL-1 and were able to bind them, promoting C3b cleavage. Anti-factor H mAbs enhanced cell death, confirming that factor H (or FHL-1) was involved in the complement resistance of this cancer cell line(97). In SK-MEL-93-2, a human melanoma cell line, factor H was the dominant factor regulating the inactivation of cell-bound C3b and was involved in the control of the classical pathway of complement(96). An anti-factor H antibody also enhanced complement-mediated killing of Raji cells, a cell line obtained from a Burkitt's lymphoma(110). Members of the SIBLING family protected murine erythroleukemia, and human myeloma and breast cancer cells from complement attack, likely by sequestration of factor H to the cell surface(111,112). Factor H and FHL-1 were highly expressed by ovarian carcinomas and both proteins were abundantly present in ascites from these tumors(107). Varsano et al. showed that lung cancer cells were extremely resistant to CDC in vitro, and this resistance was much higher than that observed in normal cells such as human nasal epithelium primary cell cultures(61,62). In their studies, they also showed that neutralizing antibodies against CD46 and CD59 were not effective in increasing the susceptibility to CDC(62), whereas the same antibodies were very effective in facilitating CDC of normal respiratory epithelial cells(61). These findings suggested that lung cancer cells have other mechanisms to resist complement activation in addition to CD46 and CD59. Our group has 177 Inmunol3/2006 copia 13/12/06 16:38 Página 178 CONTROL OF COMPLEMENT ACTIVATION BY CANCER CELLS AND ITS IMPLICATIONS IN ANTIBODY-MEDIATED ... VOL. 25 NUM. 3/ 2006 TABLE I. Unconjugated therapeutic monoclonal antibodies approved for use in Oncology Name (Tradename) Company Isotype Target Cancer indication* Rituximab (Rituxan) Roche and Genentech Chimeric IgG1 CD20 NHL Trastuzumab (Herceptin) Roche and Genentech Humanized IgG1 HER2/neu Breast Merk Chimeric IgG1 EGFR Colorectal Roche and Genentech Chimeric IgG1 VEGF Colorectal and lung Millenium and Ilex Humanized IgG1 CD52 CLL Cetuximab (Erbitux) Bevacizumab (Avastin) Alemtuzumab (Campath-1H) *NHL, non-Hodgkin´s lymphoma. CLL, Chronic lymphocytic leukemia. recently demonstrated that factor H was frequently expressed in non-small cell lung cancer (NSCLC). Factor H was also secreted to the extracellular milieu and was able to bind to lung tumor cell surfaces, inhibiting the activation of complement(98). Lung cancer is not the only cancer type in which blocking of CD46 and CD55 does not make cancer cells sensitive to complement(72,113). In these cases, it would be interesting to determine whether soluble complement regulators, such as factor H or others, secreted by the cancer cells, can control complement activation at the level of C3 convertases. C4bp, the functional analogue of factor H in the classical pathway, may be also a good candidate to regulate complement activation on cancer cells at this level. In cancer patients with various non-metastatic solid tumors, the C4BP mean plasma levels were significantly higher than those in the control group(114). C4bp was able to bind to SKOV-3, SW626 and Caov-3 ovarian adenocarcinoma cell lines, which may lead to an increased control of the classical pathway activation(115). In relation to the other soluble complement regulatory proteins, some reports have described the expression of C1 inhibitor by some primary tumors and cancer cells(44,92,105,116,117), although this is not always the case(118). Clusterin expression is also upregulated in cancer progression and tumor formation. This protein is expressed in a wide variety of tissues and it is implicated in many diverse physiological processes, such as sperm maturation, lipid transportation, tissue remodeling, membrane recycling, cell–cell and cell–substratum interactions, stabilization of stressed proteins in a foldingcompetent state, promotion or inhibition of apoptosis, and complement inhibition. Trougakos et al. have reviewed the properties of clusterin and its implication in cancer(119). The wide distribution of this protein, its diverse physiological effects and the lack of specific studies on the implication of clusterin in the resistance to complement activation in cancer cells, allows only speculation on the importance of its presence in the control of complement activation in tumors. 178 THE ROLE OF COMPLEMENT IN CANCER IMMUNOTHERAPY Tumor-specific and tumor associated-antigens can be used as targets for mAb immunotherapy. An increasing number of mAbs are currently used as therapeutic drugs to control tumor growth (Table I), and many others are currently being tested in preclinical and clinical trials(120,121). mAbs normally utilize a combination of mechanisms in directing cytotoxic effects to a tumor cell. Some of these antibodies act by blocking important cancer activities, but many of them are also able to activate the immune system through ADCC or CDC(122). Chimerized and humanized mouse mAbs containing the human IgG1 Fc-region are examples of complement activating mAbs(123). The Fc-regions of membrane-bound antibodies interact with the heterooligomeric complex C1q and activate the classical pathway. Successful complement activation may have multiple consequences on the immune response against tumors. Complement activation leads to formation of the MAC, fosters opsonization, and releases powerful proinflammatory anaphylatoxins. In addition, it has been proposed that the effect of complement can synergize with other antibody-mediated mechanisms of action important for the efficacy of these antibodies. The release of the anaphylatoxins results in the recruitment of effector cells, such as natural killer cells, into the tumor. CR3 expressed by phagocytes and natural killer cells can be manipulated in such a way that it will trigger CR3-dependent cellular cytotoxicity or enhance ADCC of iC3b-coated tumor cells in the present of the yeast cell-wall β-glucan(123). In a syngeneic mouse model of metastatic lymphoma, a mAb directed against the ganglioside GD2 expressed in EL4 lymphoma cells caused both CDC and complement-dependent enhancement of ADCC(124). Rituximab, an anti-CD20 mAb, is used for the treatment of malignant lymphoma(125). However, in some cases with bulky mass and at stage IV, lymphoma cells become resistant to rituximab treatment(126). How this resistance occurs has not yet been clarified. Rituximab may exert its activity Inmunol3/2006 copia 13/12/06 16:38 Página 179 INMUNOLOGÍA through different mechanisms: inhibition of proliferation, induction of apoptosis, CDC, and ADCC(126). Both in vitro and in vivo data suggest that CDC may be one of the most important mechanisms of action. Rituximab was shown to cure immunocompetent mice challenge with murine lymphoma EL4 cells stably transfected with human CD20. However, its efficacy was completely eliminated when the same study was done in syngeneic knockout mice lacking C1q(127). The efficacy of rituximab, as for any other cancer treatment with mAbs, may be limited by the expression of complement regulatory proteins by the target cells. No differences in the expression of CD59 have been reported between normal and malignant B cells, whereas CD55 expression was shown to be different among individual patients with B-cell malignancy(128). Clinical responses to rituximab could not be predicted by the level of CD55 or CD59 in patients with chronic lymphocytic leukemia (CLL) or B-cell non-Hodgkin lymphoma(129,130). However, in vitro data suggest that there may exist an inverse correlation between the expression of these regulators and the susceptibility to CDC in B-cell lymphoma cell lines(131,132). A decrease in susceptibility to CDC with rituximab was dependent on CD55 expression and CD55 expression correlated with tumor size in lymphoma cells from patients with non-Hodgkin lymphoma(133). Knockdown of CD55 by siRNA overcame resistance to CDC in fresh lymphoma cells treated in vitro with rituximab(133). A similar result from the same authors was obtained with siRNA of CD55 in SK-BR3 breast cancer cell line treated with trastuzumab, a clinically-approved mAb against the Her2/neu receptor(133). These data confirmed previous studies with two other breast cancer cell lines, in which the blocking of CD59 and CD55 reduced the resistance of these cells to CDC after treatment with trastuzumab. ENHANCING THE EFFICACY OF ANTIBODY-BASED IMMUNOTHERAPY BY MODULATING COMPLEMENT ACTIVATION Clinical efficacy of mAb immunotherapy may be enhanced by overcoming protection with complement inhibitors and/or by improving complement-mediated effector mechanisms. Table II summarizes some of the strategies that have been proposed for that purpose. The effectiveness of membrane regulators in protecting tissues from complement injury has provided an impetus to explore the therapeutic application of blocking mCRPs to improve antitumor therapy with mAbs. Several strategies have been developed and tested experimentally in vitro and in animal models. Theoretically, the protective capacity of mCRPs can be overcome by different ways(64): blockade of the R. PIO TABLE II. Strategies to enhance the efficacy of mAb-based immunotherapy by modulating complement activation To overcome protection capacity mediated by complement regulators • Blocking the complement regulator activities • Down-regulating the expression of the complement regulators • Removing the complement regulators from the cell surfaces To improve complement-mediated effector mechanisms • Modifying mAb by isotype switching, genetic engineering or conjugation • Using mAbs as a mixture rather than as individual ones • Selecting antigens with a strong capacity to activate complement regulator activity; down-regulation of mCRP expression; or removal of the regulator from the cell surface. Numerous papers, as mentioned above and reviewed previously(64), have demonstrated the efficacy of neutralizing antibodies to block mCRPs and to improve the complement-mediated activity of mAbs able to recognize cancer-associated antigens and fix complement. However, the challenging problem is the targeting of mCRP inhibitors selectively to malignant cells, avoiding thei effects on normal tissue. Macor et al. have proposed the use of a biotin-avidin system developed by Paganelli et al.(134) to target selectively neutralizing antibodies raised against complement regulators(135). The use of therapeutic bispecific mAbs that target a tumor antigen and simultaneously block a major complement regulatory protein has also been suggested(136,137). These antibodies recognize tumor antigens and mCRPs such as CD59 or CD55(138-140). The efficacy of this strategy has been demonstrated in a syngeneic lung metastasis model of rat colorectal cancer(141). An alternative strategy to abolish the control by mCRPs is to reduce its expression on cancer cells. Some cytokines may induce downregulation of CD55 or CD59 and thus affect the effectiveness of immunotherapy with mAbs. Blok et al. investigated the effect of ten cytokines on the expression of CD46, CD55 and CD59 in human renal tumor cell lines and proximal tubular epithelial cells(142). Three out of ten cytokines tested (IL-1β, IL-4 and TGF-β1) induced consistent downregulation of mCRP expression on all tumor cell lines studied. IL-1β downregulated the expression of CD46 and CD59, IL-4 the expression of CD46, and TGF-β1 the expression of both CD46 and CD55. Besides, changes in the expression of CD55 and CD59 were associated with changes in the amount of C3 deposited and the extent of CDC, respectively(142). However, most cytokines either enhance or have no effect on mCRP expression(91,143-149). Besides, the effect of the cytokines on mCRP expression varies, even when the 179 Inmunol3/2006 copia 13/12/06 16:38 Página 180 CONTROL OF COMPLEMENT ACTIVATION BY CANCER CELLS AND ITS IMPLICATIONS IN ANTIBODY-MEDIATED ... cell lines studied are derived from the same tumor type, making the in vivo effect highly unpredictable. Another option would be the use of chemotherapeutic agents or other compounds capable to modulate the level of expression of mCRPs(150-152), although to date highly effective reagents for this purpose are still needed. In vitro studies have tested the possibility to remove CD55 and CD59 molecules from the membrane with GPIphospholipase C. Lysis of melanoma, lung carcinoma and cervical carcinoma cells increased following treatment with this enzyme(49,62,66). GPI-phospholipase C treatment increased sensitivity of CD20-positive IM9 cells to rituximab-mediated CDC(153). However, it is important to note that the treatment is not specific for mCRPs and would remove other surface molecules, complicating the interpretation of these studies. mAb-mediated immunotherapy might be also enhanced by improving complement-mediated effector mechanisms. A way to boost complement activation is the modification of the therapeutic antibodies by isotype switching, genetic engineering or conjugation. Heteroconjugates composed of antitumor antibodies and molecules such as cobra venom factor (CVF), C3b or iC3b have been used. CVF activates the complement system by forming stable C3/C5 convertases in mammalian serum(154). Combination of CVF with antibodies or F(ab)2 fragments directed against different tumor-associated antigens had an additive effect on CDC(155,156). In an orthotopic pancreatic cancer model using nude rats, CVF conjugated to a mAb with tumor-binding properties was able to increase in vivo tumor infiltration by natural killer cells and macrophages(157). The complement-activating capacity of the 17-1A mAb was enlarged by conjugating it to CVF or C3b(136). Other authors have also used antibodies conjugated to C3b or iC3b to increase lysis of tumor cells(95,158). Kennedy et al. increased complement activity by the combined used of ribuximab and the mAb 3E7, specific for iC3b. 3E7 enhanced iC3b deposition in CD20-positive Raji and ARH-77 cells treated with rituximab. Consequently, the potential of rituximab and IF5 (another anti-CD20 mAb) to facilitate CDC was substantially enhanced in the presence of 3E7(159). Bispecific antibodies may also be engineered to recruit complement effector functions(160). Finally, to promote cooperation between CDC and ADCC, Gelderman et al. proposed the use of β-glucan to activate CR3 on effector cells and induce CR3-dependent cellular cytotoxicity(123). A low initiation of CDC by mAbs may be also related to a low density of antigenic epitopes on the target cell membrane. This reduces the chance for the IgG to form dimers required for complement activation. Some data in the literature support this concern. Sensitivity to ribuximabmediated CDC in leukemia cells freshly isolated from patients 180 VOL. 25 NUM. 3/ 2006 correlated with the expression levels of CD20 (161). The acquisition of resistance to rituximab was associated to a decreased CD20 expression(162). The antitumor activities of several murine mAbs to HER-2 overexpressed on tumor cells were more effective in activating complement as a mixture rather than as individual antibodies(163). Chimeric mAbs cMOV18 and cMOV19 against two distinct epitopes of the folate receptor were only effective in combination, and neutralization of CD46 and CD59 markedly enhanced the susceptibility of tumor cells to CDC of ovarian cancer cells(135). The selective alteration of the glycosylation pattern can also enhance the lytic potential of humanized IgG1 mAbs without affecting the affinity and specificity of the antibody. Glycosylation variants of the therapeutic Lewis Y-specific humanized IgG1 antibody IGN311 were able to lyse cells more potently than the parent antibody, which may overcome problems with low density of antigenic epitopes and lower the minimally effective clinical dose(164). The molecular architecture of the antigens selected for immunotherapy seems also essential for the proper induction of CDC(165,166). CONCLUSIONS AND PERSPECTIVES Given the numerous genetic and epigenetic changes associated with carcinogenesis, it is clear that tumor cells express many neoantigens that may be recognized by the immune system. However, cancer cells can develop mechanisms to avoid immune recognition or activation(38). The elucidation of these mechanisms may provide ways to improve cancer immunotherapy. Although there are many reports that describe abnormalities in complement levels in patients with cancer, the role of complement in immune surveillance against cancer has not been unequivocally demonstrated. The complement system of patients may be activated indirectly by immune complexes, concomitant infections, or substances generated inside the tumor mass(64). We still need to characterize more potential tumor associated antigens able to stimulate complement, and to better understand the intricate mechanisms of tolerance. We will need to explore biologically relevant animal models of cancer with and without complement deficiencies to investigate the contribution of complement in cancer immunosurveillance and the role of complement regulators in the complement-mediated elimination of tumor cells. Since most complement regulators operate in a species specific manner, syngeneic animal models are probably the best option for this kind of studies. Whether or not one accepts that cancer is normally suppressed by continuous immunosurveillance, experimental and clinical evidences clearly support the idea that the Inmunol3/2006 copia 13/12/06 16:38 Página 181 INMUNOLOGÍA immune system can be mobilized to attack malignant cells that express tumor-associated antigens. Many in vitro and in vivo studies have shown that blocking the function of regulatory proteins sensitizes resistant tumor cells to complement. These results have led to the hypothesis that inhibiting the function of complement regulatory proteins expressed on tumor cells will enhance immunotherapy. At least in animal models, this has proved to be true(167,168). However, complement regulatory proteins are widely expressed on normal cells, and targeting inhibitory molecules to complement regulators expressed and/or bound to tumor cells in vivo is still a technical challenge. Besides, additional strategies used by tumor cells to prevent complement activation have received much less attention than the expression of mCRPs, even though their role in complement resistance of tumor cells is probably also important. Each tumor may be equipped with different mechanisms of cell protection from complement attack. It is conceivable that a concerted action against different protective mechanisms will be required to achieve efficient antibody- and complementmediated cancer immunotherapy(72). ACKNOWLEDGEMENTS The author thanks Dr Santiago Rodriguez de Cordoba for helping with figure 1 and for critical reading of the manuscript. Work in the author’s laboratory has been funded by «UTE project CIMA», 2004-2006 AACR-Cancer Research and Prevention Foundation Career Development Award in Translational Lung Cancer Research, Instituto de Salud Carlos III: Red de Centros de Cáncer RTICCC (C03/10), and Ministerio de Educación y Ciencia (SAF2005-01302). DISCLOSURES The author has no financial conflict of interest. CORRESPONDENCE TO: Ruben Pio Division of Oncology CIMA Building Pio XII, 55 31008 Pamplona (Spain) Email: [email protected]. REFERENCES 1. Liszewski MK, Atkinson JP. The complement system. In: Paul WE (ed). Fundamental Immunology. Third edition. New York: Raven Press; 1993. p. 917-939. R. PIO 2. Dempsey PW, Allison ME, Akkaraju S, Goodnow CC, Fearon DT. C3d of complement as a molecular adjuvant: bridging innate and acquired immunity. Science 1996;271:348-350. 3. Morgan BP, Marchbank KJ, Longhi MP, Harris CL, Gallimore AM. Complement: central to innate immunity and bridging to adaptive responses. Immunol Lett 2005;97: 171-179. 4. Carroll MC. The complement system in regulation of adaptive immunity. Nat Immunol 2004;5: 981-986. 5. Molina H, Holers VM, Li B, Fung Y, Mariathasan S, Goellner J, et al. Markedly impaired humoral immune response in mice deficient in complement receptors 1 and 2. Proc Natl Acad Sci USA 1996;93: 3357-3361. 6. Kaya Z, Afanasyeva M, Wang Y, Dohmen KM, Schlichting J, Tretter T, et al. Contribution of the innate immune system to autoimmune myocarditis: a role for complement. Nat Immunol 2001;2:739-745. 7. Fleming SD, Shea-Donohue T, Guthridge JM, Kulik L, Waldschmidt TJ, Gipson MG, et al. Mice deficient in complement receptors 1 and 2 lack a tissue injury-inducing subset of the natural antibody repertoire. J Immunol 2002;169:2126-2133. 8. Prodeus AP, Goerg S, Shen LM, Pozdnyakova OO, Chu L, Alicot EM, et al. A critical role for complement in maintenance of selftolerance. Immunity 1998;9:721-731. 9. Carroll MC. The role of complement in B cell activation and tolerance. Adv Immunol 2000;74:61-88. 10. Kishore U, Reid KB. C1q: structure, function, and receptors. Immunopharmacology 2000;49:159-170. 11. Arlaud GJ, Gaboriaud C, Thielens NM, Rossi V. Structural biology of C1. Biochem Soc Trans 2002;30:1001-1006. 12. Sunyer JO, Lambris JD. Evolution and diversity of the complement system of poikilothermic vertebrates. Immunol Rev 1998;166:3957. 13. Pangburn MK, Schreiber RD, Muller-Eberhard HJ. Formation of the initial C3 convertase of the alternative complement pathway. Acquisition of C3b-like activities by spontaneous hydrolysis of the putative thioester in native C3. J Exp Med 1981;154:856-867. 14. Lu JH, Thiel S, Wiedemann H, Timpl R, Reid KB. Binding of the pentamer/hexamer forms of mannan-binding protein to zymosan activates the proenzyme C1r2C1s2 complex, of the classical pathway of complement, without involvement of C1q. J Immunol 1990;144: 2287-2294. 15. Zhang Y, Suankratay C, Zhang XH, Lint TF, Gewurz H. Lysis via the lectin pathway of complement activation: minireview and lectin pathway enhancement of endotoxin-initiated hemolysis. Immunopharmacology 1999;42:81-90. 16. Bara S, Lint TF. The third component of complement (C3) bound to tumor target cells enhances their sensitivity to killing by activated macrophages. J Immunol 1987;138:1303-1309. 17. Ramos OF, Nilsson B, Nilsson K, Eggertsen G, Yefenof E, Klein 181 Inmunol3/2006 copia 13/12/06 16:38 Página 182 CONTROL OF COMPLEMENT ACTIVATION BY CANCER CELLS AND ITS IMPLICATIONS IN ANTIBODY-MEDIATED ... E. Elevated NK-mediated lysis of Raji and Daudi cells carrying fixed iC3b fragments. Cell Immunol 1989;119:459-469. 18. Perlmann H, Perlmann P, Schreiber RD, Muller-Eberhard HJ. Interaction of target cell-bound C3bi and C3d with human lymphocyte receptors. Enhancement of antibody-mediated cellular cytotoxicity. J Exp Med 1981;153:1592-1603. 19. Kohl J. Anaphylatoxins and infectious and non-infectious inflammatory diseases. Mol Immunol 2001;38:175-187. 20. Ziccardi RJ. A new role for C-1-inhibitor in homeostasis: control of activation of the first component of human complement. J Immunol 1982;128:2505-2508. 21. Sim RB, Day AJ, Moffatt BE, Fontaine M. Complement factor I and cofactors in control of complement system convertase enzymes. Methods Enzymol 1993;223:13-35. 22. Weiler JM, Daha MR, Austen KF, Fearon DT. Control of the amplification convertase of complement by the plasma protein beta1H. Proc Natl Acad Sci USA 1976;73:3268-3272. 23. Whaley K, Ruddy S. Modulation of the alternative complement pathways by beta 1 H globulin. J Exp Med 1976;144:1147-1163. 24. Jenne DE, Tschopp J. Molecular structure and functional characterization of a human complement cytolysis inhibitor found in blood and seminal plasma: identity to sulfated glycoprotein 2, a constituent of rat testis fluid. Proc Natl Acad Sci USA 1989;86: 7123-7127. 25. Podack ER, Muller-Eberhard HJ. Isolation of human S-protein, an inhibitor of the membrane attack complex of complement. J Biol Chem 1979;254:9808-9814. 26. Fearon DT. Regulation of the amplification C3 convertase of human complement by an inhibitory protein isolated from human erythrocyte membrane. Proc Natl Acad Sci USA 1979;76:5867-5871. 27. Iida K, Nussenzweig V. Functional properties of membraneassociated complement receptor CR1. J Immunol 1983;130:18761880. 28. Liszewski MK, Post TW, Atkinson JP. Membrane cofactor protein (MCP or CD46): newest member of the regulators of complement activation gene cluster. Annu Rev Immunol 1991;9:431-455. VOL. 25 NUM. 3/ 2006 33. Rodriguez de Cordoba S, Lublin DM, Rubinstein P, Atkinson JP. Human genes for three complement components that regulate the activation of C3 are tightly linked. J Exp Med 1985;161:11891195. 34. Rodriguez de Cordoba S, Diaz-Guillen MA, Heine-Suner D. An integrated map of the human regulator of complement activation (RCA) gene cluster on 1q32. Mol Immunol 1999;36:803-808. 35. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell 2000;100: 57-70. 36. Hakomori S. Tumor malignancy defined by aberrant glycosylation and sphingo(glyco)lipid metabolism. Cancer Res 1996;56:53095318. 37. Warren L, Buck CA, Tuszynski GP. Glycopeptide changes and malignant transformation. A possible role for carbohydrate in malignant behavior. Biochim Biophys Acta 1978;516:97-127. 38. Pardoll D. Does the immune system see tumors as foreign or self? Annu Rev Immunol 2003;21:807-839. 39. Smyth MJ, Godfrey DI, Trapani JA. A fresh look at tumor immunosurveillance and immunotherapy. Nat Immunol 2001;2: 293-299. 40. Jakobisiak M, Lasek W, Golab J. Natural mechanisms protecting against cancer. Immunol Lett 2003;90:103-122. 41. Guidi L, Baroni R, Bartoloni C, Pellegrino M, Tricerri A, Marciano M, et al. Immune complexes in solid tumours precipitable by 3.5% polyethylene glycol: analysis of some nonspecific components. Diagn Clin Immunol 1988;5:284-288. 42. Zurlo JJ, Schechter GP, Fries LF. Complement abnormalities in multiple myeloma. Am J Med 1989;87:411-420. 43. Baatrup G, Qvist N, Junker A, Larsen KE, Zimmermann-Nielsen C. Activity and activation of the complement system in patients being operated on for cancer of the colon. Eur J Surg 1994;160:503510. 44. Bjorge L, Hakulinen J, Vintermyr OK, Jarva H, Jensen TS, Iversen OE, Meri S. Ascitic complement system in ovarian cancer. Br J Cancer 2005;92:895-905. 29. Lublin DM, Atkinson JP. Decay-accelerating factor: biochemistry, molecular biology, and function. Annu Rev Immunol 1989;7:35-58. 45. Niculescu F, Rus HG, Retegan M, Vlaicu R. Persistent complement activation on tumor cells in breast cancer. Am J Pathol 1992;140: 1039-1043. 30. Meri S, Morgan BP, Davies A, Daniels RH, Olavesen MG, Waldmann H, Lachmann PJ. Human protectin (CD59), an 18,000-20,000 MW complement lysis restricting factor, inhibits C5b-8 catalysed insertion of C9 into lipid bilayers. Immunology 1990;71:1-9. 46. Lucas SD, Karlsson-Parra A, Nilsson B, Grimelius L, Akerstrom G, Rastad J, Juhlin C. Tumor-specific deposition of immunoglobulin G and complement in papillary thyroid carcinoma. Hum Pathol 1996;27:1329-1335. 31. Rollins SA, Sims PJ. The complement-inhibitory activity of CD59 resides in its capacity to block incorporation of C9 into membrane C5b-9. J Immunol 1990;144:3478-3483. 47. Ytting H, Jensenius JC, Christensen IJ, Thiel S, Nielsen HJ. Increased activity of the mannan-binding lectin complement activation pathway in patients with colorectal cancer. Scand J Gastroenterol 2004;39:674-679. 32. Yamamoto H, Blaas P, Nicholson-Weller A, Hansch GM. Homologous species restriction of the complement-mediated killing of nucleated cells. Immunology 1990;70:422-426. 182 48. Ytting H, Christensen IJ, Thiel S, Jensenius JC, Nielsen HJ. Serum mannan-binding lectin-associated serine protease 2 levels in Inmunol3/2006 copia 13/12/06 16:38 Página 183 INMUNOLOGÍA colorectal cancer: relation to recurrence and mortality. Clin Cancer Res 2005;11:1441-1446. 49. Gelderman KA, Blok VT, Fleuren GJ, Gorter A. The inhibitory effect of CD46, CD55, and CD59 on complement activation after immunotherapeutic treatment of cervical carcinoma cells with monoclonal antibodies or bispecific monoclonal antibodies. Lab Invest 2002;82:483-493. 50. Niehans GA, Cherwitz DL, Staley NA, Knapp DJ, Dalmasso AP. Human carcinomas variably express the complement inhibitory proteins CD46 (membrane cofactor protein), CD55 (decay-accelerating factor), and CD59 (protectin). Am J Pathol 1996;149:129-142. 51. Nishioka K, Kawamura K, Hirayama T, Kawashima T, Shimada K. The complement system in tumor immunity: significance of elevated levels of complement in tumor bearing hosts. Ann N Y Acad Sci 1976;276:303-315. 52. Gminski J, Mykala-Ciesla J, Machalski M, Drozdz M, Najda J. Immunoglobulins and complement components levels in patients with lung cancer. Rom J Intern Med 1992;30:39-44. 53. Carli M, Bucolo C, Pannunzio MT, Ongaro G, Businaro R, Revoltella R. Fluctuation of serum complement levels in children with neuroblastoma. Cancer 1979;43:2399-2404. 54. Maness PF, Orengo A. Serum complement levels in patients with digestive tract carcinomas and other neoplastic diseases. Oncology 1977;34:87-89. 55. Matsutani M, Suzuki T, Hori T, Terao H, Takakura K, Nishioka K. Cellular immunity and complement levels in hosts with brain tumours. Neurosurg Rev 1984;7:29-35. 56. Fust G, Miszlay Z, Czink E, Varga L, Paloczi K, Szegedi G, Hollan SR. C1 and C4 abnormalities in chronic lymphocytic leukaemia and their significance. Immunol Lett 1987;14:255-259. R. PIO 62. Varsano S, Rashkovsky L, Shapiro H, Ophir D, Mark-Bentankur T. Human lung cancer cell lines express cell membrane complement inhibitory proteins and are extremely resistant to complementmediated lysis; a comparison with normal human respiratory epithelium in vitro, and an insight into mechanism(s) of resistance. Clin Exp Immunol 1998;113:173-182. 63. Gorter A, Meri S. Immune evasion of tumor cells using membranebound complement regulatory proteins. Immunol Today 1999;20: 576-582. 64. Fishelson Z, Donin N, Zell S, Schultz S, Kirschfink M. Obstacles to cancer immunotherapy: expression of membrane complement regulatory proteins (mCRPs) in tumors. Mol Immunol 2003;40:109-123. 65. Weichenthal M, Siemann U, Neuber K, Breitbart EW. Expression of complement regulator proteins in primary and metastatic malignant melanoma. J Cutan Pathol 1999;26:217-221. 66. Brasoveanu LI, Altomonte M, Fonsatti E, Colizzi F, Coral S, Nicotra MR, et al. Levels of cell membrane CD59 regulate the extent of complement-mediated lysis of human melanoma cells. Lab Invest 1996;74:33-42. 67. Loberg RD, Wojno KJ, Day LL, Pienta KJ. Analysis of membranebound complement regulatory proteins in prostate cancer. Urology 2005;66:1321-1326. 68. Xu C, Jung M, Burkhardt M, Stephan C, Schnorr D, Loening S, Jung K, et al. Increased CD59 protein expression predicts a PSA relapse in patients after radical prostatectomy. Prostate 2005;62: 224-232. 69. Jarvis GA, Li J, Hakulinen J, Brady KA, Nordling S, Dahiya R, Meri S. Expression and function of the complement membrane attack complex inhibitor protectin (CD59) in human prostate cancer. Int J Cancer 1997;71:1049-1055. 57. Schlesinger M, Broman I, Lugassy G. The complement system is defective in chronic lymphatic leukemia patients and in their healthy relatives. Leukemia 1996;10:1509-1513. 70. Watson NF, Spendlove I, Madjd Z, McGilvray R, Green AR, Ellis IO, et al. Expression of the stress-related MHC class I chain-related protein MICA is an indicator of good prognosis in colorectal cancer patients. Int J Cancer 2006;118:1445-1452. 58. Varga L, Czink E, Miszlai Z, Paloczi K, Banyai A, Szegedi G, Fust G. Low activity of the classical complement pathway predicts short survival of patients with chronic lymphocytic leukaemia. Clin Exp Immunol 1995;99:112-116. 71. Bjorge L, Vedeler CA, Ulvestad E, Matre R. Expression and function of CD59 on colonic adenocarcinoma cells. Eur J Immunol 1994;24: 1597-1603. 59. Mangano A, Messina L, Birgillito S, Stivala F, Bernardini A. Complement and its fractions (C3-C4) pattern in subjects with neoplasia. J Immunopharmacol 1984;6:147-162. 72. Donin N, Jurianz K, Ziporen L, Schultz S, Kirschfink M, Fishelson Z. Complement resistance of human carcinoma cells depends on membrane regulatory proteins, protein kinases and sialic acid. Clin Exp Immunol 2003;131:254-263. 60. Jurianz K, Ziegler S, Garcia-Schuler H, Kraus S, Bohana-Kashtan O, Fishelson Z, Kirschfink M. Complement resistance of tumor cells: basal and induced mechanisms. Mol Immunol 1999;36:929939. 73. Chen S, Caragine T, Cheung NK, Tomlinson S. CD59 expressed on a tumor cell surface modulates decay-accelerating factor expression and enhances tumor growth in a rat model of human neuroblastoma. Cancer Res 2000;60:3013-3018. 61. Varsano S, Frolkis I, Rashkovsky L, Ophir D, Fishelson Z. Protection of human nasal respiratory epithelium from complement-mediated lysis by cell-membrane regulators of complement activation. Am J Respir Cell Mol Biol 1996;15:731-737. 74. Coral S, Fonsatti E, Sigalotti L, De Nardo C, Visintin A, Nardi G, et al. Overexpression of protectin (CD59) down-modulates the susceptibility of human melanoma cells to homologous complement. J Cell Physiol 2000;185:317-323. 183 Inmunol3/2006 copia 13/12/06 16:38 Página 184 CONTROL OF COMPLEMENT ACTIVATION BY CANCER CELLS AND ITS IMPLICATIONS IN ANTIBODY-MEDIATED ... 75. Loberg RD, Day LL, Dunn R, Kalikin LM, Pienta KJ. Inhibition of Decay-Accelerating Factor (CD55) Attenuates Prostate Cancer Growth and Survival In Vivo. Neoplasia 2006;8:69-78. 76. Mustafa T, Klonisch T, Hombach-Klonisch S, Kehlen A, Schmutzler C, Koehrle J, et al. Expression of CD97 and CD55 in human medullary thyroid carcinomas. Int J Oncol 2004;24:285-294. 77. Durrant LG, Chapman MA, Buckley DJ, Spendlove I, Robins RA. Armitage NC. Enhanced expression of the complement regulatory protein CD55 predicts a poor prognosis in colorectal cancer patients. Cancer Immunol Immunother 2003;52:638-642. 78. Madjd Z, Durrant LG, Bradley R, Spendlove I, Ellis IO, Pinder SE. Loss of CD55 is associated with aggressive breast tumors. Clin Cancer Res 2004;10:2797-2803. 79. Liu J, Miwa T, Hilliard B, Chen Y, Lambris JD, Wells AD, Song WC. The complement inhibitory protein DAF (CD55) suppresses T cell immunity in vivo. J Exp Med 2005;201:567-577. VOL. 25 NUM. 3/ 2006 metalloproteinase to a functionally active soluble form. Eur J Immunol 2004;34:2620-2629. 89. Brasoveanu LI, Fonsatti E, Visintin A, Pavlovic M, Cattarossi I, Colizzi F, et al. Melanoma cells constitutively release an anchorpositive soluble form of protectin (sCD59) that retains functional activities in homologous complement-mediated cytotoxicity. J Clin Invest 1997;100:1248-1255. 90. Hindmarsh EJ, Marks RM. Decay-accelerating factor is a component of subendothelial extracellular matrix in vitro, and is augmented by activation of endothelial protein kinase C. Eur J Immunol 1998;28:1052-1062. 91. Nasu J, Mizuno M, Uesu T, Takeuchi K, Inaba T, Ohya S, et al. Cytokine-stimulated release of decay-accelerating factor (DAF;CD55) from HT-29 human intestinal epithelial cells. Clin Exp Immunol 1998;113:379-385. 80. Madjd Z, Durrant LG, Pinder SE, Ellis IO, Ronan J, Lewis S, et al. Do poor-prognosis breast tumours express membrane cofactor proteins (CD46)? Cancer Immunol Immunother 2005;54:149-156. 92. Jurianz K, Ziegler S, Donin N, Reiter Y, Fishelson Z, Kirschfink M. K562 erythroleukemic cells are equipped with multiple mechanisms of resistance to lysis by complement. Int J Cancer 2001;93:848-854. 81. Rushmere NK, Knowlden JM, Gee JM, Harper ME, Robertson JF, Morgan BP, Nicholson RI. Analysis of the level of mRNA expression of the membrane regulators of complement, CD59, CD55 and CD46, in breast cancer. Int J Cancer 2004;108:930-936. 93. Kawada M, Mizuno M, Nasu J, Uesu T, Okazaki H, Okada H, et al. Release of decay-accelerating factor into stools of patients with colorectal cancer by means of cleavage at the site of glycosylphosphatidylinositol anchor. J Lab Clin Med 2003;142: 306-312. 82. Medof ME, Walter EI, Rutgers JL, Knowles DM, Nussenzweig V. Identification of the complement decay-accelerating factor (DAF) on epithelium and glandular cells and in body fluids. J Exp Med 1987;165:848-864. 94. Kohno H, Mizuno M, Nasu J, Makidono C, Hiraoka S, Inaba T, et al. Stool decay-accelerating factor as a marker for monitoring the disease activity during leukocyte apheresis therapy in patients with refractory ulcerative colitis. J Gastroenterol Hepatol 2005;20: 73-78. 83. Davies A, Simmons DL, Hale G, Harrison RA, Tighe H, Lachmann PJ, Waldmann H. CD59, an LY-6-like protein expressed in human lymphoid cells, regulates the action of the complement membrane attack complex on homologous cells. J Exp Med 1989;170:637-654. 84. Seya T, Hara T, Iwata K, Kuriyama S, Hasegawa T, Nagase Y, Miyagawa S, et al. Purification and functional properties of soluble forms of membrane cofactor protein (CD46) of complement: identification of forms increased in cancer patients' sera. Int Immunol 1995;7:727-736. 85. Sadallah S, Lach E, Schwarz S, Gratwohl A, Spertini O, Schifferli JA. Soluble complement receptor 1 is increased in patients with leukemia and after administration of granulocyte colony-stimulating factor. J Leukoc Biol 1999;65:94-101. 86. Li L, Spendlove I, Morgan J, Durrant LG. CD55 is over-expressed in the tumour environment. Br J Cancer 2001;84:80-86. 87. Morgan J, Spendlove I, Durrant LG. The role of CD55 in protecting the tumour environment from complement attack. Tissue Antigens 2002;60:213-223. 88. Hakulinen J, Junnikkala S, Sorsa T, Meri S. Complement inhibitor membrane cofactor protein (MCP; CD46) is constitutively shed from cancer cell membranes in vesicles and converted by a 184 95. Reiter Y, Fishelson Z. Targeting of complement to tumor cells by heteroconjugates composed of antibodies and of the complement component C3b. J Immunol 1989;142:2771-2777. 96. Ollert MW, David K, Bredehorst R, Vogel CW. Classical complement pathway activation on nucleated cells. Role of factor H in the control of deposited C3b. J Immunol 1995;155:4955-4962. 97. Junnikkala S, Jokiranta TS, Friese MA, Jarva H, Zipfel PF, Meri S. Exceptional resistance of human H2 glioblastoma cells to complementmediated killing by expression and utilization of factor H and factor H-like protein 1. J Immunol 2000;164:6075-6081. 98. Ajona D, Castano Z, Garayoa M, Zudaire E, Pajares MJ, Martinez A, et al. Expression of complement factor H by lung cancer cells: effects on the activation of the alternative pathway of complement. Cancer Res 2004;64:6310-6308. 99. Pangburn MK, Schreiber RD, Muller-Eberhard HJ. Human complement C3b inactivator: isolation, characterization, and demonstration of an absolute requirement for the serum protein beta1H for cleavage of C3b and C4b in solution. J Exp Med 1977;146: 257-270. 100.Zipfel PF, Skerka C. FHL-1/reconectin: a human complement and Inmunol3/2006 copia 13/12/06 16:38 Página 185 INMUNOLOGÍA immune regulator with cell-adhesive function. Immunol Today 1999;20:135-140. 101.Rodriguez de Cordoba S, Esparza-Gordillo J, Goicoechea de Jorge E, Lopez-Trascasa M, Sanchez-Corral P. The human complement factor H: functional roles, genetic variations and disease associations. Mol Immunol 2004;41:355-367. 102.Gasque P, Julen N, Ischenko AM, Picot C, Mauger C, Chauzy C, et al. Expression of complement components of the alternative pathway by glioma cell lines. J Immunol 1992;149:1381-1387. 103.Katz Y, Guterman M, Lahat E. Regulation of synthesis of complement proteins in HEp2 cells. Clin Immunol Immunopathol 1993;67:117123. 104.Legoedec J, Gasque P, Jeanne JF, Fontaine M. Expression of the complement alternative pathway by human myoblasts in vitro: biosynthesis of C3, factor B, factor H and factor I. Eur J Immunol 1995;25:3460-3466. 105.Gasque P, Thomas A, Fontaine M, Morgan BP. Complement activation on human neuroblastoma cell lines in vitro: route of activation and expression of functional complement regulatory proteins. J Neuroimmunol 1996;66:29-40. 106.Kinders R, Jones T, Root R, Bruce C, Murchison H, Corey M, et al. Complement factor H or a related protein is a marker for transitional cell cancer of the bladder. Clin Cancer Res 1998;4: 25112520. 107.Junnikkala S, Hakulinen J, Jarva H, Manuelian T, Bjorge L, Butzow R, et al. Secretion of soluble complement inhibitors factor H and factor H-like protein (FHL-1) by ovarian tumour cells. Br J Cancer 2002;87:1119-1127. 108.Cheng ZZ, Corey MJ, Parepalo M, Majno S, Hellwage J, Zipfel PF, et al. Complement factor H as a marker for detection of bladder cancer. Clin Chem 2005;51:856-863. 109.Maenpaa A, Junnikkala S, Hakulinen J, Timonen T, Meri S. Expression of complement membrane regulators membrane cofactor protein (CD46), decay accelerating factor (CD55), and protectin (CD59) in human malignant gliomas. Am J Pathol 1996;148: 1139-1152. 110.Corey MJ, Kinders RJ, Brown LG, Vessella RL. A very sensitive coupled luminescent assay for cytotoxicity and complementmediated lysis. J Immunol Methods 1997;207:43-51. 111.Fedarko NS, Fohr B, Robey PG, Young MF, Fisher LW. Factor H binding to bone sialoprotein and osteopontin enables tumor cell evasion of complement-mediated attack. J Biol Chem 2000;275: 16666-16672. R. PIO GJ. Expression of CD46, CD55, and CD59 on renal tumor cell lines and their role in preventing complement-mediated tumor cell lysis. Lab Invest 1996;74:1039-1049. 114.Battistelli S, Vittoria A, Cappelli R, Stefanoni M, Roviello F. Protein S in cancer patients with non-metastatic solid tumours. Eur J Surg Oncol 2005;31:798-802. 115.Holmberg MT, Blom AM, Meri S. Regulation of complement classical pathway by association of C4b-binding protein to the surfaces of SK-OV-3 and Caov-3 ovarian adenocarcinoma cells. J Immunol 2001;167:935-939. 116.Morris KM, Aden DP, Knowles BB, Colten HR. Complement biosynthesis by the human hepatoma-derived cell line HepG2. J Clin Invest 1982;70:906-913. 117.Buo L, Karlsrud TS, Dyrhaug G, Jacobsen MB, Bell H, Johansen HT, Aasen AO. Differential diagnosis of human ascites: inhibitors of the contact system and total proteins. Scand J Gastroenterol 1993;28:777-782. 118.Perricone R, De Carolis C, Giacomello F, Giacomelli R, De Sanctis G, Fontana L. Impaired human ovarian follicular fluid complement function in hereditary angioedema. Scand J Immunol 2000;51:104108. 119.Trougakos IP, Gonos ES. Clusterin/apolipoprotein J in human aging and cancer. Int J Biochem Cell Biol 2002;34:1430-1448. 120.Harris M. Monoclonal antibodies as therapeutic agents for cancer. Lancet Oncol 2004;5:292-302. 121.Adams GP, Weiner LM. Monoclonal antibody therapy of cancer. Nat Biotechnol 2005;23:1147-1157. 122.Trikha M, Yan L, Nakada MT. Monoclonal antibodies as therapeutics in oncology. Curr Opin Biotechnol 2002;13:609-614. 123.Gelderman KA, Tomlinson S, Ross GD, Gorter A. Complement function in mAb-mediated cancer immunotherapy. Trends Immunol 2004;25:158-164. 124.Imai M, Landen C, Ohta R, Cheung NK, Tomlinson S. Complementmediated mechanisms in anti-GD2 monoclonal antibody therapy of murine metastatic cancer. Cancer Res 2005;65:10562-10568. 125.Dillman RO. Treatment of low-grade B-cell lymphoma with the monoclonal antibody rituximab. Semin Oncol 2003;30:434-447. 126.Maloney DG, Smith B, Rose A. Rituximab: mechanism of action and resistance. Semin Oncol 2002;29:2-9. 127.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. 112.Jain A, Karadag A, Fohr B, Fisher LW, Fedarko NS. Three SIBLINGs (small integrin-binding ligand, N-linked glycoproteins) enhance factor H's cofactor activity enabling MCP-like cellular evasion of complement-mediated attack. J Biol Chem 2002;277:13700-13708. 128.Bellosillo B, Villamor N, Lopez-Guillermo A, Marce S, Esteve J, Campo E, et al. Complement-mediated cell death induced by rituximab in B-cell lymphoproliferative disorders is mediated in vitro by a caspase-independent mechanism involving the generation of reactive oxygen species. Blood 2001;98:2771-2777. 113.Gorter A, Blok VT, Haasnoot WH, Ensink NG, Daha MR, Fleuren 129.Bannerji R, Kitada S, Flinn IW, Pearson M, Young D, Reed JC, 185 Inmunol3/2006 copia 13/12/06 16:38 Página 186 CONTROL OF COMPLEMENT ACTIVATION BY CANCER CELLS AND ITS IMPLICATIONS IN ANTIBODY-MEDIATED ... Byrd JC. Apoptotic-regulatory and complement-protecting protein expression in chronic lymphocytic leukemia: relationship to in vivo rituximab resistance. J Clin Oncol 2003;21:1466-1471. 130.Weng WK, Levy R. Expression of complement inhibitors CD46, CD55, and CD59 on tumor cells does not predict clinical outcome after rituximab treatment in follicular non-Hodgkin lymphoma. Blood 2001;98:1352-1357. 131.Golay J, Zaffaroni L, Vaccari T, Lazzari M, Borleri GM, 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. 132.Cardarelli PM, Quinn M, Buckman D, Fang Y, Colcher D, King DJ, et al. Binding to CD20 by anti-B1 antibody or F(ab')(2) is sufficient for induction of apoptosis in B-cell lines. Cancer Immunol Immunother 2002;51:15-24. 133.Terui Y, Sakurai T, Mishima Y, Mishima Y, Sugimura N, Sasaoka C, et al. Blockade of bulky lymphoma-associated CD55 expression by RNA interference overcomes resistance to complement-dependent cytotoxicity with rituximab. Cancer Sci 2006;97:72-79. 134.Paganelli G, Magnani P, Zito F, Villa E, Sudati F, Lopalco L, et al. Three-step monoclonal antibody tumor targeting in carcinoembryonic antigen-positive patients. Cancer Res 1991;51: 5960-5966. 135.Macor P, Mezzanzanica D, Cossetti C, Alberti P, Figini M, Canevari S, Tedesco F. Complement activated by chimeric anti-folate receptor antibodies is an efficient effector system to control ovarian carcinoma. Cancer Res 2006;66:3876-3883. 136.Gelderman KA, Kuppen PJ, Bruin W, Fleuren GJ, Gorter A. Enhancement of the complement activating capacity of 17-1A mAb to overcome the effect of membrane-bound complement regulatory proteins on colorectal carcinoma. Eur J Immunol 2002;32: 128-135. 137.Gelderman KA, Lam S, Gorter A. Inhibiting complement regulators in cancer immunotherapy with bispecific mAbs. Expert Opin Biol Ther 2005;5:1593-1601. 138.Junnikkala S, Hakulinen J, Meri S. Targeted neutralization of the complement membrane attack complex inhibitor CD59 on the surface of human melanoma cells. Eur J Immunol 1994;24:611-615. 139.Harris CL, Kan KS, Stevenson GT, Morgan BP. Tumour cell killing using chemically engineered antibody constructs specific for tumour cells and the complement inhibitor CD59. Clin Exp Immunol 1997;107:364-371. 140.Blok VT, Daha MR, Tijsma O, Harris CL, Morgan BP, Fleuren GJ, Gorter A. A bispecific monoclonal antibody directed against both the membrane-bound complement regulator CD55 and the renal tumor-associated antigen G250 enhances C3 deposition and tumor cell lysis by complement. J Immunol 1998;160:3437-3443. 141.Gelderman KA, Kuppen PJ, Okada N, Fleuren GJ, Gorter A. Tumor- 186 VOL. 25 NUM. 3/ 2006 specific inhibition of membrane-bound complement regulatory protein Crry with bispecific monoclonal antibodies prevents tumor outgrowth in a rat colorectal cancer lung metastases model. Cancer Res 2004;64:4366-4372. 142.Blok VT, Gelderman KA, Tijsma OH, Daha MR, Gorter A. Cytokines affect resistance of human renal tumour cells to complementmediated injury. Scand J Immunol 2003;57:591-599. 143.Bjorge L, Jensen TS, Ulvestad E, Vedeler CA, Matre R. The influence of tumour necrosis factor-alpha, interleukin-1 beta and interferongamma on the expression and function of the complement regulatory protein CD59 on the human colonic adenocarcinoma cell line HT29. Scand J Immunol 1995;41:350-356. 144.Andoh A, Fujiyama Y, Sumiyoshi K, Sakumoto H, Bamba T. Interleukin 4 acts as an inducer of decay-accelerating factor gene expression in human intestinal epithelial cells. Gastroenterology 1996;111:911-918. 145.Blom DJ, Goslings WR, De Waard-Siebinga I, Luyten GP, Claas FH, Gorter A, Jager MJ. Lack of effect of different cytokines on expression of membrane-bound regulators of complement activity on human uveal melanoma cells. J Interferon Cytokine Res 1997;17: 695-700. 146.Varsano S, Rashkovsky L, Shapiro H, Radnay J. Cytokines modulate expression of cell-membrane complement inhibitory proteins in human lung cancer cell lines. Am J Respir Cell Mol Biol 1998;19: 522-529. 147.Pasch MC, Bos JD, Daha MR, Asghar SS. Transforming growth factor-beta isoforms regulate the surface expression of membrane cofactor protein (CD46) and CD59 on human keratinocytes. Eur J Immunol 1999;29:100-108. 148.Spiller OB, Criado-Garcia O, Rodriguez De Cordoba S, Morgan BP. Cytokine-mediated up-regulation of CD55 and CD59 protects human hepatoma cells from complement attack. Clin Exp Immunol 2000;121:234-241. 149.Varsano S, Kaminsky M, Kaiser M, Rashkovsky L. Generation of complement C3 and expression of cell membrane complement inhibitory proteins by human bronchial epithelium cell line. Thorax 2000;55:364-369. 150.Bjorge L, Matre R. Down-regulation of CD59 (protectin) expression on human colorectal adenocarcinoma cell lines by levamisole. Scand J Immunol 1995;42:512-516. 151.Di Gaetano N, Xiao Y, Erba E, Bassan R, Rambaldi A, Golay J, Introna M. Synergism between fludarabine and rituximab revealed in a follicular lymphoma cell line resistant to the cytotoxic activity of either drug alone. Br J Haematol 2001;114:800-809. 152.Andoh A, Shimada M, Araki Y, Fujiyama Y, Bamba T. Sodium butyrate enhances complement-mediated cell injury via downregulation of decay-accelerating factor expression in colonic cancer cells. Cancer Immunol Immunother 2002;50:663-672. 153.Nagajothi N, Matsui WH, Mukhina GL, Brodsky RA. Enhanced Inmunol3/2006 copia 13/12/06 16:38 Página 187 INMUNOLOGÍA cytotoxicity of rituximab following genetic and biochemical disruption of glycosylphosphatidylinositol anchored proteins. Leuk Lymphoma 2004;45:795-799. R. PIO and complement of B-cell chronic lymphocytic leukemia: further regulation by CD55 and CD59. Blood 2001;98:3383-3389. 154.Kock MA, Hew BE, Bammert H, Fritzinger DC, Vogel CW. Structure and function of recombinant cobra venom factor. J Biol Chem 2004;279:30836-30843. 162.Takei K, Yamazaki T, Sawada U, Ishizuka H, Aizawa S. Analysis of changes in CD20, CD55, and CD59 expression on established rituximab-resistant B-lymphoma cell lines. Leuk Res 2006;30:625631. 155.Juhl H, Petrella EC, Cheung NK, Bredehorst R, Vogel CW. Complement killing of human neuroblastoma cells: a cytotoxic monoclonal antibody and its F(ab')2-cobra venom factor conjugate are equally cytotoxic. Mol Immunol 1990;27:957-964. 163.Spiridon CI, Ghetie MA, Uhr J, Marches R, Li JL, Shen GL, Vitetta ES. Targeting multiple Her-2 epitopes with monoclonal antibodies results in improved antigrowth activity of a human breast cancer cell line in vitro and in vivo. Clin Cancer Res 2002;8:1720-1730. 156.Juhl H, Petrella EC, Cheung NK, Bredehorst R, Vogel CW. Additive cytotoxicity of different monoclonal antibody-cobra venom factor conjugates for human neuroblastoma cells. Immunobiology 1997;197:444-459. 164.Schuster M, Umana P, Ferrara C, Brunker P, Gerdes C, Waxenecker G, et al. Improved effector functions of a therapeutic monoclonal Lewis Y-specific antibody by glycoform engineering. Cancer Res 2005;65:7934-7941. 157.Juhl H, Sievers M, Baltzer K, Helmig F, Wolf H, Brenner W, Kalthoff H. A monoclonal antibody-cobra venom factor conjugate increases the tumor-specific uptake of a 99mTc-labeled anti-carcinoembryonic antigen antibody by a two-step approach. Cancer Res 1995;55: 5749s-5755s. 165.Ragupathi G, Liu NX, Musselli C, Powell S, Lloyd K, Livingston PO. Antibodies against tumor cell glycolipids and proteins, but not mucins, mediate complement-dependent cytotoxicity. J Immunol 2005;174:5706-5712. 158.Yefenof E, Zehavi-Feferman R, Guy R. Control of primary and secondary antibody responses by cytotoxic T lymphocytes specific for a soluble antigen. Eur J Immunol 1990;20:1849-1853. 159.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. 160.Holliger P, Wing M, Pound JD, Bohlen H, Winter G. Retargeting serum immunoglobulin with bispecific diabodies. Nat Biotechnol 1997;15:632-636. 161.Golay J, Lazzari M, Facchinetti V, Bernasconi S, Borleri G, Barbui T, et al. CD20 levels determine the in vitro susceptibility to rituximab 166.Livingston PO, Hood C, Krug LM, Warren N, Kris MG, Brezicka T, Ragupathi G. Selection of GM2, fucosyl GM1, globo H and polysialic acid as targets on small cell lung cancers for antibody mediated immunotherapy. Cancer Immunol Immunother 2005;54: 1018-1025. 167.Imai M, Ohta R, Okada N, Tomlinson S. Inhibition of a complement regulator in vivo enhances antibody therapy in a model of mammary adenocarcinoma. Int J Cancer 2004;110:875-881. 168.Gelderman KA, Hakulinen J, Hagenaars M, Kuppen PJ, Meri S, Gorter A. Membrane-bound complement regulatory proteins inhibit complement activation by an immunotherapeutic mAb in a syngeneic rat colorectal cancer model. Mol Immunol 2003;40: 1323. 187