Radiol Oncol 2006; 40(2): 95-105. review Complement resistance impairs anti-tumour therapy Thomas Konatschnig, Nicolas Geis, Stefan Schultz, Michael Kirschfink Institute of Immunology, University of Heidelberg, Germany Background. Various studies during the last two decades clearly indicate that resistance of human tumour cells to autologous complement is mainly based on the expression of membrane-bound complement regula-tory proteins (mCRP) like CD59, CD55 and CD46 with good evidence for a predominant role of CD59. Be-yond these in vitro findings the importance of this phenomenon for the patients` outcome now becomes evident from first clinical studies. Overcoming complement resistance of tumour cells is therefore considered a promising way to improve therapeutic options and prognosis in a variety of cancer diseases. In this short review two feasible approaches are discussed in more detail: (1) neutralisation of mCRP by monoclonal or recombinant antibodies and (2) gene silencing strategies to down-regulate mCRP by blocking the expression of these proteins on the RNA level using siRNA. Conclusions. As mCRP are also present on all normal tissues like endothelial cells, parenchymatous organs (liver, kidney etc.) or blood cells, mCRP blocking strategies have to be targeted selectively to malignant cells sparing the surrounding healthy tissues from the deleterious complement attack. Despite first encouraging results, translation of mCRP inhibition to improve antibody-based immunotherapy into the clinic is still a great challenge. Key words: neoplasms – drug therapy; immunology; complement inactivators Introduction The complement system is a cascade of serin proteases that plays an important role in the immune defense, linking innate and ac-quired immunity.1 Activation of comple-ment results in the release of highly potent proinflammatory molecules, the so-called anaphylatoxins, in the formation of the lytic membrane attack complex (MAC), C5b-9, as Received 15 May 2006 Accepted 5 June 2006 Correspondence to: Michael Kirschfink, D.V.M. Ph.D., Im Neuenheimer Feld 305, 69120 Heidelberg. Tel: +49 6221 56 4076/4026; Fax: +49 6221 56 5586; Email: michael.kirschfink@urz.uni-heidelberg.de well as in the opsonisation of pathogens and immune complexes for efficient phagocyto-sis. To protect themselves from unrestricted complement attack, all cells exposed to com-plement express various membrane comple-ment regulatory proteins (mCRP), such as membrane cofactor protein (MCP, CD46), decay accelerating factor (DAF, CD55) and CD59.2 In the last years, multiple studies ha-ve shown that complement resistance of tu-mour cells is a widespread phenomenon that is based on various mechanisms like se-cretion of soluble complement inhibitors or soluble forms of mCRP, respectively, into the microenvironment3-7, expression of sia-lic acid8 or complement cleaving proteases.9 96 Konatschnig T et al. / Complement in anti-tumour therapy Also rebinding of secreted soluble comple-ment inhibitors to the tumour has been ob-served.10,11 The most important mechanism, however, is the overexpression of one ore more of the membrane-bound complement regulatory proteins CD46, CD55 and CD59.12,13 Although the influence of each mCRP varies between different tumour cell lines and has to be determined separately, there is strong evidence for an exceptional role of CD59, that blocks the assembly of the membrane attack complex (MAC) by in-terfering with the insertion of C9 thereby preventing the formation of the lytic pore. The functional importance of CD59 has been underlined by several approaches: whereas the mere number of mCRP only in part correlates with tumour cell resistance to complement-mediated lysis, transfection of CD59-negative tumour cells with CD59-cDNA increases their complement resistance considerably.14-16 Moreover, many studi-es have demonstrated that neutralisation of CD59 but also of other mCRP by using mo-noclonal antibodies significantly increases the susceptibility of cancer cells to comple-ment-mediated killing.13 Complement resistance as a prognostic factor? There are only few clinical studies yet to underline the functional importance of complement resistance for tumour cell sur-vival and disease progression. Recently, Watson et al.17 showed that expression of CD59 goes along with a deterio-ration of the patients’ prognosis in colorectal carcinomas. Furthermore, the expression of CD59 correlated with local tumour progression and tissue dedifferentiation in prostate cancer.18 High levels of CD59 are associated with an earlier biochemical relapse measu-red by increasing PSA levels after radical prostatectomy. However, contradicting data Radiol Oncol 2006; 40(2): 95-105. for other tumours do not allow to generalise about the potential impact of mCRP expression levels on disease prognosis. In a study with breast cancer patients, the loss of CD59 expression could be found to go along with a reduced over-all-survival.19 Also other mCRP and their association with the disease prognosis have been studi-ed. The overexpression of CD55 seems to predict a poorer prognosis in patients with colorectal cancer.20 The 7-year survival of patients with high expression levels of CD55 was remarkably lower than that of patients with low expression levels (24% vs. 50%). For breast tumours, Madjd et al.21 found that overexpression of CD46 correla-ted with worse histological staging and a higher risk of tumour recurrence. Intere-stingly, in certain malignancies the loss of CD55 or CD59 may also result in more aggressive tumour growth (bigger tumour si-ze, worse grading, higher rate of lymph no-de metastases) and a poorer prognosis.19,22 For gastric carcinomas a correlation bet-ween CD97(EGF) and CD55, respectively, and tumour invasion into the surrounding tissue is reported.23 High expression profiles of these two molecules go along with aggressive local tumour growth and a higher pathological and clinical staging. All in all, overexpression of mCRP by cancer cells and its possible influence on patients’ mortality seems to be rather hete-rogenous and has to be examined separa-tely for each type of cancer. Impact of complement resistance on immunotherapy Complement resistance has gained signifi-cant importance with the introduction of anti-tumour immunotherapy. It not only in-fluences the course of disease but also the patients’ prognosis by impairing therapeu-tic options. Konatschnig T et al. / Complement in anti-tumour therapy 97 The rapid progress in molecular biology and recombinant antibody technology du-ring the last two decades promoted immu-notherapy of malignant diseases. Since then, anti-tumour antibodies successfully made their way from the laboratories to the clinic and meanwhile present a well-esta-blished adjuvant therapy regimen for a va-riety of cancer diseases (Table 1).24 Classical murine monoclonal antibodies derived from hybridomas according to Köhler and Milstein25 could not succeed in clinical testing because of the risk of severe anaphylactic reactions and formation of ne-utralising human anti-mouse-antibodies (HAMA) with rapid loss of effector functi-ons.26 With the advent of recombinant technology, `designer’ antibodies became a powerful tool in anti-cancer therapy. Be-yond the well-known classical antibody ef-fector functions such as antibody-depen-dent cellular cytotoxicity (ADCC) or com-plement-dependent cytotoxicity (CDC), there are additional effects on the target cells that rather depend on the epitope than on the antibody itself.27 These so-cal-led epitope-specific antibody effects can trigger apoptosis or can modulate the auto-and paracrine secretion of tumour cells, thus influencing the tumour’s microenvi-ronment.28 It is often difficult to determine which effect is most important for the anti-body’s anti-tumour response. Despite the great success of recombi-nant antibodies in cancer therapy, clinical oncologists and tumour immunologists are Table 1. Anti-tumour antibodies in clinical use MAb name Trade name Target Type Approval date Used to treat Rituximab Rituxan CD20 IgG1, Chimeric 1997 Non-Hodgkin lymphoma Trastuzumab Herceptin p185neu IgG1, Humanised 1998 Breast cancer Gemtuzumab-ozogamicin* Mylotarg CD33 IgG4, Humanised 2000 Acute myelogenous leukemia (AML) Alemtuzumab Campath CD52 IgG1, Humanised 2001 Chronic lymphocytic leukemia (CLL) In-111/Y-90-Ibritumomab-tiuxetan* Zevalin CD20 IgG1, Murine 2002 Non-Hodgkin lymphoma Daclizumab Zenapax CD25 IgG1, Chimeric 2002 Acute and Chronic leukemia I-131-Tositumomab* Bexxar CD20 IgG2, Murine 2003 Non-Hodgkin lymphoma Bevacizumab Avastin VEGF IgG1, Humanised 2004 Colorectal cancer Cetuximab Erbitux EGFR IgG1, Chimeric 2004 Colorectal cancer * conjugated monoclonal antibodies Radiol Oncol 2006; 40(2): 95-105. 98 Konatschnig T et al. / Complement in anti-tumour therapy confronted with limitations of this appro-ach. Similar to the well-known phenome-non of chemoresistance of tumours, i.e. the capacity of certain tumour cell clones to be-come refractory to cytostatic agents, there is also a phenomenon of resistance to anti-bodies.29 After repetitive treatment cycles, tumour cells get resistant against further antibody therapy. Several mechanisms may lead to antibody resistance, e.g down-regulation of the target epitope or dimini-shed effector functions. Various studies in-dicate that the up-regulation of mCRP, na-mely CD55 and CD59, is responsible for resistance against CD20 serotherapy with ri-tuximab.29-32 Blocking of these regulatory molecules can restore the tumour cells’ su-sceptibility to rituximab in vitro.30,31 The cytotoxic effects of the anti-her2/neu anti-body used in the therapy of metastased breast tumours could be augmented by blocking of mCRP in vitro.33 From these data mCRP appear as intere-sting target epitopes for new adjuvant the-rapeutic regimen. Strategies for tackling complement resistance on human tumours The significance of complement resistance of human tumours became obvious thro-ugh multiple experiments applying murine monoclonal antibodies that blocked mCRPs. 4,6,12,13,34,35 However, translation of these findings into the clinic is hampered by two major obstacles: (1) to find the most effective and secure way of mCRP neutralisiation and (2) restriction of the potentially dangerous intervention to cancer cells. Different to the benchside situation, a therapeutic strategy must be tolerable for the patient. Blocking mCRP by murine mo-noclonal antibodies is not appropriate (for reasons as discribed above). Two promi-sing approaches have been developed for Radiol Oncol 2006; 40(2): 95-105. the future clinical application, which, ho-wever, still require comprehensive preclini-cal investigation. Bispecific mCRP-blocking antibodies For antibody-based immunotherapy the possibility to generate bispecific antibodies that can recognize two different epitopes by their two different antigen binding sites widens the scope and improves the chan-ces to generate truly tumour-specific »magic bullets«.36,37 Bispecific antibodies, which allow mCRP inhibition to be restric-ted to tumour cells in vitro have been pro-duced by various means.38-41 Harris et al.40 generated chimeric anti-CD59 x anti-CD19 and anti-CD59 x anti-CD38 antibodies by chemical linkage. B cell specific binding and lysis could be observed while sparing surrounding bystander cells. Although this work served as »proof of principle«, the chemical synthesis of bispecific antibodies is a cumbersome procedure and inappro-priate for clinical testing. Blok et al.41 obta-ined murine bispecific anti-CD55 x anti-G250 antibodies applying classical hybridoma or quadroma technology with good activity against renal cell carcinomas in vitro. Recently, a bispecific monoclonal anti-CD55 x anti-MHC class I antibody proved its efficacy on human colorectal and cervix carcinoma cell lines resulting in elevated C3-deposition and augmentation of com-plement-mediated cell lysis.39 For therapeutic approaches the use of humanised or at least chimeric antibodies is mandatory. These bispecific antibodies are nowadays constructed by recombinant »antibody engineering«.42 However, despite all progress in the field of recombinant antibody technology it remains difficult to obtain continuously sufficient amounts of bispecific antibodies for in vivo testing in experimental animals or even clinical studies. The best established Konatschnig T et al. / Complement in anti-tumour therapy 99 Figure 1. Blocking of CD59 augments tumour-directed complement activation: FACS-Scan for C3 (C3d) detection on human K562 erythroleukemic cell line after preincubation with polyclonal rabbit-anti-K562 or chimeric anti-CD59-miniantibody and pooled human serum as complement source. (A) Positive control with polyclonal rabbit-anti-K562 (green line), (B) Chimeric anti-CD59-scFv-Fc (green line). (Underlied curves each show two negative controls with heat inactivated serum or with irrelevant human IgG, respectively). way to produce humanised bispecific anti-bodies takes advantage of expression vec-tors which contain the antibody genes. These vectors are commonly transfected into mammalian or insect cells that subsequ-ently secrete the recombinant antibody into the cell culture supernatant. However, this technology still suffers from difficulties in achieving stably transfected clones, varying and vanishing protein production yields, a highly inefficient heterodimerisation of the different antibody chains, and problems with the purification of the heterodimeric bispecific antibodies. Despite the fact that there are several strategies which may help to overcome these difficulties, construction and expression of recombinant bispecific antibodies remains a »high risk challenge« far away from laboratory routine and with still unpredictable outcome. We recently developed a chimeric mou-se/human anti-CD59 miniantibody (scFv-Fc) from a murine hydridoma (MEM43) that was able to trigger C3-deposition on human tumour cells via the Fc-mediated classical pathway although it failed to signi-ficantly augment complement-dependent killing (Figure 1). 43 Ziller et al.44 generated humanised anti-CD59 and anti-CD55 miniantibodies, that were able to trigger complement-mediated lysis on human lymphoma cell lines. Fur-thermore, the lytic effect of rituximab co-uld be augmented by these antibodies. Radiol Oncol 2006; 40(2): 95-105. 100 Konatschnig T et al. / Complement in anti-tumour therapy Silencing of mCRP genes Another approach for tackling complement resistance of human tumours is RNA inter-ference (RNAi). By using small interfering RNAs (siRNA) this technique offers great potential as a novel therapeutic strategy in tumour therapy but also in a wide field of other possible applications. SiRNA technology, known since 2001, is based on short double-stranded RNA oligo-mers which cause highly specific and effici-ent silencing of target genes by posttrans-criptional gene knockdown (Figure 2).45 The antisense-strand of the siRNA molecu-le is complementary to the mRNA of the target protein. SiRNAs induce the intracellular formation of a protein-complex, called „RNA-indu-ced silencing complex (RISC)« consisting of helicase and nuclease-acitivity among others. The RISC-complex induces the separation of the sense and antisense strand, mediates the recognition of the target mRNA and catalyses the degradation of bo-und mRNA. The result is the specific inhibition of target-protein synthesis. Although siRNA and its functionality in mammalian cells was detected just 5 years ago, plenty of studies demonstrating the therapeutic potential of siRNA have already been published. In vivo studies showed positive results applying siRNA for the the-rapy of neoplastic diseases46-48, the treat-ment of sepsis49 and the reduction of chole-sterol levels.50 Meanwhile the first clinical trial of siRNA therapy of the age-related ma-cula degeneration (AMD) has been started. To better exploit complement for cancer cell eradication, we tried to reduce comple-ment-resistance of neoplastic cells by block-ing mCRP function using siRNA-techno-logy. SiRNAs targeting the mCRPs CD59, CD55 and CD46 were designed and tested concerning their downregulation efficiency in vitro. In this study siRNAs were either in- I I I I I I I I I I I '......fill J siRNA: 21-nt dsRNA, 2-nt-overhangs J !_J!! ' 'A J-1- vv ^ formation of the RNA-induced silencing complex („RISC-complex") 1 i i \ i i i i i i i l i i i i i i i i t target- m RNA recognition i i i j......m 11 i j i i 11 -n target-mRN A cleavage and further degradation no target-protein synthesis Figure 2. Schematic presentation of siRNA-induced silencing mechanism. Radiol Oncol 2006; 40(2): 95-105. Konatschnig T et al. / Complement in anti-tumour therapy 101 Figure 3. Cell surface expression of mCRPs CD59, CD55 and CD46 on BT-474 breast carcinoma cells after trans-fection of the corresponding anti-mCRP siRNA individually or in combination, respectively. dividually or combined transfected into Du145 prostate carcinoma cells or BT474 breast carcinoma cells, respectively. The inhibition of target protein expression was analysed both on protein level by FACS analysis and on mRNA level by RT-PCR. Downregulation of mCRP up to 80% could be achieved (Figure 3). Complement-resi-stance of CD55-, CD46- and/or CD59-defici-ent tumour cells, subsequently evaluated by cytotoxicity assays and by analysis of C3 deposition, clearly indicated that siRNA-indu-ced inhibition of mCRP expression sensiti-sed tumour cells to complement attack.51 Despite these encouraging findings and the outstanding potency and selectivity of siRNA, promising to improve targeted can-cer therapy, the systemic administration of aqueous siRNA, even chemically stabili-zed, is still limited by unspecific side ef-fects and a lack of activity in the target tis-sue due to limited blood stability on the one hand and poor intracellular uptake on the other hand.46,52,53 The need for devices enabling systemic administration and targeted delivery to tu- mour tissue and disseminated metastatic lesions is obvious. Strategies based on viral vector delivery would be a possible approach but for safety reasons they are hitherto only of limited cli-nical use. A feasible approach, providing tissue selectivity and safe systemic delivery is based on immunoliposome-technology.54 Liposomes are widely investigated for their properties as site-specific drug carriers allo-wing higher drug doses due to fewer syste-mic side effects.55,56 Liposomes are able to alter the pharmacokinetic profile of a drug, delivering the encapsulated agent preferen-tially to solid tumours, and acting as a slow-release depot for the drug in the dise-ased tissue.57 These attributes often result in a more favourable toxicity profile and an improved therapeutic window for the use of the agent. Though conventional liposomes allow passive tumour site targeting to some de-gree, the idea of conjugation of cell-specific antibodies to liposomes (immunoliposo-mes) has been studied for selective drug delivery.58-60 Radiol Oncol 2006; 40(2): 95-105. 102 Konatschnig T et al. / Complement in anti-tumour therapy Tumour-associated antigens can be utili-sed as appropriate target molecules. Mono-clonal antibodies against tumour-associa-ted antigens have been successfully adop-ted for targeting to various types of cancer cells.61 Internalisation of immunoliposomes by receptor-mediated endocytosis into target-cells results in intracellular drug delivery. A variety of cytotoxic drugs have been delivered to target cells in vitro by using im-munoliposomes; e.g. doxorubicin, vinorel-bine, methotrexate62 and daunomycin.63 Anti-HER2 immunoliposomal doxorubicin is awaiting Phase I clinical trials. Further-more, immunoliposomes have been emplo-yed to deliver oligodeoxyribonucleotides (ODN) designed to specifically inhibit gene expression by blocking translation, splicing or transcription process in vitro, thereby providing powerful therapeutic tools again-st viral diseases and cancer.64 Moreover, in vivo knockdown of gene expression with intravenous RNA interference (RNAi) us-ing a small hairpin RNA (shRNA) expression plasmid encapsulated in immunoliposo-mes has been shown.65 To conclude, immunoliposomes contai-ning siRNA combine specific antibody-me-diated tumour recognition with gene-speci-fic downregulation of target mRNAs. Another promising approach of targeted siRNA delivery in vivo has been achieved by complexation of chemically unmodified siRNAs with polyethylenimine (PEI).66,67 Self-assembling nanoparticles constructed with polyethylenimine were adapted for siRNA. Target-specific delivery can be achi-eved by attaching peptide ligands (e.g. to bind to integrins) to the nanoparticle. Furthermore, a protamine-antibody fusion protein for systemic, cell-type specific, antibody-mediated siRNA delivery was de-veloped recently.68 This approach takes advantage of the non-covalent nucleic acid-binding properties of protamine, which ori-Radiol Oncol 2006; 40(2): 95-105. ginally nucleates DNA in sperm. In combi-nation with the site-specific delivery pro-perties of the antibody Fab fragment this fusion protein is a feasible device to admi-nister siRNA systemically. Conclusion Complement resistance is a widespread and nowadays well examined mechanism that enables tumour cells to withstand au-tologous immune attack. A magnitude of in vitro and several in vivo studies support the notion that blocking of mCRP is a feasible approach for tackling cancer cells. By me-ans of modern recombinant technologies humanised bispecific anti-mCRP-anti-tu-mour antibodies and siRNA based immu-noliposomes for mCRP gene silencing are promising strategies that could allow transferring experimental complement research to clinical application. Encouraging results from in vitro and animal studies have to be reproduced and then could widen the sco-pe of clinical anti-tumour therapy. References 1. Walport MJ. Complement - First of Two Parts. New Engl J Med 2001; 344: 1058-66. 2. Morgan Paul B, Harris Claire L. Complement regu-latory proteins. Academic Press 1999; 32-40. 3. Thomas A, Gasque P, Vaudry D, Gonzalez B, Fontaine M. Expression of a complete and functional complement system by human neuronal cells in vitro. Int Immunol 2000; 12: 1015-23. 4. Gasque P, Morgan BP, Legoedec J, Chan P, Fontaine M. Human skeletal myoblasts spontaneously activate allogeneic complement but are resistant to killing. J Immunol 1996; 156: 3402-11. 5. Goldberger G, Arnaout MA, Aden D, Kay R, Rits M, Colten HR. Biosynthesis and postsynthetic processing of human C3b/C4b inactivator (factor I) in three hepatoma cell lines. J Biol Chem 1984; 259: 6492-97. Konatschnig T et al. / Complement in anti-tumour therapy 103 6. 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: 848-54. 7. Li L, Spendlove I, Morgan J, Durrant LG. CD55 is over-expressed in the tumour environment. Br J Cancer 2001; 84: 80-6. 8. Donin N, Jurianz K, Ziporen L, Schultz S, Kirschfink M, Fishelson Z. Complement resistance of human carcinoma cells depends on membrane re-gulatory proteins, protein kinases and sialic acid. Clin Exp Immunol 2003; 131: 254-63. 9. Frade R. Structure and functions of proteases which cleave human C3 and are expressed on normal or tumor human cells: some are involved in tumorigenic and metastatic properties to human melanoma cells. Immunopharmacology 1999; 42: 39-45. 10. 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 Immu-nol 2001; 167: 935-39. 11. Fedarko NS, Fohr B, Robey PG, Young MF, Fisher LW. Factor H binding to bone sialoprotein and osteopontin enables tumour cell evasion of com-plement-mediated attack. J Biol Chem 2000; 275: 16666-72. 12. Gorter A, Meri S: Immune evasion of tumor cells using membrane-bound complement regulatory proteins. Immunol Today 1999; 20: 576-82. 13. Fishelson Z, Donin N, Zell S, Schultz S, Kirschfink M. Obstacles to cancer immunotherapy: expression of membrane complement regulatory proteins (mCRP) in tumours. Mol Immunol 2003; 40: 109-23. 14. Yu L, Caragine T, Chen S, Morgan BP, Frey AB, Tomlinson S. Protection of human breast cancer cells from complement-mediated lysis by expression of heterologous CD59. Clin Exp Immunol 1999; 15: 13-8. 15. Chen S, Caragine T, Cheung NK, Tomlinson S. CD59 expressed on a tumour cell surface modula-tes decay-accelerating factor expression and en-hances tumour growth in a rat model of human neuroblastoma. Cancer Res 2000; 60: 3013-8. 16. DeNardo C, Fonsatti E, Sigalotti L, Calabro L, Co-alizzi F, Cortini E et al. Recombinant Transmembrane CD59 (CD59-TM) confers complement resistance to GPI-anchored protein defective melano-ma cells. J Cell Physiol 2002; 190: 200-6. 17. Watson NF, Durrant LG, Madjd Z, Ellis IO, Scho-lefield JH, Spendlove I. Expression of the membrane complement regulatory protein CD59 (protec-tin) is associated with reduced survival in colorec-tal cancer patients. Cancer Immunol Immunother 2006; 55: 973-80. 18. Xu C, Jung M, Burkhardt M, Stephan C, Schnorr D, Loening S et al. Increased CD59 protein expression predicts a PSA relapse in patients after radi-cal prostatectomy. Prostate 2005; 62: 224-32. 19. Madjd Z, Pinder SE, Paish C, Ellis IO, Carmichael J, Durrant LG. Loss of CD59 expression in breast tumours correlates with poor survival. J Pathol 2003; 200: 633-9. 20. Durrant LG, Chapman MA, Buckley DJ, Spendlo-ve I, Robins RA, Armitage NC. Enhanced expression of the complement regulatory protein CD55 predicts a poor prognosis in colorectal cancer pati-ents. Cancer Immunol Immunother 2003; 52: 638-42. 21. Madjd Z, Durrant LG, Pinder SE, Ellis IO, Ronan J, Lewis S et al. Do poor-prognosis breast tumours express membrane cofactor proteins (CD46)? Can-cer Immunol Immunother 2005; 54: 149-56. 22. Madjd Z, Durrant LG, Bradley R, Spendlove I, El-lis IO, Pinder SE. Loss of CD55 is associated with aggressive breast tumours. Clin Cancer Res 2004; 10: 2797-803. 23. Liu Y, Chen L, Peng S, Chen Z, Gimm O, Finke R, Hoang-Vu C. The expression of CD97EGF and its ligand CD55 on marginal epithelium is related to higher stage and depth of tumour invasion of ga-stric carcinomas. Oncol Rep 2005; 14: 1413-20. 24. Ross JS, Gray K, Gray GS, Worland PJ, Rolfe M. Anticancer Antibodies. Am J Clin Pathol 2003; 119: 472-85. 25. Köhler G, Milstein C. Continuous cultures of fu-sed cells secreting antibodies of predefined speci-fity. Nature 1975; 256: 495-7. 26. Courtenay-Luck NS, Epenetos AA, Moorre R, Lar-che M, Pectasides D, Dhokia B et al. Development of primary and secondary immune responses to mouse monoclonal antibodies used in the diagno-sis and therapy of malignant neoplasmas. Cancer Res 1986; 46: 6489-93. 27. Jazirehi AR, Bonavida B. Cellular and molecular signal transduction pathways modulated by rituxi-mab (rituxan, anti-CD20 mAb) in non-Hodgkin`s lymphoma: implications in chemosensitization and therapeutic intervention. Oncogene 2005; 24: 2121-43. Radiol Oncol 2006; 40(2): 95-105. 104 Konatschnig T et al. / Complement in anti-tumour therapy 28. Alas S, Emmanouilides C, Bonavida B. Inhibition of interleukin 10 by Rituximab results in down-re-gulation of Bcl-2 and sensitization of B-cell non-Hodgkin’s lymphoma to apoptosis. Clin Cancer Res 2001; 7: 709-23. 29. Treon SP, Mitsiades C, Mitsiades N, Young G, Doss D, Schlossman R et al. Tumour cell expression of CD59 is associated with resistance to CD20 serotherapy in patients with B-cell malignancies. J Immunother 2001; 24: 263-71. 30. Golay J, Zaffaroni L, Vaccari T, Lazarri M, Borleri GM, Bernasconi S et al. Biologic response of B lymphoma cells to anti-CD20 monoclonal anti-body rituximab in vitro: CD55 and CD59 regulate complement-mediated cell lysis. Blood 2000; 95: 3900-08. 31. Golay J, Lazzari M, Facchineti V, Bernasconi S, Borleri G, Barbui T et al. CD20 levels determine the in vitro susceptibility to rituximab and comple-ment of B-cell chronic lymphocytic leukemia: further regulation by CD55 and CD59. Blood 2001; 98: 3383-9. 32. Bannerji R, Kitada S, Flinn IW, Pearson M, Young D, Reed JC et al. Apoptotic-regulatory and comple-ment-protecting protein expression in chronic lymphocytic leukemia: relationship to in vivo ritu-ximab resistance. J Clin Oncol 2003; 21: 1466-71. 33. Jurianz K, Maslak S, Garcia-Schüler H, Fishelson Z, Kirschfink M. Neutralisation of complement re-gulatory proteins augments lysis of breast carcino-ma cells targeted with rhumAb anti-HER2. Immu-nopharmacology 1999; 42: 209-18. 34. Cheung NK, Walter EI, Smith-Mensah WH, Ra-tnoff WD, Tykocinski ML, Medof ME. Decay-acce-lerating factor protects human tumor cells from complement-mediated cytotoxicity in vitro. J Clin Invest 1988; 81: 1122-8. 35. Bjorge L, Hakulinen J, Wahlström T, Matre R, Meri S. Complement-regulatory proteins in ovarian malignancies. Int J Cancer 1997; 70: 14-25. 36. Weiner LM, Adams GP. New approaches to anti-body therapy. Oncogene 2000; 19: 6144-51. 37. Kipriyanov SM, Le Gall F. Recent advances in the generation of bispecific antibodies for tumour im-munotherapy. Curr Opin Drug Discov Devel 2004; 7: 233-42. 38. Gelderman KA, Lam S, Gorter A. Inhibiting com-plement regulators in cancer immunotherapy with bispecific mAbs. Expert Opin Biol Ther 2005; 5: 1593-601. 39. 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 mono-clonal antibodies or bispecific monoclonal antibo-dies. Lab Invest 2002; 82: 483-93. 40. Harris CL, Kan KS, Stevenson GT, Morgan BP. Tu-mour cell killing using chemically engineered anti-body constructs specific for tumour cells and the complement inhibitor CD59. Clin Exp Immunol 1997; 107: 364-71. 41. Blok VT, Daha MR, Tijsma O, Harris CL, Morgan BP, Fleuren GJ et al. A bispecific monoclonal anti-body directed against both the membrane-bound complement regulator CD55 and the renal tumo-ur-associated antigen G250 enhances C3 deposition and tumour cell lysis by complement. J Immu-nol 1998; 160: 3437-43. 42. Kipriyanov SM, Little M. Generation of Recombi-nant Antibodies. Mol Biotech 1999; 12: 173-201. 43. Konatschnig T, Schultz S, Kirschfink M. Targeting complement resistance on human tumours: Clo-ning, expression and functional characterisation of a novel chimeric anti-CD59 miniantibody. Mol Immumol 2006; 43: 185. 44. Ziller F, Macor P, Bulla R, Sblattero D, Marzari R, Tedesco F. Controlling complement resistance in cancer by using human monoclonal antibodies that neutralize complement-regulatory proteins CD55 and CD59. Eur J Immunol 2005; 35: 2175-83. 45. Elbashir SM, Harborth J, Lendeckel W, Yalcin A, Weber K, Tuschl T. Duplexes of 21-nucleotide RNAs mediate RNA interference in cultured mam-malian cells. Nature 2001; 411: 494-8. 46. Filleur S, Courtin A, Ait-Si-Ali S, Guglielmi J, Mer-le C, Harel-Bellan A et al. SiRNA-mediated inhibition of vascular endothelial growth factor severely limits tumour resistance to antiangiogenic throm-bospondin-1 and slows tumour vascularization and growth. Cancer Res 2003; 63: 3919-22. 47. Li MJ, McMahon R, Snyder DS, Yee JK, Rossi JJ. Specific killing of Ph+ chronic myeloid leukemia cells by a lentiviral vector-delivered anti-bcr/abl small hairpin RNA. Oligonucleotides 2003; 13: 401-9. 48. Verma UN, Surabhi RM, Schmaltieg A, Becerra C, Gaynor RB. Small interfering RNAs directed aga-inst beta-catenin inhibit the in vitro and in vivo growth of colon cancer cells. Clin Cancer Res 2003; 9: 1291-300. Radiol Oncol 2006; 40(2): 95-105. Konatschnig T et al. / Complement in anti-tumour therapy 105 49. Sorensen, D.R., Leirdal, M. & Sioud, M. Gene si-lencing by systemic delivery of synthetic siRNAs in adult mice. J Mol Biol 2003; 327: 761-6. 50. Soutschek J, Akinc A, Bramlage B, Charisse K, Constien R, Donoghue M et al. Therapeutic silen-cing of an endogenous gene by systemic administration of modified siRNAs. Nature 2004; 432: 173-8. 51. Zell S, Geis N, Rutz R, Giese T, Schultz S, Kirschfink M. Inhibition of mCRP (CD55, CD46 and CD59) expression by siRNA sensitizes tumor cells to complement attack. Mol Immumol 2006; 43: 138. 52. Tolentino MJ, Brucker AJ, Fosnot J, Ying GS, Wu IH, Malik G et al. Intravitreal injection of vascular endothelial growth factor small interfering RNA inhibits growth and leakage in a nonhuman primate, laser-induced model of choroidal neovascu-larization. Retina 2004; 24: 660. 53. Layzer JM, McCaffrey AP, Tanner AK, Huang Z, Kay MA, Sullenger BA. In vivo activity of nuclea-se-resistant siRNAs. RNA 2004; 10: 766-71. 54. Park JW, Benz CC, Martin FJ. Future directions of liposome- and immunoliposome-based cancer the-rapeutics. Semin Oncol 2004; 31: 196-205. 55. Lasic DD, Papahadjopoulos D. Liposomes revisi-ted. Science 1995; 267: 1275-6. 56. Gregoriadis G. Engineering liposomes for drug de-livery: progress and problems. Trends Biotechnol 1995; 13: 527-37. 57. Drummond D.C., Meyer O.M., Hong K., Kirpotin D.B., Papahadjopoulos D. Optimizing liposomes for delivery of chemotherapeutic agents to solid tumours. Pharmacol Rev 1999; 51: 691-743. 58. Park JW, Hong K, Kirpotin DB, Papahadjopoulos D, Benz CC. Immunoliposomes for cancer trea-tment. Adv Pharmacol 1997; 40: 399-435. 59. Vingerhoeds MH, Storm G, Crommelin DJ. Immu-noliposomes in vivo. Immunomethods 1994; 4: 259-72. 60. Allen TM. Long-circulating (sterically stabilized) li-posomes for targeted drug delivery. Trends Pharma-col Sci 1994; 15: 215-20. 61. Noble CO, Kirpotin DB, Hayes ME, Mamot C, Hong K, Park JW et al. Development of ligand-tar-geted liposomes for cancer therapy. Expert Opin Ther Targets 2004; 8: 335-53. 62. Mamot C, Drummond DC, Greiser U, Hong K, Kirpotin DB, Marks JD et al. Epidermal growth factor receptor (EGFR)-targeted immunoliposo-mes mediate specific and efficient drug delivery to EGFR- and EGFRvIII-overexpressing tumour cells. Cancer Res 2003; 63: 3154-61. 63. Huwyler J, Yang J, Pardridge WM. Receptor mediated delivery of daunomycin using immunoliposo-mes: pharmacokinetics and tissue distribution in the rat. J Pharmacol Exp Ther 1997; 282: 1541-46. 64. Meyer O, Kirpotin D, Hong K, Sternberg B, Park JW, Woodle MC et al. Cationic liposomes coated with polyethylene glycol as carriers for oligonucle-otides. J Biol Chem 1998; 273: 15621-7. 65. Zhang Y, Zhang YF, Bryant J, Charles A, Boado RJ, Pardridge WM. Intravenous RNA interference gene therapy targeting the human epidermal growth factor receptor prolongs survival in intracranial brain cancer. Clin Cancer Res 2004; 10: 3667-77. 66. Schiffelers RM, Ansari A, Xu J, Zhou Q, Tang Q, Storm G et al. Cancer siRNA therapy by tumour selective delivery with ligand-targeted sterically stabilized nanoparticle. Nucleic Acids Res 2004; 32: 149. 67. Aigner A. Gene silencing through RNA interferen-ce (RNAi) in vivo: Strategies based on the direct application of siRNAs. J Biotechnol 2006 Jan 10; [Epub ahead of print]. 68. Song E, Zhu P, Lee SK, Chowdhury D, Kussman S, Dykxhoorn DM et al. Antibody mediated in vivo delivery of small interfering RNAs via cell-surface receptors. Nat Biotechnol 2005; 23: 709-17. Radiol Oncol 2006; 40(2): 95-105. 136 Slovenian abstracts Radiol Oncol 2006; 40(2): 95-105. Odpornost na komplement ovira onkološko zdravljenje Konatschnig T, Geis N, Scultz S, Kirschfink M Izhodišča. Različne in vitro raziskave, ki so bile narejene v zadnjih dveh desetletjih, jasno kažejo, da je odpornost človeških tumorskih celic na avtologni komplement pogojena z na membrano vezanimi regulatornimi proteini komplementa (mCRP). Takšna proteina sta CD55 in CD46, najpomembnejšo vlogo pa ima CD59. Ta imunska dogajanja zelo vplivajo na potek bolezni, kar potrjujejo novejše klinične raziskave. Odpraviti odpornost na komplement obeta izboljšanje zdravljenja bolnikov z različnim rakom, s tem pa tudi izboljšanje napovedi izhoda bolezni. V pričujočem kratkem preglednem članku podrobneje predstavljamo: (1) nevtrali-zacijo proteinov mCRP z monoklonskimi ali rekombinantnimi protitelesi in (2) strategijo »utišanja« genov za proteine mCRP z delovanjem na nivoju RNA ob uporabi siRNA. Zaključki. Ker so proteini mCRP prisotni v vseh normalnih tkivih endotelnih celic paren-himskih organov (jetra, ledvica, itd...) in v krvnih celicah, je zelo pomembno, da je blokiranje delovanja proteinov mCRP selektivno in da tako ne prizadene zdravega tkiva. Čeprav so prvi rezultati ohrabrujoči, je vplivanje na delovanje proteinov MCRP, da bi izboljšali imunoterapijo, še vedno velik izziv v klinični praksi. Radiol Oncol 2006; 40(2): 107-13. Katepsini cisteinske skupine in njihovi inhibitorji pri raku glave in vratu: pregled raziskovalnega dela na Onkološkem inštitutu Ljubljana in Kliniki za otorinolaringologijo Kliničnega centra Ljubljana Strojan P Za odločitev o vrsti in intenzivnosti terapije, potrebne za uspešno ozdravitev raka, kot tudi za napoved izida bolezni je potrebna natančna ocena agresivnosti bolezni. Hipoteza, ki predpostavlja napovedni in prognostični pomen posameznih katepsinov in njihovih inhibitorjev, temelji na vpletenosti enih in drugih v obcelične proteolitične procese. Ti so sestavni del večine aktivnosti, povezanih z življenjem normalne celice, kot tudi procesov, povezanih z razgradnjo zunajceličnega matriksa med procesom invazije in zasevanja tu-morskih celic. Vlogo katepsinov in njihovih inhibitorjev pri raku lahko razčlenimo na naslednje skupine: markerji za presejanje; markerji za napoved prisotnosti zasevkov v področnih bezgavkah; markerji za napoved odgovora na zdravljenje in ponovitev bolezni; prognostični markerji. Čeprav je raziskav s področja katepsinov in njihovih endogenih inhibitorjev pri raku glave in vratu malo, rezultati opravičujejo nadaljna preučevanja. V pričujočem pregledu smo predstavili naše izkušnje in rezultate iz desetletnega obdobja klinično usmerjenega razsikovalnega dela in podali mnenje o njihovi napovedni in prognostični vlogi za potrebe vsakodnevne klinične prakse. Radiol Oncol 2006; 40(2): 133-8.