全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...
Antibodies  2012 

Natural Killer (NK)- and T-Cell Engaging Antibody-Derived Therapeutics

DOI: 10.3390/antib1010088

Keywords: NK-cell, T-cell, effector cell, tumor therapy, Fc receptor, bispecific antibody

Full-Text   Cite this paper   Add to My Lib

Abstract:

Unmodified antibodies (abs) have been successful in the treatment of hematologic malignancies, but less so for the treatment of solid tumors. They trigger anti-tumor effects through their Fc-domains, and one way to improve their efficacy is to optimize their interaction with the effectors through Fc-engineering. Another way to empower abs is the design of bispecific abs and related fusion proteins allowing a narrower choice of effector cells. Here we review frequently chosen classes of effector cells, as well as common trigger molecules. Natural Killer (NK)- and T-cells are the most investigated populations in therapeutical approaches with bispecific agents until now. Catumaxomab, the first bispecific ab to receive drug approval, targets the tumor antigen Epithelial Cell Adhesion Molecule (EpCAM) and recruits T-cells via a binding site for the cell surface protein CD3. The next generation of recombinant ab-derivatives replaces the broadly reactive Fc-domain by a binding domain for a single selected trigger. Blinatumomab is the first clinically successful member of this class, targeting cancer cells via CD19 and engaging T-cells by CD3. Other investigators have developed related recombinant fusion proteins to recruit effectors, such as NK-cells and macrophages. The first such agents currently in preclinical and clinical development will be discussed.

References

[1]  Rahlff, J.; Trusch, M.; Haag, F.; Bacher, U.; Horst, A.; Schluter, H.; Binder, M. Antigen-specificity of oligoclonal abnormal protein bands in multiple myeloma after allogeneic stem cell transplantation. Cancer Immunol. Immunother. 2012.
[2]  Sahin, U.; Tureci, O.; Schmitt, H.; Cochlovius, B.; Johannes, T.; Schmits, R.; Stenner, F.; Luo, G.; Schobert, I.; Pfreundschuh, M. Human neoplasms elicit multiple specific immune responses in the autologous host. Proc. Natl. Acad. Sci. USA 1995, 92, 11810–11813.
[3]  Scanlan, M.J.; Chen, Y.T.; Williamson, B.; Gure, A.O.; Stockert, E.; Gordan, J.D.; Tureci, O.; Sahin, U.; Pfreundschuh, M.; Old, L.J. Characterization of human colon cancer antigens recognized by autologous antibodies. Int. J. Cancer 1998, 76, 652–658, doi:10.1002/(SICI)1097-0215(19980529)76:5<652::AID-IJC7>3.0.CO;2-P.
[4]  Fridman, W.H.; Pages, F.; Sautes-Fridman, C.; Galon, J. The immune contexture in human tumours: Impact on clinical outcome. Nat. Rev. Cancer 2012, 12, 298–306, doi:10.1038/nrc3245.
[5]  Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674, doi:10.1016/j.cell.2011.02.013.
[6]  Alley, S.C.; Okeley, N.M.; Senter, P.D. Antibody-drug conjugates: Targeted drug delivery for cancer. Curr. Opin. Chem. Biol. 2011, 14, 529–537.
[7]  Carter, P. Improving the efficacy of antibody-based cancer therapies. Nat. Rev. Cancer 2001, 1, 118–129, doi:10.1038/35101072.
[8]  Dalken, B.; Giesubel, U.; Knauer, S.K.; Wels, W.S. Targeted induction of apoptosis by chimeric granzyme B fusion proteins carrying antibody and growth factor domains for cell recognition. Cell Death Differ. 2006, 13, 576–585, doi:10.1038/sj.cdd.4401773.
[9]  Kovtun, Y.V.; Audette, C.A.; Ye, Y.; Xie, H.; Ruberti, M.F.; Phinney, S.J.; Leece, B.A.; Chittenden, T.; Blattler, W.A.; Goldmacher, V.S. Antibody-drug conjugates designed to eradicate tumors with homogeneous and heterogeneous expression of the target antigen. Cancer Res. 2006, 66, 3214–3221.
[10]  Sievers, E.L.; Appelbaum, F.R.; Spielberger, R.T.; Forman, S.J.; Flowers, D.; Smith, F.O.; Shannon-Dorcy, K.; Berger, M.S.; Bernstein, I.D. Selective ablation of acute myeloid leukemia using antibody-targeted chemotherapy: A phase I study of an anti-CD33 calicheamicin immunoconjugate. Blood 1999, 93, 3678–3684.
[11]  Stasi, R. Gemtuzumab ozogamicin: An anti-CD33 immunoconjugate for the treatment of acute myeloid leukaemia. Expert Opin. Biol. Ther. 2008, 8, 527–540, doi:10.1517/14712598.8.4.527.
[12]  Zwaan, C.M.; Reinhardt, D.; Zimmerman, M.; Hasle, H.; Stary, J.; Stark, B.; Dworzak, M.; Creutzig, U.; Kaspers, G.J. Salvage treatment for children with refractory first or second relapse of acute myeloid leukaemia with gemtuzumab ozogamicin: Results of a phase II study. Br. J. Haematol. 2010, 148, 768–776, doi:10.1111/j.1365-2141.2009.08011.x.
[13]  Younes, A.; Yasothan, U.; Kirkpatrick, P. Brentuximab vedotin. Nat. Rev. Drug Discov. 2012, 11, 19–20, doi:10.1038/nrd3629.
[14]  Redner, R.L. Why doesn't imatinib cure chronic myeloid leukemia? Oncologist 2010, 15, 182–186, doi:10.1634/theoncologist.2009-0297.
[15]  Horton, H.M.; Bernett, M.J.; Pong, E.; Peipp, M.; Karki, S.; Chu, S.Y.; Richards, J.O.; Vostiar, I.; Joyce, P.F.; Repp, R.; et al. Potent in vitro and in vivo activity of an Fc-engineered anti-CD19 monoclonal antibody against lymphoma and leukemia. Cancer Res. 2008, 68, 8049–8057.
[16]  Ferrara, C.; Grau, S.; Jager, C.; Sondermann, P.; Brunker, P.; Waldhauer, I.; Hennig, M.; Ruf, A.; Rufer, A.C.; Stihle, M.; et al. Unique carbohydrate-carbohydrate interactions are required for high affinity binding between FcgammaRIII and antibodies lacking core fucose. Proc. Natl. Acad. Sci. USA 2011, 108, 12669–12674.
[17]  Shinkawa, T.; Nakamura, K.; Yamane, N.; Shoji-Hosaka, E.; Kanda, Y.; Sakurada, M.; Uchida, K.; Anazawa, H.; Satoh, M.; Yamasaki, M.; et al. The absence of fucose but not the presence of galactose or bisecting N-acetylglucosamine of human IgG1 complex-type oligosaccharides shows the critical role of enhancing antibody-dependent cellular cytotoxicity. J. Biol. Chem. 2003, 278, 3466–3473.
[18]  Vidal, L.; Gafter-Gvili, A.; Salles, G.; Dreyling, M.H.; Ghielmini, M.; Hsu Schmitz, S.F.; Pettengell, R.; Witzens-Harig, M.; Shpilberg, O. Rituximab maintenance for the treatment of patients with follicular lymphoma: An updated systematic review and meta-analysis of randomized trials. J. Natl. Cancer Inst. 2011, 103, 1799–1806.
[19]  Preithner, S.; Elm, S.; Lippold, S.; Locher, M.; Wolf, A.; da Silva, A.J.; Baeuerle, P.A.; Prang, N.S. High concentrations of therapeutic IgG1 antibodies are needed to compensate for inhibition of antibody-dependent cellular cytotoxicity by excess endogenous immunoglobulin G. Mol. Immunol. 2006, 43, 1183–1193, doi:10.1016/j.molimm.2005.07.010.
[20]  Lopez-Guillermo, A.; Mercadal, S. The clinical use of antibodies in haematological malignancies. Ann. Oncol. 2007, 18 Suppl. 9, ix51–ix57, doi:10.1093/annonc/mdm294.
[21]  Indik, Z.K.; Park, J.G.; Hunter, S.; Schreiber, A.D. The molecular dissection of Fc gamma receptor mediated phagocytosis. Blood 1995, 86, 4389–4399.
[22]  Boruchov, A.M.; Heller, G.; Veri, M.C.; Bonvini, E.; Ravetch, J.V.; Young, J.W. Activating and inhibitory IgG Fc receptors on human DCs mediate opposing functions. J. Clin. Invest. 2005, 115, 2914–2923, doi:10.1172/JCI24772.
[23]  Nimmerjahn, F.; Ravetch, J.V. Antibodies, Fc receptors and cancer. Curr. Opin. Immunol. 2007, 19, 239–245, doi:10.1016/j.coi.2007.01.005.
[24]  Selvaraj, P.; Fifadara, N.; Nagarajan, S.; Cimino, A.; Wang, G. Functional regulation of human neutrophil Fc gamma receptors. Immunol. Res. 2004, 29, 219–230, doi:10.1385/IR:29:1-3:219.
[25]  Xiang, Z.; Cutler, A.J.; Brownlie, R.J.; Fairfax, K.; Lawlor, K.E.; Severinson, E.; Walker, E.U.; Manz, R.A.; Tarlinton, D.M.; Smith, K.G. FcgammaRIIb controls bone marrow plasma cell persistence and apoptosis. Nat. Immunol. 2007, 8, 419–429, doi:10.1038/ni1440.
[26]  Nimmerjahn, F.; Ravetch, J.V. Fcgamma receptors: Old friends and new family members. Immunity 2006, 24, 19–28, doi:10.1016/j.immuni.2005.11.010.
[27]  Fanger, M.W.; Segal, D.M.; Romet-Lemonne, J.L. Bispecific antibodies and targeted cellular cytotoxicity. Immunol. Today 1991, 12, 51–54, doi:10.1016/0167-5699(91)90156-N.
[28]  Keler, T.; Graziano, R.F.; Mandal, A.; Wallace, P.K.; Fisher, J.; Guyre, P.M.; Fanger, M.W.; Deo, Y.M. Bispecific antibody-dependent cellular cytotoxicity of HER2/neu-overexpressing tumor cells by Fc gamma receptor type I-expressing effector cells. Cancer Res. 1997, 57, 4008–4014.
[29]  Muller, D.; Kontermann, R.E. Recombinant bispecific antibodies for cellular cancer immunotherapy. Curr. Opin. Mol. Ther. 2007, 9, 319–326.
[30]  Ochiai, K.; Wang, B.; Rieger, A.; Kilgus, O.; Maurer, D.; Fodinger, D.; Kinet, J.P.; Stingl, G.; Tomioka, H. A review on Fc epsilon RI on human epidermal Langerhans cells. Int. Arch. Allergy Immunol. 1994, 104 Suppl. 1, 63–64, doi:10.1159/000236756.
[31]  Prussin, C.; Metcalfe, D.D. 5. IgE, mast cells, basophils, and eosinophils. J. Allergy Clin. Immunol. 2006, 117 Suppl. 2. Mini-Primer, S450–S456, doi:10.1016/j.jaci.2005.11.016.
[32]  von Bubnoff, D.; Novak, N.; Kraft, S.; Bieber, T. The central role of FcepsilonRI in allergy. Clin. Exp. Dermatol. 2003, 28, 184–187, doi:10.1046/j.1365-2230.2003.01209.x.
[33]  Kikutani, H.; Yokota, A.; Uchibayashi, N.; Yukawa, K.; Tanaka, T.; Sugiyama, K.; Barsumian, E.L.; Suemura, M.; Kishimoto, T. Structure and function of Fc epsilon receptor II (Fc epsilon RII/CD23): A point of contact between the effector phase of allergy and B cell differentiation. Ciba Found. Symp. 1989, 147, 23–31. discussion 31-35.
[34]  Hamre, R.; Farstad, I.N.; Brandtzaeg, P.; Morton, H.C. Expression and modulation of the human immunoglobulin A Fc receptor (CD89) and the FcR gamma chain on myeloid cells in blood and tissue. Scand. J. Immunol. 2003, 57, 506–516, doi:10.1046/j.1365-3083.2003.01220.x.
[35]  Stockmeyer, B.; Dechant, M.; van Egmond, M.; Tutt, A.L.; Sundarapandiyan, K.; Graziano, R.F.; Repp, R.; Kalden, J.R.; Gramatzki, M.; Glennie, M.J.; et al. Triggering Fc alpha-receptor I (CD89) recruits neutrophils as effector cells for CD20-directed antibody therapy. J. Immunol. 2000, 165, 5954–5961.
[36]  Stockmeyer, B.; Valerius, T.; Repp, R.; Heijnen, I.A.; Buhring, H.J.; Deo, Y.M.; Kalden, J.R.; Gramatzki, M.; van de Winkel, J.G. Preclinical studies with Fc(gamma)R bispecific antibodies and granulocyte colony-stimulating factor-primed neutrophils as effector cells against HER-2/neu overexpressing breast cancer. Cancer Res. 1997, 57, 696–701.
[37]  Honeychurch, J.; Tutt, A.L.; Valerius, T.; Heijnen, I.A.; Van De Winkel, J.G.; Glennie, M.J. Therapeutic efficacy of FcgammaRI/CD64-directed bispecific antibodies in B-cell lymphoma. Blood 2000, 96, 3544–3552.
[38]  Ranft, K.; Thepen, T.; Fischer, R.; Barth, S.; Stocker, M. Recombinant bispecific single chain antibody fragments induce Fc gamma-receptor-mediated elimination of CD30+ lymphoma cells. Cancer Lett. 2009, 282, 187–194, doi:10.1016/j.canlet.2009.03.011.
[39]  Sundarapandiyan, K.; Keler, T.; Behnke, D.; Engert, A.; Barth, S.; Matthey, B.; Deo, Y.M.; Graziano, R.F. Bispecific antibody-mediated destruction of Hodgkin's lymphoma cells. J. Immunol. Methods 2001, 248, 113–123, doi:10.1016/S0022-1759(00)00347-1.
[40]  Ely, P.; Wallace, P.K.; Givan, A.L.; Graziano, R.F.; Guyre, P.M.; Fanger, M.W. Bispecific-armed, interferon gamma-primed macrophage-mediated phagocytosis of malignant non-Hodgkin's lymphoma. Blood 1996, 87, 3813–3821.
[41]  Borchmann, P.; Schnell, R.; Fuss, I.; Manzke, O.; Davis, T.; Lewis, L.D.; Behnke, D.; Wickenhauser, C.; Schiller, P.; Diehl, V.; et al. Phase 1 trial of the novel bispecific molecule H22xKi-4 in patients with refractory Hodgkin lymphoma. Blood 2002, 100, 3101–3107, doi:10.1182/blood-2001-12-0295.
[42]  Fury, M.G.; Lipton, A.; Smith, K.M.; Winston, C.B.; Pfister, D.G. A phase-I trial of the epidermal growth factor receptor directed bispecific antibody MDX-447 without and with recombinant human granulocyte-colony stimulating factor in patients with advanced solid tumors. Cancer Immunol. Immunother. 2008, 57, 155–163.
[43]  Posey, J.A.; Raspet, R.; Verma, U.; Deo, Y.M.; Keller, T.; Marshall, J.L.; Hodgson, J.; Mazumder, A.; Hawkins, M.J. A pilot trial of GM-CSF and MDX-H210 in patients with erbB-2-positive advanced malignancies. J. Immunother. 1999, 22, 371–379, doi:10.1097/00002371-199907000-00011.
[44]  James, N.D.; Atherton, P.J.; Jones, J.; Howie, A.J.; Tchekmedyian, S.; Curnow, R.T. A phase II study of the bispecific antibody MDX-H210 (anti-HER2 x CD64) with GM-CSF in HER2+ advanced prostate cancer. Br. J. Cancer 2001, 85, 152–156, doi:10.1054/bjoc.2001.1878.
[45]  Guettinger, Y.; Barbin, K.; Peipp, M.; Bruenke, J.; Dechant, M.; Horner, H.; Thierschmidt, D.; Valerius, T.; Repp, R.; Fey, G.H.; Stockmeyer, B. A recombinant bispecific single-chain fragment variable specific for HLA class II and Fc alpha RI (CD89) recruits polymorphonuclear neutrophils for efficient lysis of malignant B lymphoid cells. J. Immunol. 2010, 184, 1210–1217.
[46]  Stadick, H.; Stockmeyer, B.; Kuhn, R.; Schrott, K.M.; Kalden, J.R.; Glennie, M.J.; van de Winkel, J.G.; Gramatzki, M.; Valerius, T.; Elsasser, D. Epidermal growth factor receptor and g250: Useful target antigens for antibody mediated cellular cytotoxicity against renal cell carcinoma? J. Urol. 2002, 167, 707–712.
[47]  Valerius, T.; Stockmeyer, B.; van Spriel, A.B.; Graziano, R.F.; van den Herik-Oudijk, I.E.; Repp, R.; Deo, Y.M.; Lund, J.; Kalden, J.R.; Gramatzki, M.; et al. FcalphaRI (CD89) as a novel trigger molecule for bispecific antibody therapy. Blood 1997, 90, 4485–4492.
[48]  van Egmond, M.; Hanneke van Vuuren, A.J.; van de Winkel, J.G. The human Fc receptor for IgA (Fc alpha RI, CD89) on transgenic peritoneal macrophages triggers phagocytosis and tumor cell lysis. Immunol. Lett. 1999, 68, 83–87, doi:10.1016/S0165-2478(99)00034-6.
[49]  van Egmond, M.; van Vuuren, A.J.; Morton, H.C.; van Spriel, A.B.; Shen, L.; Hofhuis, F.M.; Saito, T.; Mayadas, T.N.; Verbeek, J.S.; van de Winkel, J.G. Human immunoglobulin A receptor (FcalphaRI, CD89) function in transgenic mice requires both FcR gamma chain and CR3 (CD11b/CD18). Blood 1999, 93, 4387–4394.
[50]  Bonde, A.K.; Tischler, V.; Kumar, S.; Soltermann, A.; Schwendener, R.A. Intratumoral macrophages contribute to epithelial-mesenchymal transition in solid tumors. BMC Cancer 2012, 12, 35, doi:10.1186/1471-2407-12-35.
[51]  Maniecki, M.B.; Etzerodt, A.; Ulhoi, B.P.; Steiniche, T.; Borre, M.; Dyrskjot, L.; Orntoft, T.F.; Moestrup, S.K.; Moller, H.J. Tumor-promoting macrophages induce the expression of the macrophage-specific receptor CD163 in malignant cells. Int. J. Cancer 2012.
[52]  Willingham, S.B.; Volkmer, J.P.; Gentles, A.J.; Sahoo, D.; Dalerba, P.; Mitra, S.S.; Wang, J.; Contreras-Trujillo, H.; Martin, R.; Cohen, J.D.; et al. The CD47-signal regulatory protein alpha (SIRPa) interaction is a therapeutic target for human solid tumors. Proc. Natl. Acad. Sci. USA 2012, 109, 6662–6667.
[53]  Chao, M.P.; Alizadeh, A.A.; Tang, C.; Jan, M.; Weissman-Tsukamoto, R.; Zhao, F.; Park, C.Y.; Weissman, I.L.; Majeti, R. Therapeutic antibody targeting of CD47 eliminates human acute lymphoblastic leukemia. Cancer Res. 2011, 71, 1374–1384.
[54]  Edris, B.; Weiskopf, K.; Volkmer, A.K.; Volkmer, J.P.; Willingham, S.B.; Contreras-Trujillo, H.; Liu, J.; Majeti, R.; West, R.B.; Fletcher, J.A.; et al. Antibody therapy targeting the CD47 protein is effective in a model of aggressive metastatic leiomyosarcoma. Proc. Natl. Acad. Sci. USA 2012, 109, 6656–6661.
[55]  Chao, M.P.; Jaiswal, S.; Weissman-Tsukamoto, R.; Alizadeh, A.A.; Gentles, A.J.; Volkmer, J.; Weiskopf, K.; Willingham, S.B.; Raveh, T.; Park, C.Y.; et al. Calreticulin is the dominant pro-phagocytic signal on multiple human cancers and is counterbalanced by CD47. Sci. Transl. Med. 2010, 2, 63–ra94, doi:10.1126/scitranslmed.3001375.
[56]  Gardai, S.J.; McPhillips, K.A.; Frasch, S.C.; Janssen, W.J.; Starefeldt, A.; Murphy-Ullrich, J.E.; Bratton, D.L.; Oldenborg, P.A.; Michalak, M.; Henson, P.M. Cell-surface calreticulin initiates clearance of viable or apoptotic cells through trans-activation of LRP on the phagocyte. Cell 2005, 123, 321–334, doi:10.1016/j.cell.2005.08.032.
[57]  Chao, M.P.; Weissman, I.L.; Majeti, R. The CD47-SIRPalpha pathway in cancer immune evasion and potential therapeutic implications. Curr. Opin. Immunol. 2012, 24, 225–232.
[58]  Kellner, C.; Bruenke, J.; Stieglmaier, J.; Schwemmlein, M.; Schwenkert, M.; Singer, H.; Mentz, K.; Peipp, M.; Lang, P.; Oduncu, F.; et al. A novel CD19-directed recombinant bispecific antibody derivative with enhanced immune effector functions for human leukemic cells. J. Immunother. 2008, 31, 871–884, doi:10.1097/CJI.0b013e318186c8b4.
[59]  Kugler, M.; Stein, C.; Kellner, C.; Mentz, K.; Saul, D.; Schwenkert, M.; Schubert, I.; Singer, H.; Oduncu, F.; Stockmeyer, B.; et al. A recombinant trispecific single-chain Fv derivative directed against CD123 and CD33 mediates effective elimination of acute myeloid leukaemia cells by dual targeting. Br. J. Haematol. 2010, 150, 574–586, doi:10.1111/j.1365-2141.2010.08300.x.
[60]  Stevenson, F.K.; Stevenson, G.T. Follicular lymphoma and the immune system: From pathogenesis to antibody therapy. Blood 2012, 119, 3659–3667, doi:10.1182/blood-2011-11-367730.
[61]  Linsley, P.S.; Brady, W.; Grosmaire, L.; Aruffo, A.; Damle, N.K.; Ledbetter, J.A. Binding of the B cell activation antigen B7 to CD28 costimulates T cell proliferation and interleukin 2 mRNA accumulation. J. Exp. Med. 1991, 173, 721–730, doi:10.1084/jem.173.3.721.
[62]  Rudd, C.E.; Schneider, H. Unifying concepts in CD28, ICOS and CTLA4 co-receptor signalling. Nat. Rev. Immunol. 2003, 3, 544–556, doi:10.1038/nri1131.
[63]  van der Merwe, P.A.; Bodian, D.L.; Daenke, S.; Linsley, P.; Davis, S.J. CD80 (B7-1) binds both CD28 and CTLA-4 with a low affinity and very fast kinetics. J. Exp. Med. 1997, 185, 393–403, doi:10.1084/jem.185.3.393.
[64]  Hodi, F.S.; O'Day, S.J.; McDermott, D.F.; Weber, R.W.; Sosman, J.A.; Haanen, J.B.; Gonzalez, R.; Robert, C.; Schadendorf, D.; Hassel, J.C.; et al. Improved survival with ipilimumab in patients with metastatic melanoma. N. Engl. J. Med. 2010, 363, 711–723.
[65]  Khong, H.T.; Wang, Q.J.; Rosenberg, S.A. Identification of multiple antigens recognized by tumor-infiltrating lymphocytes from a single patient: Tumor escape by antigen loss and loss of MHC expression. J. Immunother. 2004, 27, 184–190, doi:10.1097/00002371-200405000-00002.
[66]  Cabrera, T.; Lara, E.; Romero, J.M.; Maleno, I.; Real, L.M.; Ruiz-Cabello, F.; Valero, P.; Camacho, F.M.; Garrido, F. HLA class I expression in metastatic melanoma correlates with tumor development during autologous vaccination. Cancer Immunol. Immunother. 2007, 56, 709–717, doi:10.1007/s00262-006-0226-7.
[67]  Lanier, L.L. Follow the leader: NK cell receptors for classical and nonclassical MHC class I. Cell 1998, 92, 705–707, doi:10.1016/S0092-8674(00)81398-7.
[68]  Ferrucci, P.F.; Tosti, G.; di Pietro, A.; Passoni, C.; Pari, C.; Tedeschi, I.; Cataldo, F.; Martinoli, C.; Testori, A. Newly identified tumor antigens as promising cancer vaccine targets for malignant melanoma treatment. Curr. Top. Med. Chem. 2012, 12, 11–31, doi:10.2174/156802612798919213.
[69]  Brahmer, J.R.; Drake, C.G.; Wollner, I.; Powderly, J.D.; Picus, J.; Sharfman, W.H.; Stankevich, E.; Pons, A.; Salay, T.M.; McMiller, T.L.; et al. Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: Safety, clinical activity, pharmacodynamics, and immunologic correlates. J. Clin. Oncol. 2010, 28, 3167–3175.
[70]  Madrenas, J.; Chau, L.A.; Teft, W.A.; Wu, P.W.; Jussif, J.; Kasaian, M.; Carreno, B.M.; Ling, V. Conversion of CTLA-4 from inhibitor to activator of T cells with a bispecific tandem single-chain Fv ligand. J. Immunol. 2004, 172, 5948–5956.
[71]  Curran, M.A.; Kim, M.; Montalvo, W.; Al-Shamkhani, A.; Allison, J.P. Combination CTLA-4 blockade and 4-1BB activation enhances tumor rejection by increasing T-cell infiltration, proliferation, and cytokine production. PLoS One 2011, 6, e19499.
[72]  Curran, M.A.; Montalvo, W.; Yagita, H.; Allison, J.P. PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors. Proc. Natl. Acad. Sci. USA 2010, 107, 4275–4280.
[73]  Fisher, T.S.; Kamperschroer, C.; Oliphant, T.; Love, V.A.; Lira, P.D.; Doyonnas, R.; Bergqvist, S.; Baxi, S.M.; Rohner, A.; Shen, A.C.; et al. Targeting of 4-1BB by monoclonal antibody PF-05082566 enhances T-cell function and promotes anti-tumor activity. Cancer Immunol. Immunother. 2012.
[74]  Melero, I.; Shuford, W.W.; Newby, S.A.; Aruffo, A.; Ledbetter, J.A.; Hellstrom, K.E.; Mittler, R.S.; Chen, L. Monoclonal antibodies against the 4-1BB T-cell activation molecule eradicate established tumors. Nat. Med. 1997, 3, 682–685.
[75]  Attarwala, H. TGN1412: From discovery to disaster. J. Young Pharm. 2010, 2, 332–336, doi:10.4103/0975-1483.66810.
[76]  Suntharalingam, G.; Perry, M.R.; Ward, S.; Brett, S.J.; Castello-Cortes, A.; Brunner, M.D.; Panoskaltsis, N. Cytokine storm in a phase 1 trial of the anti-CD28 monoclonal antibody TGN1412. N. Engl. J. Med. 2006, 355, 1018–1028, doi:10.1056/NEJMoa063842.
[77]  Otz, T.; Grosse-Hovest, L.; Hofmann, M.; Rammensee, H.G.; Jung, G. A bispecific single-chain antibody that mediates target cell-restricted, supra-agonistic CD28 stimulation and killing of lymphoma cells. Leukemia 2009, 23, 71–77, doi:10.1038/leu.2008.271.
[78]  Bruenke, J.; Barbin, K.; Kunert, S.; Lang, P.; Pfeiffer, M.; Stieglmaier, K.; Niethammer, D.; Stockmeyer, B.; Peipp, M.; Repp, R.; et al. Effective lysis of lymphoma cells with a stabilised bispecific single-chain Fv antibody against CD19 and FcgammaRIII (CD16). Br. J. Haematol. 2005, 130, 218–228, doi:10.1111/j.1365-2141.2005.05414.x.
[79]  Bryceson, Y.T.; Chiang, S.C.; Darmanin, S.; Fauriat, C.; Schlums, H.; Theorell, J.; Wood, S.M. Molecular mechanisms of natural killer cell activation. J. Innate Immun. 2011, 3, 216–226, doi:10.1159/000325265.
[80]  de Bruyn, M.; Bremer, E.; Helfrich, W. Antibody-based fusion proteins to target death receptors in cancer. Cancer Lett. 2011.
[81]  de Bruyn, M.; Wei, Y.; Wiersma, V.R.; Samplonius, D.F.; Klip, H.G.; van der Zee, A.G.; Yang, B.; Helfrich, W.; Bremer, E. Cell surface delivery of TRAIL strongly augments the tumoricidal activity of T cells. Clin. Cancer Res. 2011, 17, 5626–5637, doi:10.1158/1078-0432.CCR-11-0303.
[82]  Majeti, R.; Becker, M.W.; Tian, Q.; Lee, T.L.; Yan, X.; Liu, R.; Chiang, J.H.; Hood, L.; Clarke, M.F.; Weissman, I.L. Dysregulated gene expression networks in human acute myelogenous leukemia stem cells. Proc. Natl. Acad. Sci. USA 2009, 106, 3396–3401.
[83]  ten Cate, B.; de Bruyn, M.; Wei, Y.; Bremer, E.; Helfrich, W. Targeted elimination of leukemia stem cells; a new therapeutic approach in hemato-oncology. Curr. Drug Targets 2011, 11, 95–110.
[84]  Kontermann, R.E. Bispecific Antibodies: Developments and current perspectives. In Bispecific Antibodies, 1st; Kontermann, R.E., Ed.; Springer-Verlag: Berlin, Germany, 2011; p. 373.
[85]  Haas, C.; Krinner, E.; Brischwein, K.; Hoffmann, P.; Lutterbuse, R.; Schlereth, B.; Kufer, P.; Baeuerle, P.A. Mode of cytotoxic action of T cell-engaging BiTE antibody MT110. Immunobiology 2009, 214, 441–453, doi:10.1016/j.imbio.2008.11.014.
[86]  Loffler, A.; Kufer, P.; Lutterbuse, R.; Zettl, F.; Daniel, P.T.; Schwenkenbecher, J.M.; Riethmuller, G.; Dorken, B.; Bargou, R.C. A recombinant bispecific single-chain antibody, CD19 x CD3, induces rapid and high lymphoma-directed cytotoxicity by unstimulated T lymphocyte. Blood 2000, 95, 2098–2103.
[87]  Bargou, R.; Leo, E.; Zugmaier, G.; Klinger, M.; Goebeler, M.; Knop, S.; Noppeney, R.; Viardot, A.; Hess, G.; Schuler, M.; et al. Tumor regression in cancer patients by very low doses of a T cell-engaging antibody. Science 2008, 321, 974–977.
[88]  Nagorsen, D.; Baeuerle, P.A. Immunomodulatory therapy of cancer with T cell-engaging BiTE antibody blinatumomab. Exp. Cell Res. 2011, 317, 1255–1260, doi:10.1016/j.yexcr.2011.03.010.
[89]  Topp, M.S.; Kufer, P.; Gokbuget, N.; Goebeler, M.; Klinger, M.; Neumann, S.; Horst, H.A.; Raff, T.; Viardot, A.; Schmid, M.; et al. Targeted therapy with the T-cell-engaging antibody blinatumomab of chemotherapy-refractory minimal residual disease in B-lineage acute lymphoblastic leukemia patients results in high response rate and prolonged leukemia-free survival. J. Clin. Oncol. 2011, 29, 2493–2498.
[90]  Handgretinger, R.; Zugmaier, G.; Henze, G.; Kreyenberg, H.; Lang, P.; von Stackelberg, A. Complete remission after blinatumomab-induced donor T-cell activation in three pediatric patients with post-transplant relapsed acute lymphoblastic leukemia. Leukemia 2011, 25, 181–184, doi:10.1038/leu.2010.239.
[91]  Pasquetto, M.V.; Vecchia, L.; Covini, D.; Digilio, R.; Scotti, C. Targeted drug delivery using immunoconjugates: principles and applications. J. Immunother. 2011, 34, 611–628, doi:10.1097/CJI.0b013e318234ecf5.
[92]  Hopp, J.; Hornig, N.; Zettlitz, K.A.; Schwarz, A.; Fuss, N.; Muller, D.; Kontermann, R.E. The effects of affinity and valency of an albumin-binding domain (ABD) on the half-life of a single-chain diabody-ABD fusion protein. Protein Eng. Des. Sel. 2010, 23, 827–834, doi:10.1093/protein/gzq058.
[93]  Kellner, C.; Peipp, M.; Valerius, T. Effector cell recruitment by bispecific antibodies. In Bispecific Antibodies, 1st; Kontermann, R.E., Ed.; Springer-Verlag: Berlin, Germany, 2011; p. 373.
[94]  Bluemel, C.; Hausmann, S.; Fluhr, P.; Sriskandarajah, M.; Stallcup, W.B.; Baeuerle, P.A.; Kufer, P. Epitope distance to the target cell membrane and antigen size determine the potency of T cell-mediated lysis by BiTE antibodies specific for a large melanoma surface antigen. Cancer Immunol. Immunother. 2010, 59, 1197–1209, doi:10.1007/s00262-010-0844-y.
[95]  Brischwein, K.; Schlereth, B.; Guller, B.; Steiger, C.; Wolf, A.; Lutterbuese, R.; Offner, S.; Locher, M.; Urbig, T.; Raum, T.; et al. MT110: A novel bispecific single-chain antibody construct with high efficacy in eradicating established tumors. Mol. Immunol. 2006, 43, 1129–1143, doi:10.1016/j.molimm.2005.07.034.
[96]  Cioffi, M.; Dorado, J.; Baeuerle, P.A.; Heeschen, C. EpCAM/CD3-Bispecific T-cell engaging antibody MT110 eliminates primary human pancreatic cancer stem cells. Clin. Cancer Res. 2012, 18, 465–474, doi:10.1158/1078-0432.CCR-11-1270.
[97]  Kischel, R. Characterization in primates of MCSP- and CD33-specific human BiTE antibodies for treatment of Melanoma and AML. Proc. Am. Assoc. Cancer Res. 2008, 99, 2404.
[98]  Lutterbuese, R.; Raum, T.; Kischel, R.; Lutterbuese, P.; Schlereth, B.; Schaller, E.; Mangold, S.; Rau, D.; Meier, P.; Kiener, P.A.; et al. Potent control of tumor growth by CEA/CD3-bispecific single-chain antibody constructs that are not competitively inhibited by soluble CEA. J. Immunother. 2009, 32, 341–352, doi:10.1097/CJI.0b013e31819b7c70.
[99]  Osada, T.; Hsu, D.; Hammond, S.; Hobeika, A.; Devi, G.; Clay, T.M.; Lyerly, H.K.; Morse, M.A. Metastatic colorectal cancer cells from patients previously treated with chemotherapy are sensitive to T-cell killing mediated by CEA/CD3-bispecific T-cell-engaging BiTE antibody. Br. J. Cancer 2010, 102, 124–133, doi:10.1038/sj.bjc.6605364.
[100]  Torisu-Itakura, H.; Schoellhammer, H.F.; Sim, M.S.; Irie, R.F.; Hausmann, S.; Raum, T.; Baeuerle, P.A.; Morton, D.L. Redirected lysis of human melanoma cells by a MCSP/CD3-bispecific BiTE antibody that engages patient-derived T cells. J. Immunother. 2011, 34, 597–605, doi:10.1097/CJI.0b013e3182307fd8.
[101]  Jager, M.; Schoberth, A.; Ruf, P.; Hess, J.; Hennig, M.; Schmalfeldt, B.; Wimberger, P.; Strohlein, M.; Theissen, B.; Heiss, M.M.; Lindhofer, H. Immunomonitoring results of a phase II/III study of malignant ascites patients treated with the trifunctional antibody catumaxomab (anti-EpCAM x anti-CD3). Cancer Res. 2012, 72, 24–32, doi:10.1158/0008-5472.CAN-11-2235.
[102]  Seimetz, D.; Lindhofer, H.; Bokemeyer, C. Development and approval of the trifunctional antibody catumaxomab (anti-EpCAM x anti-CD3) as a targeted cancer immunotherapy. Cancer Treat. Rev. 2010, 36, 458–467, doi:10.1016/j.ctrv.2010.03.001.
[103]  Kiewe, P.; Hasmuller, S.; Kahlert, S.; Heinrigs, M.; Rack, B.; Marme, A.; Korfel, A.; Jager, M.; Lindhofer, H.; Sommer, H.; et al. Phase I trial of the trifunctional anti-HER2 x anti-CD3 antibody ertumaxomab in metastatic breast cancer. Clin. Cancer Res. 2006, 12, 3085–3091.
[104]  Schroeder, P.; Lindemann, C.; Dettmar, K.; Brieger, J.; Gosepath, J.; Pogorzelski, B.; Seimetz, D.; Atz, J. Trifunctional antibodies induce efficient antitumour activity with immune cells from head and neck squamous cell carcinoma patients after radio-chemotherapy treatment. Clin. Transl. Oncol. 2011, 13, 889–898, doi:10.1007/s12094-011-0751-5.
[105]  Zhang, T.; Sentman, C.L. Cancer immunotherapy using a bispecific NK receptor fusion protein that engages both T cells and tumor cells. Cancer Res. 2011, 71, 2066–2076, doi:10.1158/0008-5472.CAN-10-3200.
[106]  Barber, A.; Rynda, A.; Sentman, C.L. Chimeric NKG2D expressing T cells eliminate immunosuppression and activate immunity within the ovarian tumor microenvironment. J. Immunol. 2009, 183, 6939–6947, doi:10.4049/jimmunol.0902000.
[107]  Thakur, A.; Sorenson, C.; Norkina, O.; Schalk, D.; Ratanatharathorn, V.; Lum, L.G. Activated T cells from umbilical cord blood armed with anti-CD3 x anti-CD20 bispecific antibody mediate specific cytotoxicity against CD20+ targets with minimal allogeneic reactivity: A strategy for providing antitumor effects after cord blood transplants. Transfusion 2012, 52, 63–75, doi:10.1111/j.1537-2995.2011.03232.x.
[108]  Trinchieri, G. Biology of natural killer cells. Adv. Immunol. 1989, 47, 187–376, doi:10.1016/S0065-2776(08)60664-1.
[109]  Sanchez-Correa, B.; Morgado, S.; Gayoso, I.; Bergua, J.M.; Casado, J.G.; Arcos, M.J.; Bengochea, M.L.; Duran, E.; Solana, R.; Tarazona, R. Human NK cells in acute myeloid leukaemia patients: analysis of NK cell-activating receptors and their ligands. Cancer Immunol Immunother 2011, 60, 1195–1205, doi:10.1007/s00262-011-1050-2.
[110]  Alderson, K.L.; Sondel, P.M. Clinical cancer therapy by NK cells via antibody-dependent cell-mediated cytotoxicity. J. Biomed. Biotechnol. 2011, 379123.
[111]  Lundqvist, A.; Abrams, S.I.; Schrump, D.S.; Alvarez, G.; Suffredini, D.; Berg, M.; Childs, R. Bortezomib and depsipeptide sensitize tumors to tumor necrosis factor-related apoptosis-inducing ligand: A novel method to potentiate natural killer cell tumor cytotoxicity. Cancer Res. 2006, 66, 7317–7325.
[112]  Anfossi, N.; Andre, P.; Guia, S.; Falk, C.S.; Roetynck, S.; Stewart, C.A.; Breso, V.; Frassati, C.; Reviron, D.; Middleton, D.; et al. Human NK cell education by inhibitory receptors for MHC class I. Immunity 2006, 25, 331–342, doi:10.1016/j.immuni.2006.06.013.
[113]  Leibson, P.J. Signal transduction during natural killer cell activation: Inside the mind of a killer. Immunity 1997, 6, 655–661, doi:10.1016/S1074-7613(00)80441-0.
[114]  Terunuma, H.; Deng, X.; Dewan, Z.; Fujimoto, S.; Yamamoto, N. Potential role of NK cells in the induction of immune responses: implications for NK cell-based immunotherapy for cancers and viral infections. Int. Rev. Immunol. 2008, 27, 93–110, doi:10.1080/08830180801911743.
[115]  Jaiswal, S.; Chao, M.P.; Majeti, R.; Weissman, I.L. Macrophages as mediators of tumor immunosurveillance. Trends Immunol. 2010, 31, 212–219.
[116]  Lang, P.; Barbin, K.; Feuchtinger, T.; Greil, J.; Peipp, M.; Zunino, S.J.; Pfeiffer, M.; Handgretinger, R.; Niethammer, D.; Fey, G.H. Chimeric CD19 antibody mediates cytotoxic activity against leukemic blasts with effector cells from pediatric patients who received T-cell-depleted allografts. Blood 2004, 103, 3982–3985.
[117]  Fuchs, A.; Colonna, M. The role of NK cell recognition of nectin and nectin-like proteins in tumor immunosurveillance. Semin. Cancer Biol. 2006, 16, 359–366, doi:10.1016/j.semcancer.2006.07.002.
[118]  Guerra, N.; Tan, Y.X.; Joncker, N.T.; Choy, A.; Gallardo, F.; Xiong, N.; Knoblaugh, S.; Cado, D.; Greenberg, N.M.; Raulet, D.H. NKG2D-deficient mice are defective in tumor surveillance in models of spontaneous malignancy. Immunity 2008, 28, 571–580, doi:10.1016/j.immuni.2008.02.016.
[119]  Ljunggren, H.G.; Malmberg, K.J. Prospects for the use of NK cells in immunotherapy of human cancer. Nat. Rev. Immunol. 2007, 7, 329–339, doi:10.1038/nri2073.
[120]  Lang, P.; Pfeiffer, M.; Teltschik, H.M.; Schlegel, P.; Feuchtinger, T.; Ebinger, M.; Klingebiel, T.; Bader, P.; Schlegel, P.G.; Beck, J.; et al. Natural killer cell activity influences outcome after T cell depleted stem cell transplantation from matched unrelated and haploidentical donors. Best Pract. Res. Clin. Haematol. 2011, 24, 403–411, doi:10.1016/j.beha.2011.04.009.
[121]  Locatelli, F.; Vinti, L.; Palumbo, G.; Rossi, F.; Bertaina, A.; Mastronuzzi, A.; Bernardo, M.E.; Rutella, S.; Dellabona, P.; Giorgiani, G.; et al. Strategies to optimize the outcome of children given T-cell depleted HLA-haploidentical hematopoietic stem cell transplantation. Best Pract. Res. Clin. Haematol. 2011, 24, 339–349, doi:10.1016/j.beha.2011.04.004.
[122]  Pende, D.; Marcenaro, S.; Falco, M.; Martini, S.; Bernardo, M.E.; Montagna, D.; Romeo, E.; Cognet, C.; Martinetti, M.; Maccario, R.; et al. Anti-leukemia activity of alloreactive NK cells in KIR ligand-mismatched haploidentical HSCT for pediatric patients: evaluation of the functional role of activating KIR and redefinition of inhibitory KIR specificity. Blood 2009, 113, 3119–3129.
[123]  Ruggeri, L.; Mancusi, A.; Capanni, M.; Urbani, E.; Carotti, A.; Aloisi, T.; Stern, M.; Pende, D.; Perruccio, K.; Burchielli, E.; et al. Donor natural killer cell allorecognition of missing self in haploidentical hematopoietic transplantation for acute myeloid leukemia: Challenging its predictive value. Blood 2007, 110, 433–440, doi:10.1182/blood-2006-07-038687.
[124]  Velardi, A.; Ruggeri, L.; Mancusi, A.; Aversa, F.; Christiansen, F.T. Natural killer cell allorecognition of missing self in allogeneic hematopoietic transplantation: a tool for immunotherapy of leukemia. Curr. Opin. Immunol. 2009, 21, 525–530, doi:10.1016/j.coi.2009.07.015.
[125]  Kitamoto, K.; Machida, Y.; Uchida, J.; Izumi, Y.; Shiota, M.; Nakao, T.; Iwao, H.; Yukimura, T.; Nakatani, T.; Miura, K. Effects of liposome clodronate on renal leukocyte populations and renal fibrosis in murine obstructive nephropathy. J. Pharmacol. Sci. 2009, 111, 285–292, doi:10.1254/jphs.09227FP.
[126]  Giorgini, A.; Brown, H.J.; Lock, H.R.; Nimmerjahn, F.; Ravetch, J.V.; Verbeek, J.S.; Sacks, S.H.; Robson, M.G. Fc gamma RIII and Fc gamma RIV are indispensable for acute glomerular inflammation induced by switch variant monoclonal antibodies. J. Immunol. 2008, 181, 8745–8752.
[127]  Hamerman, J.A.; Ogasawara, K.; Lanier, L.L. Cutting edge: Toll-like receptor signaling in macrophages induces ligands for the NKG2D receptor. J. Immunol. 2004, 172, 2001–2005.
[128]  Sun, J.C.; Lanier, L.L. NK cell development, homeostasis and function: Parallels with CD8 T cells. Nat. Rev. Immunol. 2011, 11, 645–657, doi:10.1038/nri3044.
[129]  Cartron, G.; Dacheux, L.; Salles, G.; Solal-Celigny, P.; Bardos, P.; Colombat, P.; Watier, H. Therapeutic activity of humanized anti-CD20 monoclonal antibody and polymorphism in IgG Fc receptor FcgammaRIIIa gene. Blood 2002, 99, 754–758, doi:10.1182/blood.V99.3.754.
[130]  Weng, W.K.; Levy, R. Two immunoglobulin G fragment C receptor polymorphisms independently predict response to rituximab in patients with follicular lymphoma. J. Clin. Oncol. 2003, 21, 3940–3947, doi:10.1200/JCO.2003.05.013.
[131]  Bibeau, F.; Lopez-Crapez, E.; Di Fiore, F.; Thezenas, S.; Ychou, M.; Blanchard, F.; Lamy, A.; Penault-Llorca, F.; Frebourg, T.; Michel, P.; et al. Impact of Fc{gamma}RIIa-Fc{gamma}RIIIa polymorphisms and KRAS mutations on the clinical outcome of patients with metastatic colorectal cancer treated with cetuximab plus irinotecan. J. Clin. Oncol. 2009, 27, 1122–1129.
[132]  Musolino, A.; Naldi, N.; Bortesi, B.; Pezzuolo, D.; Capelletti, M.; Missale, G.; Laccabue, D.; Zerbini, A.; Camisa, R.; Bisagni, G.; et al. Immunoglobulin G fragment C receptor polymorphisms and clinical efficacy of trastuzumab-based therapy in patients with HER-2/neu-positive metastatic breast cancer. J. Clin. Oncol. 2008, 26, 1789–1796.
[133]  Elsasser, D.; Stadick, H.; Stark, S.; Van de Winkel, J.G.; Gramatzki, M.; Schrott, K.M.; Valerius, T.; Schafhauser, W. Preclinical studies combining bispecific antibodies with cytokine-stimulated effector cells for immunotherapy of renal cell carcinoma. Anticancer Res. 1999, 19, 1525–1528.
[134]  Weiner, L.M.; Holmes, M.; Adams, G.P.; LaCreta, F.; Watts, P.; Garcia de Palazzo, I. A human tumor xenograft model of therapy with a bispecific monoclonal antibody targeting c-erbB-2 and CD16. Cancer Res. 1993, 53, 94–100.
[135]  Weiner, L.M.; Holmes, M.; Richeson, A.; Godwin, A.; Adams, G.P.; Hsieh-Ma, S.T.; Ring, D.B.; Alpaugh, R.K. Binding and cytotoxicity characteristics of the bispecific murine monoclonal antibody 2B1. J. Immunol. 1993, 151, 2877–2886.
[136]  Lu, H.; Shi, M.; Wang, M.; Xie, Z.; Hu, M.; Yu, M.; Shen, B.; Ma, Y.; Guo, N. In vitro and in vivo antitumor effect of a trivalent bispecific antibody targeting ErbB2 and CD16. Cancer Biol. Ther. 2008, 7, 1744–1750, doi:10.4161/cbt.7.11.6725.
[137]  Bruenke, J.; Fischer, B.; Barbin, K.; Schreiter, K.; Wachter, Y.; Mahr, K.; Titgemeyer, F.; Niederweis, M.; Peipp, M.; Zunino, S.J.; et al. A recombinant bispecific single-chain Fv antibody against HLA class II and FcgammaRIII (CD16) triggers effective lysis of lymphoma cells. Br. J. Haematol. 2004, 125, 167–179, doi:10.1111/j.1365-2141.2004.04893.x.
[138]  Singer, H.; Kellner, C.; Lanig, H.; Aigner, M.; Stockmeyer, B.; Oduncu, F.; Schwemmlein, M.; Stein, C.; Mentz, K.; Mackensen, A.; Fey, G.H. Effective elimination of acute myeloid leukemic cells by recombinant bispecific antibody derivatives directed against CD33 and CD16. J. Immunother. 2010, 33, 599–608, doi:10.1097/CJI.0b013e3181dda225.
[139]  Schubert, I.; Kellner, C.; Stein, C.; Kugler, M.; Schwenkert, M.; Saul, D.; Mentz, K.; Singer, H.; Stockmeyer, B.; Hillen, W.; et al. A single-chain triplebody with specificity for CD19 and CD33 mediates effective lysis of mixed lineage leukemia cells by dual targeting. MAbs 2011, 3, 21–30, doi:10.4161/mabs.3.1.14057.
[140]  Schubert, I.; Kellner, C.; Stein, C.; Kugler, M.; Schwenkert, M.; Saul, D.; Stockmeyer, B.; Berens, C.; Oduncu, F.S.; Mackensen, A.; et al. A recombinant triplebody with specificity for CD19 and HLA-DR mediates preferential binding to antigen double-positive cells by dual-targeting. MAbs 2012, 4, 45–56, doi:10.4161/mabs.4.1.18498.
[141]  Schubert, I.; Stein, C.; Fey, G.H. Dual-Targeting for the Elimination of Cancer Cells with Increased Selectivity. Antibodies 2012, 1, 2–18, doi:10.3390/antib1010002.
[142]  Stein, C.; Schubert, I.; Fey, G.H. Trivalent and trispecific antibody derivatives for cancer therapy. In Bispecific Antibodies, 1st; Kontermann, R.E., Ed.; Springer-Verlag: Berlin, Germany, 2011; p. 373.
[143]  Kellner, C.; Bruenke, J.; Horner, H.; Schubert, J.; Schwenkert, M.; Mentz, K.; Barbin, K.; Stein, C.; Peipp, M.; Stockmeyer, B.; et al. Heterodimeric bispecific antibody-derivatives against CD19 and CD16 induce effective antibody-dependent cellular cytotoxicity against B-lymphoid tumor cells. Cancer Lett. 2011, 303, 128–139, doi:10.1016/j.canlet.2011.01.020.
[144]  Johnson, S.; Burke, S.; Huang, L.; Gorlatov, S.; Li, H.; Wang, W.; Zhang, W.; Tuaillon, N.; Rainey, J.; Barat, B.; et al. Effector cell recruitment with novel Fv-based dual-affinity re-targeting protein leads to potent tumor cytolysis and in vivo B-cell depletion. J. Mol. Biol. 2010, 399, 436–449, doi:10.1016/j.jmb.2010.04.001.
[145]  Arndt, M.A.; Krauss, J.; Kipriyanov, S.M.; Pfreundschuh, M.; Little, M. A bispecific diabody that mediates natural killer cell cytotoxicity against xenotransplantated human Hodgkin's tumors. Blood 1999, 94, 2562–2568.
[146]  Kipriyanov, S.M.; Cochlovius, B.; Schafer, H.J.; Moldenhauer, G.; Bahre, A.; Le Gall, F.; Knackmuss, S.; Little, M. Synergistic antitumor effect of bispecific CD19 x CD3 and CD19 x CD16 diabodies in a preclinical model of non-Hodgkin's lymphoma. J. Immunol. 2002, 169, 137–44.
[147]  Schlenzka, J.; Moehler, T.M.; Kipriyanov, S.M.; Kornacker, M.; Benner, A.; Bahre, A.; Stassar, M.J.; Schafer, H.J.; Little, M.; Goldschmidt, H.; et al. Combined effect of recombinant CD19 x CD16 diabody and thalidomide in a preclinical model of human B cell lymphoma. Anticancer Drugs 2004, 15, 915–919, doi:10.1097/00001813-200410000-00013.
[148]  Champsaur, M.; Lanier, L.L. Effect of NKG2D ligand expression on host immune responses. Immunol. Rev. 2010, 235, 267–285.
[149]  Germain, C.; Larbouret, C.; Cesson, V.; Donda, A.; Held, W.; Mach, J.P.; Pelegrin, A.; Robert, B. MHC class I-related chain A conjugated to antitumor antibodies can sensitize tumor cells to specific lysis by natural killer cells. Clin. Cancer Res. 2005, 11, 7516–7522.
[150]  Dhodapkar, M.V.; Sanderson, R.D. Syndecan-1 (CD 138) in myeloma and lymphoid malignancies: A multifunctional regulator of cell behavior within the tumor microenvironment. Leuk. Lymphoma 1999, 34, 35–43.
[151]  Dhodapkar, K.M.; Krasovsky, J.; Williamson, B.; Dhodapkar, M.V. Antitumor monoclonal antibodies enhance cross-presentation ofcCellular antigens and the generation of myeloma-specific killer T cells by dendritic cells. J. Exp. Med. 2002, 195, 125–133, doi:10.1084/jem.20011097.
[152]  von Strandmann, E.P.; Hansen, H.P.; Reiners, K.S.; Schnell, R.; Borchmann, P.; Merkert, S.; Simhadri, V.R.; Draube, A.; Reiser, M.; Purr, I.; et al. A novel bispecific protein (ULBP2-BB4) targeting the NKG2D receptor on natural killer (NK) cells and CD138 activates NK cells and has potent antitumor activity against human multiple myeloma in vitro and in vivo. Blood 2006, 107, 1955–1962.
[153]  Cho, H.M.; Rosenblatt, J.D.; Tolba, K.; Shin, S.J.; Shin, D.S.; Calfa, C.; Zhang, Y.; Shin, S.U. Delivery of NKG2D ligand using an anti-HER2 antibody-NKG2D ligand fusion protein results in an enhanced innate and adaptive antitumor response. Cancer Res. 2010, 70, 10121–10130.
[154]  Jachimowicz, R.D.; Fracasso, G.; Yazaki, P.J.; Power, B.E.; Borchmann, P.; Engert, A.; Hansen, H.P.; Reiners, K.S.; Marie, M.; von Strandmann, E.P.; et al. Induction of in vitro and in vivo NK cell cytotoxicity using high-avidity immunoligands targeting prostate-specific membrane antigen in prostate carcinoma. Mol. Cancer Ther. 2011, 10, 1036–1045, doi:10.1158/1535-7163.MCT-10-1093.
[155]  Kellner, C.; Hallack, D.; Glorius, P.; Staudinger, M.; Mohseni Nodehi, S.; de Weers, M.; van de Winkel, J.G.; Parren, P.W.; Stauch, M.; Valerius, T.; et al. Fusion proteins between ligands for NKG2D and CD20-directed single-chain variable fragments sensitize lymphoma cells for natural killer cell-mediated lysis and enhance antibody-dependent cellular cytotoxicity. Leukemia 2011, 26, 830–834.
[156]  Barkholt, L.; Alici, E.; Conrad, R.; Sutlu, T.; Gilljam, M.; Stellan, B.; Christensson, B.; Guven, H.; Bjorkstrom, N.K.; Soderdahl, G.; et al. Safety analysis of ex vivo-expanded NK and NK-like T cells administered to cancer patients: A phase I clinical study. Immunotherapy 2009, 1, 753–764, doi:10.2217/imt.09.47.
[157]  Klingemann, H. G.; Martinson, J. Ex vivo expansion of natural killer cells for clinical applications. Cytotherapy 2004, 6, 15–22, doi:10.1080/14653240310004548.
[158]  Koehl, U.; Esser, R.; Zimmermann, S.; Tonn, T.; Kotchetkov, R.; Bartling, T.; Sorensen, J.; Gruttner, H.P.; Bader, P.; Seifried, E.; et al. Ex vivo expansion of highly purified NK cells for immunotherapy after haploidentical stem cell transplantation in children. Klin. Padiatr. 2005, 217, 345–350, doi:10.1055/s-2005-872520.
[159]  Schubert, I. Department Biology, University of Erlangen-Nuremberg. 2012, unpublished work.
[160]  Alici, E.; Sutlu, T.; Bjorkstrand, B.; Gilljam, M.; Stellan, B.; Nahi, H.; Quezada, H.C.; Gahrton, G.; Ljunggren, H.G.; Dilber, M.S. Autologous antitumor activity by NK cells expanded from myeloma patients using GMP-compliant components. Blood 2008, 111, 3155–3162, doi:10.1182/blood-2007-09-110312.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133

WeChat 1538708413