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  2013 

Improving the Therapeutic Potential of Human Granzyme B for Targeted Cancer Therapy

DOI: 10.3390/antib2010019

Keywords: immunotherapy, immunotoxin, human cytolytic fusion protein, bio-engineering, serpin B9, surface-charge, endosomal release

Full-Text   Cite this paper   Add to My Lib

Abstract:

Conventional cancer treatments lack specificity and often cause severe side effects. Targeted therapeutic approaches are therefore preferred, including the use of immunotoxins (ITs) that comprise cell-binding and cell death-inducing components to allow the direct and specific delivery of pro-apoptotic agents into malignant cells. The first generation of ITs consisted of toxins derived from bacteria or plants, making them immunogenic in humans. The recent development of human cytolytic fusion proteins (hCFP) consisting of human effector enzymes offers the prospect of highly-effective targeted therapies with minimal side effects. One of the most promising candidates is granzyme B (GrB) and this enzyme has already demonstrated its potential for targeted cancer therapy. However, the clinical application of GrB may be limited because it is inactivated by the overexpression in tumors of its specific inhibitor serpin B9 (PI-9). It is also highly charged, which means it can bind non-specifically to the surface of non-target cells. Furthermore, human enzymes generally lack an endogenous translocation domain, thus the endosomal release of GrB following receptor-mediated endocytosis can be inefficient. In this review we provide a detailed overview of these challenges and introduce promising solutions to increase the cytotoxic potency of GrB for clinical applications.

References

[1]  Klebanoff, C.A.; Acquavella, N.; Yu, Z.Y.; Restifo, N.P. Therapeutic cancer vaccines: Are we there yet? Immunol. Rev. 2011, 239, 27–44, doi:10.1111/j.1600-065X.2010.00979.x.
[2]  Kreitman, R.J. Immunotoxins for targeted cancer therapy. AAPS J. 2006, 8, E532–E551, doi:10.1208/aapsj080363.
[3]  Vanneman, M.; Dranoff, G. Combining immunotherapy and targeted therapies in cancer treatment. Nat. Rev. Cancer 2012, 12, 237–251, doi:10.1038/nrc3237.
[4]  Waldmann, T.A. Immunotherapy: Past, present and future. Nat. Med. 2003, 9, 269–277, doi:10.1038/nm0303-269.
[5]  Scott, A.M.; Allison, J.P.; Wolchok, J.D. Monoclonal antibodies in cancer therapy. Cancer Immun. 2012, 12, 14.
[6]  Reichert, J.M. Antibody-based therapeutics to watch in 2011. MAbs 2011, 3, 76–99, doi:10.4161/mabs.3.1.13895.
[7]  Schrama, D.; Reisfeld, R.A.; Becker, J.C. Antibody targeted drugs as cancer therapeutics. Nat. Rev. Drug Discov. 2006, 5, 147–59, doi:10.1038/nrd1957.
[8]  Carter, P. Improving the efficacy of antibody-based cancer therapies. Nat. Rev. Cancer 2001, 1, 118–129, doi:10.1038/35101072.
[9]  Rybak, S.M.; Hoogenboom, H.R.; Meade, H.M.; Raus, J.C.; Schwartz, D.; Youle, R.J. Humanization of immunotoxins. Proc. Natl. Acad. Sci. USA 1992, 89, 3165–3169.
[10]  Natsume, A.; Niwa, R.; Satoh, M. Improving effector functions of antibodies for cancer treatment: Enhancing ADCC and CDC. Drug Des. Devel. Ther. 2009, 3, 7–16.
[11]  Carter, P.J.; Senter, P.D. Antibody-drug conjugates for cancer therapy. Cancer J. 2008, 14, 154–169, doi:10.1097/PPO.0b013e318172d704.
[12]  Lambert, J.M. Drug-conjugated monoclonal antibodies for the treatment of cancer. Curr. Opin. Pharmacol. 2005, 5, 543–549, doi:10.1016/j.coph.2005.04.017.
[13]  Younes, A.; Yasothan, U.; Kirkpatrick, P. Brentuximab vedotin. Nat. Rev. Drug Discov. 2012, 11, 19–20, doi:10.1038/nrd3629.
[14]  Bross, P.F.; Beitz, J.; Chen, G.; Chen, X.H.; Duffy, E.; Kieffer, L.; Roy, S.; Sridhara, R.; Rahman, A.; Williams, G.; et al. Approval summary: Gemtuzumab ozogamicin in relapsed acute myeloid leukemia. Clin. Cancer Res. 2001, 7, 1490–1496.
[15]  Kreitman, R.J. Recombinant toxins for the treatment of cancer. Curr. Opin. Mol. Ther. 2003, 5, 44–51.
[16]  Allen, T.M. Ligand-targeted therapeutics in anticancer therapy. Nat. Rev. Cancer 2002, 2, 750–763, doi:10.1038/nrc903.
[17]  Pavlinkova, G.; Beresford, G.W.; Booth, B.J.; Batra, S.K.; Colcher, D. Pharmacokinetics and biodistribution of engineered single-chain antibody constructs of MAb CC49 in colon carcinoma xenografts. J. Nucl. Med. 1999, 40, 1536–1546.
[18]  Pirker, R.; FitzGerald, D.J.; Willingham, M.C.; Pastan, I. Enhancement of the activity of immunotoxins made with either ricin A chain or Pseudomonas exotoxin in human ovarian and epidermoid carcinoma cell lines. Cancer Res. 1988, 48, 3919–3923.
[19]  Barth, S.; Huhn, M.; Matthey, B.; Schnell, R.; Tawadros, S.; Schinkothe, T.; Lorenzen, J.; Diehl, V.; Engert, A. Recombinant anti-CD25 immunotoxin RFT5(SCFV)-ETA' demonstrates successful elimination of disseminated human Hodgkin lymphoma in SCID mice. Int. J. Cancer 2000, 86, 718–724, doi:10.1002/(SICI)1097-0215(20000601)86:5<718::AID-IJC18>3.0.CO;2-N.
[20]  Barth, S.; Huhn, M.; Matthey, B.; Tawadros, S.; Schnell, R.; Schinkothe, T.; Diehl, V.; Engert, A. Ki-4(scFv)-ETA', a new recombinant anti-CD30 immunotoxin with highly specific cytotoxic activity against disseminated Hodgkin tumors in SCID mice. Blood 2000, 95, 3909–3914.
[21]  Schnell, R.; Vitetta, E.; Schindler, J.; Borchmann, P.; Barth, S.; Ghetie, V.; Hell, K.; Drillich, S.; Diehl, V.; Engert, A. Treatment of refractory Hodgkin's lymphoma patients with an anti-CD25 ricin A-chain immunotoxin. Leukemia 2000, 14, 129–135, doi:10.1038/sj.leu.2401626.
[22]  Kreitman, R.J. Recombinant immunotoxins for the treatment of chemoresistant hematologic malignancies. Curr. Pharm. Des. 2009, 15, 2652–2664, doi:10.2174/138161209788923949.
[23]  Kreitman, R.J.; Wilson, W.H.; Bergeron, K.; Raggio, M.; Stetler-Stevenson, M.; FitzGerald, D.J.; Pastan, I. Efficacy of the anti-CD22 recombinant immunotoxin BL22 in chemotherapy-resistant hairy-cell leukemia. N. Engl. J. Med. 2001, 345, 241–247, doi:10.1056/NEJM200107263450402.
[24]  Frankel, A.E.; Powell, B.L.; Hall, P.D.; Case, L.D.; Kreitman, R.J. Phase I trial of a novel diphtheria toxin/granulocyte macrophage colony-stimulating factor fusion protein (DT388GMCSF) for refractory or relapsed acute myeloid leukemia. Clin. Cancer Res. 2002, 8, 1004–1013.
[25]  Szatrowski, T.P.; Dodge, R.K.; Reynolds, C.; Westbrook, C.A.; Frankel, S.R.; Sklar, J.; Stewart, C.C.; Hurd, D.D.; Kolitz, J.E.; Velez-Garcia, E.; et al. Lineage specific treatment of adult patients with acute lymphoblastic leukemia in first remission with anti-B4-blocked ricin or high-dose cytarabine: Cancer and Leukemia Group B Study 9311. Cancer 2003, 97, 1471–1480, doi:10.1002/cncr.11219.
[26]  Pai, L.H.; Pastan, I. Clinical trials with Pseudomonas exotoxin immunotoxins. Curr. Top. Microbiol. Immunol. 1998, 234, 83–96.
[27]  Pastan, I.; Hassan, R.; Fitzgerald, D.J.; Kreitman, R.J. Immunotoxin therapy of cancer. Nat. Rev. Cancer 2006, 6, 559–565, doi:10.1038/nrc1891.
[28]  Pastan, I.; Hassan, R.; FitzGerald, D.J.; Kreitman, R.J. Immunotoxin treatment of cancer. Annu. Rev. Med. 2007, 58, 221–237, doi:10.1146/annurev.med.58.070605.115320.
[29]  Madhumathi, J.; Verma, R.S. Therapeutic targets and recent advances in protein immunotoxins. Curr. Opin. Microbiol. 2012, 15, 300–309, doi:10.1016/j.mib.2012.05.006.
[30]  Becker, N.; Benhar, I. Antibody-Based Immunotoxins for the Treatment of Cancer. Antibodies 2012, 1, 39–69, doi:10.3390/antib1010039.
[31]  Dang, N.H.; Pro, B.; Hagemeister, F.B.; Samaniego, F.; Jones, D.; Samuels, B.I.; Rodriguez, M.A.; Goy, A.; Romaguera, J.E.; McLaughlin, P.; et al. Phase II trial of denileukin diftitox for relapsed/refractory T-cell non-Hodgkin lymphoma. Br. J. Haematol. 2007, 136, 439–447, doi:10.1111/j.1365-2141.2006.06457.x.
[32]  Mathew, M.; Verma, R.S. Humanized immunotoxins: A new generation of immunotoxins for targeted cancer therapy. Cancer Sci. 2009, 100, 1359–1365, doi:10.1111/j.1349-7006.2009.01192.x.
[33]  Tur, M.K.; Neef, I.; Jager, G.; Teubner, A.; Stocker, M.; Melmer, G.; Barth, S. Immunokinases, a novel class of immunotherapeutics for targeted cancer therapy. Curr. Pharm. Des. 2009, 15, 2693–2699, doi:10.2174/138161209788923877.
[34]  Huhn, M.; Sasse, S.; Tur, M.K.; Matthey, B.; Schinkothe, T.; Rybak, S.M.; Barth, S.; Engert, A. Human angiogenin fused to human CD30 ligand (Ang-CD30L) exhibits specific cytotoxicity against CD30-positive lymphoma. Cancer Res. 2001, 61, 8737–8742.
[35]  Rosenblum, M.G.; Barth, S. Development of novel, highly cytotoxic fusion constructs containing granzyme B: Unique mechanisms and functions. Curr. Pharm. Des. 2009, 15, 2676–2692, doi:10.2174/138161209788923958.
[36]  von Minckwitz, G.; Harder, S.; Hovelmann, S.; Jager, E.; Al-Batran, S.E.; Loibl, S.; Atmaca, A.; Cimpoiasu, C.; Neumann, A.; Abera, A.; et al. Phase I clinical study of the recombinant antibody toxin scFv(FRP5)-ETA specific for the ErbB2/HER2 receptor in patients with advanced solid malignomas. Breast Cancer Res. 2005, 7, R617–R626, doi:10.1186/bcr1264.
[37]  Engert, A.; Diehl, V.; Schnell, R.; Radszuhn, A.; Hatwig, M.T.; Drillich, S.; Schon, G.; Bohlen, H.; Tesch, H.; Hansmann, M.L.; et al. A phase-I study of an anti-CD25 ricin A-chain immunotoxin (RFT5-SMPT-dgA) in patients with refractory Hodgkin's lymphoma. Blood 1997, 89, 403–410.
[38]  Hall, P.D.; Virella, G.; Willoughby, T.; Atchley, D.H.; Kreitman, R.J.; Frankel, A.E. Antibody response to DT-GM, a novel fusion toxin consisting of a truncated diphtheria toxin (DT) linked to human granulocyte-macrophage colony stimulating factor (GM), during a phase I trial of patients with relapsed or refractory acute myeloid leukemia. Clin. Immunol. 2001, 100, 191–17, doi:10.1006/clim.2001.5066.
[39]  Posey, J.A.; Khazaeli, M.B.; Bookman, M.A.; Nowrouzi, A.; Grizzle, W.E.; Thornton, J.; Carey, D.E.; Lorenz, J.M.; Sing, A.P.; Siegall, C.B.; et al. A phase I trial of the single-chain immunotoxin SGN-10 (BR96 sFv-PE40) in patients with advanced solid tumors. Clin. Cancer Res. 2002, 8, 3092–3099.
[40]  Scadden, D.T.; Schenkein, D.P.; Bernstein, Z.; Luskey, B.; Doweiko, J.; Tulpule, A.; Levine, A.M. Immunotoxin combined with chemotherapy for patients with AIDS-related non-Hodgkin's lymphoma. Cancer 1998, 83, 2580–2587, doi:10.1002/(SICI)1097-0142(19981215)83:12<2580::AID-CNCR25>3.0.CO;2-C.
[41]  Mischak, R.P.; Foxall, C.; Rosendorf, L.L.; Knebel, K.; Scannon, P.J.; Spitler, L.E. Human antibody responses to components of the monoclonal antimelanoma antibody ricin A chain immunotoxin XomaZyme-MEL. Mol. Biother. 1990, 2, 104–109.
[42]  Tur, M.K.; Neef, I.; Jost, E.; Galm, O.; Jager, G.; Stocker, M.; Ribbert, M.; Osieka, R.; Klinge, U.; Barth, S. Targeted restoration of down-regulated DAPK2 tumor suppressor activity induces apoptosis in Hodgkin lymphoma cells. J. Immunother. 2009, 32, 431–441, doi:10.1097/CJI.0b013e31819f1cb6.
[43]  Hwang, W.Y.; Foote, J. Immunogenicity of engineered antibodies. Methods 2005, 36, 3–10, doi:10.1016/j.ymeth.2005.01.001.
[44]  Boulianne, G.L.; Hozumi, N.; Shulman, M.J. Production of functional chimaeric mouse/human antibody. Nature 1984, 312, 643–646, doi:10.1038/312643a0.
[45]  Morrison, S.L.; Johnson, M.J.; Herzenberg, L.A.; Oi, V.T. Chimeric human antibody molecules: Mouse antigen-binding domains with human constant region domains. Proc. Natl. Acad. Sci. USA 1984, 81, 6851–6855.
[46]  Jones, P.T.; Dear, P.H.; Foote, J.; Neuberger, M.S.; Winter, G. Replacing the complementarity-determining regions in a human antibody with those from a mouse. Nature 1986, 321, 522–525, doi:10.1038/321522a0.
[47]  Verhoeyen, M.; Milstein, C.; Winter, G. Reshaping human antibodies: Grafting an antilysozyme activity. Science 1988, 239, 1534–1536.
[48]  Hu, W.G.; Yin, J.; Chau, D.; Negrych, L.M.; Cherwonogrodzky, J.W. Humanization and characterization of an anti-ricin neutralization monoclonal antibody. PLoS One 2012, 7, e45595.
[49]  Lonberg, N. Fully human antibodies from transgenic mouse and phage display platforms. Curr. Opin. Immunol. 2008, 20, 450–459, doi:10.1016/j.coi.2008.06.004.
[50]  Menoret, S.; Iscache, A.L.; Tesson, L.; Remy, S.; Usal, C.; Osborn, M.J.; Cost, G.J.; Bruggemann, M.; Buelow, R.; Anegon, I. Characterization of immunoglobulin heavy chain knockout rats. Eur. J. Immunol. 2010, 40, 2932–2941, doi:10.1002/eji.201040939.
[51]  Hansen, J.K.; Weldon, J.E.; Xiang, L.; Beers, R.; Onda, M.; Pastan, I. A recombinant immunotoxin targeting CD22 with low immunogenicity, low nonspecific toxicity, and high antitumor activity in mice. J. Immunother. 2010, 33, 297–304, doi:10.1097/CJI.0b013e3181cd1164.
[52]  Liu, W.; Onda, M.; Lee, B.; Kreitman, R.J.; Hassan, R.; Xiang, L.; Pastan, I. Recombinant immunotoxin engineered for low immunogenicity and antigenicity by identifying and silencing human B-cell epitopes. Proc. Natl. Acad. Sci. USA 2012, 109, 11782–11787.
[53]  Tsutsumi, Y.; Onda, M.; Nagata, S.; Lee, B.; Kreitman, R.J.; Pastan, I. Site-specific chemical modification with polyethylene glycol of recombinant immunotoxin anti-Tac(Fv)-PE38 (LMB-2) improves antitumor activity and reduces animal toxicity and immunogenicity. Proc. Natl. Acad. Sci. USA 2000, 97, 8548–8553.
[54]  Oratz, R.; Speyer, J.L.; Wernz, J.C.; Hochster, H.; Meyers, M.; Mischak, R.; Spitler, L.E. Antimelanoma monoclonal antibody-ricin A chain immunoconjugate (XMMME-001-RTA) plus cyclophosphamide in the treatment of metastatic malignant melanoma: Results of a phase II trial. J. Biol. Response Mod. 1990, 9, 345–354.
[55]  Siegall, C.B.; Haggerty, H.G.; Warner, G.L.; Chace, D.; Mixan, B.; Linsley, P.S.; Davidson, T. Prevention of immunotoxin-induced immunogenicity by coadministration with CTLA4Ig enhances antitumor efficacy. J. Immunol. 1997, 159, 5168–5173.
[56]  Igney, F.H.; Krammer, P.H. Immune escape of tumors: Apoptosis resistance and tumor counterattack. J. Leuk. Biol. 2002, 71, 907–920.
[57]  Vesely, M.D.; Kershaw, M.H.; Schreiber, R.D.; Smyth, M.J. Natural innate and adaptive immunity to cancer. Annu. Rev. Immunol. 2011, 29, 235–271, doi:10.1146/annurev-immunol-031210-101324.
[58]  Griffith, T.S.; Chin, W.A.; Jackson, G.C.; Lynch, D.H.; Kubin, M.Z. Intracellular regulation of TRAIL-induced apoptosis in human melanoma cells. J. Immunol. 1998, 161, 2833–2840.
[59]  Sutton, V.R.; Wowk, M.E.; Cancilla, M.; Trapani, J.A. Caspase activation by granzyme B is indirect, and caspase autoprocessing requires the release of proapoptotic mitochondrial factors. Immunity 2003, 18, 319–329, doi:10.1016/S1074-7613(03)00050-5.
[60]  Campos, L.; Rouault, J.P.; Sabido, O.; Oriol, P.; Roubi, N.; Vasselon, C.; Archimbaud, E.; Magaud, J.P.; Guyotat, D. High expression of bcl-2 protein in acute myeloid leukemia cells is associated with poor response to chemotherapy. Blood 1993, 81, 3091–3096.
[61]  Ambrosini, G.; Adida, C.; Altieri, D.C. A novel anti-apoptosis gene, survivin, expressed in cancer and lymphoma. Nat. Med. 1997, 3, 917–921, doi:10.1038/nm0897-917.
[62]  Russell, J.H.; Ley, T.J. Lymphocyte-mediated cytotoxicity. Annu. Rev. Immunol. 2002, 20, 323–370, doi:10.1146/annurev.immunol.20.100201.131730.
[63]  Boivin, W.A.; Cooper, D.M.; Hiebert, P.R.; Granville, D.J. Intracellular versus extracellular granzyme B in immunity and disease: Challenging the dogma. Lab. Invest. 2009, 89, 1195–1220, doi:10.1038/labinvest.2009.91.
[64]  Raja, S.M.; Wang, B.; Dantuluri, M.; Desai, U.R.; Demeler, B.; Spiegel, K.; Metkar, S.S.; Froelich, C.J. Cytotoxic cell granule-mediated apoptosis. Characterization of the macromolecular complex of granzyme B with serglycin. J. Biol. Chem. 2002, 277, 49523–49530.
[65]  Grossman, W.J.; Revell, P.A.; Lu, Z.H.; Johnson, H.; Bredemeyer, A.J.; Ley, T.J. The orphan granzymes of humans and mice. Curr. Opin. Immunol. 2003, 15, 544–552, doi:10.1016/S0952-7915(03)00099-2.
[66]  Bots, M.; Medema, J.P. Granzymes at a glance. J. Cell Sci. 2006, 119, 5011–5014, doi:10.1242/jcs.03239.
[67]  Galvin, J.P.; Spaeny-Dekking, L.H.; Wang, B.; Seth, P.; Hack, C.E.; Froelich, C.J. Apoptosis induced by granzyme B-glycosaminoglycan complexes: Implications for granule-mediated apoptosis in vivo. J. Immunol. 1999, 162, 5345–5350.
[68]  Grujic, M.; Braga, T.; Lukinius, A.; Eloranta, M.L.; Knight, S.D.; Pejler, G.; Abrink, M. Serglycin-deficient cytotoxic T lymphocytes display defective secretory granule maturation and granzyme B storage. J. Biol. Chem. 2005, 280, 33411–33418.
[69]  Lanier, L.L. NK cell recognition. Annu. Rev. Immunol. 2005, 23, 225–274, doi:10.1146/annurev.immunol.23.021704.115526.
[70]  Kurschus, F.C.; Jenne, D.E. Delivery and therapeutic potential of human granzyme B. Immunol. Rev. 2010, 235, 159–171.
[71]  Andrade, F.; Casciola-Rosen, L.A.; Rosen, A. Granzyme B-induced cell death. Acta Haematol. 2004, 111, 28–41, doi:10.1159/000074484.
[72]  Heibein, J.A.; Goping, I.S.; Barry, M.; Pinkoski, M.J.; Shore, G.C.; Green, D.R.; Bleackley, R.C. Granzyme B-mediated cytochrome c release is regulated by the Bcl-2 family members bid and Bax. J. Exp. Med. 2000, 192, 1391–1402, doi:10.1084/jem.192.10.1391.
[73]  Pinkoski, M.J.; Waterhouse, N.J.; Heibein, J.A.; Wolf, B.B.; Kuwana, T.; Goldstein, J.C.; Newmeyer, D.D.; Bleackley, R.C.; Green, D.R. Granzyme B-mediated apoptosis proceeds predominantly through a Bcl-2-inhibitable mitochondrial pathway. J. Biol. Chem. 2001, 276, 12060–12067.
[74]  Metkar, S.S.; Wang, B.; Ebbs, M.L.; Kim, J.H.; Lee, Y.J.; Raja, S.M.; Froelich, C.J. Granzyme B activates procaspase-3 which signals a mitochondrial amplification loop for maximal apoptosis. J. Cell Biol. 2003, 160, 875–885, doi:10.1083/jcb.200210158.
[75]  Talanian, R.V.; Yang, X.; Turbov, J.; Seth, P.; Ghayur, T.; Casiano, C.A.; Orth, K.; Froelich, C.J. Granule-mediated killing: Pathways for granzyme B-initiated apoptosis. J. Exp. Med. 1997, 186, 1323–1331, doi:10.1084/jem.186.8.1323.
[76]  Waterhouse, N.J.; Sedelies, K.A.; Trapani, J.A. Role of Bid-induced mitochondrial outer membrane permeabilization in granzyme B-induced apoptosis. Immunol. Cell Biol. 2006, 84, 72–78, doi:10.1111/j.1440-1711.2005.01416.x.
[77]  Hengartner, M.O. The biochemistry of apoptosis. Nature 2000, 407, 770–776, doi:10.1038/35037710.
[78]  Stahnke, B.; Thepen, T.; Stocker, M.; Rosinke, R.; Jost, E.; Fischer, R.; Tur, M.K.; Barth, S. Granzyme B-H22(scFv), a human immunotoxin targeting CD64 in acute myeloid leukemia of monocytic subtypes. Mol. Cancer Ther. 2008, 7, 2924–2932, doi:10.1158/1535-7163.MCT-08-0554.
[79]  Schiffer, S. Fraunhofer Institute for Molecular Biology and Applied Ecology IME. unpublished work, Aachen, Germany, 2012.
[80]  Liu, Y.; Cheung, L.H.; Hittelman, W.N.; Rosenblum, M.G. Targeted delivery of human pro-apoptotic enzymes to tumor cells: In vitro studies describing a novel class of recombinant highly cytotoxic agents. Mol. Cancer Ther. 2003, 2, 1341–1350.
[81]  Liu, Y.; Zhang, W.; Niu, T.; Cheung, L.H.; Munshi, A.; Meyn, R.E., Jr.; Rosenblum, M.G. Targeted apoptosis activation with GrB/scFvMEL modulates melanoma growth, metastatic spread, chemosensitivity, and radiosensitivity. Neoplasia 2006, 8, 125–135, doi:10.1593/neo.05556.
[82]  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.
[83]  Medema, J.P.; de Jong, J.; Peltenburg, L.T.; Verdegaal, E.M.; Gorter, A.; Bres, S.A.; Franken, K.L.; Hahne, M.; Albar, J.P.; Melief, C.J.; et al. Blockade of the granzyme B/perforin pathway through overexpression of the serine protease inhibitor PI-9/SPI-6 constitutes a mechanism for immune escape by tumors. Proc. Natl. Acad. Sci. USA 2001, 98, 11515–11520.
[84]  Jiang, X.; Ellison, S.J.; Alarid, E.T.; Shapiro, D.J. Interplay between the levels of estrogen and estrogen receptor controls the level of the granzyme inhibitor, proteinase inhibitor 9 and susceptibility to immune surveillance by natural killer cells. Oncogene 2007, 26, 4106–4114, doi:10.1038/sj.onc.1210197.
[85]  Kurschus, F.C.; Kleinschmidt, M.; Fellows, E.; Dornmair, K.; Rudolph, R.; Lilie, H.; Jenne, D.E. Killing of target cells by redirected granzyme B in the absence of perforin. FEBS Lett. 2004, 562, 87–92, doi:10.1016/S0014-5793(04)00187-5.
[86]  Liu, Y.; Cheung, L.H.; Thorpe, P.; Rosenblum, M.G. Mechanistic studies of a novel human fusion toxin composed of vascular endothelial growth factor (VEGF)121 and the serine protease granzyme B: Directed apoptotic events in vascular endothelial cells. Mol. Cancer Ther. 2003, 2, 949–959.
[87]  Zhang, L.; Zhao, J.; Wang, T.; Yu, C.J.; Jia, L.T.; Duan, Y.Y.; Yao, L.B.; Chen, S.Y.; Yang, A.G. HER2-targeting recombinant protein with truncated pseudomonas exotoxin A translocation domain efficiently kills breast cancer cells. Cancer Biol. Ther. 2008, 7, 1226–1231.
[88]  Wang, T.; Zhao, J.; Ren, J.L.; Zhang, L.; Wen, W.H.; Zhang, R.; Qin, W.W.; Jia, L.T.; Yao, L.B.; Zhang, Y.Q.; et al. Recombinant immunoproapoptotic proteins with furin site can translocate and kill HER2-positive cancer cells. Cancer Res. 2007, 67, 11830–11839.
[89]  Kanatani, I.; Lin, X.; Yuan, X.; Manorek, G.; Shang, X.; Cheung, L.H.; Rosenblum, M.G.; Howell, S.B. Targeting granzyme B to tumor cells using a yoked human chorionic gonadotropin. Cancer Chemother. Pharmacol. 2011, 68, 979–990, doi:10.1007/s00280-011-1573-4.
[90]  Hegde, R.; Srinivasula, S.M.; Zhang, Z.; Wassell, R.; Mukattash, R.; Cilenti, L.; DuBois, G.; Lazebnik, Y.; Zervos, A.S.; Fernandes-Alnemri, T.; et al. Identification of Omi/HtrA2 as a mitochondrial apoptotic serine protease that disrupts inhibitor of apoptosis protein-caspase interaction. J. Biol. Chem. 2002, 277, 432–438.
[91]  Liu, X.; Kim, C.N.; Yang, J.; Jemmerson, R.; Wang, X. Induction of apoptotic program in cell-free extracts: Requirement for dATP and cytochrome c. Cell 1996, 86, 147–157, doi:10.1016/S0092-8674(00)80085-9.
[92]  Han, J.; Goldstein, L.A.; Gastman, B.R.; Froelich, C.J.; Yin, X.M.; Rabinowich, H. Degradation of Mcl-1 by granzyme B: Implications for Bim-mediated mitochondrial apoptotic events. J. Biol. Chem. 2004, 279, 22020–22029.
[93]  Alimonti, J.B.; Shi, L.; Baijal, P.K.; Greenberg, A.H. Granzyme B induces BID-mediated cytochrome c release and mitochondrial permeability transition. J. Biol. Chem. 2001, 276, 6974–6982, doi:10.1074/jbc.M008444200.
[94]  Waterhouse, N.J.; Sedelies, K.A.; Browne, K.A.; Wowk, M.E.; Newbold, A.; Sutton, V.R.; Clarke, C.J.; Oliaro, J.; Lindemann, R.K.; Bird, P.I.; et al. A central role for Bid in granzyme B-induced apoptosis. J. Biol. Chem. 2005, 280, 4476–4482.
[95]  Cullen, S.P.; Adrain, C.; Luthi, A.U.; Duriez, P.J.; Martin, S.J. Human and murine granzyme B exhibit divergent substrate preferences. J. Cell Biol. 2007, 176, 435–444, doi:10.1083/jcb.200612025.
[96]  Chowdhury, D.; Lieberman, J. Death by a thousand cuts: Granzyme pathways of programmed cell death. Annu. Rev. Immunol. 2008, 26, 389–420, doi:10.1146/annurev.immunol.26.021607.090404.
[97]  Trapani, J.A.; Sutton, V.R. Granzyme B: Pro-apoptotic, antiviral and antitumor functions. Curr. Opin. Immunol. 2003, 15, 533–543, doi:10.1016/S0952-7915(03)00107-9.
[98]  Bleackley, R.C. A molecular view of cytotoxic T lymphocyte induced killing. Biochem. Cell Biol. 2005, 83, 747–51, doi:10.1139/o05-146.
[99]  Sun, J.; Bird, C.H.; Sutton, V.; McDonald, L.; Coughlin, P.B.; De Jong, T.A.; Trapani, J.A.; Bird, P.I. A cytosolic granzyme B inhibitor related to the viral apoptotic regulator cytokine response modifier A is present in cytotoxic lymphocytes. J. Biol. Chem. 1996, 271, 27802–27809.
[100]  Poe, M.; Blake, J.T.; Boulton, D.A.; Gammon, M.; Sigal, N.H.; Wu, J.K.; Zweerink, H.J. Human cytotoxic lymphocyte granzyme B. Its purification from granules and the characterization of substrate and inhibitor specificity. J. Biol. Chem. 1991, 266, 98–103.
[101]  Zhao, J.; Zhang, L.H.; Jia, L.T.; Zhang, L.; Xu, Y.M.; Wang, Z.; Yu, C.J.; Peng, W.D.; Wen, W.H.; Wang, C.J.; et al. Secreted antibody/granzyme B fusion protein stimulates selective killing of HER2-overexpressing tumor cells. J. Biol. Chem. 2004, 279, 21343–21348.
[102]  Caputo, A.; Garner, R.S.; Winkler, U.; Hudig, D.; Bleackley, R.C. Activation of recombinant murine cytotoxic cell proteinase-1 requires deletion of an amino-terminal dipeptide. J. Biol. Chem. 1993, 268, 17672–17675.
[103]  Dalken, B.; Jabulowsky, R.A.; Oberoi, P.; Benhar, I.; Wels, W.S. Maltose-binding protein enhances secretion of recombinant human granzyme B accompanied by in vivo processing of a precursor MBP fusion protein. PLoS One 2010, 5, e14404.
[104]  Kam, C.M.; Hudig, D.; Powers, J.C. Granzymes (lymphocyte serine proteases): Characterization with natural and synthetic substrates and inhibitors. Biochim. Biophys. Acta 2000, 1477, 307–323, doi:10.1016/S0167-4838(99)00282-4.
[105]  Griffiths, G.M.; Isaaz, S. Granzymes A and B are targeted to the lytic granules of lymphocytes by the mannose-6-phosphate receptor. J. Cell Biol. 1993, 120, 885–896, doi:10.1083/jcb.120.4.885.
[106]  Edwards, K.M.; Davis, J.E.; Browne, K.A.; Sutton, V.R.; Trapani, J.A. Anti-viral strategies of cytotoxic T lymphocytes are manifested through a variety of granule-bound pathways of apoptosis induction. Immunol. Cell Biol. 1999, 77, 76–89, doi:10.1046/j.1440-1711.1999.00799.x.
[107]  Giesubel, U.; Dalken, B.; Mahmud, F.; Wels, W.S. Cell binding, internalization and cytotoxic activity of human granzyme B expressed in the yeast Pichia pastoris. Biochem. J. 2006, 394, 563–573, doi:10.1042/BJ20050687.
[108]  Xia, Z.; Kam, C.M.; Huang, C.; Powers, J.C.; Mandle, R.J.; Stevens, R.L.; Lieberman, J. Expression and purification of enzymatically active recombinant granzyme B in a baculovirus system. Biochem. Biophys. Res. Commun. 1998, 243, 384–389, doi:10.1006/bbrc.1998.8102.
[109]  Smyth, M.J.; McGuire, M.J.; Thia, K.Y. Expression of recombinant human granzyme B. A processing and activation role for dipeptidyl peptidase I. J. Immunol. 1995, 154, 6299–6305.
[110]  Godal, R.; Keilholz, U.; Uharek, L.; Letsch, A.; Asemissen, A.M.; Busse, A.; Na, I.K.; Thiel, E.; Scheibenbogen, C. Lymphomas are sensitive to perforin-dependent cytotoxic pathways despite expression of PI-9 and overexpression of bcl-2. Blood 2006, 107, 3205–3211, doi:10.1182/blood-2005-07-2880.
[111]  Soriano, C.; Mukaro, V.; Hodge, G.; Ahern, J.; Holmes, M.; Jersmann, H.; Moffat, D.; Meredith, D.; Jurisevic, C.; Reynolds, P.N.; et al. Increased proteinase inhibitor-9 (PI-9) and reduced granzyme B in lung cancer: Mechanism for immune evasion? Lung Cancer 2012, 77, 38–45, doi:10.1016/j.lungcan.2012.01.017.
[112]  Ray, M.; Hostetter, D.R.; Loeb, C.R.; Simko, J.; Craik, C.S. Inhibition of Granzyme B by PI-9 protects prostate cancer cells from apoptosis. Prostate 2012, 72, 846–855, doi:10.1002/pros.21486.
[113]  Rousalova, I.; Krepela, E.; Prochazka, J.; Cermak, J.; Benkova, K. Expression of proteinase inhibitor-9/serpinB9 in non-small cell lung carcinoma cells and tissues. Int. J. Oncol. 2010, 36, 275–283.
[114]  Jiang, X.G.; Patterson, N.M.; Ling, Y.; Xie, J.W.; Helferich, W.G.; Shapiro, D.J. Low Concentrations of the Soy Phytoestrogen Genistein Induce Proteinase Inhibitor 9 and Block Killing of Breast Cancer Cells by Immune Cells. Endocrinology 2008, 149, 5366–5373, doi:10.1210/en.2008-0857.
[115]  van Houdt, I.S.; Oudejans, J.J.; van den Eertwegh, A.J.; Baars, A.; Vos, W.; Bladergroen, B.A.; Rimoldi, D.; Muris, J.J.; Hooijberg, E.; Gundy, C.M.; et al. Expression of the apoptosis inhibitor protease inhibitor 9 predicts clinical outcome in vaccinated patients with stage III and IV melanoma. Clin. Cancer Res. 2005, 11, 6400–6407.
[116]  Oudejans, J.J.; Harijadi, H.; Kummer, J.A.; Tan, I.B.; Bloemena, E.; Middeldorp, J.M.; Bladergroen, B.; Dukers, D.F.; Vos, W.; Meijer, C.J. High numbers of granzyme B/CD8-positive tumour-infiltrating lymphocytes in nasopharyngeal carcinoma biopsies predict rapid fatal outcome in patients treated with curative intent. J. Pathol. 2002, 198, 468–475, doi:10.1002/path.1236.
[117]  Bots, M.; Liesbeth, V.A.N.B.; Rademaker, M.T.; Offringa, R.; Medema, J.P. Serpins prevent granzyme-induced death in a species-specific manner. Immunol. Cell Biol. 2006, 84, 79–86, doi:10.1111/j.1440-1711.2005.01417.x.
[118]  Huntington, J.A.; Read, R.J.; Carrell, R.W. Structure of a serpin-protease complex shows inhibition by deformation. Nature 2000, 407, 923–926, doi:10.1038/35038119.
[119]  Bladergroen, B.A.; Strik, M.C.; Bovenschen, N.; van Berkum, O.; Scheffer, G.L.; Meijer, C.J.; Hack, C.E.; Kummer, J.A. The granzyme B inhibitor, protease inhibitor 9, is mainly expressed by dendritic cells and at immune-privileged sites. J. Immunol. 2001, 166, 3218–3225.
[120]  Buzza, M.S.; Hosking, P.; Bird, P.I. The granzyme B inhibitor, PI-9, is differentially expressed during placental development and up-regulated in hydatidiform moles. Placenta 2006, 27, 62–69, doi:10.1016/j.placenta.2004.11.009.
[121]  Kannan-Thulasiraman, P.; Shapiro, D.J. Modulators of inflammation use nuclear factor-kappa B and activator protein-1 sites to induce the caspase-1 and granzyme B inhibitor, proteinase inhibitor 9. J. Biol. Chem. 2002, 277, 41230–41239, doi:10.1074/jbc.M200379200.
[122]  El Haddad, N.; Moore, R.; Heathcote, D.; Mounayar, M.; Azzi, J.; Mfarrej, B.; Batal, I.; Ting, C.; Atkinson, M.; Sayegh, M.H.; et al. The novel role of SERPINB9 in cytotoxic protection of human mesenchymal stem cells. J. Immunol. 2011, 187, 2252–2260, doi:10.4049/jimmunol.1003981.
[123]  Heutinck, K.M.; Kassies, J.; Florquin, S.; Ten Berge, I.J.; Hamann, J.; Rowshani, A.T. SerpinB9 expression in human renal tubular epithelial cells is induced by triggering of the viral dsRNA sensors TLR3, MDA5 and RIG-I. Nephrol. Dial. Transplant. 2012, 27, 2746–2754, doi:10.1093/ndt/gfr690.
[124]  Rowshani, A.T.; Strik, M.C.; Molenaar, R.; Yong, S.L.; Wolbink, A.M.; Bemelman, F.J.; Hack, C.E.; Ten Berge, I.J. The granzyme B inhibitor SERPINB9 (protease inhibitor 9) circulates in blood and increases on primary cytomegalovirus infection after renal transplantation. J. Infect. Dis. 2005, 192, 1908–1911, doi:10.1086/497606.
[125]  Classen, C.F.; Bird, P.I.; Debatin, K.M. Modulation of the granzyme B inhibitor proteinase inhibitor 9 (PI-9) by activation of lymphocytes and monocytes in vitro and by Epstein-Barr virus and bacterial infection. Clin. Exp. Immunol. 2006, 143, 534–542, doi:10.1111/j.1365-2249.2006.03006.x.
[126]  Buzza, M.S.; Hirst, C.E.; Bird, C.H.; Hosking, P.; McKendrick, J.; Bird, P.I. The granzyme B inhibitor, PI-9, is present in endothelial and mesothelial cells, suggesting that it protects bystander cells during immune respons. Cell. Immunol. 2001, 210, 21–29, doi:10.1006/cimm.2001.1806.
[127]  Barrie, M.B.; Stout, H.W.; Abougergi, M.S.; Miller, B.C.; Thiele, D.L. Antiviral cytokines induce hepatic expression of the granzyme B inhibitors, proteinase inhibitor 9 and serine proteinase inhibitor 6. J. Immunol. 2004, 172, 6453–6459.
[128]  Horie, O.; Saigo, K.; Murayama, T.; Ryo, R. Differential expression of proteinase inhibitor-9 and granzyme B mRNAs in activated immunocompetent cells. Tohoku J. Exp. Med. 2005, 205, 103–113, doi:10.1620/tjem.205.103.
[129]  Hirst, C.E.; Buzza, M.S.; Bird, C.H.; Warren, H.S.; Cameron, P.U.; Zhang, M.; Ashton-Rickardt, P.G.; Bird, P.I. The intracellular granzyme B inhibitor, proteinase inhibitor 9, is up-regulated during accessory cell maturation and effector cell degranulation, and its overexpression enhances CTL potency. J. Immunol. 2003, 170, 805–815.
[130]  Rowshani, A.T.; Florquin, S.; Bemelman, F.; Kummer, J.A.; Hack, C.E.; Ten Berge, I.J. Hyperexpression of the granzyme B inhibitor PI-9 in human renal allografts: A potential mechanism for stable renal function in patients with subclinical rejection. Kidney Int. 2004, 66, 1417–1422, doi:10.1111/j.1523-1755.2004.00903.x.
[131]  Walker, P.R.; Saas, P.; Dietrich, P.Y. Role of Fas ligand (CD95L) in immune escape: The tumor cell strikes back. J. Immunol. 1997, 158, 4521–4524.
[132]  Buehring, G.C.; Eby, E.A.; Eby, M.J. Cell line cross-contamination: How aware are Mammalian cell culturists of the problem and how to monitor it? In Vitro Cell. Dev. Biol. Anim. 2004, 40, 211–215, doi:10.1290/1543-706X(2004)40<211:CLCHAA>2.0.CO;2.
[133]  Bird, C.H.; Sutton, V.R.; Sun, J.; Hirst, C.E.; Novak, A.; Kumar, S.; Trapani, J.A.; Bird, P.I. Selective regulation of apoptosis: The cytotoxic lymphocyte serpin proteinase inhibitor 9 protects against granzyme B-mediated apoptosis without perturbing the Fas cell death pathway. Mol. Cell. Biol. 1998, 18, 6387–6398.
[134]  Schiffer, S. Efficacy of an adapted Granzyme B-based anti-CD30 cytolytic fusion protein against PI-9-positive classical Hodgkin lymphoma cells in a murine model. Blood Cancer J. 2012. submitted for publication.
[135]  ten Berge, R.L.; Meijer, C.J.; Dukers, D.F.; Kummer, J.A.; Bladergroen, B.A.; Vos, W.; Hack, C.E.; Ossenkoppele, G.J.; Oudejans, J.J. Expression levels of apoptosis-related proteins predict clinical outcome in anaplastic large cell lymphoma. Blood 2002, 99, 4540–4546, doi:10.1182/blood.V99.12.4540.
[136]  Tiacci, E.; Doring, C.; Brune, V.; van Noesel, C.J.; Klapper, W.; Mechtersheimer, G.; Falini, B.; Kuppers, R.; Hansmann, M.L. Analyzing primary Hodgkin and Reed-Sternberg cells to capture the molecular and cellular pathogenesis of classical Hodgkin lymphoma. Blood 2012, 120, 4609–4620, doi:10.1182/blood-2012-05-428896.
[137]  Fritsch, K. Die Bedeutung des Proteinase Inhibitor 9 für die Apoptoseresistenz in leuk?mischen Zelllinien. Ph.D. Thesis, Albert-Ludwigs-University, Freiburg im Breisgau.
[138]  Bossard, C.; Belhadj, K.; Reyes, F.; Martin-Garcia, N.; Berger, F.; Kummer, J.A.; Briere, J.; Baglin, A.C.; Cheze, S.; Bosq, J.; et al. Expression of the granzyme B inhibitor PI9 predicts outcome in nasal NK/T-cell lymphoma: Results of a Western series of 48 patients treated with first-line polychemotherapy within the Groupe d'Etude des Lymphomes de l'Adulte (GELA) trials. Blood 2007, 109, 2183–2189, doi:10.1182/blood-2006-07-033142.
[139]  Bladergroen, B.A.; Meijer, C.J.; ten Berge, R.L.; Hack, C.E.; Muris, J.J.; Dukers, D.F.; Chott, A.; Kazama, Y.; Oudejans, J.J.; van Berkum, O.; et al. Expression of the granzyme B inhibitor, protease inhibitor 9, by tumor cells in patients with non-Hodgkin and Hodgkin lymphoma: A novel protective mechanism for tumor cells to circumvent the immune system? Blood 2002, 99, 232–237, doi:10.1182/blood.V99.1.232.
[140]  Mahrus, S.; Kisiel, W.; Craik, C.S. Granzyme M is a regulatory protease that inactivates proteinase inhibitor 9, an endogenous inhibitor of granzyme B. J. Biol. Chem. 2004, 279, 54275–54282.
[141]  Madison, E.L.; Goldsmith, E.J.; Gerard, R.D.; Gething, M.J.; Sambrook, J.F. Serpin-resistant mutants of human tissue-type plasminogen activator. Nature 1989, 339, 721–724, doi:10.1038/339721a0.
[142]  Potempa, J.; Korzus, E.; Travis, J. The serpin superfamily of proteinase inhibitors: Structure, function, and regulation. J. Biol. Chem. 1994, 269, 15957–15960.
[143]  Marszal, E.; Shrake, A. Serpin crystal structure and serpin polymer structure. Arch. Biochem. Biophys. 2006, 453, 123–129, doi:10.1016/j.abb.2006.03.006.
[144]  Silverman, G.A.; Bird, P.I.; Carrell, R.W.; Church, F.C.; Coughlin, P.B.; Gettins, P.G.; Irving, J.A.; Lomas, D.A.; Luke, C.J.; Moyer, R.W.; et al. The serpins are an expanding superfamily of structurally similar but functionally diverse proteins. Evolution, mechanism of inhibition, novel functions, and a revised nomenclature. J. Biol. Chem. 2001, 276, 33293–33296.
[145]  Sun, J.; Whisstock, J.C.; Harriott, P.; Walker, B.; Novak, A.; Thompson, P.E.; Smith, A.I.; Bird, P.I. Importance of the P4' residue in human granzyme B inhibitors and substrates revealed by scanning mutagenesis of the proteinase inhibitor 9 reactive center loop. J. Biol. Chem. 2001, 276, 15177–15184.
[146]  Hedstrom, L. Serine protease mechanism and specificity. Chem. Rev. 2002, 102, 4501–4524, doi:10.1021/cr000033x.
[147]  Ye, S.; Cech, A.L.; Belmares, R.; Bergstrom, R.C.; Tong, Y.; Corey, D.R.; Kanost, M.R.; Goldsmith, E.J. The structure of a Michaelis serpin-protease complex. Nat. Struct. Biol. 2001, 8, 979–983.
[148]  Rotonda, J.; Garcia-Calvo, M.; Bull, H.G.; Geissler, W.M.; McKeever, B.M.; Willoughby, C.A.; Thornberry, N.A.; Becker, J.W. The three-dimensional structure of human granzyme B compared to caspase-3, key mediators of cell death with cleavage specificity for aspartic acid in P1. Chem. Biol. 2001, 8, 357–368.
[149]  Kortemme, T.; Baker, D. A simple physical model for binding energy hot spots in protein-protein complexes. Proc. Natl. Acad. Sci. USA 2002, 99, 14116–14121, doi:10.1073/pnas.202485799.
[150]  Losasso, V.; Schiffer, S.; Barth, S.; Carloni, P. Design of human granzyme B variants resistant to serpin B9. Proteins 2012, 80, 2514–2522.
[151]  Jabulowsky, R.A.; Oberoi, P.; Bahr-Mahmud, H.; Dalken, B.; Wels, W.S. Surface Charge-Modification Prevents Sequestration and Enhances Tumor-Cell Specificity of a Recombinant Granzyme B-TGF alpha Fusion Protein. Bioconjug. Chem. 2012, 23, 1567–1576.
[152]  Motyka, B.; Korbutt, G.; Pinkoski, M.J.; Heibein, J.A.; Caputo, A.; Hobman, M.; Barry, M.; Shostak, I.; Sawchuk, T.; Holmes, C.F.; et al. Mannose 6-phosphate/insulin-like growth factor II receptor is a death receptor for granzyme B during cytotoxic T cell-induced apoptosis. Cell 2000, 103, 491–500, doi:10.1016/S0092-8674(00)00140-9.
[153]  Veugelers, K.; Motyka, B.; Goping, I.S.; Shostak, I.; Sawchuk, T.; Bleackley, R.C. Granule-mediated killing by granzyme B and perforin requires a mannose 6-phosphate receptor and is augmented by cell surface heparan sulfate. Mol. Biol. Cell 2006, 17, 623–633.
[154]  Kurschus, F.C.; Bruno, R.; Fellows, E.; Falk, C.S.; Jenne, D.E. Membrane receptors are not required to deliver granzyme B during killer cell attack. Blood 2005, 105, 2049–2058, doi:10.1182/blood-2004-06-2180.
[155]  Trapani, J.A.; Sutton, V.R.; Thia, K.Y.; Li, Y.Q.; Froelich, C.J.; Jans, D.A.; Sandrin, M.S.; Browne, K.A. A clathrin/dynamin- and mannose-6-phosphate receptor-independent pathway for granzyme B-induced cell death. J. Cell Biol. 2003, 160, 223–233, doi:10.1083/jcb.200210150.
[156]  Bretthauer, R.K.; Castellino, F.J. Glycosylation of Pichia pastoris-derived proteins. Biotechnol. Appl. Biochem. 1999, 30, 193–200.
[157]  Raja, S.M.; Metkar, S.S.; Froelich, C.J. Cytotoxic granule-mediated apoptosis: Unraveling the complex mechanism. Curr. Opin. Immunol. 2003, 15, 528–532, doi:10.1016/S0952-7915(03)00111-0.
[158]  Gross, C.; Koelch, W.; DeMaio, A.; Arispe, N.; Multhoff, G. Cell surface-bound heat shock protein 70 (Hsp70) mediates perforin-independent apoptosis by specific binding and uptake of granzyme B. J. Biol. Chem. 2003, 278, 41173–41181.
[159]  Bird, C.H.; Sun, J.; Ung, K.; Karambalis, D.; Whisstock, J.C.; Trapani, J.A.; Bird, P.I. Cationic sites on granzyme B contribute to cytotoxicity by promoting its uptake into target cells. Mol. Cell. Biol. 2005, 25, 7854–7867.
[160]  Shi, L.; Keefe, D.; Durand, E.; Feng, H.; Zhang, D.; Lieberman, J. Granzyme B binds to target cells mostly by charge and must be added at the same time as perforin to trigger apoptosis. J. Immunol. 2005, 174, 5456–5461.
[161]  Metkar, S.S.; Wang, B.; Aguilar-Santelises, M.; Raja, S.M.; Uhlin-Hansen, L.; Podack, E.; Trapani, J.A.; Froelich, C.J. Cytotoxic cell granule-mediated apoptosis: Perforin delivers granzyme B-serglycin complexes into target cells without plasma membrane pore formation. Immunity 2002, 16, 417–428, doi:10.1016/S1074-7613(02)00286-8.
[162]  Onda, M.; Nagata, S.; Tsutsumi, Y.; Vincent, J.J.; Wang, Q.; Kreitman, R.J.; Lee, B.; Pastan, I. Lowering the isoelectric point of the Fv portion of recombinant immunotoxins leads to decreased nonspecific animal toxicity without affecting antitumor activity. Cancer Res. 2001, 61, 5070–5077.
[163]  Buzza, M.S.; Zamurs, L.; Sun, J.; Bird, C.H.; Smith, A.I.; Trapani, J.A.; Froelich, C.J.; Nice, E.C.; Bird, P.I. Extracellular matrix remodeling by human granzyme B via cleavage of vitronectin, fibronectin, and laminin. J. Biol. Chem. 2005, 280, 23549–23558.
[164]  Ronday, H.K.; van der Laan, W.H.; Tak, P.P.; de Roos, J.A.; Bank, R.A.; TeKoppele, J.M.; Froelich, C.J.; Hack, C.E.; Hogendoorn, P.C.; Breedveld, F.C.; et al. Human granzyme B mediates cartilage proteoglycan degradation and is expressed at the invasive front of the synovium in rheumatoid arthritis. Rheumatology (Oxford) 2001, 40, 55–61, doi:10.1093/rheumatology/40.1.55.
[165]  Tak, P.P.; Spaeny-Dekking, L.; Kraan, M.C.; Breedveld, F.C.; Froelich, C.J.; Hack, C.E. The levels of soluble granzyme A and B are elevated in plasma and synovial fluid of patients with rheumatoid arthritis (RA). Clin. Exp. Immunol. 1999, 116, 366–370, doi:10.1046/j.1365-2249.1999.00881.x.
[166]  Goldbach-Mansky, R.; Suson, S.; Wesley, R.; Hack, C.E.; El-Gabalawy, H.S.; Tak, P.P. Raised granzyme B levels are associated with erosions in patients with early rheumatoid factor positive rheumatoid arthritis. Ann. Rheum. Dis. 2005, 64, 715–721, doi:10.1136/ard.2003.007039.
[167]  Chamberlain, C.M.; Ang, L.S.; Boivin, W.A.; Cooper, D.M.; Williams, S.J.; Zhao, H.; Hendel, A.; Folkesson, M.; Swedenborg, J.; Allard, M.F.; et al. Perforin-independent extracellular granzyme B activity contributes to abdominal aortic aneurysm. Am. J. Pathol. 2010, 176, 1038–1049, doi:10.2353/ajpath.2010.090700.
[168]  Saito, Y.; Kondo, H.; Hojo, Y. Granzyme B as a novel factor involved in cardiovascular diseases. J. Cardiol. 2011, 57, 141–147, doi:10.1016/j.jjcc.2010.10.001.
[169]  Kurschus, F.C.; Fellows, E.; Stegmann, E.; Jenne, D.E. Granzyme B delivery via perforin is restricted by size, but not by heparan sulfate-dependent endocytosis. Proc. Natl. Acad. Sci. USA 2008, 105, 13799–13804, doi:10.1073/pnas.0801724105.
[170]  Thomas, G. Furin at the cutting edge: From protein traffic to embryogenesis and disease. Nat. Rev. Mol. Cell Biol. 2002, 3, 753–766, doi:10.1038/nrm934.
[171]  Molloy, S.S.; Anderson, E.D.; Jean, F.; Thomas, G. Bi-cycling the furin pathway: From TGN localization to pathogen activation and embryogenesis. Trends Cell Biol. 1999, 9, 28–35, doi:10.1016/S0962-8924(98)01382-8.
[172]  Ogata, M.; Fryling, C.M.; Pastan, I.; FitzGerald, D.J. Cell-mediated cleavage of Pseudomonas exotoxin between Arg279 and Gly280 generates the enzymatically active fragment which translocates to the cytosol. J. Biol. Chem. 1992, 267, 25396–25401.
[173]  Theuer, C.P.; FitzGerald, D.; Pastan, I. A recombinant form of Pseudomonas exotoxin directed at the epidermal growth factor receptor that is cytotoxic without requiring proteolytic processing. J. Biol. Chem. 1992, 267, 16872–16877.
[174]  Wolf, P.; Elsasser-Beile, U. Pseudomonas exotoxin A: From virulence factor to anti-cancer agent. Int. J. Med. Microbiol. 2009, 299, 161–176, doi:10.1016/j.ijmm.2008.08.003.
[175]  Collier, R.J. Understanding the mode of action of diphtheria toxin: A perspective on progress during the 20th century. Toxicon 2001, 39, 1793–1803, doi:10.1016/S0041-0101(01)00165-9.
[176]  Zhan, H.; Choe, S.; Huynh, P.D.; Finkelstein, A.; Eisenberg, D.; Collier, R.J. Dynamic transitions of the transmembrane domain of diphtheria toxin: Disulfide trapping and fluorescence proximity studies. Biochemistry 1994, 33, 11254–11263.
[177]  Plank, C.; Oberhauser, B.; Mechtler, K.; Koch, C.; Wagner, E. The influence of endosome-disruptive peptides on gene transfer using synthetic virus-like gene transfer systems. J. Biol. Chem. 1994, 269, 12918–12924.
[178]  Vives, E.; Brodin, P.; Lebleu, B. A truncated HIV-1 Tat protein basic domain rapidly translocates through the plasma membrane and accumulates in the cell nucleus. J. Biol. Chem. 1997, 272, 16010–16017, doi:10.1074/jbc.272.25.16010.
[179]  Gius, D.R.; Ezhevsky, S.A.; Becker-Hapak, M.; Nagahara, H.; Wei, M.C.; Dowdy, S.F. Transduced p16INK4a peptides inhibit hypophosphorylation of the retinoblastoma protein and cell cycle progression prior to activation of Cdk2 complexes in late G1. Cancer Res. 1999, 59, 2577–2580.
[180]  Vocero-Akbani, A.M.; Heyden, N.V.; Lissy, N.A.; Ratner, L.; Dowdy, S.F. Killing HIV-infected cells by transduction with an HIV protease-activated caspase-3 protein. Nat. Med. 1999, 5, 29–33, doi:10.1038/4710.
[181]  Falnes, P.O.; Wesche, J.; Olsnes, S. Ability of the Tat basic domain and VP22 to mediate cell binding, but not membrane translocation of the diphtheria toxin A-fragment. Biochemistry 2001, 40, 4349–4358, doi:10.1021/bi002443l.
[182]  Ziegler, A.; Seelig, J. Interaction of the protein transduction domain of HIV-1 TAT with heparan sulfate: Binding mechanism and thermodynamic parameters. Biophys. J. 2004, 86, 254–263, doi:10.1016/S0006-3495(04)74101-6.
[183]  Noguchi, H.; Ueda, M.; Matsumoto, S.; Kobayashi, N.; Hayashi, S. BETA2/NeuroD protein transduction requires cell surface heparan sulfate proteoglycans. Hum. Gene Ther. 2007, 18, 10–17, doi:10.1089/hum.2006.118.
[184]  Richard, J.P.; Melikov, K.; Brooks, H.; Prevot, P.; Lebleu, B.; Chernomordik, L.V. Cellular uptake of unconjugated TAT peptide involves clathrin-dependent endocytosis and heparan sulfate receptors. J. Biol. Chem. 2005, 280, 15300–15306.
[185]  Tilstra, J.; Rehman, K.K.; Hennon, T.; Plevy, S.E.; Clemens, P.; Robbins, P.D. Protein transduction: Identification, characterization and optimization. Biochem. Soc. Trans. 2007, 35, 811–815, doi:10.1042/BST0350811.
[186]  Zahid, M.; Lu, X.; Mi, Z.; Robbins, P.D. Cationic and tissue-specific protein transduction domains identification, characterization, and therapeutic application. Adv. Genet. 2010, 69, 83–95, doi:10.1016/S0065-2660(10)69007-4.
[187]  Fuchs, H.; Bachran, C.; Heisler, I.; Sutherland, M. A Closer Look at Protein Transduction Domains as a Tool in Drug Delivery. Curr. Nanosci. 2005, 1, 117–124, doi:10.2174/1573413054065367.
[188]  Kabouridis, P.S. Biological applications of protein transduction technology. Trends Biotechnol. 2003, 21, 498–503, doi:10.1016/j.tibtech.2003.09.008.
[189]  van den Berg, A.; Dowdy, S.F. Protein transduction domain delivery of therapeutic macromolecules. Curr. Opin. Biotechnol. 2011, 22, 888–893, doi:10.1016/j.copbio.2011.03.008.
[190]  Snyder, E.L.; Meade, B.R.; Dowdy, S.F. Anti-cancer protein transduction strategies: Reconstitution of p27 tumor suppressor function. J. Control Release 2003, 91, 45–51, doi:10.1016/S0168-3659(03)00212-8.
[191]  Plescia, J.; Salz, W.; Xia, F.; Pennati, M.; Zaffaroni, N.; Daidone, M.G.; Meli, M.; Dohi, T.; Fortugno, P.; Nefedova, Y.; et al. Rational design of shepherdin, a novel anticancer agent. Cancer Cell 2005, 7, 457–468, doi:10.1016/j.ccr.2005.03.035.
[192]  Hong, F.D.; Clayman, G.L. Isolation of a peptide for targeted drug delivery into human head and neck solid tumors. Cancer Res. 2000, 60, 6551–6556.
[193]  Wang, J.L.; Zhang, Z.J.; Choksi, S.; Shan, S.; Lu, Z.; Croce, C.M.; Alnemri, E.S.; Korngold, R.; Huang, Z. Cell permeable Bcl-2 binding peptides: A chemical approach to apoptosis induction in tumor cells. Cancer Res. 2000, 60, 1498–1502.
[194]  Curnis, F.; Arrigoni, G.; Sacchi, A.; Fischetti, L.; Arap, W.; Pasqualini, R.; Corti, A. Differential binding of drugs containing the NGR motif to CD13 isoforms in tumor vessels, epithelia, and myeloid cells. Cancer Res. 2002, 62, 867–874.
[195]  Snyder, E.L.; Saenz, C.C.; Denicourt, C.; Meade, B.R.; Cui, X.S.; Kaplan, I.M.; Dowdy, S.F. Enhanced targeting and killing of tumor cells expressing the CXC chemokine receptor 4 by transducible anticancer peptides. Cancer Res. 2005, 65, 10646–10650, doi:10.1158/0008-5472.CAN-05-0118.
[196]  Keller, J.; Heisler, I.; Tauber, R.; Fuchs, H. Development of a novel molecular adapter for the optimization of immunotoxins. J. Control Release 2001, 74, 259–261, doi:10.1016/S0168-3659(01)00329-7.
[197]  Heisler, I.; Keller, J.; Tauber, R.; Sutherland, M.; Fuchs, H. A cleavable adapter to reduce nonspecific cytotoxicity of recombinant immunotoxins. Int. J. Cancer 2003, 103, 277–282, doi:10.1002/ijc.10809.
[198]  Hetzel, C.; Bachran, C.; Tur, M.K.; Fuchs, H.; Stocker, M. Improved immunotoxins with novel functional elements. Curr. Pharm Des. 2009, 15, 2700–2711, doi:10.2174/138161209788923930.
[199]  Hetzel, C.; Bachran, C.; Fischer, R.; Fuchs, H.; Barth, S.; Stocker, M. Small cleavable adapters enhance the specific cytotoxicity of a humanized immunotoxin directed against CD64-positive cells. J. Immunother. 2008, 31, 370–376, doi:10.1097/CJI.0b013e31816a2d23.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133

WeChat 1538708413