Novel delivery methods to achieve immunomodulation David J Gould and Yuti Chernajovsky Immunomodulation in infectious diseases, cancer, cardiovascular disease and autoimmunity can now be targeted by sophisticated protein design, altering cellular responses by increasing therapeutic cell numbers ex vivo and then reimplanting, or altering cell function by gene transfer of cells ex vivo. In the last year, vaccination has been applied to modulate responses to autoantigens, allergens, viral or cancer antigens. The application of these technologies has entered the clinical arena and is having a positive impact on the treatment and prevention of human diseases.
are being developed that can be applied to the delivery of all these types of molecules at disease sites. For the goal to be fully achieved, cell-targeting strategies require still further development.
Addresses Bone and Joint Research Unit, William Harvey Research Institute, Barts and The London, Queen Mary’s School of Medicine and Dentistry, University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom
Protein delivery systems
Corresponding author: Chernajovsky, Yuti (
[email protected]) Current Opinion in Pharmacology 2007, 7:445–450 This review comes from a themed issue on Immunomodulation Edited by Gordon Duff and Anthony G Wilson
We concentrate this review on the engineering of molecules and cells that are helping the development of novel therapeutics (see Table 1). We are not going into the details of the methods of gene therapy as this is beyond the scope of this review.
Antibodies against tumour antigens have been used for both diagnostic and therapeutic purposes (see Figure 1a). The cellular immune response against cancers can be potentiated by delivering cytokines to sites of tumour growth. For this purpose, fusion proteins between anti-tumour antigen antibodies and cytokines (immunocytokines) have been developed [1]. A humanised anti-ganglioside GD2 antibody fused with IL-2 was investigated in phase I clinical trials in melanoma [2] and prostate cancer patients [3]. In both cases increased cellular immune responses were reported.
Available online 3rd July 2007 1471-4892/$ – see front matter # 2007 Elsevier Ltd. All rights reserved. DOI 10.1016/j.coph.2007.05.002
Introduction Immunomodulation can impact many pathological processes including vaccination, autoimmunity, cancer and transplantation. Using naturally occurring compounds from steroids to antibodies and cytokines has been advantageous for disease management. Yet, their use is limited due to systemic effects on healthy tissue that can lead to unwanted side effects. In this review, we want to focus on new molecular and cellular technologies that are advancing the overall aim of targeting therapy to particular disease sites or mechanisms of disease and hence are more specific. Intracellular delivery systems will be required for all molecules that have intracellular function. For example, nucleic acid molecules including encoding genes, oligonucleotides and RNA molecules must enter cells and target the nucleus when transcription is the target. Proteins with intracellular function require delivery into cells. Regardless of the molecules for delivery, a common requirement is the evasion of endosomal uptake that may cause degradation and denaturation. Several approaches www.sciencedirect.com
Immunocytokines provide increased half-life to cytokines, but the antibody moiety has a decreased half-life when compared with the non-fused protein. This is probably due to the interactions of immunocytokines with cytokine receptors that are widely expressed throughout the body. In this respect, engineering latent cytokines [4,5] overcome the interaction with cytokine receptors. Latent cytokines are fusion proteins between the latent-associated peptide (LAP) from transforming growth factor (TGF) b followed by a matrix metalloproteinase (MMP) cleavage site and the cytokine of interest. The LAP dimer encloses the cytokine in a shell-like structure conferring extended half-life to the cytokine that cannot interact with its receptor(s) until released from LAP at sites of high MMP activity found in pathologies with local inflammation and active tissue remodeling such as autoimmune disease, atherosclerosis and cancer. Protein transduction domains (PTDs) are peptide sequences that can penetrate cell membranes independent of interaction with specific receptors or transporters. The first characterised PTD is the third a helix of Antennapedia homeodomain protein [6] whilst a region of HIV Tat containing a basic domain is probably the most widely applied PTD [7] (for review on PTDs see Reference [8]). The uptake mechanism for Tat is via a lipid raft-dependent macropinocytosis mechanism [9]. Following cell entry Tat targets molecules to the nucleus Current Opinion in Pharmacology 2007, 7:445–450
446 Immunomodulation
Table 1 Delivery systems Delivery system Antibody Latent molecules Protein transduction domains DNA vaccination Replicating viruses DNA/RNA complexes T bodies
Application
References
Targeting cytokines and toxins to specific Ag expressing cells/tissues Specific activity of proteins at sites of MMP activity Intracellular delivery of proteins, DNA, RNA to any cell type Recombinant vaccination through DNA expression of antigens Specific proliferation of viruses in tumour cells Delivery of RNA or DNA to cells avoiding endosomal degradation T cells engineered with chimeric receptors that target them to antigen bearing tumours or other cell types/tissues
[1,2,3] [4,5] [8,10,11,13] [26] [17,21] [38,39] [43,44,45,46]
because of a strong nuclear localisation signal unless this signal is overridden by a nuclear export signal in the cargo molecule. In view of PTDs’ lack of immunogenicity, and versatility as carriers of a variety of molecules (proteins, peptides, siRNA, DNA–protein complexes and viruses) of unlimited size, their clinical application seems likely in the near future. The versatility of these molecules is illustrated in a recent study where PTDs were used to deliver pre-mRNA to correct aberrant splicing of a gene
[10]. Also Bim, an antagonist of Bcl-2-mediated cell survival, was delivered to tumour cells with Tat that significantly slowed down tumour growth in murine models of pancreatic cancer and melanoma [11]. Another potential strategy for cancer treatment utilising a PTD is delivery of HSV-TK fused with Tat (TK-Tat), and in this in vitro study TK-Tat localised to the nucleus was stable and cells were sensitive to ganciclovir treatment with good bystander killing observed [12]. PTDs have been
Figure 1
Schematic representation of the structure of antibodies, scFv and chimeric scFv signaling receptors. Panel a shows the structure of an antibody with its variable heavy (VH), variable light (VL) domains that comprise the antigen recognition/binding site with their respective constant regions (CH and CL). The whole structure is kept together by disulfide bonds. Panel b shows the structure of a scFv where the VH and VL are fused via a 15 aminoacid linker of the sequence (GGGGS)3. Panel c shows the structure of scFv chimeric receptor where the scFv is extracellular and is normally linked to the cytoplasmic signaling domain of a TCR chain such as the FcRg or the z chain through a spacer or hinge region. Current Opinion in Pharmacology 2007, 7:445–450
www.sciencedirect.com
Novel delivery methods to achieve immunomodulation Gould and Chernajovsky 447
employed for immunomodulation in experimental models of inflammation; for example, the delivery of the Tat-IkBa superrepressor in a rat pleurisy model caused a reduction in both leucocyte recruitment and production of proinflammatory cytokines [13]. One interesting strategy is the use of a short amphipathic peptide carrier, Pep-1, which can be linked non-covalently to cargo molecules through Fmoc, which then dissociates immediately after it has crossed the cell membrane [14] and the liberated cargo can then distribute with normal tropism without influence from the attached PTD. Human papilloma virus (HPV) has proven hard to eradicate because of ineffective means of in vitro culture. Cloning of the HPV capsid gene (L1) and its expression in heterologous systems [15] has allowed the development of effective vaccines. Hence, prophylactic vaccination to HPV types 6/11/16 and18 to prevent cervical cancer is becoming a reality [16].
New viral gene delivery systems Somatic gene therapy was originally thought to be safer using non-replicating viruses. However, for cancer treatment scientists have now developed attenuated viruses that are capable of replicating more efficiently in cancer cells than in normal cells. These oncolytic viruses could in principle infect neighbouring cancer cells and lyse them sparing the normal tissue. A wide variety of RNA and DNA viruses are being investigated [17]. Replicating poxvirus can deliver antiangiogenic factors both as secreted factors or as shRNA [18,19] or cytokines such as GM-CSF [20]. Recently, the data of a phase I clinical trial in prostate cancer using intravenous injection of a replication-selective, prostate-specific antigen-targeted oncolytic adenovirus were published with some positive results [21]. The use of oncolytic viruses, the relevance of the animal models and the environmental safety issues involved raised some concerns [22]. The immune system appears to be dealing quite effectively with these oncolytic viruses because most humans are preimmune to these viruses either via natural exposure or by vaccination. Whether the antiviral immune response will allow for effective cancer therapy will need to be resolved in clinical trials.
Non-viral delivery systems Plasmid DNA has no innate mechanism to enter cells, but direct injection in skeletal muscle achieves transfection and gene expression in most species from rodents [23] to humans [24]. This expression has led to the effective application of plasmid vectors in numerous vaccination studies conducted in rodents. DNA-based vaccines have several advantages including their simple preparation and stability. Vaccination is more flexible because the plasmid encoding the antigen can be combined with other genes that modify the immune response. Plasmid DNA also has immunostimulatory properties due to unmethylated CpG www.sciencedirect.com
repeats that interact with TLR9 receptors expressed on antigen-presenting cells (APC), although this is not a prerequisite for vaccination as responses are also observed in TLR9-knockout mice [25]. Interestingly, the effectiveness of DNA vaccines in small animals has not translated to primates and humans and proof of activity in clinical trials has only recently been reported. In a phase I trial a DNA vaccine for bird flu was shown to be safe and achieved antibody responses at tested doses [26]. Transfection of plasmid DNA in skeletal muscle is efficiently enhanced in rodents by the use of electroporation that opens pores in the cells to permit direct entry of DNA [27]. Electroporation can enhance DNA vaccination [28], but there are few reports using electroporation in large animals. However, hydrodynamic delivery of plasmid DNA or siRNA into the vasculature of an occluded limb has proven equally effective in primates and rodents [29]. Nucleic acid molecules also need to be delivered intracellularly for function to the nucleus (for gene transcription, or for transcriptional decoy effects) or to the cytoplasm for inhibition of RNA translation or to target mRNA degradation (antisense RNA, ribozymes). RNA interference (RNAi) is an endogenous mechanism whereby double stranded RNA is processed by the RNAse III-like protein Dicer to produce short interfering RNA (siRNA) that are incorporated into the RNAinduced silencing complex (RISC) [30]. Synthetic siRNA are structurally related to endogenous microRNAs (miRNA) and can be used for sequence specific silencing. Bevasiranib, an siRNA targeting VEGF mRNA, is already in phase II clinical trials for the treatment of wet agerelated macular degeneration [31]. These small molecules were applied in a variety of experimental models in saline, complexed with lipids or conjugated with molecules for improved pharmacokinetics or targeting [32]. Another interesting aspect of these molecules relevant to immunomodulation is their interaction with Toll-like receptor (TLR)-7 through which they can co-deliver an immunostimulatory response. In a recent study, immunostimulation was further enhanced with an siRNA targeting expression of IL-10 in combination with the TLR7 stimulation by the molecule [33]. siRNA was used in arthritis models for targeting TNFa. siRNA was delivered to knee joints in combination with electroporation [34,35] or systemically complexed with cationic liposomes [36]. Both approaches required readministration. This transient action is clearly a shortcoming for long-term effects in the treatment of chronic diseases such as rheumatoid arthritis. An alternative is the use of short hairpin RNA (shRNA) molecules expressed long term from gene delivery vectors. shRNA are additionally processed in the nucleus by an RNAse III protein (Drosha) producing pre-miRNA which are exported to the cytoplasm for further processing by Dicer to produce mature miRNA. Current Opinion in Pharmacology 2007, 7:445–450
448 Immunomodulation
Ribozymes are catalytic RNA molecules that cleave mRNA sequences. Their design is more complex than siRNA. Recently, a hammerhead ribozyme targeting TNFa was shown to inhibit a model of arthritis when delivered intravenously before onset of the disease [37]. Naked DNA and RNA molecules can be combined with a variety of polycations in polyplexes for delivery to cells. The polycations used have different characteristics including ease of DNA unpackaging that is influenced by their molecular weight, toxicity, stability and ability to facilitate endosomal escape. Polyethylenimine (PEI), for example, is a polycation with strong endosome escape properties. To decrease the toxicity of polycations and retain their stability, reducible polycations are used that are specifically cleaved within cells to release the DNA. Synthetic vectors based on reducible (thiolcontaining) polycations consisting of histidine and polylysine residues were shown to efficiently deliver DNA, mRNA and siRNA in a variety of different cells [38]. The reducible approach has also been used to coat PEI complexes [39]. To target liposomes, phage peptide libraries are used to screen for tissue-binding peptides in vivo, for example, in cancer [40] or synovial endothelium in arthritis [41]. These selected peptides can be conjugated to liposomes in order to target their cell of interest.
Cell delivery systems The principle of immunosurveillance has always been central to immunotherapy approaches for the treatment of cancer. Despite the myriad of mechanisms by which cancer cells evade immunosurveillance, it has now been shown that infusion of autologous tumour antigen-specific T cells expanded ex vivo are therapeutic [42]. Another method for T cell therapy of cancer is the development of T bodies [43] in which chimeric receptors with extracellular scFv (see Figure 1b) of antibodies against tumour antigens are grafted onto the cytoplasmic and signaling domains of T cell receptor (TCR) subunits (see Figure 1c). The direct recognition of antigen by the scFv obviates the need for antigen processing and presentation by the major histocompatibility complex. In a clinical trial using T bodies, targeting of the tumour sites was poor and survival of the engineered cells very limited [44]. This may be due in part to the fact that the scFv used was of mouse origin. Similar work has also been done by cloning a and b chains of TCR from tumour infiltrating lymphocytes with some degree of success in melanoma [45,46]. It is of interest that the fate of the endogenous TCR chains of the transduced cells is unknown. Whether the endogenous TCR chains recognise autoantigens by themselves or by combination with the transduced TCR chains could have consequences for autoimmunity and needs a long-term follow-up of these patients. Current Opinion in Pharmacology 2007, 7:445–450
Mesenchymal stromal cells (MSC) have immunosuppressive properties. Transplantation of MSC has been shown to inhibit steroid-resistant grade III–IV graft versus host disease (GvHD) in patients receiving allogeneic stem cell therapy [47,48]. MSC seem to affect the cytokine secretion profile of dendritic cells (DCs), naive and effector T helper cells, and natural killer (NK) cells to induce a more anti-inflammatory or tolerant phenotype [49]. GvHD is mediated by T cells from donor origin. T cells can be retrovirally transduced, after cell cycle activation ex vivo with anti-CD28/CD3 or phytohaemaglutinin, to express the ‘suicidal’ gene thymidine kinase from Herpes simplex virus (HSV tk) that renders dividing T cells sensitive to ganciclovir, and this approach has been used in clinical trials [50,51]. Recently, it has been shown that using IL-7-mediated proliferation of T cells preserves the effector function of T cells more effectively than TCR-mediated protocols for viral transduction [52].
Conclusions The ingenuity of scientists and the exponential growth in the understanding of molecular mechanisms involved in different pathologies have increased substantially the possibilities for immunomodulation. The potential of novel approaches such as gene therapy and drug design enable targeting of therapeutics with better therapeutic index. The limitation of these therapies resides in our better understanding of the long-term outcome of delivering biologicals and manipulating the immune system. The immune system is finely tuned in health to react to foreign pathogens and oncogenic changes of cells in the body. Tilting the immune system with therapeutic agents to behave in a particular way is not without consequences. The challenge for immunomodulating therapies will be to obtain long-lived therapeutic outcomes with shorter therapeutic interventions. This may require the combination of immunotherapies alone or with stem cell therapies.
Acknowledgements We acknowledge support of the Arthritis Research Campaign, UK, EUFP6 (Genostem), the British Heart Foundation and The Wellcome Trust. We are grateful to G Adams for the editing of the manuscript.
References and recommended reading Papers of particular interest, published within the annual period of review, have been highlighted as: of special interest of outstanding interest 1.
Lo KM, Lan Y, Lauder S, Zhang J, Brunkhorst B, Qin G, Verma R, Courtenay-Luck N, Gillies SD: huBC1-IL12, an immunocytokine which targets EDB-containing oncofetal fibronectin in tumours and tumour vasculature, shows potent anti-tumour activity in human tumour models. Cancer Immunol Immunother 2007, 56:447-457.
2.
King DM, Albertini MR, Schalch H, Hank JA, Gan J, Surfus J, Mahvi D, Schiller JH, Warner T, Kim K et al.: Phase I clinical trial of the immunocytokine EMD 273063 in melanoma patients. J Clin Oncol 2004, 22:4463-4473. First report of clinical trial in cancer using immunocytokines. www.sciencedirect.com
Novel delivery methods to achieve immunomodulation Gould and Chernajovsky 449
3.
Ko YJ, Bubley GJ, Weber R, Redfern C, Gold DP, Finke L, Kovar A, Dahl T, Gillies SD: Safety, pharmacokinetics, and biological pharmacodynamics of the immunocytokine EMD 273066 (huKS-IL2): results of a phase I trial in patients with prostate cancer. J Immunother 2004, 27:232-239.
4.
Adams G, Vessillier S, Dreja H, Chernajovsky Y: Targeting cytokines to inflammation sites. Nat Biotechnol 2003, 21:1314-1320. First description of latent cytokine engineering using IFN b as the therapeutic molecule.
20. Kim JH, Oh JY, Park BH, Lee DE, Kim JS, Park HE, Roh MS, Je JE, Yoon JH, Thorne SH et al.: Systemic armed oncolytic and immunologic therapy for cancer with JX-594, a targeted poxvirus expressing GM-CSF. Mol Ther 2006, 14:361-370. 21. Small EJ, Carducci MA, Burke JM, Rodriguez R, Fong L, van Ummersen L, Yu DC, Aimi J, Ando D, Working P et al.: A phase I trial of intravenous CG7870, a replication-selective, prostatespecific antigen-targeted oncolytic adenovirus, for the treatment of hormone-refractory, metastatic prostate cancer. Mol Ther 2006, 14:107-117. First report on the clinical use of prostate specific oncolytic virus. 23 patients were treated, 5 patients had a decrease in serum prostatespecific antigen (PSA) of 25% to 49% following a single treatment, including 3 of 8 patients at the highest dose levels.
5.
Vessillier S, Adams G, Chernajovsky Y: Latent cytokines: development of novel cleavage sites and kinetic analysis of their differential sensitivity to MMP-1 and MMP-3. Protein Eng Des Sel 2004, 17:829-835.
6.
Derossi D, Joliot AH, Chassaing G, Prochiantz A: The third helix of the Antennapedia homeodomain translocates through biological membranes. J Biol Chem 1994, 269:10444-10450.
22. Chernajovsky Y, Layward L, Lemoine N: Fighting cancer with oncolytic viruses. BMJ 2006, 332:170-172.
7.
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.
23. Wolff JA, Malone RW, Williams P, Chong W, Acsadi G, Jani A, Felgner PL: Direct gene transfer into mouse muscle in vivo. Science 1990, 247:1465-1468. Original report of long term expression from muscle by in vivo DNA injection.
8.
Chauhan A, Tikoo A, Kapur AK, Singh M: The taming of the cell penetrating domain of the HIV Tat: myths and realities. J Control Release 2007, 117:148-162.
9.
Wadia JS, Stan RV, Dowdy SF: Transducible TAT-HA fusogenic peptide enhances escape of TAT-fusion proteins after lipid raft macropinocytosis. Nat Med 2004, 10:310-315. Elucidation of Tat-PTD mechanism of entry via a lipid raft-dependent macropinocytosis.
24. Isner JM, Baumgartner I, Rauh G, Schainfeld R, Blair R, Manor O, Razvi S, Symes JF: Treatment of thromboangiitis obliterans (Buerger’s disease) by intramuscular gene transfer of vascular endothelial growth factor: preliminary clinical results. J Vasc Surg 1998, 28:964-973. Important pioneering use of plasmid DNA delivery to express VEGF for the treatment of human disease. This work was expanded also to diabetic limb ischemia and coronary heart disease.
10. El-Andaloussi S, Johansson HJ, Lundberg P, Langel U: Induction of splice correction by cell-penetrating peptide nucleic acids. J Gene Med 2006, 8:1262-1273.
25. Spies B, Hochrein H, Vabulas M, Huster K, Busch DH, Schmitz F, Heit A, Wagner H: Vaccination with plasmid DNA activates dendritic cells via Toll-like receptor 9 (TLR9) but functions in TLR9-deficient mice. J Immunol 2003, 171:5908-5912.
11. Kashiwagi H, McDunn JE, Goedegebuure PS, Gaffney MC, Chang K, Trinkaus K, Piwnica-Worms D, Hotchkiss RS, Hawkins WG: TAT-Bim induces extensive apoptosis in cancer cells. Ann Surg Oncol 2007.
26. Drape RJ, Macklin MD, Barr LJ, Jones S, Haynes JR, Dean HJ: Epidermal DNA vaccine for influenza is immunogenic in humans. Vaccine 2006, 24:4475-4481.
12. Cao L, Si J, Wang W, Zhao X, Yuan X, Zhu H, Wu X, Zhu J, Shen G: Intracellular localization and sustained prodrug cell killing activity of TAT-HSVTK fusion protein in hepatocelullar carcinoma cells. Mol Cells 2006, 21:104-111. 13. Blackwell NM, Sembi P, Newson JS, Lawrence T, Gilroy DW, Kabouridis PS: Reduced infiltration and increased apoptosis of leukocytes at sites of inflammation by systemic administration of a membrane-permeable IkappaBalpha repressor. Arthritis Rheum 2004, 50:2675-2684. 14. Morris MC, Depollier J, Mery J, Heitz F, Divita G: A peptide carrier for the delivery of biologically active proteins into mammalian cells. Nat Biotechnol 2001, 19:1173-1176. 15. Heim K, Widschwendter A, Szedenik H, Geier A, Christensen ND, Bergant A, Concin N, Hopfl R: Specific serologic response to genital human papilloma virus types in patients with vulvar precancerous and cancerous lesions. Am J Obstet Gynecol 2005, 192:1073-1083. 16. Villa LL, Costa RL, Petta CA, Andrade RP, Paavonen J, Iversen OE, Olsson SE, Hoye J, Steinwall M, Riis-Johannessen G et al.: High sustained efficacy of a prophylactic quadrivalent human papillomavirus types 6/11/16/18 L1 virus-like particle vaccine through 5 years of follow-up. Br J Cancer 2006, 95:1459-1466. First long term study on the efficacy of HPV vaccination.
27. Mir LM, Bureau MF, Gehl J, Rangara R, Rouy D, Caillaud JM, Delaere P, Branellec D, Schwartz B, Scherman D: High-efficiency gene transfer into skeletal muscle mediated by electric pulses. Proc Natl Acad Sci USA 1999, 96:4262-4267. 28. Prud’homme GJ, Glinka Y, Khan AS, Draghia-Akli R: Electroporation-enhanced nonviral gene transfer for the prevention or treatment of immunological, endocrine and neoplastic diseases. Curr Gene Ther 2006, 6:243-273. 29. Hagstrom JE, Hegge J, Zhang G, Noble M, Budker V, Lewis DL, Herweijer H, Wolff JA: A facile nonviral method for delivering genes and siRNAs to skeletal muscle of mammalian limbs. Mol Ther 2004, 10:386-398. First demonstration of effectiveness of hydrodynamic DNA delivery both in rodents and monkeys. 30. Fire A, Xu S, Montgomery MK, Kostas SA, Driver SE, Mello CC: Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature 1998, 391:806-811. 31. Michels S, Schmidt-Erfurth U, Rosenfeld PJ: Promising new treatments for neovascular age-related macular degeneration. Expert Opin Investig Drugs 2006, 15:779-793. 32. Bumcrot D, Manoharan M, Koteliansky V, Sah DW: RNAi therapeutics: a potential new class of pharmaceutical drugs. Nat Chem Biol 2006, 2:711-719.
17. Liu TC, Kirn D: Systemic efficacy with oncolytic virus therapeutics: clinical proof-of-concept and future directions. Cancer Res 2007, 67:429-432.
33. Furset G, Sioud M: Design of bifunctional siRNAs: combining immunostimulation and gene-silencing in one single siRNA molecule. Biochem Biophys Res Commun 2007, 352:642-649.
18. Thorne SH, Tam BY, Kirn DH, Contag CH, Kuo CJ: Selective intratumoural amplification of an antiangiogenic vector by an oncolytic virus produces enhanced antivascular and antitumour efficacy. Mol Ther 2006, 13:938-946.
34. Inoue A, Takahashi KA, Mazda O, Terauchi R, Arai Y, Kishida T, Shin-Ya M, Asada H, Morihara T, Tonomura H et al.: Electrotransfer of small interfering RNA ameliorated arthritis in rats. Biochem Biophys Res Commun 2005, 336:903-908.
19. Yoo JY, Kim JH, Kwon YG, Kim EC, Kim NK, Choi HJ, Yun CO: VEGF-specific Short Hairpin RNA-expressing oncolytic adenovirus elicits potent inhibition of angiogenesis and tumour growth. Mol Ther 2007, 15:295-302.
35. Schiffelers RM, Xu J, Storm G, Woodle MC, Scaria PV: Effects of treatment with small interfering RNA on joint inflammation in mice with collagen-induced arthritis. Arthritis Rheum 2005, 52:1314-1318.
www.sciencedirect.com
Current Opinion in Pharmacology 2007, 7:445–450
450 Immunomodulation
36. Khoury M, Louis-Plence P, Escriou V, Noel D, Largeau C, Cantos C, Scherman D, Jorgensen C, Apparailly F: Efficient new cationic liposome formulation for systemic delivery of small interfering RNA silencing tumour necrosis factor alpha in experimental arthritis. Arthritis Rheum 2006, 54:1867-1877. 37. Kumar R, Dammai V, Yadava PK, Kleinau S: Gene targeting by ribozyme against TNF-alpha mRNA inhibits autoimmune arthritis. Gene Ther 2005, 12:1486-1493. 38. Read ML, Singh S, Ahmed Z, Stevenson M, Briggs SS, Oupicky D, Barrett LB, Spice R, Kendall M, Berry M et al.: A versatile reducible polycation-based system for efficient delivery of a broad range of nucleic acids. Nucleic Acids Res 2005, 33:e86. 39. Carlisle RC, Etrych T, Briggs SS, Preece JA, Ulbrich K, Seymour LW: Polymer-coated polyethylenimine/DNA complexes designed for triggered activation by intracellular reduction. J Gene Med 2004, 6:337-344. 40. Akita N, Maruta F, Seymour LW, Kerr DJ, Parker AL, Asai T, Oku N, Nakayama J, Miyagawa S: Identification of oligopeptides binding to peritoneal tumours of gastric cancer. Cancer Sci 2006, 97:1075-1081. 41. Lee L, Buckley C, Blades MC, Panayi G, George AJ, Pitzalis C: Identification of synovium-specific homing peptides by in vivo phage display selection. Arthritis Rheum 2002, 46:2109-2120. First description of peptide-specific targeting to synovial endothelium. 42. Mackensen A, Meidenbauer N, Vogl S, Laumer M, Berger J, Andreesen R: Phase I study of adoptive T-cell therapy using antigen-specific CD8+ T cells for the treatment of patients with metastatic melanoma. J Clin Oncol 2006, 24:5060-5069. 43. Eshhar Z, Bach N, Fitzer-Attas CJ, Gross G, Lustgarten J, Waks T, Schindler DG: The T-body approach: potential for cancer immunotherapy. Springer Semin Immunopathol 1996, 18:199-209. 44. Kershaw MH, Westwood JA, Parker LL, Wang G, Eshhar Z, Mavroukakis SA, White DE, Wunderlich JR, Canevari S, RogersFreezer L et al.: A phase I study on adoptive immunotherapy using gene-modified T cells for ovarian cancer. Clin Cancer Res 2006, 12:6106-6115. 45. Morgan RA, Dudley ME, Wunderlich JR, Hughes MS, Yang JC, Sherry RM, Royal RE, Topalian SL, Kammula US, Restifo NP et al.:
Current Opinion in Pharmacology 2007, 7:445–450
Cancer regression in patients after transfer of genetically engineered lymphocytes. Science 2006, 314:126-129. Reports the successful use of engineered T cells with TCR genes targeting cancer antigens. 46. Johnson LA, Heemskerk B, Powell DJ Jr, Cohen CJ, Morgan RA, Dudley ME, Robbins PF, Rosenberg SA: Gene transfer of tumour-reactive TCR confers both high avidity and tumour reactivity to nonreactive peripheral blood mononuclear cells and tumour-infiltrating lymphocytes. J Immunol 2006, 177:6548-6559. 47. Le Blanc K, Rasmusson I, Sundberg B, Gotherstrom C, Hassan M, Uzunel M, Ringden O: Treatment of severe acute graft-versushost disease with third party haploidentical mesenchymal stem cells. Lancet 2004, 363:1439-1441. First use of MSC in human clinical trials. 48. Ringden O, Uzunel M, Rasmusson I, Remberger M, Sundberg B, Lonnies H, Marschall HU, Dlugosz A, Szakos A, Hassan Z et al.: Mesenchymal stem cells for treatment of therapy-resistant graft-versus-host disease. Transplantation 2006, 81:1390-1397. 49. Aggarwal S, Pittenger MF: Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood 2005, 105:1815-1822. Analysis of the possible mechanisms mediating the immunosuppressive effects of MSC in humans. 50. Ciceri F, Bonini C, Gallo-Stampino C, Bordignon C: Modulation of GvHD by suicide-gene transduced donor T lymphocytes: clinical applications in mismatched transplantation. Cytotherapy 2005, 7:144-149. 51. Recchia A, Bonini C, Magnani Z, Urbinati F, Sartori D, Muraro S, Tagliafico E, Bondanza A, Stanghellini MT, Bernardi M et al.: Retroviral vector integration deregulates gene expression but has no consequence on the biology and function of transplanted T cells. Proc Natl Acad Sci USA 2006, 103:1457-1462. 52. Qasim W, Mackey T, Sinclair J, Chatziandreou I, Kinnon C, Thrasher AJ, Gaspar HB: Lentiviral vectors for T-cell suicide gene therapy: preservation of T-cell effector function after cytokine-mediated transduction. Mol Ther 2007, 15:355-360. The use of lentiviral vectors in humans has been argued for several years. This study is an important step forward in their possible use for the treatment of GvHD.
www.sciencedirect.com