701
Views
11
CrossRef citations to date
0
Altmetric
Commentary

Bacterial vectors for active immunotherapy reach clinical and industrial stages

, , , &
Pages 1454-1458 | Received 02 Jul 2012, Accepted 10 Jul 2012, Published online: 16 Aug 2012

Abstract

Active immunotherapy based on live attenuated bacterial vectors has matured in terms of industrial development and develops through a combination of three phenomena. First, active immunotherapy that stimulates an antigen-specific cytotoxic T-cell immune response has become a reality after several years of work. Second, there is still a need to identify vectors that can deliver antigens to the cytosol of antigen-presenting cells in vivo. Third, the recent progress in the understanding of bacterial lifestyle and in developing genetic engineering tools has enabled the design of bioengineered bugs that are capable of delivering antigens. Here, we review the mechanisms by which clinical bacterial vectors deliver antigens into the cytosol of antigen-presenting cells and summarize the development strategy of the three identified firms in this field.

Why Have Bacterial Vectors Reached the Clinical and Industrial Stage?

Active cellular immunotherapy is emerging

Advances in basic immunology have led to an improved understanding of the interactions between the immune system, tumors and pathogens. To re-engage the immune system in its fight against “bad” cells, active immunotherapy focuses on the development of agents that activate the immune system to target and kill these cells. Strategies to stimulate effector immune cells include ex vivo or in vivo vaccination with specific antigens, treatment with cytokines (e.g., IL-2), enhancement of antigen presentation (by stimulation of toll-like receptors (TLRs), administration of dendritic cells (DCs) or the use of CD40-targeted agonistic antibodies) and blocking inhibitory signals (antibodies against CTLA4). At the moment, more than 200 active therapeutic vaccine trials across a wide spectrum of cancers and infectious diseases are listed at ClinicalTrials.gov. The first active immunotherapeutic approach that reached the market, in 2010, was Sipuleucel-T (Provenge, Dendreon), which consists of autologous peripheral blood mononuclear cells loaded ex vivo with a fusion protein of prostatic acid phosphatase (PAP) and granulocyte-macrophage colony-stimulating factor (GM-CSF) to treat patients with advanced prostate cancer.

Need for in vivo antigen delivery

Among the different strategies to stimulate the antigen-specific T-cell response, the ex vivo loading of DCs (as in Provenge’s strategy) appears to be too difficult to generalize because of logistical problems. Rather, many firms have conducted clinical trials using in vivo strategies based on the development of vectors that deliver antigens or nucleic acids encoding antigens to the cytosol of antigen-presenting cells (APCs). These vectors include viruses, virus-like particles, plasmid DNA, peptides, chimeric proteins and bacteria.Citation1

Bacterial lifestyles have become better understood, and molecular engineering has assisted in the development of bacterial vaccines

“Killed” or attenuated bacteria have long been used for prophylactic vaccines. Hence, extensive clinical data are available, and firms have expertise in preparing and formulating this kind of vaccine. Microbial pathogens were the earliest non-surgical cancer treatment. In the nineteenth century, William Coley developed the first bacterial-based cancer treatment, which was composed of killed gram-positive streptococci and gram-negative Serratia marcescens injected directly into the tumor.2 However, safety concerns and the advent of radiotherapy and chemotherapy treatments led to a decline in the use of this treatment. Nevertheless, live Bacillus Calmette-Guerin (BCG) is still in clinical use for bladder cancer treatment. In this kind of bacteria-mediated cancer immunotherapy, many of the antitumor effects induced by bacteria are nonspecific, including angiostatic effects induced by pro-inflammatory cytokines, such as IL-12 and IFN-gamma, direct tumor necrosis by tumor necrosis factor (TNF) α, induction of epitope spreading induced by the death of tumor cells from these indirect mechanisms and the direct killing of tumor cells infected by bacteria. An important molecular component of DCs that can be targeted for immunotherapy is the TLR, which recognizes microbes. Indeed, the activation of DCs is likely to contribute to therapeutic effects that are associated with the nonspecific activation of anti-tumor immunity by pathogen-associated molecular patterns (PAMPs), and several TLR ligands are currently being tested in clinical trials.Citation3

Bacteria can also mediate antigen-specific responses if they are engineered to express and deliver antigens to the correct compartment, i.e. the cytosol of APCs, during the vaccination process which became, in this case, a controlled infection. The understanding of the molecular mechanisms whereby DCs augment the stimulation of naive T-cells is rapidly growing. Nevertheless, host perception of the level of danger linked to this infection directly influences the type and intensity of the immune response.Citation4 Because the challenge of CD8+ T-cell-mediated immunotherapy is to overcome tolerogenic events (the majority of tumor antigens bear strong similarity to self-proteins), bacterial vectors have the advantage of delivering both an antigenic message and a strong danger signal mediated by their PAMPs. PAMPs, such as lipopolysaccharide, lipoproteins, flagellin and DNA containing unmethylated cytosine-phosphate-guanine (CpG), can bind to their respective TLRs on APCs, such as DCs, leading to upregulation of co-stimulatory molecules, the maturation and migration of APCs to secondary lymph nodes and the production of pro-inflammatory cytokines.

Over the past 40 years, advances in bacterial engineering, molecular biology and our understanding of pathogenic bacterial lifestyles have significantly accelerated the rational design of bacterial vectors that induce the in vivo delivery of heterologous antigens inside APCs. Bacterial vaccine vectors offer multiple advantages: (1) there are several well-characterized virulence-attenuating mutations; (2) the number, the amount and the in vivo location of antigen expression can be regulated; (3) multiple vaccine delivery routes are possible; and (4) they potently stimulate the innate and adaptive immune systems.

How to Deliver an Antigen Inside APCs by Means of a Bacterium In Vivo?

Currently, two major engineered but natural processes are being explored at preclinical or clinical stages to achieve the delivery of antigenic proteins inside APCs. The first is based on the use of the type III secretion system to deliver bacterial toxins to a cell’s cytoplasm. The second uses pathogens with natural intracellular tropism. At the industrial development stage, the first strategy uses primarily Salmonella sp or P. aeruginosa, and the second uses Listeria monocytogenes.

Type III-mediated delivery

The type III secretion system (T3SS) is a critical virulence factor used by a broad range of pathogens (e.g., Salmonella, Shigella, Yersinia and Pseudomonas) to deliver toxins into the cytoplasm of host cells. Toxins are addressed to the T3SS thanks to a short signaling sequence and when fusing an antigen of interest to this sort sequence, recombinant protein can be engaged by the T3SS and delivered to the cytosol of DCs, which are often targeted by pathogens to disrupt the immune response and favor natural infection. Therefore, this strategy is an effective way to deliver antigens directly inside APCs.Citation5-Citation9

Intracellular microbes bearing antigens

L. monocytogenes is a facultative, intracellular, gram-positive rod that invades the intestinal mucosa and is captured by phagocytes. Listeria can modify phagosomes into large compartments termed spacious Listeria-associated phagosomes (SLAPS). Listeria secretes listeriolysin O (LLO) and phospholipase C, which induces the degradation of the phagolysosome membrane and the release of 5–10% of the bacteria into the cytoplasm, where they multiply and become motile via the expression of ActA, an actin polymerase that enables actin mobilization. The use of live recombinant strains of L. monocytogenes to deliver antigens began with the pioneering work of Dr. Yvonne Paterson in Pennsylvania. Her work demonstrated that a recombinant L. monocytogenes that expressed and secreted the model antigen influenza virus nucleoprotein (NP) fused to LLO could efficiently infect APCs, and this expressed NP protein was processed by the endogenous antigen-processing pathway, facilitating the presentation of NP epitopes in the context of MHC class I molecules.Citation10 Since then, the L. monocytogenes platform has rapidly evolved to become one of the most efficacious approaches for antigen delivery for infectious diseasesCitation11 and cancer (For a review see.Citation12)

Salmonella is a gram-negative bacterium and intracellular pathogen that causes salmonellosis. Its intracellular survival and replication are restricted to the endosomal compartment of eukaryotic cells, such as macrophages. Several publications have illustrated the use of live attenuated Salmonella strains to elicit mucosal and systemic immune responses against antigens from other infectious bacteria, viruses, tumor antigens or tumor-promoting growth factors.Citation13 Despite the numerous preclinical studies using salmonella strains, few have advanced to human clinical trials, and no clinical efficacy has been reported.

How to Transform a Pathogen into an Innovative Therapeutic Product?

Strategies for attenuating bacterial virulence

According to the “patho-biotechnology” approach,Citation14 live strains of bacteria have to be engineered to significantly improve their clinical effectiveness (safety and efficacy) and technological robustness. The primary goal is to develop bacterial strains that retain their capacity to synthesize/deliver antigens and induce robust immune responses against antigens but exhibit reduced pathogenicity.

In this context, two main strategies have been explored: live attenuated bacteria and killed but metabolically active (KBMA) bacteria.

Live attenuated vaccines

Over the past few decades, advances in bacterial genetics combined with the emergence of improved molecular biology tools have allowed the dissection of the mechanisms of bacterial virulence and have specified the complexity of host–pathogen interactions.Citation15

The use of well-defined, non-reverting attenuating deletions allows for the development of live attenuated bacteria with no risk of reversion to a virulent state. To date, few strains of attenuated live bacteria vaccines have proven acceptable for human use in preventing infectious diseases. Attenuation is primarily based on the deletion of genes involved in particular metabolic pathways or critical virulence factors. One of the best-characterized attenuations in a metabolic pathway is the deletion of the aroA gene, which encodes 3-phosphoshikimate 1-carboxyvinyltransferase, an enzyme that is involved in aromatic amino acid synthesis. This attenuation renders Salmonella, Listeria and Pseudomonas species auxotrophic for aromatic amino acids, lowering their capacity for in vivo replication and eliminating their pathogenicity.Citation6,Citation16

Multiple virulence-attenuated vaccine vectors have also been developed. For example, in Salmonella, the most widely studied strains have mutations in the phoP/phoQ regulator system or in Salmonella pathogenicity island 2 (SPI2). Salmonella virulence factors encoded in the phoP locus induce the formation of spacious vacuoles from phagosomes, which allows bacterial persistence and growth in these organelles. The most commonly used deletions to decrease the virulence of Listeria vectors are the actA deletion, which blocks its ability to spread from cell to cell, thus limiting bacterial dissemination, and the inlB deletion, which blocks the direct uptake of Listeria by hepatocytes, thus minimizing its potential to cause liver-related toxicity.Citation17

It is important to maintain a subtle balance between minimal pathogenicity and maximal immunogenicity with live bacterial vectors because attenuation procedures could impact the course of infection and hence the immune response. For example, oral administration of a PhoP-null Salmonella mutant induces the innate immune response, whereas an aroA mutant elicits a stronger TH1-like T-cell response.Citation18 However, an engineered strain of L. monocytogenes that enhances the NLRC4 inflammasome is too attenuated and fails to induce protective immunity based on antigen-specific CD8+ T-cells.Citation19 To counteract the poor infectivity linked to attenuation, Wang et al. have demonstrated that satisfactory T-cell immune responses with an aroA-attenuated P. aeruginosa vector could be restored by mimicking the intrabody dissemination using multiple loci injections.Citation20

Killed but metabolically active vaccine

The killed but metabolically active (KBMA) approach recently reviewed,Citation21 is based on the Intercept Blood system® developed by Cerus Corporation (Concord, CA) to inactivate undetected microbes contaminating plasma and platelet blood products.Citation22 Briefly, it consists of a combination of a synthetic psoralen (amotosalen hydrochloride, S59) and illumination with long-wave UV (UVA) light, which induces covalent monoadducts and crosslinks of DNA and RNA. Brockstedt et al. used this technology and envisioned “a new vaccine paradigm” in which they combined the safety of a killed vaccine with the potency of a live vaccine. To obtain such a result, they removed genes required for nucleotide excision repair (uvrAB), which is the primary mechanism by which bacteria repair psoralen-induced crosslinks. Accordingly, these gene deletions render microbial-based vaccines more sensitive to photochemical inactivation with amotosalen and UVA illumination. Therefore, the number of crosslinks required to “kill” the bacterium is very low (theoretically, only one crosslink is sufficient to block bacterial replication). However, bacterial genes can still be expressed, a fact that allows KBMA bacteria to retain the essential properties of live bacteria without the ability to multiply. Thus, this innovative vaccine approach, using a Listeria vectorial strain, has been demonstrated to elicit functional T-cells and long-term protective immunity that correlate with vaccine efficacy in mouse models of infectious diseases and cancer.Citation23

Stable and strong heterologous protein expression

The use of bacterial systems for the production of heterologous recombinant proteins is widespread in academic and industrial sectors. Experience in the fields of molecular biology and biochemistry in codon optimization, cloning of genes, tag fusions to enable detection, secretion, co-expression of multiple proteins and genome modeling has contributed to the continuous improvement of bacterial vaccines. Plasmids are commonly used to express large quantities of recombinant proteins in bacterial hosts. However, plasmid maintenance in bacteria often requires antibiotic resistance markers, which have been discouraged in clinical applications by the Food and Drug Administration. Therefore, two strategies have been explored to produce antigen delivery systems without antibiotic resistance genes. The first approach involves plasmid stabilization using complementation systems or balanced lethal systems with the strain. The second strategy consists of inserting the heterologous gene into the bacterial chromosome either by homologous recombinationCitation24 or by phage-specific insertion.Citation25

The plasmid-based strategy has the advantage of achieving high copy numbers of heterologous genes. Mechanisms of complementation have been described previously. In L. monocytogenes, one mechanism is based on the complementation of the pfrA-deficient strain with an episomal copy of pfrA,Citation24 and the second method uses d-alanine racemase (dal) complementation.Citation26 In Salmonella, a variety of vectors have been developed based on complementation of genetic defects in the biosynthetic pathways for DNA precursors, amino acids and cell walls biosynthetic pathways (for a review see reference Citation27). In plasmid-based balanced lethal systems, plasmids encode an essential protein that is required for bacterial growth and replication, thereby forcing the bacterium to retain the plasmid.Citation28

Chromosomal integration is an obvious solution to achieve the stable expression of heterologous genes, but the fact that this might entail a single copy of the transgene of interest could constitute a major drawback of this approach. The addition of multiple copies of genes of interest could potentially overcome this drawback.

Complex antigens can be delivered

We demonstrated that large proteins can be correctly secreted and translocated by the T3SS of P. aeruginosa when fused to the N-terminal domain of the natural exotoxin, ExoS.Citation29 In this system, expression was controlled by bypassing the natural regulation of T3SS expression using a strong IPTG-inducible promoter to govern the expression of the ExsA natural T3SS transcription activator protein in trans. Furthermore, using that platform, it will likely be possible to develop more sophisticated expression systems that would allow, for example, the expression and delivery of multiple antigenic proteins.Citation20 These properties suggest that immunization protocols with whole antigenic proteins are feasible and simultaneously provide a panel of putative mono- or multi-antigenic epitopes. This approach offers the potential for a broad spectrum of applications for vaccination, bypassing the limitations associated with the use of short peptides that are restricted to particular major histocompatibility complex (MHC) class I alleles.

Good manufacturing practices

One of the advantages of bacterial vaccine vectors is that bacteria can be easily grown and can be purified and processed inexpensively at an industrial level. However, gene deletion for attenuation often renders the bacteria more difficult to culture. Moreover, to satisfy current good manufacturing practices for drug production, bacteria need to be grown in a chemically defined medium. These two problems could be simultaneously solved if bacterial strains can be progressively adapted to grow in an optimized CD medium, as was achieved for the P. aeruginosa platform, CLIN-1.Citation20

Stabilization/formulation

Each therapeutic agent must be stable during the period of time between its production and its administration to a patient. Moreover, it is important that reconstituting the product is easy at the time of delivery to the patient. With bacteria, it should be possible to achieve stabilization using special media or lyophilization. Here again, recent knowledge concerning the lifestyle of bacteria could aid the pharmaceutic development of live bacterial vectors. For example, in trans expression of the stress tolerance gene, betL, from L. monocytogenes increases resistance to relevant stressors, including osmo-, cryo- and baro-stress, and this gene has been shown to improve tolerance to spray- and freeze-drying in other bacterial strains.Citation30

Which Companies Develop Bacteria-based Active Vaccines?

Three companies developing live bacteria-based vehicles for in vivo antigen delivery inside APCs have been identified. Each of these companies uses engineered L. monocytogenes or P. aeruginosa strains. One project using Salmonella typhimurium was also started in 2008 (VION Pharmaceuticals) but appears to have been terminated.

ADVAXIS SA and ADUROBIOTECH SA have completed the most advanced trials with L. monocytogenes.

ADAVAXIS has 2 ongoing phase 2 trials with their ADXS-HPV product, which is a live Listeria vaccine that secretes the HPV-16 E7 antigen fused to a non-hemolytic fragment of the Lm protein listeriolysin O (LLO) (http://clinicaltrials.gov/ct2/show/NCT01116245?term=advaxis&rank=1 and http://clinicaltrials.gov/ct2/show/NCT01266460?term=listeria&rank=6). The results of the phase 1 trial indicate that the vaccine is safe and that an efficacy signal can be observed.Citation31

ADUROBIOTECH is also conducting a phase 2 clinical trial with CRS-207, a live attenuated Listeria vaccine against mesothelin tumor-associated antigen, for use in pancreatic cancer (http://www.clinicaltrials.gov/ct2/show/NCT01417000?term=NCT01417000&rank=1). CRS-207 was evaluated in a phase 1 trial in end-stage patients with cancers known to express mesothelin, an antigen that is overexpressed in a range of solid tumors.Citation32 The trial revealed a mesothelin-specific T-cell response in multiple patients, and despite an expected survival of 3–5 mo for all subjects, 6 out of 17 lived for at least 15 mo.

APCure is a new French firm that develops (preclinical stage) live P. aeruginosa type III-mediated vectorization of antigens from the Merkel virus, which is responsible for Merkel cell carcinoma.

Perspectives: What is More Exciting than Using Microbes for the Benefit of Mankind?

It is interesting to note that all three of these firms are closely associated with academic laboratories that have explored both the physiology of host/bacteria interactions and bacterial interactions with the immune system and have developed high-level genetic engineering tools. For the Pseudomonas-based vaccine, type III-mediated virulence has been extensively studied by many laboratories throughout the world, but the first demonstration that this bacterium could inject proteins inside immune cells (neutrophils, macrophages, DCs) and that this could help in the development of a novel antigen delivery tool was made by researchers at Grenoble University, from which the APCure firm spun off (Patent number WO2005/049644). This emphasizes the fact that only with in-depth knowledge of the ways in which bacteria interact with their natural hosts and disseminate during infection and a firm understanding of how to engineer bacteria to suit our needs can we develop an innovative therapeutic product. The future of live bacteria-based vaccines is linked to the success of the first line of developed products and to the abilities of scientists to integrate complex data into a synthetic biology approach. Neil S. Forbes have highlighted how synthetic biology techniques can be used to solve many of the key challenges that are associated with bacterial therapies, such as toxicity, stability and efficiency, and to fine-tune their beneficial features, thus enabling the development of ”perfect” cancer therapies.Citation33

References

  • Bachmann MF, Jennings GT. Vaccine delivery: a matter of size, geometry, kinetics and molecular patterns. Nat Rev Immunol 2010; 10:787 - 96; http://dx.doi.org/10.1038/nri2868; PMID: 20948547
  • Banchereau J, Palucka AK. Dendritic cells as therapeutic vaccines against cancer. Nat Rev Immunol 2005; 5:296 - 306; http://dx.doi.org/10.1038/nri1592; PMID: 15803149
  • Blander JM, Sander LE. Beyond pattern recognition: five immune checkpoints for scaling the microbial threat. Nat Rev Immunol 2012; 12:215 - 25; http://dx.doi.org/10.1038/nri3167; PMID: 22362354
  • Chamekh M. Immunomodulation using genetically engineered bacteria for type III-mediated delivery of heterologous antigens and cytokines: potential application in vaccine and therapeutical developments. Immunopharmacol Immunotoxicol 2010; 32:1 - 4; http://dx.doi.org/10.3109/08923970902817890; PMID: 19778176
  • Epaulard O, Derouazi M, Margerit C, Marlu R, Filopon D, Polack B, et al. Optimization of a type III secretion system-based Pseudomonas aeruginosa live vector for antigen delivery. Clin Vaccine Immunol 2008; 15:308 - 13; http://dx.doi.org/10.1128/CVI.00278-07; PMID: 18094108
  • Epaulard O, Toussaint B, Quenee L, Derouazi M, Bosco N, Villiers C, et al. Anti-tumor immunotherapy via antigen delivery from a live attenuated genetically engineered Pseudomonas aeruginosa type III secretion system-based vector. Mol Ther 2006; 14:656 - 61; http://dx.doi.org/10.1016/j.ymthe.2006.06.011; PMID: 17010670
  • Hegazy WA, Xu X, Metelitsa L, Hensel M. Evaluation of Salmonella enterica type III secretion system effector proteins as carriers for heterologous vaccine antigens. Infect Immun 2012; 80:1193 - 202; http://dx.doi.org/10.1128/IAI.06056-11; PMID: 22252866
  • Evans DT, Chen LM, Gillis J, Lin KC, Harty B, Mazzara GP, et al. Mucosal priming of simian immunodeficiency virus-specific cytotoxic T-lymphocyte responses in rhesus macaques by the Salmonella type III secretion antigen delivery system. J Virol 2003; 77:2400 - 9; http://dx.doi.org/10.1128/JVI.77.4.2400-2409.2003; PMID: 12551977
  • Pan ZK, Ikonomidis G, Pardoll D, Paterson Y. Regression of established tumors in mice mediated by the oral administration of a recombinant Listeria monocytogenes vaccine. Cancer Res 1995; 55:4776 - 9; PMID: 7585503
  • Paterson Y, Johnson RS. Progress towards the use of Listeria monocytogenes as a live bacterial vaccine vector for the delivery of HIV antigens. Expert Rev Vaccines 2004; 3:Suppl S119 - 34; http://dx.doi.org/10.1586/14760584.3.4.S119; PMID: 15285711
  • Guirnalda P, Wood L, Paterson Y. Listeria monocytogenes and its products as agents for cancer immunotherapy. Adv Immunol 2012; 113:81 - 118; http://dx.doi.org/10.1016/B978-0-12-394590-7.00004-X; PMID: 22244580
  • Shahabi V, Maciag PC, Rivera S, Wallecha A. Live, attenuated strains of Listeria and Salmonella as vaccine vectors in cancer treatment. Bioeng Bugs 2010; 1:235 - 43; http://dx.doi.org/10.4161/bbug.1.4.11243; PMID: 21327055
  • Sleator RD, Hill C. ‘Bioengineered Bugs’ - a patho-biotechnology approach to probiotic research and applications. Med Hypotheses 2008; 70:167 - 9; http://dx.doi.org/10.1016/j.mehy.2007.03.008; PMID: 17452084
  • Spreng S, Dietrich G, Weidinger G. Rational design of Salmonella-based vaccination strategies. Methods 2006; 38:133 - 43; http://dx.doi.org/10.1016/j.ymeth.2005.09.012; PMID: 16414270
  • Stocker BA. Auxotrophic Salmonella typhi as live vaccine. Vaccine 1988; 6:141 - 5; http://dx.doi.org/10.1016/S0264-410X(88)80017-3; PMID: 3291451
  • Brockstedt DG, Giedlin MA, Leong ML, Bahjat KS, Gao Y, Luckett W, et al. Listeria-based cancer vaccines that segregate immunogenicity from toxicity. Proc Natl Acad Sci U S A 2004; 101:13832 - 7; http://dx.doi.org/10.1073/pnas.0406035101; PMID: 15365184
  • VanCott JL, Chatfield SN, Roberts M, Hone DM, Hohmann EL, Pascual DW, et al. Regulation of host immune responses by modification of Salmonella virulence genes. Nat Med 1998; 4:1247 - 52; http://dx.doi.org/10.1038/3227; PMID: 9809547
  • Sauer JD, Pereyre S, Archer KA, Burke TP, Hanson B, Lauer P, et al. Listeria monocytogenes engineered to activate the Nlrc4 inflammasome are severely attenuated and are poor inducers of protective immunity. Proc Natl Acad Sci U S A 2011; 108:12419 - 24; http://dx.doi.org/10.1073/pnas.1019041108; PMID: 21746921
  • Wang Y, Gouëllec AL, Chaker H, Asrih H, Polack B, Toussaint B. Optimization of antitumor immunotherapy mediated by type III secretion system-based live attenuated bacterial vectors. J Immunother 2012; 35:223 - 34; http://dx.doi.org/10.1097/CJI.0b013e31824747e5; PMID: 22421940
  • Dubensky TW Jr., Skoble J, Lauer P, Brockstedt DG. Killed but metabolically active vaccines. Curr Opin Biotechnol 2012; http://dx.doi.org/10.1016/j.copbio.2012.04.005; PMID: 22608846
  • Irsch J, Lin L. Pathogen Inactivation of Platelet and Plasma Blood Components for Transfusion Using the INTERCEPT Blood System™. Transfus Med Hemother 2011; 38:19 - 31; http://dx.doi.org/10.1159/000323937; PMID: 21779203
  • Brockstedt DG, Bahjat KS, Giedlin MA, Liu W, Leong M, Luckett W, et al. Killed but metabolically active microbes: a new vaccine paradigm for eliciting effector T-cell responses and protective immunity. Nat Med 2005; 11:853 - 60; http://dx.doi.org/10.1038/nm1276; PMID: 16041382
  • Gunn GR, Zubair A, Peters C, Pan ZK, Wu TC, Paterson Y. Two Listeria monocytogenes vaccine vectors that express different molecular forms of human papilloma virus-16 (HPV-16) E7 induce qualitatively different T cell immunity that correlates with their ability to induce regression of established tumors immortalized by HPV-16. J Immunol 2001; 167:6471 - 9; PMID: 11714814
  • Lauer P, Chow MY, Loessner MJ, Portnoy DA, Calendar R. Construction, characterization, and use of two Listeria monocytogenes site-specific phage integration vectors. J Bacteriol 2002; 184:4177 - 86; http://dx.doi.org/10.1128/JB.184.15.4177-4186.2002; PMID: 12107135
  • Verch T, Pan ZK, Paterson Y. Listeria monocytogenes-based antibiotic resistance gene-free antigen delivery system applicable to other bacterial vectors and DNA vaccines. Infect Immun 2004; 72:6418 - 25; http://dx.doi.org/10.1128/IAI.72.11.6418-6425.2004; PMID: 15501772
  • Spreng S, Viret JF. Plasmid maintenance systems suitable for GMO-based bacterial vaccines. Vaccine 2005; 23:2060 - 5; http://dx.doi.org/10.1016/j.vaccine.2005.01.009; PMID: 15755571
  • Curtiss R 3rd, Galan JE, Nakayama K, Kelly SM. Stabilization of recombinant avirulent vaccine strains in vivo. Res Microbiol 1990; 141:797 - 805; http://dx.doi.org/10.1016/0923-2508(90)90113-5; PMID: 1966252
  • Polack B, Vergnaud S, Paclet MH, Lamotte D, Toussaint B, Morel F. Protein delivery by Pseudomonas type III secretion system: Ex vivo complementation of p67(phox)-deficient chronic granulomatous disease. Biochem Biophys Res Commun 2000; 275:854 - 8; http://dx.doi.org/10.1006/bbrc.2000.3399; PMID: 10973811
  • Sheehan VM, Sleator RD, Fitzgerald GF, Hill C. Heterologous expression of BetL, a betaine uptake system, enhances the stress tolerance of Lactobacillus salivarius UCC118. Appl Environ Microbiol 2006; 72:2170 - 7; http://dx.doi.org/10.1128/AEM.72.3.2170-2177.2006; PMID: 16517668
  • Maciag PC, Radulovic S, Rothman J. The first clinical use of a live-attenuated Listeria monocytogenes vaccine: a Phase I safety study of Lm-LLO-E7 in patients with advanced carcinoma of the cervix. Vaccine 2009; 27:3975 - 83; http://dx.doi.org/10.1016/j.vaccine.2009.04.041; PMID: 19389451
  • Singh R, Wallecha A. Cancer immunotherapy using recombinant Listeria monocytogenes: transition from bench to clinic. Hum Vaccin 2011; 7:497 - 505; http://dx.doi.org/10.4161/hv.7.5.15132; PMID: 21422819
  • Forbes NS. Engineering the perfect (bacterial) cancer therapy. Nat Rev Cancer 2010; 10:785 - 94; http://dx.doi.org/10.1038/nrc2934; PMID: 20944664

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.