Light emitting microorganisms and cells for diagnosis and therapy of diseases associated with wounded or inflamed tissue

Abstract
Described is the use of a microorganism or cell containing a DNA sequence encoding a detectable protein or a protein capable of inducing a detectable signal, e.g., a luminescent or fluorescent protein for the preparation of a diagnostic composition for diagnosis and/or visualization of wounded or inflamed tissue or a disease associated therewith. Moreover, therapeutic uses are described, wherein the microorganism or cell additionally contain an expressible DNA sequence encoding a protein suitable for therapy, e.g. an enzyme causing cell death or digestion of debris.
Description

This application is the National Stage of International Application. No. PCT/EP03/05907, filed 5 Jun. 2003, and claims benefit of priority under 35 U.S.C. §365(b) to European Patent Application No. 02012552.2, filed 5 Jun. 2002. The subject matter of the above-noted applications is incorporated herein by reference in its entirety.


The present invention relates to the use of a microorganism or cell containing a DNA sequence encoding a detectable protein or a protein capable of inducing a detectable signal, e.g. a luminescent or fluorescent protein, for the preparation of a diagnostic composition for diagnosis and/or visualization of wounded or inflamed tissue or a disease associated therewith. The present invention also relates to therapeutic uses wherein said microorganism or cell additionally contain an expressible DNA sequence encoding a protein suitable for therapy, e.g. an enzyme causing cell death or digestion of debris.


Bacteremias may arise from traumatic injuries and surgical procedures as well as from physiological functions, such as chewing or tooth brushing. Blood cultures taken before and after invasive procedures and physiological functions from healthy human subjects show that while the premanipulation blood samples are sterile, bacteria are present in the blood in varying frequencies depending on the procedures. A potential consequence of bacteremia is colonization of susceptible sites. However, despite the occurrence of transient bacteremias, only a certain percentage of high-risk patients develop bacterial colonization of potentially susceptible sites. A number of investigators have suggested that bacteria from the blood circulation can colonize inflamed tissues in animal models and on the surface of implanted materials. The inconsistency in the pathological changes in humans following a bacteremia may also be due to the resistance of host immune system, the variability in the concentration of bacteria in the blood subsequent to different bacteremia events, and the virulence of any given bacterial strain.


A number of investigators have focused on the nature of the implanted materials as the factor that influences the ability of bacteria to adhere. Materials such as sutures and surgical clips which are used for closure of wounds, are potential sites of bacterial colonization. Infection of these materials may impede wound healing and/or place patients at increased risk of secondary infections. A variety of wound closure materials have been manufactured with varying affinities for bacteria. Certain wound closure materials, such as braided sutures, have been associated with a higher incidence of infection. The multifilament nature of this type of suture material lends itself to increased susceptibility to bacterial colonization as well as causing a wicking effect that allows penetration of bacteria across the tissues. Mere permanent implantable materials have demonstrated a similar affinity for bacteria. Prosthetic heart valves and joints may be at increased risk of bacterial colonization. It is commonly believed that this higher susceptibility is caused by the inherent ability of bacteria to adhere more readily to the implant surfaces. An alternative explanation may be that inflammation in the tissues surrounding the implants provides an environment that is more suitable for bacterial colonization. In addition to these given possibilities, another factor that may influence the susceptibility of a site, with regards to colonization with bacteria could be the degree of inflammatory status of the affected tissues. Implanted materials may create transient or chronic sites of inflammation in the body.


Presence of implanted materials is not a requirement for bacterial colonization. Alteration of natural anatomical structures that may arise from disease conditions may produce surfaces that are easier to colonize by bacteria. It had been suggested that for the occurrence of infective endocarditis (IE), the valve surface must be altered in order to produce a suitable site for bacterial attachment and colonization. Additionally, the microorganisms have to reach this site and adhere, since it is not possible to produce IE in experimental animals with injections of bacteria unless the valvular surface is damaged. Lesions with high turbulence create conditions that lead to bacterial colonization, whereas defects with a large surface area or low flow are seldom implicated in IE.


However, so far, it could not be proven that transient bacteremias actually cause colonization of inflamed or wounded tissue, since there was no model available allowing the tracing of bacteria in a living organism, i.e. allowing to explain the temporal and spatial relationship between bacterial infections and diseased tissue sites. Moreover, unfortunately, so far the early diagnosis and therapy of inflamed or wounded tissues or diseases associated therewith, e.g., an atherosclerotic disease, endocarditis, pericarditis etc., are unsatisfactory.


Therefore, it is the object of the present invention to provide a means for the efficient and reliable diagnosis as well as the therapy of wounded or inflamed tissue or a disease associated therewith which overcomes the disadvantages of the diagnostic and therapeutic approaches presently used.


According to the present invention this is achieved by the subject matters defined in the claims. In the experiments leading to the present invention it has been found that inflamed tissues, e.g. near implanted material, permit bacterial colonization. Therefore, it is generally possible to visualize inflamed tissues through use of the system of the present invention described below. It could be shown that expression of genes encoding light-emitting proteins in bacteria provides a genetic tool that allows the tracing of the bacteria in a living host, i.e. the evaluation of the dynamics of an infection process in a living host. The external detection of light-emitting bacteria allowed the inventors to non-invasively study the spatial and temporal relationships between infections and the manifested disease conditions. For generation of the light-emitting bacteria, the bacterial luxab operon was used which encodes the enzyme luciferase which catalyzes the oxidation of reduced flavin mononucleotide (FMNE2), in the presence of the substrate, decanal. This reaction then yields FMN, decanoic acid, water and a photon of light. The light photons can then be captured by radiographs, luminometers, or by low light imagers. Recently, the entire bacterial luxcdabe operon, which encodes the substrate as well as the luciferase enzyme, has been used for detection of bacteria in living animals. The advantage of this system is that it does not require exogenously added substrate, which makes it ideal for in vivo studies.


In the studies leading to the present invention, the colonization of wounded and inflamed tissue by bacteria initially present in the circulating blood could be demonstrated and it could be shown that tissues that are irritated by implanted materials such as sutures, wound closure clips and prosthetic devices are more susceptible to bacterial colonization subsequent to bacteremias. The data obtained from experiments with the attenuated S. typhimurium shows that following an intravenous injection, bacteria disseminate throughout the body of the live animals. Therefore, it is reasonable to suggest that the bacteria reach the wounded or inflamed sites via the circulation. These findings described in detail in the examples, below, open the way for (a) designing multifunctional viral vectors useful for the detection of wounded or inflamed tissue based on signals like light emission or signals that can be visualised by MRI and (b) the development of bacterium- and mammalian cell-based wounded or inflamed tissue targeting systems in combination with therapeutic gene constructs for the treatment of diseases associated with wounded or inflamed tissue such as, e.g., an atherosclerotic disease. These systems have the following advantages: (a) They target the wounded or inflamed tissue specifically without affecting normal tissue; (b) the expression and secretion of the therapeutic gene constructs are, preferably, under the control of an inducible promoter, enabling secretion to be switched on or off; and (c) the location of the delivery system inside the tissue can be verified by direct visualisation before activating gene expression and protein delivery. Finally, there are a number of diagnostic methods that could be enhanced or advantageously replaced by the diagnostic approach of the present invention. For example, conventional angiography and MRA techniques and MRA techniques both image blood flowing through the lumen of a vessel to visualize plaque, rather than imaging the plaque directly. MRA is particularly sensitive to turbulence caused by the plaque and, as a result, is often inaccurate. These shortcomings can be overcome by the diagnostic uses of the present invention.


Accordingly, the present invention relates to the use of a microorganism or cell containing a DNA sequence encoding a detectable protein or a protein capable of inducing a detectable signal for the preparation of a diagnostic composition for diagnosis and/or visualization of wounded or inflamed tissue or a disease associated therewith. In addition, said microorganism is also useful for therapy, since following visualization of wounded or inflamed tissue compounds suitable for therapy can be applied, e.g. by topical administration, such as, e.g., acylated iridoid glycosides from Scrophularia nodosa, cortisol, corticosteroid analogs, colchicine, methotrexate, non-steroidal anti-inflammatory drugs (NSAIDs), leflunomide, etanercept, minocycline, cyclosporine, thalidomide, a cytotoxic agent, 6-mercaptopurine, azathioprine, antibiotics or one or more of the proteins listed below.


The present invention also relates to the use of a microorganism or cell containing a DNA sequence encoding a detectable protein or a protein capable of inducing a detectable signal for the preparation of a pharmaceutical composition for the treatment of wounded or inflamed tissue or a disease associated therewith, wherein said micoroorganism or cell furthermore contains one or more expressible DNA sequences encoding (a) proteine(s) suitable for the therapy of wounded or inflamed tissue or diseases associated therewith.


Proteins suitable for the therapy of wounded or inflamed tissue or diseases associated therewith include transforming growth factor (TGF-alpha), platelet-derived growth factor (PDG-F), keratinocyte growth factor (KGF) and insulin-like growth factor-1 (IGF-1), insulin-like growth factor-binding proteins (IGFBPs), IL-4, IL-8, endothelin-1 (ET-1), connective tissue growth factor (CTGF), TNF-alpha, vascular endothelial growth factor (VEGF), cyclooxygenase, cyclooxygenase-2 inhibitor, infliximab (a chimeric anti-TNF-alpha monoclonal antibody), IL-10, lipase, protease, lysozyme, pro-apoptotic factor, peroxisome proliferator-activated receptor (PPAR) agonist etc.


Any microorganism or cell is useful for the diagnostic and therapeutic uses of the present invention, provided that it replicates in the organism, is not pathogenic for the organism e.g. attenuated and, is recognized by the immune system of the organism, etc. The terms “microorganism” and “cell” as used herein refer to microorganisms and cells which are per se not targeted to wounded or inflamed tissues (i.e. they cannot differentiate between wounded or inflamed tissues and the non-wounded or non-inflamed counterpart tissues) since the results of the experiments leading to the present invention show that microorganisms and cells accumulate in wounded or inflamed tissues due to the fact that in this environment they are not exposed to attack by the immune system of the host. The microorganisms and cells accumulate for a specific time, e.g. 3 to 5 days, as long as the vascularization/lymphatic system has not been restored.


In a preferred embodiment, the microorganism or cell contains a DNA sequence encoding a luminescent and/or fluorescent protein. As used herein, the term “DNA sequence encoding a luminescent or fluorescent protein” also comprises a DNA sequence encoding a luminescent and fluorescent protein as fusion protein.


In an alternative preferred embodiment of the use of the present invention, the microorganism or cell contains a DNA sequence encoding a protein capable of inducing a signal detectable by magnetic resonance imaging (MRI), e.g. a metal binding protein. Furthermore, the protein can bind a contrasting agent, chromophore, or a compound required for visualization of tissues.


Suitable devices for analysing the localization or distribution of luminescent and/or fluorescent proteins in a tissue are well known to the person skilled in the art and, furthermore described in the literature cited above as well as the examples, below.


Preferably, for transfecting the cells the DNA sequences encoding a detectable protein or a protein capable of inducing a detectable signal, e.g., a luminescent or fluorescent protein, are present in a vector or an expression vector. A person skilled in the art is familiar with examples thereof. The DNA sequences can also be contained in a recombinant virus containing appropriate expression cassettes. Suitable viruses that may be used include baculovirus, vaccinia, sindbis virus, Sendai virus, adenovirus, an AAV virus or a parvovirus, such as MVM or H-1. The vector may also be a retrovirus, such as MoMULV, MoMuLV, HaMuSV, MuMTV, RSV or GaLV. For expression in mammals, a suitable promoter is e.g. human cytomegalovirus “immediate early promoter” (pCMV). Furthermore, tissue and/or organ specific promoters are useful. Preferably, the DNA sequences encoding a detectable protein or a protein capable of inducing a detectable signal are operatively linked with a promoter allowing high expression. Such promoters, e.g. inducible promoters are well-known to the person skilled in the art.


For generating the above described DNA sequences and for constructing expression vectors or viruses which contain said DNA sequences, it is possible to use general methods known in the art. These methods include e.g. in vitro recombination techniques, synthetic methods and in vivo recombination methods as described in Sambrook et al., Molecular Cloning, A Laboratory Manual, 2nd edition (1989) Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., for example. Methods of transfecting cells, of phenotypically selecting transfectants and of expressing the DNA sequences by using the above described vectors are known in the art.


The person skilled in the art knows DNA sequences encoding luminescent or fluorescent proteins that can be used for carrying out the present invention. During the past decade, the identification and isolation of structural genes encoding light-emitting proteins from bacterial luciferase from Vibrio harveyi (Belas et al., Science 218 (1982), 791-793) and from Vibrio fischerii (Foran and Brown, Nucleic acids Reds. 16 (1988), 177), firefly luciferase (de Wet et al., Mol. Cell. Biol. 7 (1987), 725-737), aequorin from Aequorea victoria (Prasher et al., Biochem. 26 (1987), 1326-1332), Renilla luciferase from Renilla reniformis (Lorenz et al., PNAS USA 88 (1991), 4438-4442) and green fluorescent protein from Aequorea victoria (Prasher et al., Gene 111 (1987), 229-233) have been described that allow the tracing of bacteria or viruses based on light emission. Transformation and expression of these genes in bacteria allows detection of bacterial colonies with the aid of the low light imaging camera or individual bacteria under the fluorescent microscope (Engebrecht et al., Science 227 (1985), 1345-1347; Legocki et al., PNAS 83 (1986), 9080-9084; Chalfie et al., Science 263 (1994), 802-805).


Luciferase genes have been expressed in a variety of organisms. Promoter activation based on light emission, using luxAB fused to the nitrogenase promoter, was demonstrated in Rhizobia residing within the cytoplasm of cells of infected root nodules by low light imaging (Legocki et al., PNAS 83 (1986), 9080-9084; O'Kane et al., J. Plant Mol. Biol. 10 (1988), 387-399). Fusion of the lux A and lux B genes resulted in a fully functional luciferase protein (Escher et al., PNAS 86 (1989), 6528-6532). This fusion gene (Fab2) was introduced into Bacillus subtilis and Bacillus megatherium under the xylose promoter and then fed into insect larvae and was injected into the hemolymph of worms. Imaging of light emission was conducted using a low light video camera. The movement and localization of pathogenic bacteria in transgenic arabidopsis plants, which carry the pathogen-activated PAL promoter-bacterial luciferase fusion gene construct, was demonstrated by localizing Pseudomonas or Ervinia spp. infection under the low light imager as well as in tomato plant and stacks of potatoes (Giacomin and Szalay, Plant Sci. 116 (1996), 59-72).


Thus, in a more preferred embodiment, the luminescent or fluorescent protein present in the above described microorganism or cell is luciferase, RFP or GFP.


All of the luciferases expressed in bacteria require exogenously added substrates such as decanal or coelenterazine for light emission. In contrast, while visualization of GFP fluorescence does not require a substrate, an excitation light source is needed. More recently, the gene cluster encoding the bacterial luciferase and the proteins for providing decanal within the cell, which includes luxCDABE was isolated from Xenorhabdus luminescens (Meighen and Szittner, J. Bacteriol. 174 (1992), 5371-5381) and Photobacterium leiognathi (Lee et al., Eur. J. Biochem. 201 (1991), 161-167) and transferred into bacteria resulting in continuous light emission independent of exogenously added substrate (Fernandez-Pinas and Wolk, Gene 150 (1994), 169-174). Bacteria containing the complete lux operon sequence, when injected intraperitoneally, intramuscularly, or intravenously, allowed the visualization and localization of bacteria in live mice indicating that the luciferase light emission can penetrate the tissues and can be detected externally (Contag et al., Mol. Microbiol. 18 (1995), 593-603).


Thus, in an even more preferred embodiment, the microorganism or cell containing a DNA sequence encoding a luciferase additionally contains a gene encoding a substrate for a luciferase.


Preferably, the microorganism is a bacterium. Particularly preferred is attenuated Salmonella thyphimurium, attenuated Vibrio cholerae, attenuated Listeria monocytogenes or E. coli.


Alternatively, viruses such as Vaccinia virus, AAV, a retrovirus etc. are also useful for the diagnostic and therapeutic uses of the present invention. Preferably, the virus is Vaccinia virus.


Preferably, the cell for the uses of the present invention is a mammalian cell such as a stem cell which can be autologous or heterologous concerning the organism.


In a further preferred embodiment, the microorganism or cell useful in the present invention contains a ruc-gfp expression cassette which contains the Renilla luciferase (ruc) and Aequorea gfp cDNA sequences under the control of a strong synthetic early/late (PE/L) promoter of Vaccinia or the luxCDABE cassette.


In a preferred use of the microorganisms and cells described above the protein suitable for the therapy of diseases associated with wounded or inflamed tissue like atherosclerotic diasease is an enzyme causing cell death or an enzyme causing the digestion of debris, e.g. in the interior of an atherosclerotic plaque causing the plaque to collapse under the force of the intraluminal blood pressure. Suitable enzymes include a lipase, protease, lysozyme, proapoptotic factor, PPAR-agonist etc. If the inflammatory component of atherosclerosis should be treated suitable compounds are cortisol, corticosteroid analogs, cyclooxygenase and cyclooxygenase-2 inhibitors, colchicine, methotrexate, NSAIDs, leflunomide, etanercept, minocycline, cyclosporine, thalidomide, infliximab, IL-10, 6-mercaptopurine, azathioprine or a cytotoxic agent. Some of these compounds might be in the form of pro-drugs.


Accordingly, the protein expressed by a microorganism of the invention can be an enzyme converting an inactive substance (pro-drug) administered to the organism into an active substance.


Preferably, the gene encoding an enzyme as discussed above is directed by an inducible promoter additionally ensuring that, e.g., the conversion of the pro-drug into the active substance only occurs in the target tissue, e.g., an IPTG-, antibiotic-, heat-, pH-, light-, metal-, aerobic-, host cell-, drug-, cell cycle- or tissue specific-inducible promoter. Moreover, the delivery system of the present invention even allows the application of compounds which could so far not be used for therapy due to their high toxicity when systemically applied or due to the fact that they cannot be administered, e.g., intravenously in sufficiently high dosages to achieve levels inside, e.g., sinuses, abscesses or across the blood brain barrier. Such compounds include thalidomide, cytotoxic drugs, antibiotics etc.


Furthermore, the microorganism or cell of the present invention can contain a BAC (Bacterial Artificial Chromosome) or MAC (Mammalian Artificial Chromosome) encoding several or all proteins of a specific pathway, e.g. woundhealing-pathway, such as TNF-alpha, COX-2, CTGF etc. Additionally, the cell can be a cyber cell or cyber virus encoding these proteins.


For administration, the microorganisms or cells described above are preferably combined with suitable pharmaceutical carriers. Examples of suitable pharmaceutical carriers are well known in the art and include phosphate buffered saline solutions, water, emulsions, such as oil/water emulsions, various types of wetting agents, sterile solutions etc. Such carriers can be formulated by conventional methods and can be administered to the subject at a suitable dose. Administration of the microorganisms or cells may be effected by different ways, e.g. by intravenous, intraperetoneal, subcutaneous, intramuscular, topical or intradermal administration. The preferred route of administration is intravenous injection. The route of administration, of course, depends on the nature of the tissue and the kind of microorganisms or cells contained in the pharmaceutical composition. The dosage regimen will be determined by the attending physician and other clinical factors. As is well known in the medical arts, dosages for any one patient depends on many factors, including the patient's size, body surface area, age, sex, the particular compound to be administered, time and route of administration, the kind and localisation of the tissue, general health and other drugs being administered concurrently.


A preferred therapeutical use is the preparation of a pharamaceutical composition for the treatment of endocarditis, pericarditis, imflammatory bowel disease (e.g. Crohn's disease or Ulcerative colitis), low back pain (herniated nucleus pulposis), temporal arteritis, polyarteritis nodosa or an arthritic disease.


In the past few years, there has been many reports showing evidence for Chlamydia pneumoniae, Heliobacter pylori, CMV, HSV and other infectious agents inside atherosclerotic plaques. The presence of these infectious agents within atherosclerotic plaque suggests that the interior of the plaque is a protected environment that permits replication, otherwise these infectious agents would be cleared by the immune system. Moreover, there is considerable evidence that an inflammatory process is present within the interior of atherosclerotic plaque. Accordingly, it is reasonable to assume that this disease can be diagnosed and treated by the microorganisms or cells of the present invention that—after intravenous injection—will penetrate into the atherosclerotic plaque where they start to replicate. After a suitable period of time, the plaque can be imaged using, e.g., light sensitive cameras or suitable MRI equipment. Further, said microorganisms or cells can additionally produce an enzyme, e.g. an enzyme as described above, resulting in the elimination of plaques. Thus, a further preferred use is the diagnosis and treatment of an atherosclerotic disease.


A further preferred use is the diagnosis and treatment of coronary artery disease, peripheral vascular disease or cerebral artery disease. Therapeutic treatments according to the present invention might replace treatments like balloon angioplasty, stent placement, coronary artery bypass graft, carotid endarterectomy, aorto-femoral bypass graft and other invasive procedures. Moreover, plaque in inaccessible regions, such as the basilar and middle cerebral arteries can be treated using the therapeutic approach of the present invention.


For the therapy of wounds, fractures, surgical incisions and burns the microorganisms of the present invention are preferably combined with proteins like transforming growth factor (TGF-alpha), platelet-derived growth factor (PDG-F), keratinocyte growth factor (KGF) and insulin-like growth factor-1 (IGF-1), insulin-like growth factor-binding proteins (IGFBPs), IL-4, IL-8, endothelin-1 (ET-1), connective tissue growth factor (CTGF), TNF-alpha, vascular endothelial growth factor (VEGF), cyclooxygenase, cyclooxygenase-2 inhibitor, infliximab (a chimeric anti-TNF-alpha monoclonal antibody), IL-10, lipase, protease, lysozyme, pro-apoptotic factor, peroxisome proliferator-activated receptor (PPAR) agonist (or contain expressible DNA-sequences encoding said proteins). For the treatment of infectious diseases, the microorganisms of the present invention are preferably applied in combination with antibiotics. For the treatment of auto-immune and inflammatory diseases, including reumathoid arthritis, inflammatory bowel disease and multiple sclerosis, the microorganisms of the present invention are preferably applied in combination with cortisol, corticosteroid analogs, cyclooxygenase and cyclooxygenase-2 inhibitors, colchicine, methotrexate, NSAIDs, leflunomide, etanercept, minocycline, cyclosporine, thalidomide, infliximab, IL-10, 6-mercaptopurine, azathioprine or a cytotoxic agent. For the therapy of diseases like atherosclerosis, the microorganisms of the present invention are preferably applied in combination with lipases, lysozymes, pro-apoptopic factors, PPAR-agonists (or the corresponding DNA-sequences) or an agent listed above with respect to the treatment of inflammatory diseases. For the treatment of Alzheimer's disease, the microorganisms of the present invention are preferably applied in combination with one or more agents listed above with respect to auto-immune- or inflammatory diseases.


Finally, the above described microorganisms and cells are useful for (a) monitoring the efficacy of an antibiotic regimen, preferably based on light extinction or (b) comparing the resistance of various sutures and implantable materials to bacterial colonization.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1: Visualization of Bacteria Intravenously Injected into Nude Mice


Nude mice were injected with 1×107 attenuated Salmonella typhimurium (A) or 1×107 attenuated Vibrio cholera (B). Both strains were transformed with pLITE201 carrying the lux operon. Photon collection was for one minute 20 min after bacterial injections.



FIG. 2: Visualization of S. typhimurium in the Same Animal Over a 5-Day Observation Period


Nude mice were injected with 1×107 attenuated S. typhimurium. On the first observation period, bacteria were disseminated throughout the body of the animal (A). Two days later, bacteria were cleared from the animal with the exception of the incision wound and the ear tag region as indicated by the arrows (B). On day 5, the animal had been able to clear the organism from the wounded regions (C).



FIG. 3: Visualization of V. cholera in the Same Animal Over an 8-Day Observation Period


Nude mice were injected with 1×107 attenuated V. cholera. On the first observation period, bacteria were visualized in the liver region of the animal (A). Five days later, bacteria were cleared from the entire animal with the exception of the incision wound as indicated by the arrows (B). On day 8, the animal had been able to clear the organism from the wound (C).



FIG. 4: Visualization of V. cholera in an Immunocompetent C57 Mouse


1×107 attenuated V. cholera were intravenously injected into the animal. Light-emitting bacteria colonized the ear tag on the forth day after bacterial injection (indicated by the white arrow).



FIG. 5: Visualization of Light Emitting Bacteria in the Liver of Rats


Sprague Dawley rats were intravenously injected with 1×108 attenuated E. coli transformed with the plasmid DNA pLITE201 carrying the luxcdabe operon. Photons were collected immediately after infection for one minute under the low light imager (Night Owl). Light emitting bacteria were visualized in the liver of the whole live animal.



FIG. 6: Colonization of Rat Hearts with Light Emitting Bacteria


Intravenous injection of the rats with 1×108 attenuated E. coli transformed with the plasmid pLITE201 carrying the luxedabe operon did not lead to colonization of the hearts of control animals, which had not been catheterized (A). Similar induction of bacteremias in rats catheterized through the right carotid artery lead to the colonization of the heart with light emitting bacteria (B).



FIG. 7: Detection of Residual Bacteria in the Organs of Rats


Three days following intravenous injection of the rats with 1×108 attenuated E. coli, the hearts, livers, and spleens were excised and cultured overnight. Light emitting bacteria were visualized under the low light imager (Hamamatsu) in all specimens from the catheterized rats (A-C), while in the control animals, bacteria were detected in the liver (A) and spleen (B) but not the heart (C).





The present invention is explained by the following examples.


EXAMPLE 1
Materials and Methods

(A) Bacterial Strains


The strains used were a non-pathogenic laboratory strain Escherichia coli, strain DH5α, attenuated Salmonella typhimurium (SL7207 hisG46, DEL407 [aroA544::Tn101] and attenuated Vibrio cholerae (Bengal 2 Serotyp 0139, M010 DattRSI).


(B) Plasmid Constructs


The plasmid DNA pLITE201 containing the luxcdabe gene cassette was obtained from Dr. F. Marines (Voisey and Marines, Biotech. 24 (1998) 56-58).


(C) Recipient Animals


Five- to six-week-old male BALB/c nu/nu mice (25-30 g body weight) and Sprague Dawley rats (300-325 g body weight) were purchased from Harlan (Frederick, Md., USA). CS7BL/6J mice were obtained from Jackson Laboratories (Bar Harbor, Me., USA). All animal experiments were carried out in accordance with protocols approved by the Lorna Linda University animal research committee. The animals containing recombinant DNA materials and attenuated pathogens were kept in the Loma Linda University animal care facility at biosafety level two.


(D) Detection of Luminescence


Immediately before imaging, the animals were anesthetized with intraperitoneal injections of sodium pentobarbital (Nembutal® Sodium solution, Abbot Laboratories, North Chicago, Ill.; 60 mg/kg body weight). The animals were placed inside the dark box for photon counting and recording superimposed images (ARGUS 100 Low Light Imaging System, Hamamatsu, Hamamatsu, Japan and Night Owl, Berthold Technologies, GmbH and Co. KG, Bad Wildbad, Germany). Photon collection was for one minute from ventral and dorsal views of the animals. A photographic image was then recorded and the low light image was superimposed over the photographic image to demonstrate the location of luminescent activity.


EXAMPLE 2
Colonization of Cutaneous Wounds by Intravenously Injected Light Emitting Bacteria in Live Animals

To determine the fate of intravenously injected luminescent bacteria in the animals, 1×107 bacteria carrying the pLITE201 plasmid DNA in 50 μl were injected into the left femoral vein of nude mice under anesthesia. To expose the femoral vein, a 1-cm incision was made with a surgical blade. Following closure of the incision with 6-0 sutures, the mice were monitored under the low light imager and photon emissions were collected for one minute. Imaging of each animal was repeated at various time intervals to study the dissemination of the light-emitting bacteria throughout the body of the animals. It was found that the distribution pattern of light emission following an intravenous injection of bacteria into the mice was bacterial-strain-dependent. Injection of attenuated S. typhimurium caused wide dissemination of the bacteria throughout the body of the animals (FIG. 1A). This pattern of distribution was visible within 5 minutes after bacterial injection and continued to be detected at the one-hour observation period. Injection of attenuated V. cholera into the bloodstream, however, resulted in light emission that was localized to the liver within 5 minutes after bacterial injection and remained visible in the liver at the one-hour observation period (FIG. 1B).


The difference in the bacterial distribution patterns suggests a difference in the interaction of these strains with the host once inside the animal. Imaging the same animals 48 h after bacterial injection showed that all of the detectable light emission from the earlier time had diminished and was eliminated completely from the injected animal with the exception of the inflamed wounded tissues such as the incision wound and the ear tag region. Inflammation in these tissues was identified by their red and edematous appearance. Light emission was detected in the incision wound and/or in the inflamed ear tag region up to 5 to 8 days postinjection, which was confirmed by longer photon collection times, i.e. 10 minutes (FIG. 2A-C and FIG. 3A-C). The absence of light emission was not due to the loss of the plasmid DNA or the silencing of gene expression in the bacteria. In other experiments light emission in animals could be consistently detected for up to 50 days. Similar data were obtained in immunocompetent C57BU6J mice (FIG. 4), showing that these observations are not limited to animals with altered immune systems. Careful examination of individually excised organs as well as blood samples from infected animals confirmed the absence of luminescence in these normal uninjured tissues. Furthermore, the experimental data demonstrated that colonization of the injured tissues is a common occurrence in mice. Twenty-four of 29 incision wounds (82.8%) and 12 of 29 ear tags (41.4%) in the mice were colonized by intravenously injected bacteria. Wound colonization by intravenously injected bacteria occurred following injection of V. cholera in concentrations as low as 1×105 bacterial cells.


EXAMPLE 3
Colonization of Catheterized Rat Hearts Subsequent to Femoral Vein Injection of Light-Emitting Bacteria

Surgical heart defects were created according to the procedures previously described (Santoro and Levison, Infect. Immun. 19(3)(1978), 915-918; Overholser et al., J. Infect. Dis. 155(1)(1987), 107-112). Briefly, animals were anesthetized with sodium pentobarbital (60 mg/kg i.p.). A midline neck incision was made to expose the tight carotid artery. A polypropylene catheter was introduced and advanced until resistance was met indicating insertion to the level of the aortic valve. The catheter was then secured using a 10-0 suture (AROSurgical Instrument Corporation, Japan) and the incision was closed using 4-0 silk sutures (American Cyanamide Company, Wayne, N.J.). Placement of the catheter causes irritation and subsequent inflammation of the aortic valve (Santoro and Levison, 1978). Control animals did not undergo the catheterization procedure. Bacteremias were induced by injection of 1×108 light-emitting bacterial cells of E. coli via the femoral vein. When observed immediately after infection under the low light imager, bacterial colonization was visible in the liver region (FIG. 5). Three days later, while catheterized animals consistently demonstrated colonization of the heart with light emitting bacteria, control animals showed no sign of light emission from the heart (FIG. 6). To determine if low and undetectable levels of bacteria were present in the tissues, the heart, liver and spleen were excised from each animal and cultured overnight. The livers and spleens of the rats, which are organs that are directly involved in bacterial clearance, in both groups showed presence of light emitting bacteria. Strong light emission was detected in the catheterized heart in contrast to the control heart, which had complete absence of emitted light (FIG. 7). No bacteria were detected on the cultured catheters.


These findings indicate that while light-emitting bacteria injected into the bloodstream via the femoral vein were cleared from normal tissues, injured or inflamed tissues in immunocompromised and immunocompetent animals provided sites that continued to retain bacteria for an extended period of time.

Claims
  • 1. A method for diagnosis by imaging wounded or inflamed tissue inside of a live subject, comprising: identifying a subject suspected of having an internal wound or inflammation to be tested for the presence or absence of internal wounded tissue or internal inflamed tissue;systemically administering to the subject in whom the presence or absence of a wounded tissue or inflamed tissue is to be detected, a bacterium, wherein: the bacterium encodes a protein that is detectable by imaging in the subject or encodes a protein that induces a signal detectable by imaging;the bacterium replicates in the subject;the bacterium is not pathogenic to the subject and is recognized by the immune system of the subject;the bacterium is not targeted; andafter a sufficient time for the bacterium to accumulate in wounded or inflamed tissues inside of the subject, imaging the detectable bacterium inside of the live subject to detect accumulation of the bacterium in the subject, and thereby imaging wounded or inflamed tissues inside of the live subject, wherein imaging the accumulation indicates the location of wounded tissue or inflamed tissue inside of the subject.
  • 2. The method of claim 1, wherein the bacterium encodes a protein(s) for the therapy of the imaged wounded or inflamed tissue.
  • 3. The method of claim 1, wherein the bacterium encodes a luminescent or fluorescent protein.
  • 4. The method of claim 1, wherein the bacterium encodes a luciferase, red fluorescent protein or green fluorescent protein.
  • 5. The method of claim 4, wherein the bacterium encodes a luciferase and a protein(s) for the production of a substrate for a luciferase.
  • 6. A method for detecting wounded or inflamed tissue inside a subject, comprising: identifying a subject to be tested for the presence or absence of wounded tissue or inflamed tissue therein;systemically administering to the subject in whom the presence or absence of a wounded tissue or inflamed tissue is to be detected, a bacterium, wherein: the bacterium is detectable by imaging in the subject;the bacterium replicates in the subject;the bacterium is not pathogenic to the subject and is recognized by the immune system of the subject;the bacterium is not targeted; andafter a sufficient time for the bacterium to accumulate in wounded or inflamed tissues inside of the subject, imaging the detectable bacterium inside of the subject to detect accumulation of the bacterium in the subject, and thereby detecting wounded or inflamed tissues inside of the subject, wherein: detection of the accumulation indicates the location of wounded tissue or inflamed tissue inside of the subject; and the bacterium encodes a protein that induces a signal detectable by magnetic resonance imaging (MRI) or that binds to contrasting agent, chromophore or a ligand.
  • 7. The method of claim 1, wherein the bacterium is selected among an attenuated Salmonella typhimurium, an attenuated Vibrio cholerae, an attenuated Listeria monocytogenes and E. coli.
  • 8. The method of claim 2, wherein the protein for the therapy is an enzyme that causes cell death or an enzyme that causes the digestion of debris.
  • 9. The method of claim 2, wherein the subject in whom the presence or absence of wounded tissue or inflamed tissue is detected has a disease selected among endocarditis, pericarditis, inflammatory bowel disease, low back pain (herniated nucleus pulposis), temporal arteritis, polyarteritis nodosa and an arthritic disease.
  • 10. A method for detecting wounded or inflamed tissue inside of a subject, comprising: identifying a subject to be tested for the presence or absence of wounded tissue or inflamed tissue therein;systemically administering to the subject in whom the presence or absence of a wounded tissue or inflamed tissue is to be detected, a bacterium, wherein: the bacterium is detectable by imaging in the subject;the bacterium replicates in the subject;the bacterium is not pathogenic to the subject and is recognized by the immune system of the subject;the bacterium is not targeted; andafter a sufficient time for the bacterium to accumulate in wounded or inflamed tissues inside of the subject, imaging the detectable bacterium inside of the subject to detect accumulation of the bacterium in the subject, and thereby detecting wounded or inflamed tissues inside of the subject, wherein: detection of the accumulation indicates the location of wounded tissue or inflamed tissue inside of the subject;the bacterium encodes a protein(s) for the therapy of the detected imaged wounded or inflamed tissue; andthe subject for whom the presence or absence of wounded tissue or inflamed tissue is detected has an atherosclerotic disease.
  • 11. The method of claim 2, wherein the subject for whom the presence or absence of wounded tissue or inflamed tissue is detected has a disease that is selected among coronary artery disease, peripheral vascular disease and cerebral artery disease.
  • 12. A method for detecting wounded or inflamed tissue inside of a subject, comprising: identifying a subject to be tested for the presence or absence of wounded tissue or inflamed tissue therein;systemically administering to the subject in whom the presence or absence of a wounded tissue or inflamed tissue is to be detected, a bacterium, wherein: the bacterium is detectable by imaging in the subject;the bacterium replicates in the subject;the bacterium is not pathogenic to the subject and is recognized by the immune system of the subject;the bacterium is not targeted; andafter a sufficient time for the bacterium to accumulate in wounded or inflamed tissues inside of the subject, imaging the detectable bacterium inside of the subject to detect accumulation of the bacterium in the subject, and thereby detecting wounded or inflamed tissues inside of the subject, wherein: detection of the accumulation indicates the location of wounded tissue or inflamed tissue inside of the subject; andthe detecting is performed by magnetic resonance imaging (MRI).
  • 13. The method of claim 2, wherein the bacterium contains an inducible promoter that regulates the expression of the therapeutic protein.
  • 14. A method for detecting wounded or inflamed tissue inside of a subject, comprising: identifying a subject to be tested for the presence or absence of wounded tissue or inflamed tissue therein;systemically administering to the subject in whom the presence or absence of a wounded tissue or inflamed tissue is to be detected, a bacterium, wherein: the bacterium is detectable by imaging in the subject;the bacterium replicates in the subject;the bacterium is not pathogenic to the subject and is recognized by the immune system of the subject;the bacterium is not targeted; andafter a sufficient time for the bacterium to accumulate in wounded or inflamed tissues inside of the subject, imaging the detectable bacterium inside of the subject to detect accumulation of the bacterium in the subject, and thereby detecting wounded or inflamed tissues inside of the subject, wherein: detection of the accumulation indicates the location of wounded tissue or inflamed tissue inside of the subject;the disease is an atherosclerotic disease; andthe therapeutic protein is selected from among a lipase, protease, lysozyme, proapoptotic factor and PPAR-agonist.
  • 15. The method of claim 1, further comprising: administering a therapeutic agent for the therapy of a wounded tissue, inflamed tissue or a disease associated therewith.
  • 16. A method for detecting wounded or inflamed tissue inside of a subject, comprising: intravenously or intramuscularly administering to a subject in whom the presence or absence of a wounded tissue or inflamed tissue is to be detected, a bacterium, wherein: the bacterium is detectable in the subject;the bacterium replicates in the subject;the bacterium is not pathogenic to the subject and is recognized by the immune system of the subject;the bacterium is not targeted; andafter a sufficient time for the bacterium to accumulate in wounded or inflamed tissues inside of the subject, monitoring or imaging the detectable bacterium inside of the subject to detect accumulation of the bacterium in the subject, and thereby detecting wounded or inflamed tissues inside of the subject.
  • 17. The method of claim 16, further comprising: administering a therapeutic agent for the therapy of a wounded tissue, inflamed tissue or a disease associated therewith.
  • 18. The method of claim 16, wherein the bacterium is selected among an attenuated Salmonella typhimurium, an attenuated Vibrio cholerae, an attenuated Listeria monocytogenes and E. coli.
  • 19. The method of claim 16, wherein the bacterium encodes a protein(s) for the therapy of the detected wounded or inflamed tissue.
  • 20. The method of claim 16, wherein the bacterium encodes a luminescent or fluorescent protein.
  • 21. The method of claim 16, wherein the bacterium encodes a luciferase, red fluorescent protein or green fluorescent protein.
  • 22. The method of claim 21, wherein the bacterium encodes a luciferase and a protein(s) for the production of a substrate for a luciferase.
  • 23. The method of claim 19, wherein the protein for the therapy is an enzyme that causes cell death or an enzyme that causes the digestion of debris.
  • 24. The method of claim 17, wherein the subject in whom the presence or absence of wounded tissue or inflamed tissue is detected has a disease selected among endocarditis, pericarditis, inflammatory bowel disease, low back pain (herniated nucleus pulposis), temporal arteritis, polyarteritis nodosa and an arthritic disease.
Priority Claims (1)
Number Date Country Kind
02012552 Jun 2002 EP regional
PCT Information
Filing Document Filing Date Country Kind 371c Date
PCT/EP03/05907 6/5/2003 WO 00 6/27/2005
Publishing Document Publishing Date Country Kind
WO03/104485 12/18/2003 WO A
US Referenced Citations (124)
Number Name Date Kind
4442203 Varshavsky Apr 1984 A
4722848 Paoletti et al. Feb 1988 A
4778759 Szalay et al. Oct 1988 A
5221623 Legocki et al. Jun 1993 A
5283187 Aebischer et al. Feb 1994 A
5300436 Goldstein et al. Apr 1994 A
5550050 Holland et al. Aug 1996 A
5639275 Baetge et al. Jun 1997 A
5646298 Powell et al. Jul 1997 A
5650135 Contag et al. Jul 1997 A
5650148 Gage et al. Jul 1997 A
5653975 Baetge et al. Aug 1997 A
5656481 Baetge et al. Aug 1997 A
5676943 Baetge et al. Oct 1997 A
5693533 Raney et al. Dec 1997 A
5704910 Humes Jan 1998 A
5718902 Yilma et al. Feb 1998 A
5750103 Cherksey May 1998 A
5756455 Kinzler et al. May 1998 A
5762959 Soon-Shiong et al. Jun 1998 A
5795790 Schinstine et al. Aug 1998 A
5798113 Dionne et al. Aug 1998 A
5800828 Dionne et al. Sep 1998 A
5800829 Dionne et al. Sep 1998 A
5830702 Portnoy et al. Nov 1998 A
5833975 Paoletti et al. Nov 1998 A
5833979 Schinstine et al. Nov 1998 A
5834001 Dionne et al. Nov 1998 A
5837234 Gentile et al. Nov 1998 A
5840576 Schinstine et al. Nov 1998 A
5842431 Wu Dec 1998 A
5853385 Emerich et al. Dec 1998 A
5853717 Schinstine et al. Dec 1998 A
5861290 Goldsmith et al. Jan 1999 A
5976796 Szalay et al. Nov 1999 A
6007806 Lathe et al. Dec 1999 A
6025155 Hadlaczky et al. Feb 2000 A
6045802 Schlom et al. Apr 2000 A
6077697 Hadlaczky et al. Jun 2000 A
6080849 Bermudes et al. Jun 2000 A
6093700 Mastrangelo et al. Jul 2000 A
6099848 Frankel et al. Aug 2000 A
6106826 Brandt et al. Aug 2000 A
6150170 Powell et al. Nov 2000 A
6190657 Pawelek et al. Feb 2001 B1
6217847 Contag et al. Apr 2001 B1
6232523 Tan et al. May 2001 B1
6235967 Tan et al. May 2001 B1
6235968 Tan et al. May 2001 B1
6251384 Tan et al. Jun 2001 B1
6265557 Diamond et al. Jul 2001 B1
6359189 Fleischmann Mar 2002 B1
6416754 Brown et al. Jul 2002 B1
6428968 Molnar-Kimber et al. Aug 2002 B1
6455673 Collier Sep 2002 B1
6491905 Sorscher et al. Dec 2002 B1
6503703 Palese et al. Jan 2003 B1
6511967 Weissleder et al. Jan 2003 B1
6548068 Schlom et al. Apr 2003 B1
6589531 Andino-Pavlovsky et al. Jul 2003 B1
6596279 Paoletti et al. Jul 2003 B1
6627190 Wold et al. Sep 2003 B2
6649143 Contag et al. Nov 2003 B1
6649159 Yang et al. Nov 2003 B2
6652849 Brown et al. Nov 2003 B2
6685935 Pawelek et al. Feb 2004 B1
6713293 Grummt et al. Mar 2004 B1
6743967 Hadlaczky et al. Jun 2004 B2
6759038 Tan et al. Jul 2004 B2
6916462 Contag et al. Jul 2005 B2
6984374 Szalay et al. Jan 2006 B2
7588767 Szalay et al. Sep 2009 B2
7588771 Szalay et al. Sep 2009 B2
7662398 Szalay et al. Feb 2010 B2
7763420 Stritzker et al. Jul 2010 B2
20010008025 Hadlaczky et al. Jul 2001 A1
20010029023 Szalay et al. Oct 2001 A1
20020054865 Fujimori et al. May 2002 A1
20020160410 Hadlaczky et al. Oct 2002 A1
20020160970 Hadlaczky et al. Oct 2002 A1
20030009015 Ulrich et al. Jan 2003 A1
20030031628 Zhao et al. Feb 2003 A1
20030031681 Mc Cart et al. Feb 2003 A1
20030033617 Hadlaczky et al. Feb 2003 A1
20030044384 Roberts et al. Mar 2003 A1
20030059400 Szalay Mar 2003 A1
20030083293 Hadlaczky et al. May 2003 A1
20030086906 Mastrangelo et al. May 2003 A1
20030101480 Hadlaczky et al. May 2003 A1
20030103941 Crombleholme et al. Jun 2003 A1
20030133949 Szalay et al. Jul 2003 A1
20030161788 Zhao et al. Aug 2003 A1
20030165465 Roberts et al. Sep 2003 A1
20030165477 Balloul et al. Sep 2003 A1
20030198627 Arts et al. Oct 2003 A1
20030213007 Slattery et al. Nov 2003 A1
20030228261 Szalay et al. Dec 2003 A1
20030228330 Falkner et al. Dec 2003 A1
20040076622 Studeny et al. Apr 2004 A1
20040143861 Hadlaczky et al. Jul 2004 A1
20040213741 Szalay et al. Oct 2004 A1
20040234455 Szalay et al. Nov 2004 A1
20050025745 Fujimori et al. Feb 2005 A1
20050031643 Szalay et al. Feb 2005 A1
20050063993 Schlom et al. Mar 2005 A1
20050069491 Yu et al. Mar 2005 A1
20060051370 Szalay et al. Mar 2006 A1
20070025981 Szalay et al. Feb 2007 A1
20070202572 Szalay et al. Aug 2007 A1
20070212727 Szalay et al. Sep 2007 A1
20080193373 Stritzker et al. Aug 2008 A1
20090081639 Hill et al. Mar 2009 A1
20090117047 Szalay et al. May 2009 A1
20090117048 Szalay et al. May 2009 A1
20090117049 Szalay et al. May 2009 A1
20090123382 Szalay et al. May 2009 A1
20090136917 Szalay et al. May 2009 A1
20090155287 Chen et al. Jun 2009 A1
20090162288 Chen et al. Jun 2009 A1
20090180955 Stritzker et al. Jul 2009 A1
20090180987 Stritzker et al. Jul 2009 A1
20100008946 Szalay et al. Jan 2010 A1
20100062016 Szalay et al. Mar 2010 A1
20110064650 Szalay Mar 2011 A1
Foreign Referenced Citations (73)
Number Date Country
709336 Apr 1995 AU
44 25 382 Aug 1997 DE
0 037 441 Oct 1981 EP
0 037 441 May 1984 EP
0 861 093 Sep 1998 EP
1 020 197 Jul 2000 EP
1 146 125 Oct 2001 EP
1 281 772 Feb 2003 EP
1 281 777 Feb 2003 EP
1 281 767 May 2003 EP
1 369 491 Dec 2003 EP
1489164 Dec 2004 EP
1 254 250 Mar 2005 EP
1 512 746 Mar 2005 EP
1 526 185 Apr 2005 EP
55035004 Mar 1980 JP
09-502993 Mar 1997 JP
2000-502884 Mar 2000 JP
2000-295987 Oct 2000 JP
2002097144 Apr 2002 JP
WO 8400381 Feb 1984 WO
WO 8800617 Jan 1988 WO
WO 8911294 Nov 1989 WO
WO 9013658 Nov 1990 WO
9107989 Jun 1991 WO
WO 9222327 Dec 1992 WO
9410302 May 1994 WO
9531105 Nov 1995 WO
9611279 Apr 1996 WO
WO 9640238 Dec 1996 WO
9718841 May 1997 WO
WO 9735997 Oct 1997 WO
WO 9740183 Oct 1997 WO
WO 9805274 Feb 1998 WO
WO 9814605 Apr 1998 WO
WO 9835695 Aug 1998 WO
WO 9930648 Jun 1999 WO
9932646 Jul 1999 WO
WO 0023471 Apr 2000 WO
0047237 Aug 2000 WO
WO 0069448 Nov 2000 WO
WO 0071718 Nov 2000 WO
WO 0073479 Dec 2000 WO
0105229 Jan 2001 WO
0112234 Feb 2001 WO
0114579 Mar 2001 WO
0118195 Mar 2001 WO
0120989 Mar 2001 WO
0124637 Apr 2001 WO
0125399 Apr 2001 WO
WO 0148231 Jul 2001 WO
0155444 Aug 2001 WO
WO 02071062 Sep 2002 WO
03006069 Jan 2003 WO
03014380 Feb 2003 WO
03045153 Jun 2003 WO
03057007 Jul 2003 WO
03063593 Aug 2003 WO
03092600 Nov 2003 WO
03102168 Dec 2003 WO
03102169 Dec 2003 WO
03104485 Dec 2003 WO
WO 2004030631 Apr 2004 WO
2004044175 May 2004 WO
WO 2004069178 Aug 2004 WO
2005047458 May 2005 WO
2005057488 Jun 2005 WO
2005072622 Aug 2005 WO
PCTUS200715829 Jul 2007 WO
PCTUS200722172 Oct 2007 WO
WO 2008099001 Aug 2008 WO
WO 2008100292 Aug 2008 WO
WO 2009126189 Oct 2009 WO
Related Publications (1)
Number Date Country
20050249670 A1 Nov 2005 US