Open Access

Tumor‑targeting bacterial therapy: A potential treatment for oral cancer (Review)

  • Authors:
    • Sai Liu
    • Xiaoping Xu
    • Xin Zeng
    • Longjiang Li
    • Qianming Chen
    • Jing Li
  • View Affiliations

  • Published online on: September 11, 2014     https://doi.org/10.3892/ol.2014.2525
  • Pages: 2359-2366
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Certain obligate or facultative anaerobic bacteria, which exhibit an inherent ability to colonize solid tumors in vivo, may be used in tumor targeting. As genetically manipulated bacteria may actively and specifically penetrate into the tumor tissue, bacterial therapy is becoming a promising approach in the treatment of tumors. However, to the best of our knowledge, no reports have been published thus far regarding the bacterial treatment of oral cancer, one of the most common types of cancer worldwide. In this review, the progress in the understanding of bacterial strategies used in tumor‑targeted therapy is discussed and particular bacterial strains that may have great therapeutic potential in oral squamous cell carcinoma (OSCC) tumor‑targeted therapy are predicted as determined by previous studies.

1. Brief history of tumor-targeting bacterial therapy

The possibility of using bacteria in the treatment of cancer has been recognized for more than a century (1,2). Although it has potential as a novel treatment, the usage of bacteria to target tumors has limitations due to potential biosafety and other deleterious effects, including intrinsic bacterial toxicity, lowered targeting efficiency, genetic instability, and complicated interactions with other therapies (37). The original observation of spontaneous tumor regression from concurrent clostridial infection was reported in 1813 (8,9). The first patient with cancer to be purposefully infected with bacteria was possibly cured by German physician Busch in 1868 (2,10). Over 20 years later, in 1890, Coley, a New York physician, found that several patients with inoperable tumors exhibited tumor regression subsequent to being inoculated with Streptococcus pyogenes. However, the effect was not as great as to eradicate the disease (11). In 1935, Connell observed tumor regression in advanced cancer during therapy using sterile filtrates from Clostridium histolyticum; the author attributed these results to the production of enzymes (12). In 1947, the first study concerning the deliberate injection of Clostridium was published (13). Nonetheless, this field was stagnant due to certain drawbacks (14). It was not until 1976, when Morales, Eidinger and Bruce reported successful treatment of bladder cancer with bacillus Calmette-Guérin (BCG), that this field began to increase rapidly (15). Since then, a number of investigative reports, experimental studies and reviews have been published in this area. Due to these efforts, certain attenuated and engineered obligatory anaerobic bacteria, such as Clostridium, Bifidobacterium, Salmonella, Mycobacterium, Bacillus and Listeria, are known to specifically act as antitumor agents, and colonize hypoxic and necrotic regions, which are present in solid tumors while normally absent in other parts of the body.

2. Strategies using bacteria to target tumors

The hypothesis that living bacteria may function as anticancer therapeutic agents was first advanced in the middle of the twentieth century. Due to the obstacles of hypoxia and necrosis, accessing tumor tissue with traditional treatments has proved difficult. However, bacteria may actively migrate away from the vasculature and penetrate deep into tumor tissue and accumulate (Fig. 1A). Three classes of anaerobic and facultative anaerobes have been examined for use in anticancer therapy (16,17): Bifidobacteria, facultative intracellular bacteria and strictly anaerobic bacteria.

The ideal criteria for the selection of therapeutic bacteria (1820) are as follows: Non-toxic to the host; selective for a specific type of tumor; has the ability to penetrate deeply into the tumor where ordinary treatment does not reach; non-immunogenic (does not trigger an immune response immediately but may be cleared by the host); harmless to normal tissue; able to be manipulated easily; and has a drug carrier that may be controlled. In addition to studies of bacteria designed to induce immune responses (21) and mediate antiangiogenesis therapy (22), a recent study has focused on the usage of bacterial products as anticancer agents (23). Three main strategies in bacterial cancer treatment are discussed in this review: i) Bacteria as tumor markers; ii) Bacteria engineered to express anticancer agents (Fig. 1B); and iii) Bacteria for oncolytic therapy (Fig. 1C).

Bacteria as tumor markers

As replicating anaerobic bacteria are able to selectively target tumors, the use of these bacteria may be an innovative approach for locating tumors that is simple and direct, but practical and effective. Two types of non-bacterial material have served as tumor markers: Viral vectors, including adenovirus, adeno-associated virus, herpes simplex virus (HSV)-1, HSV amplicon, Sindbis, poliovirus replicon and lentivirus/Moloney murine leukemia virus; and non-viral vectors, such as therapeutic DNA, microRNA, short hairpin (sh)RNA, small interfering (si)RNA and oligodeoxynucleotides (ODNs) (2427). However, anaerobic bacteria are preferable to these other two types of tumor marker due to increased mobility (Table I). Once the marker has been administered, a number of methods may be used to locate the tumor, including bioluminescence, fluorescence and magnetic resonance imaging (MRI), as well as positron emission tomography (6). Bacteria may be detected using light, MRI or positron emission tomography (28,29).

Table I

Materials used as tumor markers.

Table I

Materials used as tumor markers.

A, Viral vectors

ExamplesAdvantagesDisadvantages
• Adenovirus• High transfection efficiency• Generation of immune response
• Adeno-associated virus• Efficient in initiating gene expression• Toxicity
• HSV-1• Possibility of proto-oncogene activation
• HSV amplicon• Specific targeting
• Sindbis
• Poliovirus replicon• High production cost
• Lentivirus/MoMLV
• Limitations in deliverable gene size

B, Non-viral vectors

Therapeutic DNA, RNAsa and ODNs
  • Easy to prepare and to scale-up

  • Flexible with regard to the size of the DNA

  • Do not elicit an immune response

  • Less immunogenic

  • Ease of chemical modification

  • Low cost

  • Can be used in different combinations

  • Low transfection efficiency

  • Less efficient in initiating gene expression

Anaerobic bacteria
  • Specific targeting

  • High deliverable gene size

  • Motility

  • Can penetrate deep into tumor

  • Easy to manipulate

  • Low cost

  • Environmental sensing

  • Controlled propagation

  • Immunostimulation

  • Toxicity

  • Genetic instability

a Including microRNAs, short hairpin RNAs and small interfering RNAs.

{ label (or @symbol) needed for fn[@id='tfn2-ol-08-06-2359'] } HSV, herpes simplex virus; MoMLV, Moloney murine leukemia virus; ODNs, oligodeoxynucleotides.

Bacteria engineered to express anticancer agents

Bacteria exhibit the ability to manufacture and deliver specific materials; these can be artificially coupled to certain anticancer agents (Fig. 1B) (28). The most common current carriers employed in gene therapy are viral vectors, such as retrovirus, adenovirus, viral vaccines, herpes simplex virus and adeno-associated virus. Non-viral delivery systems have been gradually established with the development of technology; currently, the gene therapy field has evolved to encompass not only the delivery of therapeutic DNA, but also of microRNA, shRNA, siRNA and ODNs (20,30,31). However, non-viral gene delivery systems exhibit lower transfection potency, resulting in lowered ability to traverse the various obstacles encountered during treatment (27). Conversely, bacteria have great advantages in the drug carrier field. Two predominant mechanisms have been investigated: The direct expression of antitumor proteins and the transfer of eukaryotic expression vectors into infected cancer cells. In direct expression, four categories of anticancer therapies may be utilized: Proteins with physiological activity against tumors, cytotoxic agents, antiangiogenic agents or enzymes that convert the nonfunctional prodrug to an anticancer drug. In the transfer of eukaryotic expression vectors, gene-silencing shRNAs (32), cytokines and growth factors, and tumor antigens have been investigated (Table II) (7). Furthermore, the number of useful agents is increasing due to new developments in combinatorial synthesis and the advent of metagenomics, which is an unlimited source of novel anticancer bacterial products.

Table II

Molecules that may be used as anticancer agents through direct expression by bacteria.

Table II

Molecules that may be used as anticancer agents through direct expression by bacteria.

CategoryAnticancer moleculeRefs
Cytotoxic agentsCly A(34,35)
FASL(36)
TRAIL(37)
TNFα(38,39)
CytokinesCCL21(41)
IL-2(41,42,43)
IL-18(43,44)
LIGHT(44,45)
Antigens and antibodiesCtxB-PSA fusion protein(46)
CPV-OmpA fusion protein(47)
NY-ESO-1 tumor antigen(48)
RAF1(49)
Single chain HIF1α antibodies(50)
DNA transferEndostatin(53,57)
Thrombospondin-1(54)
TRAIL and SMAC(53)
Stat3(54,55,57)
Bcl2(56,57,58)
FLT3L(58)
GM-CSF(57)
IL-12(58,61)
AFP(62)
VEGFR2(63)
EnzymesE. coli CD(64,65)
HSV-TK(66)

[i] Cly A (also known as HlyeE), Cytolysin A; FASL, FAS ligand; TRAIL, TNF-related apoptosis-inducing ligand; TNFα, tumor necrosis factor-α; CCL, collagen cross-linking; IL, interleukin; PSA, prostate-specific antigen; CtxB, cholera toxin subunit B; CPV, canine parvovirus; HIF1α, hypoxia-inducible factor 1-alpha; FLT3L, FMS-like tyrosine kinase 3 ligand; GM-CSF, granulocyte/macrophage colony stimulating factor; AFP, α-fetoprotein; VEGFR, vascular endothelial growth factor receptor; CD, cytosine deaminase; HSV-TK, herpes simplex virus thymidine kinase.

Bacterial oncolytic therapy

The employment of bacteria in oncolytic therapy is the initial treatment and most direct method to kill tumor cells. Clostridial spores are the main components in oncolytic therapy and have been thoroughly analyzed (22,33,67). Bacterial-based cancer therapies using Clostridium spores have the advantage of overcoming the obstacles of hypoxia and necrosis (68). Clostridium spp. are strictly anaerobic and only colonize areas devoid of oxygen; therefore, when Clostridium spp. are systematically injected into solid tumors, spores germinate and multiply in the hypoxic/necrotic regions. Parker et al were the first to demonstrate clostridial oncolysis and tumor regression in mouse tumors by injecting a Clostridium spore suspension into transplanted mouse sarcomas 69). However, during follow-up studies, spore treatment with wild-type Clostridium was not sufficient to eradicate solid tumors (17,70,71). Thus, genetic engineering and repetitive screens are required to enhance the tumor oncolytic capacity of Clostridium. M-55, which was isolated from a non-pathogenic Clostridium oncolyticum strain by Carey et al (72,73), broke this impasse. Since then, multitudinous recombinant Clostridium strains have been used in tumor treatment. Among these, C. histolyticium, C. tetani, C. oncolyticum, C. oncolyticum (sporogenes), C. beijerinckii (acetobutylicum) and C. novyi-NT have been the most commonly investigated (9,74).

3. Advantages and problems of tumor-targeting bacterial therapy

As novel tumor-targeting therapies are introduced, tumor-targeting bacteria have an irreplaceable status due to their unique traits (3). Firstly, it is unsuitable for various types of tumor. Solid tumors are seldom homogeneous; however, almost all tumors have the same microenvironment of low oxygen tension or hypoxia, an environment obligate anaerobes prefer. Furthermore, as bacteria may be easily manipulated, bacteria may be engineered to overcome the limitations that hamper current cancer therapies. In addition, bacteria are highly mobile and actively move away from the vasculature, penetrate deeply and accumulate in tumor tissue. Bacterial therapy achieves adequate tissue penetration, which other treatments, including chemotherapy and radiation, do not (Fig. 1A).

However, certain human trials have shown that the flaws of bacterial therapy cannot be ignored (37). As mentioned above, the investigation of bacteria for tumor targeting was stagnant for a long time due to intrinsic bacterial toxicity. In addition, the wild-type bacteria used for therapy, such as Bifidobacterium longum, Salmonella, Listeria and Escherichia coli, exerted no marked targeting efficiency or oncolytic effect, which reduces the effect of cancer therapy. Furthermore, bacteria exhibit intrinsic genetic instability. Although advanced recombinant DNA technology has rendered it possible to overcome numerous hurdles, bacterial plasmids are not stable and may be lost during bacterial growth.

4. Methods and tools used to overcome treatment issues

Several approaches have been employed as attempts to overcome the difficulties mentioned above. Considerable efforts have been recently invested, and synthetic biology techniques are being improved to optimize bacterial therapy and to resolve key challenges. The use of live, attenuated and engineered bacterial strains may abate toxicity (75). The antitumor effects of bacterial treatment were found to be increased by the application of engineered Clostridium strains. Saccharolytic Clostridium, C. novyi-NT and E. coli have been repeatedly screened to become non-toxic with higher tumor colonization (76). C. oncolyticum M-55 engineered by Carey et al (77), was the first bacterial strain to be genetically manipulated to express an exotic gene, and was used without any side effects. However, the recombinant strains did not function as expected. A non-toxic strain of Clostridium nocyi was developed by deleting the virulence gene through heat treatment (78). In addition, a number of genes have been successfully expressed (3350), which has improved bacterial targeting efficiency and oncolytic effects, as explained previously. A recent study examined and characterized the dynamics of plasmid instability using attenuated strains of S. typhimurium in vivo, which produced good results (79).

Bacterial treatment has also achieved gratifying outcomes when administered in combination with other treatments, including antivascular agents, chemotherapeutic drugs, heat shock proteins, heavy metals and radiation. Combined bacteriolytic therapy is a proposed method for cancer treatment that has been relatively successful thus far (68). The combination of particular species with low-dose radiotherapy dampened the tumor immune escape mechanism (79). In addition, Salmonella with a modified lipid A (strain VNP20009) was found to be non-toxic and successfully colonized the tumors (81). Although the precise immunological mechanism of BCG therapy remains unclear, increasing numbers of reaction types have been found to be induced by BCG complexes, including infections of urothelial cells or bladder cancer cells, induction of immune responses and induction of antitumor effects (82).

5. Analyzing potential OSCC tumor-targeting bacteria groups

Oral cancer, a subtype of head and neck cancer, is defined as cancerous tissue growth located in the oral cavity. Several types of oral cancers have been classified, but 90% of cases worldwide are oral squamous cell carcinoma (OSCC) (83). When the tumor is small enough, a commonly recommended treatment is surgical removal if the outcome would be functionally satisfactory. However, in circumstances in which the tumor is inoperable, radiation therapy with or without chemotherapy is a common treatment option (84). Despite recent advances in diagnosis and therapy, the five-year survival rate of patients with OSCC is only 50% (85). Oral cancer is unusual in conferring a high risk of second primary tumors. This heightened risk may last 5–10 years or occasionally longer (86). Therefore, novel targeting strategies are required to prevent and treat oral cancer. Among the candidate methods of postoperative treatments, tumor-targeting bacterial therapy is expected to have the greatest potential and may even become the main method due to the tumor-targeting specificity.

Specific bacterial species colonize different host locations (87). However, the different roles of the majority of these bacteria have not been determined (88), and may be causal, coincidental or potentially protective. In the human mouth, ~500–1,000 types of bacteria have been detected with various functions; ~110 types constitute the vast majority of oral bacteria (data not shown) (89,90). Three species, Capnocytophaga gingivalis, Pevotella melaninogenica and Streptococcus mitis, have been found to act as diagnostic markers, predicting 80% of oral cancers (86). Although considerable progress has been achieved in elucidating the etiology of oral cancer, the mechanism underlying the association between oral bacteria and oral cancer remains unknown. Further investigation is certainly warranted, but in terms of tumor-targeting therapy, as long as bacteria thrive in OSCC, modern molecular techniques using bacteria may be applied. In addition, artificial modification may further optimize bacteria to meet specific treatment requirements.

Bacteria commonly used in tumor-targeting therapy include Bifidobacterium, Caulobacter, Clostridium, Escherichia, Listeria, Proteus, Salmonella, Streptococcus, Mycobacterium and Shigella. As a vector in tumor-targeting treatment, Salmonella typhimurium VNP2009, an attenuated mutant of S. typhimurium, was first considered due to its significant native toxicity against murine tumors (91). In addition, S. typhimurium was analyzed in a first-in-man phase I clinical trial for toxicity and anticancer activity (92). However, S. typhimurium is not considered part of the native oral microbiota, which indicates that this species may have a poor OSCC tumor-targeting effect. Six prevalent genera in the OSCC library (93) have been identified: Streptococcus, Gemella, Rothia, Peptostreptococcus, Porphyromonas and Lactobacillus (94). In the present review, the bacteria commonly used in tumor-targeting therapy were compared with the following: Human bacterial flora in the mouth, bacteria with colony-forming units (CFU)/ml ≥105 flora in the human mouth, the genera most prevalent in the OSCC library and three species of oral cancer diagnostic markers (86), respectively. Through this analysis, Bifidobacterium, Streptococcus, Caulobacter and Clostridium species were found to have potential for use in OSCC therapy, as these bacteria are part of the normal flora of the mouth, and have previously been used in tumor-targeting therapy (Fig. 2A); Bifidobacterium and Streptococcus were present at CFU/ml ≥105 commensal flora in the mouth (Fig. 2B). Streptococcus may have the most promising OSCC tumor-targeting therapeutic effect, as it is one of the genera most prevalent in the OSCC library and is used as an oral cancer diagnostic marker (Fig. 2C and D).

Promising bacteria used as part of the three main strategies in oral cancer therapy have been discussed in the present review. In order to identify suitable bacteria as diagnostic tools to predict oral cancer, the available information was searched and three of the six most prevalent genera were found in the OSCC library. C. gingivalis, P. melaninogenica and S. mitis predict >80% of oral cancers. In addition, Candida spp., which is commonly detected in oral cancer, has been reported to serve as a precancerous diagnostic marker (95). Among the three genera, S. mitis may be the best candidate for application as an OSCC tumor-targeting vector due to previous analysis (Fig. 2C and D).

The strategy of employing bacteria engineered to express anticancer agents may be easily used in oral cancer therapy if the correct carriers are selected. Bifidobacterium, Streptococcus and Caulobacter are all suitable, but Streptococcus exhibits the most promising therapeutic capacity in this strategy (Fig. 2C and D). Although Clostridium is not part of the human bacterial flora of the mouth whose presense is not <105 CFU/ml, it is present in the oral cavity (Fig. 2A), which suggests that Clostridia spp. may also be used in OSCC bacterial oncolytic therapy.

However, this is only conjecture according to analysis of the existing data; further experiments are required to verify these hypotheses.

6. Conclusion and future perspectives

In the field of cancer treatment, bacterial therapies show great promise, due to the potential tumor-targeting antitumor capability and the ability to deliver therapeutic genes. Currently, one issue in tumor-targeting therapy is selecting the appropriate carrier. The most commonly used carriers are viral vectors, such as retrovirus, adenovirus, viral vaccines, herpes simplex virus and adeno-associated virus. However, the safety, immunogenicity and the limitations of viral vectors are not yet fully understood, and there appears to be no perfect solution for these problems. Thus, as a novel method, bacterial therapy may aid in cancer treatment.

Through this review, Bifidobacterium, Streptococcus, Caulobacter and Clostridium spp. were found to be suitable for application in OSCC tumor-targeting therapy. Streptococcus exhibited the most promising therapeutic application. Engineered bacteria may further alter mutant bacterial strains to express anticancer agents. Thus, tumor-targeting bacterial therapy has the greatest potential of all candidate methods for oral cancer postoperative treatments.

Acknowledgements

This review was supported by the National Natural Science Foundation of China (grant nos. 81302371 and 81072218), the Foundation for Innovative Research Groups of the National Natural Science Foundation of China (grant no. 81321002), Projects of International Cooperation and Exchanges National Natural Science Foundation of China (grant no. 2012DFA31370) and the Doctoral Program of the Ministry of Education of China (grant no. 20110181110055).

References

1 

Nowotny A: Antitumor effects of endotoxins. Handbook of Endotoxin. Berry LJ: 3. Elsevier Science; Amsterdam: pp. 389–448. 1985

2 

Busch W: Aus der Sitzung der medicinischen Section vom 13 November 1867. Berl Klin Wochenschr. 5:1371868.(In German).

3 

Mengesha A: Use of non-pathogenic bacteria as vectors for targeted gene expression in cancer gene therapy (unpublished PhD thesis). Maastricht University. 2009.

4 

Theys J, Barbé S, Landuyt W, et al: Tumor-specific gene delivery using genetically engineered bacteria. Curr Gene Ther. 3:207–221. 2003.

5 

Lavigne MD and Górecki DC: Emerging vectors and targeting methods for nonviral gene therapy. Expert Opin Emerg Drugs. 11:541–557. 2006.

6 

Ptak C and Petronis A: Epigenetics and complex disease: from etiology to new therapeutics. Annu Rev Pharmacol Toxicol. 48:257–276. 2008.

7 

Patyar S, Joshi R, Byrav DS, et al: Review bacteria in cancer therapy: a novel experimental strategy. J Biomed Sci. 17:212010.

8 

Van Mellaert L, Barbé S and Anné J: Clostridium spores as anti-tumour agents. Trends Microbiol. 14:190–196. 2006.

9 

Wei MQ, Mengesha A, Good D and Anné J: Bacterial targeted tumour therapy-dawn of a new era. Cancer Lett. 259:16–27. 2008.

10 

Linnebacher M, Maletzki C, Klier U and Klar E: Bacterial immunotherapy of gastrointestinal tumors. Langenbecks Arch Surg. 397:557–568. 2012.

11 

Coley WB: The treatment of malignant tumors by repeated inoculations of erysipelas: with a report of ten original cases. Am J Med Sci. 105:487–510. 1893.

12 

Connell HC: The Study and Treatment of Cancer by Proteolytic Enzymes: Preliminary Report. Can Med Assoc J. 33:364–370. 1935.

13 

Bhat JV and Barker HA: Clostridium lacto-acetophilum Nov. Spec and the Role of Acetic Acid in the Butyric Acid Fermentation of Lactate. J Bacteriol. 54:381–391. 1947.

14 

Takícs J and Imreh E: Incidence of clostridia in meat in the complementary bacteriological meat examinations. Journal. 421:1977.

15 

Morales A, Eidinger D and Bruce AW: Intracavitary Bacillus Calmette-Guerin in the treatment of superficial bladder tumors. J Urol. 116:180–183. 1976.

16 

Bernardes N, Chakrabarty AM and Fialho AM: Engineering of bacterial strains and their products for cancer therapy. Appl Microbiol Biotechnol. 97:5189–5199. 2013.

17 

Xu J, Liu XS, Zhou SF and Wei MQ: Combination of immunotherapy with anaerobic bacteria for immunogene therapy of solid tumours. Gene Ther Mol Biol. 13:36–52. 2009.

18 

Inoue M, Mukai M, Hamanaka Y, et al: Targeting hypoxic cancer cells with a protein prodrug is effective in experimental malignant ascites. Int J Oncol. 25:713–720. 2004.

19 

Forbes NS: Profile of a bacterial tumor killer. Nat Biotechnol. 24:1484–1485. 2006.

20 

Schmidt-Wolf GD and Schmidt-Wolf IG: Non-viral and hybrid vectors in human gene therapy: an update. Trends Mol Med. 9:67–72. 2003.

21 

Cebra JJ: Influences of microbiota on intestinal immune system development. Am J Clin Nutr. 69:1046S–1051S. 1999.

22 

Gardlik R, Behuliak M, Palffy R, Celec P and Li C: Gene therapy for cancer: bacteria-mediated anti-angiogenesis therapy. Gene Ther. 18:425–431. 2011.

23 

Jain KK: Use of bacteria as anticancer agents. Expert Opin Biol Ther. 1:291–300. 2001.

24 

Thomas CE, Ehrhardt A and Kay MA: Progress and problems with the use of viral vectors for gene therapy. Nat Rev Genet. 4:346–358. 2003.

25 

Morille M, Passirani C, Vonarbourg A, Clavreul A and Benoit JP: Progress in developing cationic vectors for non-viral systemic gene therapy against cancer. Biomaterials. 29:3477–3496. 2008.

26 

Ji SR, Liu C, Zhang B, et al: Carbon nanotubes in cancer diagnosis and therapy. Biochim Biophys Acta. 1806:29–35. 2010.

27 

McCrudden CM and McCarthy HO: Cancer gene therapy - key biological concepts in the design of multifunctional non-viral delivery systems. Gene Therapy - Tools and Potential Applications. Martin Molina F: InTech; Rijeka: pp. 213–248. 2013

28 

Forbes NS: Engineering the perfect (bacterial) cancer therapy. Nat Rev Cancer. 10:785–794. 2010.

29 

van der Meel R, Gallagher WM, Oliveira S, et al: Recent advances in molecular imaging biomarkers in cancer: application of bench to bedside technologies. Drug Discov Today. 15:102–114. 2010.

30 

Kay MA, Glorioso JC and Naldini L: Viral vectors for gene therapy: the art of turning infectious agents into vehicles of therapeutics. Nat Med. 7:33–40. 2001.

31 

Atkinson H and Chalmers R: Delivering the goods: viral and non-viral gene therapy systems and the inherent limits on cargo DNA and internal sequences. Genetica. 138:485–498. 2010.

32 

Xu DQ, Zhang L, Kopecko DJ, et al: Bacterial delivery of siRNAs: a new approach to solid tumor therapy. siRNA and miRNA Gene Silencing. Sioud M: Springer; New York, NY: pp. 1–27. 2009

33 

Barbé S, Van Mellaert L and Anné J: The use of clostridial spores for cancer treatment. J Appl Microbiol. 101:571–578. 2006.

34 

Ganai S, Arenas RB, Sauer JP, Bentley B and Forbes NS: In tumors Salmonella migrate away from vasculature toward the transition zone and induce apoptosis. Cancer Gene Ther. 18:457–466. 2011.

35 

Jiang SN, Phan TX, Nam TK, et al: Inhibition of tumor growth and metastasis by a combination of Escherichia coli-mediated cytolytic therapy and radiotherapy. Mol Ther. 18:635–642. 2010.

36 

Loeffler M, Le’Negrate G, Krajewska M and Reed JC: Inhibition of tumor growth using Salmonella expressing Fas ligand. J Natl Cancer Inst. 100:1113–1116. 2008.

37 

Schrama D, Reisfeld RA and Becker JC: Antibody targeted drugs as cancer therapeutics. Nat Rev Drug Discovery. 5:147–159. 2006.

38 

Theys J, Nuyts S, Landuyt W, et al: Stable Escherichia coli-Clostridium acetobutylicum shuttle vector for secretion of murine tumor necrosis factor alpha. Appl Environ Microbiol. 65:4295–4300. 1999.

39 

Nuyts S, Theys J, Landuyt W, van Mellaert L, Lambin P and Anné J: Increasing specificity of anti-tumor therapy: cytotoxic protein delivery by non-pathogenic clostridia under regulation of radio-induced promoters. Anticancer Res. 21:857–861. 2001.

40 

Saltzman DA: Cancer immunotherapy based on the killing of Salmonella typhimurium-infected tumour cells. Expert Opin Biol Ther. 5:443–449. 2005.

41 

Loeffler M, Le’Negrate G, Krajewska M and Reed JC: Salmonella typhimurium engineered to produce CCL21 inhibit tumor growth. Cancer Immunol Immunother. 58:769–775. 2009.

42 

Theys J, Pennington O, Dubois L, et al: Repeated cycles of Clostridium-directed enzyme prodrug therapy result in sustained antitumour effects in vivo. Br J Cancer. 95:1212–1219. 2006.

43 

Barbé S, Van Mellaert L, Theys J, et al: Secretory production of biologically active rat interleukin-2 by Clostridium acetobutylicum DSM792 as a tool for anti-tumor treatment. FEMS Microbiol Lett. 246:67–73. 2005.

44 

Loeffler M, Le’Negrate G, Krajewska M and Reed JC: IL-18-producing Salmonella inhibit tumor growth. Cancer Gene Ther. 15:787–794. 2008.

45 

Loeffler M, Le’Negrate G, Krajewska M and Reed JC: Attenuated Salmonella engineered to produce human cytokine LIGHT inhibit tumor growth. Proc Natl Acad Sci USA. 104:12879–12883. 2007.

46 

Fensterle J, Bergmann B, Yone C, et al: Cancer immunotherapy based on recombinant Salmonella enterica serovar Typhimurium aro A strains secreting prostate-specific antigen and cholera toxin subunit B. Cancer Gene Ther. 15:85–93. 2008.

47 

Lee SR, Kim SH, Jeong KJ, et al: Multi-immunogenic outer membrane vesicles derived from an MsbB-deficient Salmonella enterica serovar typhimurium mutant. J Microbiol Biotechnol. 19:1271–1279. 2009.

48 

Karbach J, Neumann A, Brand K, et al: Phase I Clinical Trial of Mixed Bacterial Vaccine (Coley’s Toxins) in Patients with NY-ESO-1 Expressing Cancers: Immunological Effects and Clinical Activity. Clin Cancer Res. 18:5449–5459. 2012.

49 

Gentschev I, Fensterle J, Schmidt A, et al: Use of a recombinant Salmonella enterica serovar Typhimurium strain expressing C-Raf for protection against C-Raf induced lung adenoma in mice. BMC Cancer. 5:152005.

50 

Groot AJ, Mengesha A, van der Wall E, van Diest PJ, Theys J and Vooijs M: Functional antibodies produced by oncolytic clostridia. Biochem Biophys Res Commun. 364:985–989. 2007.

51 

Ao A, Wang H, Kamarajugadda S and Lu J: Involvement of estrogen-related receptors in transcriptional response to hypoxia and growth of solid tumors. Proc Natl Acad Sci USA. 105:7821–7826. 2008.

52 

Salyers AA and Shoemaker NB: Resistance gene transfer in anaerobes: new insights, new problems. Clin Infect Dis. 23(Suppl 1): S36–S43. 1996.

53 

Lee CH, Wu CL and Shiau AL: Endostatin gene therapy delivered by Salmonella choleraesuis in murine tumor models. J Gene Med. 6:1382–1393. 2004.

54 

Lee CH, Wu CL and Shiau AL: Systemic administration of attenuated Salmonella choleraesuis carrying thrombospondin-1 gene leads to tumor-specific transgene expression, delayed tumor growth and prolonged survival in the murine melanoma model. Cancer Gene Ther. 12:175–184. 2004.

55 

Hoffman RM: Tumor-seeking Salmonella amino acid auxotrophs. Curr Opin Biotechnol. 22:917–923. 2011.

56 

Leschner S and Weiss S: Salmonella - allies in the fight against cancer. J Mol Med (Berl). 88:763–773. 2010.

57 

Li X, Li Y, Wang B, et al: Delivery of the co-expression plasmid pEndo-Si-Stat3 by attenuated Salmonella serovar typhimurium for prostate cancer treatment. J Cancer Res Clin Oncol. 139:971–980. 2013.

58 

Basu D and Herlyn M: Salmonella typhimurium as a novel RNA interference vector for cancer gene therapy. Cancer Biol Ther. 7:151–152. 2008.

59 

Hawkins LK, Lemoine NR and Kirn D: Oncolytic biotherapy: a novel therapeutic platform. Lancet Oncol. 3:17–26. 2002.

60 

Fux CA, Costerton JW, Stewart PS and Stoodley P: Survival strategies of infectious biofilms. Trends Microbiol. 13:34–40. 2005.

61 

Fujimori M, Taniguchi SI, Amano J, et al: Anaerobic bacterium as a drug for cancer gene therapy US Patent 7,740,835. Filed February 23, 2004; issued June 22, 2010.

62 

Fu W, Lan H, Li S, et al: Synergistic antitumor efficacy of suicide/ePNP gene and 6-methylpurine 2′-deoxyriboside via Salmonella against murine tumors. Cancer Gene Ther. 15:474–484. 2008.

63 

Wei MQ, Ellem KA, Dunn P, et al: Facultative or obligate anaerobic bacteria have the potential for multimodality therapy of solid tumours. Eur J Cancer. 43:490–496. 2007.

64 

Liu SC, Minton NP, Giaccia AJ and Brown JM: Anticancer efficacy of systemically delivered anaerobic bacteria as gene therapy vectors targeting tumor hypoxia/necrosis. Gene Ther. 9:291–296. 2002.

65 

Fuchita M, Ardiani A, Zhao L, et al: Bacterial cytosine deaminase mutants created by molecular engineering show improved 5-fluorocytosine-mediated cell killing in vitro and in vivo. Cancer Res. 69:4791–4799. 2009.

66 

Bermudes D, Low B and Pawelek J: Tumor-targeted Salmonella. Adv Exp Med Biol. 57–63. 2002.

67 

Minton NP, Mauchline ML, Lemmon MJ, et al: Chemotherapeutic tumour targeting using clostridial spores. FEMS Microbiol Rev. 17:357–364. 1995.

68 

Umer B, Good D, Anné J, Duan W and Wei MQ: Clostridial spores for cancer therapy: targeting solid tumour microenvironment. J Toxicol. 2012:8627642012.

69 

Parker RC, Plummer HC, et al: Effect of histolyticus infection and toxin on transplantable mouse tumors. Proc Soc Exp Biol Med. 66:461–467. 1947.

70 

Gericke D and Engelbart K: Oncolysis by Clostridia. II Experiments on a tumor spectrum with a variety of Clostridia in combination with heavy metal. Cancer Res. 24:217–221. 1964.

71 

Dietzel F and Gericke D: Intensification of the oncolysis by Clostridia by means of radio-frequency hyperthermy in experiments on animals-dependence on dosage and on intervals (author’s transl). Strahlentherapie. 153:263–266. 1977.(In German).

72 

Brown JM: Tumor hypoxia in cancer therapy. Methods Enzymol. 435:297–321. 2007.

73 

Brown JM and Wilson WR: Exploiting tumour hypoxia in cancer treatment. Nat Rev Cancer. 4:437–447. 2004.

74 

Mengesha A, Dubois L, Paesmans K, et al: Clostridia in anti-tumour therapy. Clostridia: Molecular Biology in the Post-Genomic Era. Brüggemann H and Gottschalk G: Caister Academic Press; Norfolk, UK: pp. 199–214. 2009

75 

Vassaux G, Nitcheu J, Jezzard S and Lemoine NR: Bacterial gene therapy strategies. J Pathol. 208:290–298. 2006.

76 

Dang LH, Bettegowda C, Agrawal N, et al: Targeting vascular and avascular compartments of tumors with C. novyi-NT and anti-microtubule agents. Cancer Biol Ther. 3:326–337. 2004.

77 

Carey R, Holland J, Whang H, Neter E and Bryant B: Clostridial oncolysis in man. Eur J Cancer. 3:37–46. 1967.

78 

St Jean AT, Zhang M and Forbes NS: Bacterial therapies: completing the cancer treatment toolbox. Curr Opin Biotechnol. 19:511–517. 2008.

79 

Danino T, Lo J, Prindle A, Hasty J and Bhatia SN: In Vivo Gene Expression Dynamics of Tumor-Targeted Bacteria. ACS Synth Biol. 1:465–470. 2012.

80 

Avogadri F, Mittal D, Saccheri F, et al: Intra-tumoral Salmonella typhimurium induces a systemic anti-tumor immune response that is directed by low-dose radiation to treat distal disease. Eur J Immunol. 38:1937–1947. 2008.

81 

Jia LJ, Wei DP, Sun QM, et al: Oral delivery of tumor-targeting Salmonella for cancer therapy in murine tumor models. Cancer Sci. 98:1107–1112. 2007.

82 

Kawai K, Miyazaki J, Joraku A, Nishiyama H and Akaza H: Bacillus Calmette-Guerin (BCG) immunotherapy for bladder cancer: Current understanding and perspectives on engineered BCG vaccine. Cancer Sci. 104:22–27. 2013.

83 

Choi S and Myers JN: Molecular pathogenesis of oral squamous cell carcinoma: implications for therapy. J Dent Res. 87:14–32. 2008.

84 

Subramani K and Ahmed W: Emerging nanotechnologies in dentistry: Processes, materials and applications. William Andrew; Norwich, NY, USA: 2011

85 

Vira D, Basak SK, Lai C, et al: Immunohistochemical variations in the expression of cancer stem cell and macrophage markers in primary and recurrent oral squamous cell carcinomas. Cancer Res. 72:53552012.

86 

Mager DL: Bacteria and cancer: cause, coincidence or cure? A review. J Transl Med. 4:142006.

87 

Scully C and Bagan J: Oral squamous cell carcinoma overview. Oral Oncol. 45:301–308. 2009.

88 

Kurago ZB, Lam-ubol A, Stetsenko A, et al: Lipopolysaccharide-squamous cell carcinoma-monocyte interactions induce cancer-supporting factors leading to rapid STAT3 activation. Head Neck Pathol. 2:1–12. 2008.

89 

Gunn HD: Tissue targeted antigenic activation of the immune response to cancers US Patent 8,034,359. Filed September 19, 2008; issued October 11, 2011.

90 

Hu Y-J, Wang Q, Jiang Y-T, et al: Characterization of oral bacterial diversity of irradiated patients by high-throughput sequencing. Int J Oral Sci. 5:21–25. 2013.

91 

Chen G, Tang B, Yang BY, et al: Tumor-targeting Salmonella typhimurium, a natural tool for activation of prodrug 6MePdR and their combination therapy in murine melanoma model. Appl Microbiol Biotechnol. 97:4393–4401. 2013.

92 

Chorobik P, Czaplicki D, Ossysek K and Bereta J: Salmonella and cancer: from pathogens to therapeutics. Acta Biochim Pol. 60:285–297. 2013.

93 

Hooper SJ, Crean SJ, Lewis MA, Spratt DA, Wade WG and Wilson MJ: Viable bacteria present within oral squamous cell carcinoma tissue. J Clin Microbiol. 44:1719–1725. 2006.

94 

Pushalkar S, Mane SP, Ji X, et al: Microbial diversity in saliva of oral squamous cell carcinoma. FEMS Immunol Med Microbiol. 61:269–277. 2011.

95 

Gall F, Colella G, Di Onofrio V, et al: Candida spp. in oral cancer and oral precancerous lesions. New Microbiol. 36:283–288. 2013.

Related Articles

Journal Cover

December-2014
Volume 8 Issue 6

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Liu S, Xu X, Zeng X, Li L, Chen Q and Li J: Tumor‑targeting bacterial therapy: A potential treatment for oral cancer (Review). Oncol Lett 8: 2359-2366, 2014
APA
Liu, S., Xu, X., Zeng, X., Li, L., Chen, Q., & Li, J. (2014). Tumor‑targeting bacterial therapy: A potential treatment for oral cancer (Review). Oncology Letters, 8, 2359-2366. https://doi.org/10.3892/ol.2014.2525
MLA
Liu, S., Xu, X., Zeng, X., Li, L., Chen, Q., Li, J."Tumor‑targeting bacterial therapy: A potential treatment for oral cancer (Review)". Oncology Letters 8.6 (2014): 2359-2366.
Chicago
Liu, S., Xu, X., Zeng, X., Li, L., Chen, Q., Li, J."Tumor‑targeting bacterial therapy: A potential treatment for oral cancer (Review)". Oncology Letters 8, no. 6 (2014): 2359-2366. https://doi.org/10.3892/ol.2014.2525