1
|
Kefayat A, Bahrami M, Karami M, Rostami S, Ghahremani F. Veillonella parvula as an anaerobic lactate-fermenting bacterium for inhibition of tumor growth and metastasis through tumor-specific colonization and decrease of tumor's lactate level. Sci Rep 2024; 14:21008. [PMID: 39251652 PMCID: PMC11385575 DOI: 10.1038/s41598-024-71140-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 08/26/2024] [Indexed: 09/11/2024] Open
Abstract
High tumor's lactate level directly associates with high tumor growth, metastasis, and patients' poor prognosis. Therefore, many studies have focused on the decrease of tumor's lactate as a novel cancer treatment. In the present study for the first time, a strictly anaerobic lactate-fermenting bacterium, Veillonella parvula, was employed for the decrease of tumor's lactate level. At first, 4T1 breast tumor-bearing BALB/c mice were administered with 106 V. parvula bacteria intravenously, orally, intraperitoneally, and intratumorally. Then, the bacteria biodistribution was evaluated. The best administration route according to tumor colonization was selected and its safety was assessed. Then, the therapeutic effect of V. parvula administration through the best route was investigated according to 4T1 murine breast tumor's growth and metastasis in vivo. In addition, histopathological and immunohistochemistry evaluations were done to estimate microscopic changes at the inner of the tumor and tumor's lactate level was measured after V. parvula administration. V. parvula exhibited considerable tumor-targeting and colonization efficacy, 24 h after intravenous administration. Normal organs were free of the bacteria after 72 h and no side effect was observed. Tumor colonization by V. parvula significantly decreased the tumors' lactate level for about 46% in comparison with control tumors which caused 44.3% and 51.6% decline (P < 0.05) in the mean tumors' volume and liver metastasis of the treatment group in comparison with the control group, respectively. The treatment group exhibited 35% inhibition in the cancer cell proliferation in comparison with the control according to the Ki-67 immunohistochemistry staining. Therefore, intravenous administration of V. parvula is a tumor-specific and safe treatment which can significantly inhibit tumors' growth and metastasis by decreasing the tumor lactate level.
Collapse
Affiliation(s)
- Amirhosein Kefayat
- Department of Oncology, Isfahan University of Medical Sciences, Isfahan, 81746-73461, Iran
| | - Mahshid Bahrami
- Department of Radiology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mojtaba Karami
- Department of Dermatology, Tehran University of Medical Sciences, Tehran, Iran
| | - Soodabeh Rostami
- Nosocomial Infection Research Center, Isfahan University of Medical Sciences, Isfahan, 81746-73461, Iran
| | - Fatemeh Ghahremani
- Department of Medical Physics and Radiotherapy, Arak University of Medical Sciences, Sardasht, Meydan Basij, Arāk, 38481-76941, Iran.
| |
Collapse
|
2
|
Zhou Y, Li Q, Wu Y, Zhang W, Ding L, Ji C, Li P, Chen T, Feng L, Tang BZ, Huang X. Synergistic Brilliance: Engineered Bacteria and Nanomedicine Unite in Cancer Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2313953. [PMID: 38400833 DOI: 10.1002/adma.202313953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/21/2024] [Indexed: 02/26/2024]
Abstract
Engineered bacteria are widely used in cancer treatment because live facultative/obligate anaerobes can selectively proliferate at tumor sites and reach hypoxic regions, thereby causing nutritional competition, enhancing immune responses, and producing anticancer microbial agents in situ to suppress tumor growth. Despite the unique advantages of bacteria-based cancer biotherapy, the insufficient treatment efficiency limits its application in the complete ablation of malignant tumors. The combination of nanomedicine and engineered bacteria has attracted increasing attention owing to their striking synergistic effects in cancer treatment. Engineered bacteria that function as natural vehicles can effectively deliver nanomedicines to tumor sites. Moreover, bacteria provide an opportunity to enhance nanomedicines by modulating the TME and producing substrates to support nanomedicine-mediated anticancer reactions. Nanomedicine exhibits excellent optical, magnetic, acoustic, and catalytic properties, and plays an important role in promoting bacteria-mediated biotherapies. The synergistic anticancer effects of engineered bacteria and nanomedicines in cancer therapy are comprehensively summarized in this review. Attention is paid not only to the fabrication of nanobiohybrid composites, but also to the interpromotion mechanism between engineered bacteria and nanomedicine in cancer therapy. Additionally, recent advances in engineered bacteria-synergized multimodal cancer therapies are highlighted.
Collapse
Affiliation(s)
- Yaofeng Zhou
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang, 330047, P. R. China
| | - Qianying Li
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang, 330047, P. R. China
| | - Yuhao Wu
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang, 330047, P. R. China
| | - Wan Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, P. R. China
| | - Lu Ding
- Department of Cardiology, Jiangxi Hypertension Research Institute, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, P. R. China
| | - Chenlin Ji
- School of Engineering, Westlake University, Hangzhou, 310030, P. R. China
| | - Ping Li
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang, 330047, P. R. China
| | - Tingtao Chen
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, 330036, P. R. China
| | - Lili Feng
- Key Laboratory of Superlight Materials and Surface Technology Ministry of Education, College of Material Sciences and Chemical Engineering, Harbin Engineering University, Harbin, 150001, P. R. China
| | - Ben Zhong Tang
- School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong, 518172, P. R. China
| | - Xiaolin Huang
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang, 330047, P. R. China
| |
Collapse
|
3
|
Santos‐Beneit F. What is the role of microbial biotechnology and genetic engineering in medicine? Microbiologyopen 2024; 13:e1406. [PMID: 38556942 PMCID: PMC10982607 DOI: 10.1002/mbo3.1406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 02/26/2024] [Accepted: 03/12/2024] [Indexed: 04/02/2024] Open
Abstract
Microbial products are essential for developing various therapeutic agents, including antibiotics, anticancer drugs, vaccines, and therapeutic enzymes. Genetic engineering techniques, functional genomics, and synthetic biology unlock previously uncharacterized natural products. This review highlights major advances in microbial biotechnology, focusing on gene-based technologies for medical applications.
Collapse
Affiliation(s)
- Fernando Santos‐Beneit
- Institute of Sustainable ProcessesValladolidSpain
- Department of Chemical Engineering and Environmental Technology, School of Industrial EngineeringUniversity of ValladolidValladolidSpain
| |
Collapse
|
4
|
Williams JS, Higgins AT, Stott KJ, Thomas C, Farrell L, Bonnet CS, Peneva S, Derrick AV, Hay T, Wang T, Morgan C, Dwyer S, D'Ambrogio J, Hogan C, Smalley MJ, Parry L, Dyson P. Enhanced bacterial cancer therapy delivering therapeutic RNA interference of c-Myc. Cell Biosci 2024; 14:38. [PMID: 38521952 PMCID: PMC10961001 DOI: 10.1186/s13578-024-01206-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 02/06/2024] [Indexed: 03/25/2024] Open
Abstract
BACKGROUND Bacterial cancer therapy was first trialled in patients at the end of the nineteenth century. More recently, tumour-targeting bacteria have been harnessed to deliver plasmid-expressed therapeutic interfering RNA to a range of solid tumours. A major limitation to clinical translation of this is the short-term nature of RNA interference in vivo due to plasmid instability. To overcome this, we sought to develop tumour-targeting attenuated bacteria that stably express shRNA by virtue of integration of an expression cassette within the bacterial chromosome and demonstrate therapeutic efficacy in vitro and in vivo. RESULTS The attenuated tumour targeting Salmonella typhimurium SL7207 strain was modified to carry chromosomally integrated shRNA expression cassettes at the xylA locus. The colorectal cancer cell lines SW480, HCT116 and breast cancer cell line MCF7 were used to demonstrate the ability of these modified strains to perform intracellular infection and deliver effective RNA and protein knockdown of the target gene c-Myc. In vivo therapeutic efficacy was demonstrated using the Lgr5creERT2Apcflx/flx and BlgCreBrca2flx/flp53flx/flx orthotopic immunocompetent mouse models of colorectal and breast cancer, respectively. In vitro co-cultures of breast and colorectal cancer cell lines with modified SL7207 demonstrated a significant 50-95% (P < 0.01) reduction in RNA and protein expression with SL7207/c-Myc targeted strains. In vivo, following establishment of tumour tissue, a single intra-peritoneal administration of 1 × 106 CFU of SL7207/c-Myc was sufficient to permit tumour colonisation and significantly extend survival with no overt toxicity in control animals. CONCLUSIONS In summary we have demonstrated that tumour tropic bacteria can be modified to safely deliver therapeutic levels of gene knockdown. This technology has the potential to specifically target primary and secondary solid tumours with personalised therapeutic payloads, providing new multi-cancer detection and treatment options with minimal off-target effects. Further understanding of the tropism mechanisms and impact on host immunity and microbiome is required to progress to clinical translation.
Collapse
Affiliation(s)
- Jason S Williams
- Institute of Life Science, School of Medicine, Swansea University, Singleton Park, Swansea, SA2 8PP, UK
| | - Adam T Higgins
- European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Maindy Road, Cathays, Cardiff, CF24 4HQ, UK
| | - Katie J Stott
- European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Maindy Road, Cathays, Cardiff, CF24 4HQ, UK
| | - Carly Thomas
- Institute of Life Science, School of Medicine, Swansea University, Singleton Park, Swansea, SA2 8PP, UK
| | - Lydia Farrell
- Institute of Life Science, School of Medicine, Swansea University, Singleton Park, Swansea, SA2 8PP, UK
| | - Cleo S Bonnet
- Institute of Life Science, School of Medicine, Swansea University, Singleton Park, Swansea, SA2 8PP, UK
| | - Severina Peneva
- Institute of Life Science, School of Medicine, Swansea University, Singleton Park, Swansea, SA2 8PP, UK
| | - Anna V Derrick
- Institute of Life Science, School of Medicine, Swansea University, Singleton Park, Swansea, SA2 8PP, UK
| | - Trevor Hay
- European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Maindy Road, Cathays, Cardiff, CF24 4HQ, UK
| | - Tianqi Wang
- European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Maindy Road, Cathays, Cardiff, CF24 4HQ, UK
| | - Claire Morgan
- Institute of Life Science, School of Medicine, Swansea University, Singleton Park, Swansea, SA2 8PP, UK
| | - Sarah Dwyer
- European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Maindy Road, Cathays, Cardiff, CF24 4HQ, UK
| | - Joshua D'Ambrogio
- European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Maindy Road, Cathays, Cardiff, CF24 4HQ, UK
| | - Catherine Hogan
- European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Maindy Road, Cathays, Cardiff, CF24 4HQ, UK
| | - Matthew J Smalley
- European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Maindy Road, Cathays, Cardiff, CF24 4HQ, UK
| | - Lee Parry
- European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Maindy Road, Cathays, Cardiff, CF24 4HQ, UK.
| | - Paul Dyson
- Institute of Life Science, School of Medicine, Swansea University, Singleton Park, Swansea, SA2 8PP, UK.
| |
Collapse
|
5
|
Lim D, Kim K, Duysak T, So E, Jeong JH, Choy HE. Bacterial cancer therapy using the attenuated fowl-adapted Salmonella enterica serovar Gallinarum. Mol Ther Oncolytics 2023; 31:100745. [PMID: 38053546 PMCID: PMC10694566 DOI: 10.1016/j.omto.2023.100745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2023] Open
Abstract
We report here a novel anti-cancer therapy based on an avian-host-specific serotype Salmonella enterica serovar Gallinarum (S. Gallinarum) deficient in ppGpp synthesis. To monitor the tumor targeting, a bioluminescent ΔppGpp S. Gallinarum was constructed and injected intravenously into mice bearing syngeneic and human xenograft tumors. Strong bioluminescent signals were detected specifically in all grafted tumors at 2 days post-injection (dpi). The bacterial counts in normal and tumor tissue at 1 dpi revealed that ΔppGpp S. Gallinarum reached >108 CFU/g in tumor tissue and 106-107 CFU/g in endothelial organs; counts were much lower in other organs. At 16 dpi, ΔppGpp S. Gallinarum counts in tumor tissue decreased to ∼106 CFU/g, while those in the other organs became undetectable. A strong anti-cancer effect was observed after the injection of ΔppGpp S. Gallinarum into BALB/c mice grafted with CT26 colon cancer cells. This could be attributed to reduced virulence, which allowed the administration of at least a 10-fold greater dose (108 CFU) of ΔppGpp S. Gallinarum than other attenuated strains of S. enterica serovar Typhimurium (≤107 CFU). An advantage of the avian-specific S. Gallinarum as a cancer therapeutic should be a reduced capacity to cause infections or harm in humans.
Collapse
Affiliation(s)
- Daejin Lim
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Kwangsoo Kim
- Odysseus Bio, Basic Medical Research Building, Chonnam National University Medical College, 322 Seoyangro, Hwasun, Jeonnam 58128, Republic of Korea
- Department of Microbiology, Chonnam National University Medical School, Gwangju 61468, Republic of Korea
| | - Taner Duysak
- Department of Microbiology, Chonnam National University Medical School, Gwangju 61468, Republic of Korea
| | - EunA. So
- Department of Microbiology, Chonnam National University Medical School, Gwangju 61468, Republic of Korea
| | - Jae-Ho Jeong
- Department of Microbiology, Chonnam National University Medical School, Gwangju 61468, Republic of Korea
| | - Hyon E. Choy
- Odysseus Bio, Basic Medical Research Building, Chonnam National University Medical College, 322 Seoyangro, Hwasun, Jeonnam 58128, Republic of Korea
- Department of Microbiology, Chonnam National University Medical School, Gwangju 61468, Republic of Korea
| |
Collapse
|
6
|
Choi E, Murray B, Choi S. Biofilm and Cancer: Interactions and Future Directions for Cancer Therapy. Int J Mol Sci 2023; 24:12836. [PMID: 37629016 PMCID: PMC10454087 DOI: 10.3390/ijms241612836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/13/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
There is a growing body of evidence supporting the significant role of bacterial biofilms in the pathogenesis of various human diseases, including cancer. Biofilms are polymicrobial communities enclosed within an extracellular matrix composed of polysaccharides, proteins, extracellular DNA, and lipids. This complex matrix provides protection against antibiotics and host immune responses, enabling the microorganisms to establish persistent infections. Moreover, biofilms induce anti-inflammatory responses and metabolic changes in the host, further facilitating their survival. Many of these changes are comparable to those observed in cancer cells. This review will cover recent research on the role of bacterial biofilms in carcinogenesis, especially in colorectal (CRC) and gastric cancers, emphasizing the shared physical and chemical characteristics of biofilms and cancer. This review will also discuss the interactions between bacteria and the tumor microenvironment, which can facilitate oncogene expression and cancer progression. This information will provide insight into developing new therapies to identify and treat biofilm-associated cancers, such as utilizing bacteria as delivery vectors, using bacteria to upregulate immune function, or more selectively targeting biofilms and cancer for their shared traits.
Collapse
Affiliation(s)
- Euna Choi
- Department of Biology, Union University, Jackson, TN 38305, USA; (E.C.); (B.M.)
| | - Ben Murray
- Department of Biology, Union University, Jackson, TN 38305, USA; (E.C.); (B.M.)
| | - Sunga Choi
- Department of Bioinformatics and Biosystems, Seongnam Campus of Korea Polytechnics, Seongnam-si 13122, Republic of Korea
| |
Collapse
|
7
|
Constitutive Expression of a Cytotoxic Anticancer Protein in Tumor-Colonizing Bacteria. Cancers (Basel) 2023; 15:cancers15051486. [PMID: 36900277 PMCID: PMC10000871 DOI: 10.3390/cancers15051486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 02/23/2023] [Accepted: 02/23/2023] [Indexed: 03/03/2023] Open
Abstract
Bacterial cancer therapy is a promising next-generation modality to treat cancer that often uses tumor-colonizing bacteria to deliver cytotoxic anticancer proteins. However, the expression of cytotoxic anticancer proteins in bacteria that accumulate in the nontumoral reticuloendothelial system (RES), mainly the liver and spleen, is considered detrimental. This study examined the fate of the Escherichia coli strain MG1655 and an attenuated strain of Salmonella enterica serovar Gallinarum (S. Gallinarum) with defective ppGpp synthesis after intravenous injection into tumor-bearing mice (~108 colony forming units/animal). Approximately 10% of the injected bacteria were detected initially in the RES, whereas approximately 0.01% were in tumor tissues. The bacteria in the tumor tissue proliferated vigorously to up to 109 colony forming units/g tissue, whereas those in the RES died off. RNA analysis revealed that tumor-associated E. coli activated rrnB operon genes encoding the rRNA building block of ribosome needed most during the exponential stage of growth, whereas those in the RES expressed substantially decreased levels of this gene and were cleared soon presumably by innate immune systems. Based on this finding, we engineered ΔppGpp S. Gallinarum to express constitutively a recombinant immunotoxin comprising TGFα and the Pseudomonas exotoxin A (PE38) using a constitutive exponential phase promoter, the ribosomal RNA promoter rrnB P1. The construct exerted anticancer effects on mice grafted with mouse colon (CT26) or breast (4T1) tumor cells without any notable adverse effects, suggesting that constitutive expression of cytotoxic anticancer protein from rrnB P1 occurred only in tumor tissue.
Collapse
|
8
|
The Use of Bacteria in Cancer Treatment: A Review from the Perspective of Cellular Microbiology. Emerg Med Int 2022; 2022:8127137. [PMID: 35978704 PMCID: PMC9377996 DOI: 10.1155/2022/8127137] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 07/15/2022] [Accepted: 07/16/2022] [Indexed: 11/17/2022] Open
Abstract
Cellular microbiology, which is the interaction between harmful microbes and infected cells, is important in the determination of the bacterial infection processes and in the progression of data of different cellular mechanisms. The therapeutic role of bacteria has gained attention since the known methods such as radiation, chemotherapy, and immunotherapy have got drawbacks. Bacteria have demonstrated a favorable impact in treating cancer through eradication of tumors. Bacteria, in cancer treatment, have proven to be promising and have been shown in some of the previous work that it can successfully suppress the growth of tumors. In this paper, we analyzed the difficulties and settlement for using bacteria in cancer therapy as well the mechanisms in which bacteria works in order to achieve tumor eradication. Future works may focus on the use of bacteria along with other treatments in order to achieve effective tumor therapy.
Collapse
|
9
|
Amara S, Yang LV, Tiriveedhi V, Muzaffar M. Complex Role of Microbiome in Pancreatic Tumorigenesis: Potential Therapeutic Implications. Cells 2022; 11:1900. [PMID: 35741028 PMCID: PMC9221309 DOI: 10.3390/cells11121900] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 06/05/2022] [Accepted: 06/08/2022] [Indexed: 12/12/2022] Open
Abstract
Pancreatic cancer (PC) is the fourth leading cause of cancer-related mortality with limited diagnostic and therapeutic options. Although immunotherapy has shown promise in the treatment of several cancers, its role in pancreatic cancer is rather limited. Several studies have focused on determining the role of the tumor microenvironment with cancer-cell-intrinsic events and tumor-infiltrating immune cellular properties. However, in the past decade, there has been emerging research aimed at delineating the role of the host microbiome, including the metabolites from microbes and host responses, on pancreatic tumorigenesis. Importantly, there is emerging evidence suggesting the beneficial role of a gut microbiome transplant to improve immunotherapeutic outcomes in cancer patients. In this review, we summarize the recent understanding of the role of the microbiome in pancreatic cancer progression, along with its clinical diagnostic and therapeutic implications.
Collapse
Affiliation(s)
- Suneetha Amara
- Division of Hematology/Oncology, Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA; (L.V.Y.); (M.M.)
| | - Li V. Yang
- Division of Hematology/Oncology, Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA; (L.V.Y.); (M.M.)
| | - Venkataswarup Tiriveedhi
- Department of Biological Sciences, Tennessee State University, Nashville, TN 37209, USA;
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37212, USA
| | - Mahvish Muzaffar
- Division of Hematology/Oncology, Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA; (L.V.Y.); (M.M.)
| |
Collapse
|
10
|
Engineered microbial systems for advanced drug delivery. Adv Drug Deliv Rev 2022; 187:114364. [PMID: 35654214 DOI: 10.1016/j.addr.2022.114364] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 04/06/2022] [Accepted: 05/25/2022] [Indexed: 12/11/2022]
Abstract
The human body is a natural habitat for a multitude of microorganisms, with bacteria being the major constituent of the microbiota. These bacteria colonize discrete anatomical locations that provide suitable conditions for their survival. Many bacterial species, both symbiotic and pathogenic, interact with the host via biochemical signaling. Based on these attributes, commensal and attenuated pathogenic bacteria have been engineered to deliver therapeutic molecules to target specific diseases. Recent advances in synthetic biology have enabled us to perform complex genetic modifications in live bacteria and bacteria-derived particles, which simulate micron or submicron lipid-based vectors, for the targeted delivery of therapeutic agents. In this review, we highlight various examples of engineered bacteria or bacteria-derived particles that encapsulate, secrete, or surface-display therapeutic molecules for the treatment or prevention of various diseases. The review highlights recent studies on (i) the production of therapeutics by microbial cell factories, (ii) disease-triggered release of therapeutics by sense and respond systems, (iii) bacteria targeting tumor hypoxia, and (iv) bacteria-derived particles as chassis for drug delivery. In addition, we discuss the potential of such drug delivery systems to be translated into clinical therapies.
Collapse
|
11
|
Divyashree M, Prakash SK, Aditya V, Aljabali AA, Alzahrani KJ, Azevedo V, Góes-Neto A, Tambuwala MM, Barh D. Bugs as drugs: neglected but a promising future therapeutic strategy in cancer. Future Oncol 2022; 18:1609-1626. [PMID: 35137604 DOI: 10.2217/fon-2021-1137] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Effective cancer treatment is an urgent need due to the rising incidence of cancer. One of the most promising future strategies in cancer treatment is using microorganisms as cancer indicators, prophylactic agents, immune activators, vaccines or vectors in antitumor therapy. The success of bacteria-mediated chemotherapy will be dependent on the balance of therapeutic benefit and the control of bacterial infection in the body. Additionally, protozoans and viruses have the potential to be used in cancer therapy. This review summarizes how these microorganisms interact with tumor microenvironments and the challenges of a 'bugs as drugs' approach in cancer therapy. Several standpoints are discussed, such as bacteria as vectors for gene therapy that shuttle therapeutic compounds into tumor tissues, their intrinsic antitumor activities and their combination with chemotherapy or radiotherapy. Bug-based cancer therapy is a two-edged sword and we need to find the opportunities by overcoming the challenges.
Collapse
Affiliation(s)
- Mithoor Divyashree
- Nitte University Centre for Science Education & Research (NUCSER), NITTE (Deemed to be University), Paneer Campus, Deralakatte, Mangalore, 575018, Karnataka, India
| | - Shama K Prakash
- K. S. Hegde Medical Academy, NITTE (Deemed to be University), Deralakatte, Mangalore, 575018, Karnataka, India
| | - Vankadari Aditya
- Nitte University Centre for Science Education & Research (NUCSER), NITTE (Deemed to be University), Paneer Campus, Deralakatte, Mangalore, 575018, Karnataka, India
| | - Alaa Aa Aljabali
- Department of Pharmaceutics & Pharmaceutical Technology, Yarmouk University-Faculty of Pharmacy, Irbid, 566, Jordan
| | - Khalid J Alzahrani
- Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, Taif University, Taif, 21944, Saudi Arabia
| | - Vasco Azevedo
- Department of Genetics, Laboratory of Cellular & Molecular Genetics, Ecology & Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, CEP, 31270-901, Brazil
| | - Aristóteles Góes-Neto
- Department of Microbiology, Molecular & Computational Biology of Fungi Laboratory, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, CEP, 31270-901, Brazil
| | - Murtaza M Tambuwala
- School of Pharmacy & Pharmaceutical Science, Ulster University, Coleraine, Northern Ireland, BT52 1SA, UK
| | - Debmalya Barh
- Department of Genetics, Laboratory of Cellular & Molecular Genetics, Ecology & Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, CEP, 31270-901, Brazil.,Institute of Integrative Omics & Applied Biotechnology (IIOAB), Nonakuri, Purba Medinipur WB, 721172, India
| |
Collapse
|
12
|
Afzali S, Doosti A, Heidari M, Babaei N, Keshavarz P, Nadem Z, Kahnamoei A. Effects of Staphylococcus aureus enterotoxin type A on inducing the apoptosis in cervical cancer cell line. GENE REPORTS 2021. [DOI: 10.1016/j.genrep.2021.101397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
13
|
David F, Davis AM, Gossing M, Hayes MA, Romero E, Scott LH, Wigglesworth MJ. A Perspective on Synthetic Biology in Drug Discovery and Development-Current Impact and Future Opportunities. SLAS DISCOVERY 2021; 26:581-603. [PMID: 33834873 DOI: 10.1177/24725552211000669] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The global impact of synthetic biology has been accelerating, because of the plummeting cost of DNA synthesis, advances in genetic engineering, growing understanding of genome organization, and explosion in data science. However, much of the discipline's application in the pharmaceutical industry remains enigmatic. In this review, we highlight recent examples of the impact of synthetic biology on target validation, assay development, hit finding, lead optimization, and chemical synthesis, through to the development of cellular therapeutics. We also highlight the availability of tools and technologies driving the discipline. Synthetic biology is certainly impacting all stages of drug discovery and development, and the recognition of the discipline's contribution can further enhance the opportunities for the drug discovery and development value chain.
Collapse
Affiliation(s)
- Florian David
- Department of Biology and Biological Engineering, Division of Systems and Synthetic Biology, Chalmers University of Technology, Gothenburg, Sweden
| | - Andrew M Davis
- Discovery Sciences, Biopharmaceutical R&D, AstraZeneca, Cambridge, UK
| | - Michael Gossing
- Discovery Sciences, Biopharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Martin A Hayes
- Discovery Sciences, Biopharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Elvira Romero
- Discovery Sciences, Biopharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Louis H Scott
- Discovery Sciences, Biopharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | | |
Collapse
|
14
|
Karmakar R. State of the art of bacterial chemotaxis. J Basic Microbiol 2021; 61:366-379. [PMID: 33687766 DOI: 10.1002/jobm.202000661] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 02/09/2021] [Accepted: 02/25/2021] [Indexed: 12/13/2022]
Abstract
Bacterial chemotaxis is a biased movement of bacteria toward the beneficial chemical gradient or away from a toxic chemical gradient. This movement is achieved by sensing a chemical gradient by chemoreceptors. In most of the chemotaxis studies, Escherichia coli has been used as a model organism. E. coli have about 4-6 flagella on their surfaces, and the motility is achieved by rotating the flagella. Each flagellum has reversible flagellar motors at its base, which rotate the flagella in counterclockwise and clockwise directions to achieve "run" and "tumble." The chemotaxis of bacteria is regulated by a network of interacting proteins. The sensory signal is processed and transmitted to the flagellar motor by cytoplasmic proteins. Bacterial chemotaxis plays an important role in many biological processes such as biofilm formation, quorum sensing, bacterial pathogenesis, and host infection. Bacterial chemotaxis can be applied for bioremediation, horizontal gene transfer, drug delivery, or maybe some other industry in near future. This review contains an overview of bacterial chemotaxis, recent findings of the physiological importance of bacterial chemotaxis in other biological processes, and the application of bacterial chemotaxis.
Collapse
Affiliation(s)
- Richa Karmakar
- Department of Physics, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
15
|
Evaluating the effect of spaceflight on the host-pathogen interaction between human intestinal epithelial cells and Salmonella Typhimurium. NPJ Microgravity 2021; 7:9. [PMID: 33750813 PMCID: PMC7943786 DOI: 10.1038/s41526-021-00136-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 02/03/2021] [Indexed: 01/31/2023] Open
Abstract
Spaceflight uniquely alters the physiology of both human cells and microbial pathogens, stimulating cellular and molecular changes directly relevant to infectious disease. However, the influence of this environment on host-pathogen interactions remains poorly understood. Here we report our results from the STL-IMMUNE study flown aboard Space Shuttle mission STS-131, which investigated multi-omic responses (transcriptomic, proteomic) of human intestinal epithelial cells to infection with Salmonella Typhimurium when both host and pathogen were simultaneously exposed to spaceflight. To our knowledge, this was the first in-flight infection and dual RNA-seq analysis using human cells.
Collapse
|
16
|
Liu L, Zhang J, Gu M, Li G, Ni J, Fan M. Antitumor Effect of Cycle Inhibiting Factor Expression in Colon Cancer via Salmonella VNP20009. Anticancer Agents Med Chem 2020; 20:1722-1727. [PMID: 32324525 DOI: 10.2174/1871520620666200423080622] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 01/26/2020] [Accepted: 03/06/2020] [Indexed: 12/29/2022]
Abstract
BACKGROUND Colon cancer is one of the major causes of morbidity and mortality worldwide. Cycle inhibiting factors (Cifs) have been shown to deamidate Nedd8, resulting in cell cycle arrest. OBJECTIVE To determine the antitumor effect of Cifs on colon cancer by using attenuated Salmonella typhimurium VNP20009. METHODS The VNP-SOPE2-cif and VNP-SOPE2-cif-C/A plasmids were transfected into attenuated Salmonella typhimurium VNP20009. The efficiency and specificity of the Cif promoter were validated in colon cancer SW480 cell lines. Western blotting was subsequently performed to evaluate cell cycle regulators, including P21, P27 and Wee1. In vivo, the antitumor effect of VNP20009 was evaluated in a colon cancer xenograft model. RESULTS Firstly, VNP-SOPE2-cif and VNP-SOPE2-cif-C/A were selectively expressed both in the bacterial and colon cancer cells. Cif expression in SW480 cells via the VNP tumor-targeted expression system induced the accumulation of Wee1, p21 and p27 expression. Moreover, tumor growth was significantly inhibited in the mice with VNP-SOPE2-cif compared to the mice with VNP with the empty construct. CONCLUSION These results suggest that Cif gene delivered by VNP20009 is a promising approach for the treatment of colon cancer.
Collapse
Affiliation(s)
- Liang Liu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Junhua Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Mingqiang Gu
- Department of General Surgery, Shandong Province Qianfoshan Hospital, 274200, Shandong, China
| | - Guichao Li
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jianjiao Ni
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Min Fan
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
17
|
Development of Oxytolerant Salmonella typhimurium Using Radiation Mutation Technology (RMT) for Cancer Therapy. Sci Rep 2020; 10:3764. [PMID: 32111878 PMCID: PMC7048768 DOI: 10.1038/s41598-020-60396-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 02/05/2020] [Indexed: 02/02/2023] Open
Abstract
A critical limitation of Salmonella typhimurium (S. typhimurium) as an anti-cancer agent is the loss of their invasive or replicative activities, which results in no or less delivery of anti-cancer agents inside cancer cells in cancer therapy. Here we developed an oxytolerant attenuated Salmonella strain (KST0650) from the parental KST0649 (ΔptsIΔcrr) strain using radiation mutation technology (RMT). The oxytolerant KST0650 strain possessed 20-times higher replication activity in CT26 cancer cells and was less virulent than KST0649. Furthermore, KST0650 migrated effectively into tumor tissues in mice. KST0650 was further equipped with a plasmid harboring a spliced form of the intracellular pro-apoptotic protein sATF6, and the expression of sATF6 was controlled by the radiation-inducible recN promoter. The new strain was named as KST0652, in which sATF6 protein expression was induced in response to radiation in a dose-dependent manner. This strain was effectively delivered inside cancer cells and tumor tissues via the Salmonella type III secretion system (T3SS). In addition, combination treatment with KST0652 and radiation showed a synergistic anti-tumor effect in murine tumor model with complete inhibition of tumor growth and protection against death. In conclusion, we showed that RMT can be used to effectively develop an anti-tumor Salmonella strain for delivering anti-cancer agents inside tumors.
Collapse
|
18
|
Mi Z, Feng ZC, Li C, Yang X, Ma MT, Rong PF. Salmonella-Mediated Cancer Therapy: An Innovative Therapeutic Strategy. J Cancer 2019; 10:4765-4776. [PMID: 31598148 PMCID: PMC6775532 DOI: 10.7150/jca.32650] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 07/06/2019] [Indexed: 12/31/2022] Open
Abstract
Bacterial-mediated cancer therapy (BMCT) has become a hot topic in the area of antitumor treatment. Salmonella has been recommended to specifically colonize and proliferate inside tumors and even inhibit tumor growth. Salmonella typhimurium (S. typhimurium) is one of the most promising mediators, which can be easily manipulated. S. typhimurium has been engineered and designed as cancer-targeting therapeutics, and can be improved by combining with other therapeutic methods, e.g. chemotherapy and radiotherapy, which regulate the tumor microenvironment synergistically. In view of all these strengths, the engineered attenuated strains have significant advantages for tumor diagnosis and treatment. This treatment has also been approved by the FDA for clinical trial. In this review, we summarized the recent progress and research in the field of Salmonella -mediated cancer therapy.
Collapse
Affiliation(s)
- Ze Mi
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Zhi-Chao Feng
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Cheng Li
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Xiao Yang
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Meng-Tian Ma
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Peng-Fei Rong
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| |
Collapse
|
19
|
Jazeela K, Chakraborty A, Karunasagar I, Deekshit VK. Nontyphoidal Salmonella: a potential anticancer agent. J Appl Microbiol 2019; 128:2-14. [PMID: 31038778 DOI: 10.1111/jam.14297] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 04/24/2019] [Accepted: 04/25/2019] [Indexed: 02/06/2023]
Abstract
Use of bacteria in cancer therapy, despite being considered as a potent strategy, has not really picked up the way other methods of cancer therapies have evolved. However, in recent years, the interest on use of bacteria to kill cancer cells has renewed considerably. The standard and widely followed strategies of cancer treatment often fail either due to the complexity of tumour biology or because of the accompanying side effects. In contrast, these limitations can be easily overcome in a bacteria-mediated approach. Salmonella is a bacterium, which is known for its ability to colonize solid or semisolid tumours more efficiently than any other bacteria. Among more than 2500 serovars of Salmonella, S. Typhimurium has been widely studied for its antagonistic effects on cancer cells. Here in, we review the current status of the preclinical and the clinical studies with a focus on the mechanisms that attribute the anticancer properties to nontyphoidal Salmonella.
Collapse
Affiliation(s)
- K Jazeela
- Nitte University Center for Science Education and Research, Nitte (Deemed to be University), Deralakatte, Mangaluru, Karnataka, India
| | - A Chakraborty
- Nitte University Center for Science Education and Research, Nitte (Deemed to be University), Deralakatte, Mangaluru, Karnataka, India
| | - I Karunasagar
- Nitte University Center for Science Education and Research, Nitte (Deemed to be University), Deralakatte, Mangaluru, Karnataka, India
| | - V K Deekshit
- Nitte University Center for Science Education and Research, Nitte (Deemed to be University), Deralakatte, Mangaluru, Karnataka, India
| |
Collapse
|
20
|
Gwee CP, Khoo CH, Yeap SK, Tan GC, Cheah YK. Targeted inactivation of Salmonella Agona metabolic genes by group II introns and in vivo assessment of pathogenicity and anti-tumour activity in mouse model. PeerJ 2019; 7:e5989. [PMID: 30671294 PMCID: PMC6339473 DOI: 10.7717/peerj.5989] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 10/22/2018] [Indexed: 12/13/2022] Open
Abstract
The fight against cancer has been a never-ending battle. Limitations of conventional therapies include lack of selectivity, poor penetration and highly toxic to the host. Using genetically modified bacteria as a tumour therapy agent has gained the interest of scientist from the past few decades. Low virulence and highly tolerability of Salmonella spp. in animals and humans make it as the most studied pathogen with regards to anti-tumour therapy. The present study aims to construct a genetically modified S. Agona auxotroph as an anti-tumour agent. LeuB and ArgD metabolic genes in ΔSopBΔSopD double knockout S. Agona were successfully knocked out using a Targetron gene knockout system. The knockout was confirmed by colony PCR and the strains were characterized in vitro and in vivo. The knockout of metabolic genes causes significant growth defect in M9 minimal media. Quadruple knockout ΔSopBΔSopDΔLeuBΔArgD (BDLA) exhibited lowest virulence among all of the strains in all parameters including bacterial load, immunity profile and histopathology studies. In vivo anti-tumour study on colorectal tumour bearing-BALB/c mice revealed that all strains of S. Agona were able to suppress the growth of the large solid tumour as compared with negative control and ΔLeuBΔArgD (LA) and BDLA auxotroph showed better efficacy. Interestingly, higher level of tumour growth suppression was noticed in large tumour. However, multiple administration of bacteria dosage did not increase the tumour suppression efficacy. In this study, the virulence of BDLA knockout strain was slightly reduced and tumour growth suppression efficacy was successfully enhanced, which provide a valuable starting point for the development of S. Agona as anti-tumour agent.
Collapse
Affiliation(s)
- Chin Piaw Gwee
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Chai Hoon Khoo
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Swee Keong Yeap
- China-ASEAN College of Marine Sciences, Xiamen University Malaysia, Selangor, Malaysia
| | - Geok Chin Tan
- Department of Pathology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur, Malaysia
| | - Yoke Kqueen Cheah
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| |
Collapse
|
21
|
Suh S, Leaman EJ, Zhan Y, Behkam B. Mathematical Modeling of Bacteria-Enabled Drug Delivery System Penetration into Multicellular Tumor Spheroids. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2018; 2018:6162-6165. [PMID: 30441741 DOI: 10.1109/embc.2018.8513596] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Bacteria-based cancer treatment is a promising approach to address the need for targeted tumor therapies in an effort to avoid the systemic toxicity inherent in conventional chemotherapy. A number of bacterial strains have been shown to preferentially colonize tumors and impart therapeutic benefits. However, the physical underpinnings of bacteria intratumoral transport remain poorly studied. It is hypothesized that cell Iysis in hypoxic and necrotic regions of tumors creates a niche in which some bacteria thrive. To understand if preferential growth plausibly explains the experimentally observed bacterial colonization profiles, we have developed a mathematical model incorporating transport and growth dependent on tumor cell Iysate. We fit model parameters to experimental data, showing that our formulation captures experimentally observed trends. Moreover, we find that bacteria have a higher effective diffusivity than nanoparticles alone, demonstrating transport advantages to designing bacteria-based cancer therapy. This model serves as a first step towards building computational tools for designing optimized bacteria- based chemotherapeutic delivery systems.
Collapse
|
22
|
Kocijancic D, Felgner S, Schauer T, Frahm M, Heise U, Zimmermann K, Erhardt M, Weiss S. Local application of bacteria improves safety of Salmonella -mediated tumor therapy and retains advantages of systemic infection. Oncotarget 2018. [PMID: 28637010 PMCID: PMC5564822 DOI: 10.18632/oncotarget.18392] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Cancer is a devastating disease and a large socio-economic burden. Novel therapeutic solutions are on the rise, although a cure remains elusive. Application of microorganisms represents an ancient therapeutic strategy, lately revoked and refined via simultaneous attenuation and amelioration of pathogenic properties. Salmonella Typhimurium has prevailed in preclinical development. Yet, using virulent strains for systemic treatment might cause severe side effects. In the present study, we highlight a modified strain based on Salmonella Typhimurium UK-1 expressing hexa-acylated Lipid A. We corroborate improved anti-tumor properties of this strain and investigate to which extent an intra-tumoral (i.t.) route of infection could help improve safety and retain advantages of systemic intravenous (i.v.) application. Our results show that i.t. infection exhibits therapeutic efficacy against CT26 and F1.A11 tumors similar to a systemic route of inoculation. Moreover, i.t. application allows extensive dose titration without compromising tumor colonization. Adverse colonization of healthy organs was generally reduced via i.t. infection and accompanied by less body weight loss of the murine host. Despite local application, adjuvanticity remained, and a CT26-specific CD8+ T cell response was effectively stimulated. Most interestingly, also secondary tumors could be targeted with this strategy, thereby extending the unique tumor targeting ability of Salmonella. The i.t. route of inoculation may reap the benefits of systemic infection and aid in safety assurance while directing potency of an oncolytic vector to where it is most needed, namely the primary tumor.
Collapse
Affiliation(s)
- Dino Kocijancic
- Molecular Immunology, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Sebastian Felgner
- Molecular Immunology, Helmholtz Center for Infection Research, Braunschweig, Germany.,Infection Biology of Salmonella, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Tim Schauer
- Molecular Immunology, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Michael Frahm
- Molecular Immunology, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Ulrike Heise
- Mouse-Pathology Service Unit, Helmholtz Center for Infection Research, Braunschweig, Germany
| | | | - Marc Erhardt
- Infection Biology of Salmonella, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Siegfried Weiss
- Molecular Immunology, Helmholtz Center for Infection Research, Braunschweig, Germany.,Institute of Immunology, Medical School Hannover, Hannover, Germany
| |
Collapse
|
23
|
Song J, Zhang Y, Zhang C, Du X, Guo Z, Kuang Y, Wang Y, Wu P, Zou K, Zou L, Lv J, Wang Q. A microfluidic device for studying chemotaxis mechanism of bacterial cancer targeting. Sci Rep 2018; 8:6394. [PMID: 29686328 PMCID: PMC5913277 DOI: 10.1038/s41598-018-24748-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 04/10/2018] [Indexed: 02/05/2023] Open
Abstract
Bacterial cancer targeting may become an efficacious cancer therapy, but the mechanisms underlying bacterial specificity for cancer cells need to be explored prior to adopting it as a new clinical application. To characterize the mechanism of bacterial chemotactic preference towards cancer cells, we developed a microfluidic device for in vitro study. The device consists of a cell culture chamber on both sides of a central bacteria channel, with micro-channels used as barriers between them. The device, when used as model for lung cancer, was able to provide simultaneous three-dimensional co-culture of multiple cell lines in separate culture chambers, and when used as model for bacterial chemotaxis, established constant concentration gradients of biochemical compounds in a central channel by diffusion through micro-channels. Fluorescence intensity of green fluorescence protein (GFP)-encoding bacteria was used to measure bacterial taxis behavior due to established chemotactic gradients. Using this platform, we found that Escherichia coli (E. coli) clearly illustrated the preference for lung cancer cells (NCI-H460) which was attributed to biochemical factors secreted by carcinoma cells. Furthermore, by secretome analysis and validation experiments, clusterin (CLU) was found as a key regulator for the chemotaxis of E. coli in targeting lung cancer.
Collapse
Affiliation(s)
- Jing Song
- Department of Respiratory Medicine, The Second Hospital, Dalian Medical University, Dalian, China
| | - Yu Zhang
- Department of Radiotherapy, The Second Hospital, Dalian Medical University, Dalian, China
| | - Chengqian Zhang
- Laboratory of Protein and Peptide Pharmaceuticals and Laboratory of Proteomics, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiaohui Du
- Department of Scientific Research Center, The Second Hospital, Dalian Medical University, Dalian, China
| | - Zhe Guo
- Department of Respiratory Medicine, The Second Hospital, Dalian Medical University, Dalian, China
- Department of Respiratory Medicine, The first Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Yanbin Kuang
- Department of Respiratory Medicine, The Second Hospital, Dalian Medical University, Dalian, China
| | - Yingyan Wang
- Laboratory Center for Diagnostics, Dalian Medical University, Dalian, China
| | - Peng Wu
- Laboratory of Protein and Peptide Pharmaceuticals and Laboratory of Proteomics, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Kun Zou
- Department of Radiotherapy, The First Hospital, Dalian Medical University, Dalian, China
| | - Lijuan Zou
- Department of Radiotherapy, The Second Hospital, Dalian Medical University, Dalian, China.
| | - Jianxin Lv
- Key Laboratory of Medical Genetics, Wenzhou Medical University, Wenzhou, China.
| | - Qi Wang
- Department of Respiratory Medicine, The Second Hospital, Dalian Medical University, Dalian, China.
| |
Collapse
|
24
|
Song S, Vuai MS, Zhong M. The role of bacteria in cancer therapy - enemies in the past, but allies at present. Infect Agent Cancer 2018; 13:9. [PMID: 29568324 PMCID: PMC5856380 DOI: 10.1186/s13027-018-0180-y] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 02/15/2018] [Indexed: 12/20/2022] Open
Abstract
In recent decades, bacteria’s therapeutic role has aroused attention in medicinal and pharmaceutical research. While bacteria are considered among the primary agents for causing cancer, recent research has shown intriguing results suggesting that bacteria can be effective agents for cancer treatment – they are the perfect vessels for targeted cancer therapy. Several bacterial strains/species have been discovered to possess inherent oncolytic potentials to invade and colonize solid tumors in vivo. The therapeutic strategy of using bacteria for treating cancer is considered to be effective; however, the severe side effects encountered during the treatment resulted in the abandonment of the therapy. State-of-the-art genetic engineering has been recently applied to bacteria therapy and resulted in a greater efficacy with minimum side effects. In addition, the anti-cancer potential of tumor-targeting bacteria through oral administration circumvents the use of the intravenous route and the associated adverse effects. This review aims to provide a comprehensive summary of the latest literature on the role of bacteria in cancer treatment.
Collapse
Affiliation(s)
- Shiyu Song
- 1Department of Medical Microbiology, Dalian Medical University, 9 Western Section, Lvshun South Road, Lvshunkou District, Dalian, 116044 China
| | - Miza S Vuai
- 1Department of Medical Microbiology, Dalian Medical University, 9 Western Section, Lvshun South Road, Lvshunkou District, Dalian, 116044 China.,2Department of Natural Science, State University of Zanzibar (SUZA), P.O Box 146, Zanzibar, Tanzania
| | - Mintao Zhong
- 1Department of Medical Microbiology, Dalian Medical University, 9 Western Section, Lvshun South Road, Lvshunkou District, Dalian, 116044 China
| |
Collapse
|
25
|
Saltzman D, Augustin L, Leonard A, Mertensotto M, Schottel J. Low dose chemotherapy combined with attenuated Salmonella decreases tumor burden and is less toxic than high dose chemotherapy in an autochthonous murine model of breast cancer. Surgery 2018; 163:509-514. [DOI: 10.1016/j.surg.2017.09.036] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 09/25/2017] [Accepted: 09/29/2017] [Indexed: 01/18/2023]
|
26
|
Kramer MG, Masner M, Ferreira FA, Hoffman RM. Bacterial Therapy of Cancer: Promises, Limitations, and Insights for Future Directions. Front Microbiol 2018; 9:16. [PMID: 29472896 PMCID: PMC5810261 DOI: 10.3389/fmicb.2018.00016] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 01/05/2018] [Indexed: 01/21/2023] Open
Abstract
Spontaneous tumors regression has been associated with microbial infection for 100s of years and inspired the use of bacteria for anticancer therapy. Dr. William B. Coley (1862-1936), a bone- sarcoma surgeon, was a pioneer in treating his patients with both live bacterial-based and mixture of heat-killed bacteria known as "Coley's toxins." Unfortunately, Coley was forced to stop his work which interrupted this field for about half a century. Currently, several species of bacteria are being developed against cancer. The bacterial species, their genetic background and their infectious behavior within the tumor microenvironment are thought to be relevant factors in determining their anti-tumor effectiveness in vivo. In this perspective article we will update the most promising results achieved using bacterial therapy (alone or combined with other strategies) in clinically-relevant animal models of cancer and critically discuss the impact of the bacterial variants, route of administration and mechanisms of bacteria-cancer-cell interaction. We will also discuss strategies to apply this information using modern mouse models, molecular biology, genetics and imaging for future bacterial therapy of cancer patients.
Collapse
Affiliation(s)
- M. Gabriela Kramer
- Department of Biotechnology, Instituto de Higiene, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
- Laboratory of Carbohydrates and Glycoconjugates, Department of Organic Chemistry, Facultad de Química, Universidad de la República, Montevideo, Uruguay
| | - Martín Masner
- Department of Biotechnology, Instituto de Higiene, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Fernando A. Ferreira
- Department of Biotechnology, Instituto de Higiene, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
- Laboratory of Carbohydrates and Glycoconjugates, Department of Organic Chemistry, Facultad de Química, Universidad de la República, Montevideo, Uruguay
| | - Robert M. Hoffman
- AntiCancer, Inc., San Diego, CA, United States
- Department of Surgery, University of California, San Diego, San Diego, CA, United States
| |
Collapse
|
27
|
Bascuas T, Moreno M, Grille S, Chabalgoity JA. Salmonella Immunotherapy Improves the Outcome of CHOP Chemotherapy in Non-Hodgkin Lymphoma-Bearing Mice. Front Immunol 2018; 9:7. [PMID: 29410666 PMCID: PMC5787062 DOI: 10.3389/fimmu.2018.00007] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 01/03/2018] [Indexed: 12/12/2022] Open
Abstract
We have previously shown that Salmonella immunotherapy is effective to treat B-cell non-Hodgkin lymphoma (B-NHL) in mice. However, this model involves animals with high tumor burden, whereas in the clinics B-NHL patients are usually treated with chemotherapy (CHOP: cyclophosphamide, doxorubicin, vincristine, and prednisone) as first-line therapy prior to immunotherapy. Recently, we have described a NHL-B preclinical model using CHOP chemotherapy to achieve MRD in immunocompetent animals that closely resemble patients' conditions. In this work, we assessed the efficacy of Salmonella immunotherapy in B-NHL-bearing mice undergoing chemotherapy. Salmonella administration significantly delayed tumor growth and prolonged survival of chemotherapy-treated NHL-bearing animals. Mice receiving the CHOP-Salmonella combined therapy showed increased numbers of tumor-infiltrating leukocytes and a different profile of cytokines and chemokines expressed in the tumor microenvironment. Further, Salmonella immunotherapy in CHOP-treated animals also enhanced NK cells cytotoxic activity as well as induced systemic lymphoma-specific humoral and cellular responses. Chemotherapy treatment profoundly impacted on the general health status of recipient animals, but those receiving Salmonella showed significantly better overall body condition. Altogether, the results clearly demonstrated that Salmonella immunotherapy could be safely used in individuals under CHOP treatment, resulting in a better prognosis. These results give strong support to consider Salmonella as a neoadjuvant therapy in a clinical setting.
Collapse
Affiliation(s)
- Thais Bascuas
- Laboratory for Vaccine Research, Departamento de Desarrollo Biotecnológico, Instituto de Higiene, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - María Moreno
- Laboratory for Vaccine Research, Departamento de Desarrollo Biotecnológico, Instituto de Higiene, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Sofía Grille
- Cátedra de Hematología, Hospital de Clínicas, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
- Departamento Básico de Medicina, Hospital de Clínicas, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - José A. Chabalgoity
- Laboratory for Vaccine Research, Departamento de Desarrollo Biotecnológico, Instituto de Higiene, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
28
|
Kim K, Min SY, Lim HD, You SH, Lim D, Jeong JH, Kim HJ, Rhee JH, Park K, Shin M, Kim GJ, Min JJ, Choy HE. Cell mass-dependent expression of an anticancer protein drug by tumor-targeted Salmonella. Oncotarget 2018; 9:8548-8559. [PMID: 29492216 PMCID: PMC5823552 DOI: 10.18632/oncotarget.24013] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 12/03/2017] [Indexed: 01/09/2023] Open
Abstract
Bacterial cancer therapy relies on the properties of certain bacterial species capable of targeting and proliferating within solid malignancies. If these bacteria could be loaded with antitumor proteins, the efficacy of this approach could be greatly increased. However, because most antitumor proteins are also toxic to normal tissue, they must be expressed by bacteria that specifically target and exclusively localize to tumor tissue. As a strategy for treating solid malignancies, we recently evaluated L-asparaginase (L-ASNase) delivered by tumor-targeted Salmonella. In this system, L-ASNase was expressed under the control of the araBAD promoter (PBAD) of the E. coli arabinose operon, which is induced by injection of L-arabinose. Here, we further improved the performance of recombinant Salmonella in cancer therapy by exploiting the quorum-sensing (QS) system, which uses cell mass-dependent auto-induction logic. This approach obviates the necessity of monitoring intratumoral bacterial status and inducing cargo protein expression by administration of an exogenous compound. Recombinant Salmonella in tumors expressed and secreted active L-ASNase in a cell mass-dependent manner, yielding significant anticancer effects. These results suggest that expression of a therapeutic protein under the control of the QS system represents a promising engineering platform for the production of recombinant proteins in vivo.
Collapse
Affiliation(s)
- Kwangsoo Kim
- Department of Microbiology, Chonnam National University Medical School, Jeollanam-do, Republic of Korea.,Molecular Medicine, BK21 plus, Chonnam National University Graduate School, Gwangju, Republic of Korea
| | - Sa-Young Min
- Department of Biological Sciences, College of Natural Sciences, Chonnam National University, Gwangju, Republic of Korea
| | - Ho-Dong Lim
- Department of Biological Sciences, College of Natural Sciences, Chonnam National University, Gwangju, Republic of Korea
| | - Sung-Hwan You
- Department of Biological Sciences, College of Natural Sciences, Chonnam National University, Gwangju, Republic of Korea
| | - Daejin Lim
- Department of Microbiology, Chonnam National University Medical School, Jeollanam-do, Republic of Korea.,Molecular Medicine, BK21 plus, Chonnam National University Graduate School, Gwangju, Republic of Korea
| | - Jae-Ho Jeong
- Department of Microbiology, Chonnam National University Medical School, Jeollanam-do, Republic of Korea.,Molecular Medicine, BK21 plus, Chonnam National University Graduate School, Gwangju, Republic of Korea
| | - Hyun-Ju Kim
- Department of Microbiology, Chonnam National University Medical School, Jeollanam-do, Republic of Korea.,Molecular Medicine, BK21 plus, Chonnam National University Graduate School, Gwangju, Republic of Korea
| | - Joon Haeng Rhee
- Department of Microbiology, Chonnam National University Medical School, Jeollanam-do, Republic of Korea.,Molecular Medicine, BK21 plus, Chonnam National University Graduate School, Gwangju, Republic of Korea
| | - Kyeongil Park
- Department of Microbiology, Chonnam National University Medical School, Jeollanam-do, Republic of Korea.,Molecular Medicine, BK21 plus, Chonnam National University Graduate School, Gwangju, Republic of Korea
| | - Minsang Shin
- Department of Microbiology, Kyungpook National University Medical School, Daegu, Republic of Korea
| | - Geun-Joong Kim
- Department of Biological Sciences, College of Natural Sciences, Chonnam National University, Gwangju, Republic of Korea
| | - Jung-Joon Min
- Molecular Medicine, BK21 plus, Chonnam National University Graduate School, Gwangju, Republic of Korea.,Department of Nuclear Medicine, Chonnam National University Medical School and Hwasun Hospital, Jeonnam, Republic of Korea
| | - Hyon E Choy
- Department of Microbiology, Chonnam National University Medical School, Jeollanam-do, Republic of Korea.,Molecular Medicine, BK21 plus, Chonnam National University Graduate School, Gwangju, Republic of Korea
| |
Collapse
|
29
|
Kocijancic D, Felgner S, Frahm M, Komoll RM, Iljazovic A, Pawar V, Rohde M, Heise U, Zimmermann K, Gunzer F, Hammer J, Crull K, Leschner S, Weiss S. Therapy of solid tumors using probiotic Symbioflor-2: restraints and potential. Oncotarget 2017; 7:22605-22. [PMID: 26981777 PMCID: PMC5008385 DOI: 10.18632/oncotarget.8027] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Accepted: 02/25/2016] [Indexed: 12/11/2022] Open
Abstract
To date, virulent bacteria remain the basis of most bacteria mediated cancer therapies. For clinical application attenuation is required. However, this might result in a drastically lowered therapeutic capacity. Herein we argue that the E. coli probiotic Symbioflor-2, with a history of safe application may constitute a viable tumor therapeutic candidate. We demonstrate that Symbioflor-2 displays a highly specific tumor targeting ability as determined in murine CT26 and RenCa tumor models. The excellent specificity was ascribed to reduced levels of adverse colonization. A high safety standard was demonstrated in WT and Rag1−/− mice. Thus, Symbioflor-2 may represent an ideal tumor targeting delivery system for therapeutic molecules. Moreover, Symbioflor-2 was capable of inducing CT26 tumor clearance as result of an adjuvant effect on tumor specific CD8+ T cells analogous to the Salmonella variant SL7207. However, lower therapeutic efficacy against RenCa tumors suggested a generally reduced therapeutic potency for probiotics. Interestingly, concurrent depletion of Gr-1+ or Ly6G+ cells installed therapeutic efficacy equal to SL7207, thus highlighting the role of innate effector cells in restraining the anti-tumor effects of Symbioflor-2. Collectively, our findings argue for a strategy of safe strain application and a more sustainable use of bacteria as a delivery system for therapeutic molecules.
Collapse
Affiliation(s)
- Dino Kocijancic
- Department of Molecular Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Sebastian Felgner
- Department of Molecular Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Michael Frahm
- Department of Molecular Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Ronja-Melinda Komoll
- Department of Molecular Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Aida Iljazovic
- Department of Molecular Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Vinay Pawar
- Department of Molecular Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Manfred Rohde
- Central Facility for Microscopy, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Ulrike Heise
- Mouse-Pathology Service Unit, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | | | - Florian Gunzer
- Institute of Medical Microbiology and Hygiene, Dresden University of Technology, Dresden, Germany
| | - Juliane Hammer
- Department of Molecular Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Katja Crull
- Department of Molecular Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Sara Leschner
- Department of Molecular Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Siegfried Weiss
- Department of Molecular Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Institute of Immunology, Medical School Hannover, Hannover, Germany
| |
Collapse
|
30
|
Ning BT, Yu B, Chan S, Chan JL, Huang JD, Chan GCF. Treatment of Neuroblastoma with an Engineered "Obligate" Anaerobic Salmonella typhimurium Strain YB1. J Cancer 2017; 8:1609-1618. [PMID: 28775780 PMCID: PMC5535716 DOI: 10.7150/jca.18776] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Accepted: 02/27/2017] [Indexed: 12/13/2022] Open
Abstract
Purpose Neuroblastoma is an embryonic solid tumor derived from the progenitors of the sympathetic nervous system. More than half of the patients developed metastatic disease at the time of initial diagnosis and had poor outcome with current therapeutic approaches. In recent years, some obligate and facultative anaerobic bacteria were reported to target the hypoxic and necrotic region of solid tumor models and caused tumor regression. We recently successfully constructed an "obligate" anaerobic Salmonella strain YB1 that was applied in breast cancer nude mice model by us. Here, we report the application of YB1 in neuroblastoma treatment. Methods The anti-cancer effect and side-effects of YB1 was examined in both in vitro and in vivo experiment. Previous established orthotopic neuroblastoma SCID/beige murine model using SK-NLP/luciferase cell line was adopted. ResultsIn vitro, YB1 induced apoptosis for up to 31.4% of the neuroblastoma cells under anaerobic condition, three times more than that under aerobic condition (10.9%). The expression of both Toll like Receptor 4 and 5 (TLR4 and TLR5) in cancer cells were significantly up-regulated (p<0.05, p<0.01 respectively) after the treatment of YB1 under anaerobic condition. In mouse model, YB1 preferentially accumulated inside the core of the tumors, rather than in normal tissues as our previous reported. This is suggestive of the hypoxic nature of tumor core. Tumor growth was significantly retarded in YB1 treatment group (n=6, P<0.01). Furthermore, there was no long-term organ damage noted in all the organs examined including heart, lung, liver, spleen and brain in the YB1 treated mice. Conclusion The genetic modified Salmonella strain YB1 is a promising anti-tumor strategy against the tumor bulk for neuroblastoma. Future study can be extended to other common cancer types to verify the relative efficacy on different neoplastic cells.
Collapse
Affiliation(s)
- Bo-Tao Ning
- Pediatric Intensive Care Unit, Shanghai Children's medical Center affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.,Department of Peadiatrics & Adolescent, Queen Mary Hospital, LKS Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong SAR, PR China
| | - Bin Yu
- Department of Biochemistry, LKS Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong SAR, PR China
| | - Shing Chan
- Department of Peadiatrics & Adolescent, Queen Mary Hospital, LKS Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong SAR, PR China
| | - Jian-Liang Chan
- Department of Peadiatrics & Adolescent, Queen Mary Hospital, LKS Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong SAR, PR China
| | - Jian-Dong Huang
- Department of Biochemistry, LKS Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong SAR, PR China
| | - Godfrey Chi-Fung Chan
- Department of Peadiatrics & Adolescent, Queen Mary Hospital, LKS Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong SAR, PR China.,Center of Cancer Research, LKS Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong SAR, PR China
| |
Collapse
|
31
|
Coutermarsh-Ott SL, Broadway KM, Scharf BE, Allen IC. Effect of Salmonella enterica serovar Typhimurium VNP20009 and VNP20009 with restored chemotaxis on 4T1 mouse mammary carcinoma progression. Oncotarget 2017; 8:33601-33613. [PMID: 28431394 PMCID: PMC5464893 DOI: 10.18632/oncotarget.16830] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 03/27/2017] [Indexed: 12/18/2022] Open
Abstract
A variety of bacterial strains have been evaluated as bio-therapeutic and immunomodulatory agents to treat cancer. One such strain, Salmonella enterica serovar Typhimurium VNP20009, which is attenuated by a purine auxotrophic mutation and modified lipid A, is characterized in previous models as a safely administered, tumor colonizing agent. However, earlier work tended to use less aggressive cancer cell lines and immunocompromised animal models. Here, we investigated the safety and efficacy of VNP20009 in a highly malignant murine model of human breast cancer. Additionally, as VNP20009 has recently been found to have a defective chemotaxis system, we tested whether restoring chemotaxis would improve anti-cancer properties in this model system. Exposure to VNP20009 had no significant effect on primary mammary tumor size or pulmonary metastasis, and the tumor colonizing process appeared chemotaxis independent. Moreover, tumor-bearing mice exposed to Salmonella exhibited increased morbidity that was associated with significant liver disease. Our results suggest that VNP20009 may not be safe or efficacious when used in aggressive, metastatic breast cancer models utilizing immunocompetent animals.
Collapse
Affiliation(s)
- Sheryl L. Coutermarsh-Ott
- Virginia Tech, VA-MD College of Veterinary Medicine, Department of Biomedical Sciences and Pathobiology, Blacksburg, VA, USA
| | | | - Birgit E. Scharf
- Virginia Tech, Department of Biological Sciences, Blacksburg, VA, USA
| | - Irving C. Allen
- Virginia Tech, VA-MD College of Veterinary Medicine, Department of Biomedical Sciences and Pathobiology, Blacksburg, VA, USA
| |
Collapse
|
32
|
Toxicology and efficacy of tumor-targeting Salmonella typhimurium A1-R compared to VNP 20009 in a syngeneic mouse tumor model in immunocompetent mice. Oncotarget 2017; 8:54616-54628. [PMID: 28903369 PMCID: PMC5589608 DOI: 10.18632/oncotarget.17605] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 03/21/2017] [Indexed: 12/12/2022] Open
Abstract
Salmonella typhimurium A1-R (S. typhimurium A1-R) attenuated by leu and arg auxotrophy has been shown to target multiple types of cancer in mouse models. In the present study, toxicologic and biodistribution studies of tumor-targeting S. typhimurium A1-R and S. typhimurium VNP20009 (VNP 20009) were performed in a syngeneic tumor model growing in immunocompetent BALB/c mice. Single or multiple doses of S. typhimurium A1-R of 2.5 × 105 and 5 × 105 were tolerated. A single dose of 1 × 106 resulted in mouse death. S. typhimurium A1-R (5 × 105 CFU) was eliminated from the circulation, liver and spleen approximately 3-5 days after bacterial administration via the tail vein, but remained in the tumor in high amounts. S. typhimurium A1-R was cleared from other organs much more rapidly. S. typhimurium A1-R and VNP 20009 toxicity to the spleen and liver was minimal. S. typhimurium A1-R showed higher selective targeting to the necrotic areas of the tumors than VNP20009. S. typhimurium A1-R inhibited the growth of CT26 colon carcinoma to a greater extent at the same dose of VNP20009. In conclusion, we have determined a safe dose and schedule of S. typhimurium A1-R administration in BALB/c mice, which is also efficacious against tumor growth. The results of the present report indicate similar toxicity of S. typhimurium A1-R and VNP20009, but greater antitumor efficacy of S. typhimurium A1-R in an immunocompetent animal. Since VNP2009 has already proven safe in a Phase I clinical trial, the present results indicate the high clinical potential of S. typhimurium A1-R.
Collapse
|
33
|
Broadway KM, Suh S, Behkam B, Scharf BE. Optimizing the restored chemotactic behavior of anticancer agent Salmonella enterica serovar Typhimurium VNP20009. J Biotechnol 2017; 251:76-83. [PMID: 28433721 DOI: 10.1016/j.jbiotec.2017.04.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 04/04/2017] [Accepted: 04/05/2017] [Indexed: 12/17/2022]
Abstract
Bacteria, including strains of Salmonella, have been researched and applied as therapeutic cancer agents for centuries. Salmonella are particularly of interest due to their facultative anaerobic nature, facilitating colonization of differentially oxygenated tumor regions. Additionally, Salmonella can be manipulated with relative ease, resulting in the ability to attenuate the pathogen or engineer vectors for drug delivery. It was recently discovered that the anti-cancer Salmonella enterica serovar Typhimurium strain VNP20009 is lacking in chemotactic ability, due to a non-synonymous single nucleotide polymorphism in cheY. Replacing the mutated copy of cheY with the wild-type sequence restored chemotaxis to 70% of the parental strain. We aimed to investigate further if chemotaxis of VNP20009 can be optimized. By restoring the gene msbB in VNP20009 cheY+, which confers attenuation by lipid A modification, we observed a 9% increase in swimming speed, 13% increase in swim plate performance, 19% increase in microfluidic device partitioning towards the attractant at the optimum concentration gradient, and mitigation of a non-motile cell subpopulation. We conclude that chemotaxis can be enhanced further but at the cost of changing one defining characteristic of VNP20009. A less compromised strain might be needed to employ for investigating bacterial chemotaxis in tumor interactions.
Collapse
Affiliation(s)
- Katherine M Broadway
- Department of Biological Sciences, Life Sciences I, Virginia Tech, Blacksburg, VA 24061, USA
| | - Seungbeum Suh
- Department of Mechanical Engineering, Kelly Hall, Virginia Tech, Blacksburg, VA 24061, USA
| | - Bahareh Behkam
- Department of Mechanical Engineering, Kelly Hall, Virginia Tech, Blacksburg, VA 24061, USA; School of Biomedical Engineering and Sciences, Virginia Tech, Blacksburg, VA 24061, USA
| | - Birgit E Scharf
- Department of Biological Sciences, Life Sciences I, Virginia Tech, Blacksburg, VA 24061, USA.
| |
Collapse
|
34
|
Hatzikirou H, López Alfonso JC, Leschner S, Weiss S, Meyer-Hermann M. Therapeutic Potential of Bacteria against Solid Tumors. Cancer Res 2017; 77:1553-1563. [PMID: 28202530 DOI: 10.1158/0008-5472.can-16-1621] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 11/22/2016] [Accepted: 12/21/2016] [Indexed: 11/16/2022]
Abstract
Intentional bacterial infections can produce efficacious antitumor responses in mice, rats, dogs, and humans. However, low overall success rates and intense side effects prevent such approaches from being employed clinically. In this work, we titered bacteria and/or the proinflammatory cytokine TNFα in a set of established murine models of cancer. To interpret the experiments conducted, we considered and calibrated a tumor-effector cell recruitment model under the influence of functional tumor-associated vasculature. In this model, bacterial infections and TNFα enhanced immune activity and altered vascularization in the tumor bed. Information to predict bacterial therapy outcomes was provided by pretreatment tumor size and the underlying immune recruitment dynamics. Notably, increasing bacterial loads did not necessarily produce better long-term tumor control, suggesting that tumor sizes affected optimal bacterial loads. Short-term treatment responses were favored by high concentrations of effector cells postinjection, such as induced by higher bacterial loads, but in the longer term did not correlate with an effective restoration of immune surveillance. Overall, our findings suggested that a combination of intermediate bacterial loads with low levels TNFα administration could enable more favorable outcomes elicited by bacterial infections in tumor-bearing subjects. Cancer Res; 77(7); 1553-63. ©2017 AACR.
Collapse
Affiliation(s)
- Haralampos Hatzikirou
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Center for Information Services and High Performance Computing, Technische Universität Dresden, Dresden, Germany
| | - Juan Carlos López Alfonso
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Center for Information Services and High Performance Computing, Technische Universität Dresden, Dresden, Germany
| | - Sara Leschner
- Molecular Immunology, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Siegfried Weiss
- Molecular Immunology, Helmholtz Center for Infection Research, Braunschweig, Germany.,Institute of Immunology, Medical School Hannover, Hannover, Germany
| | - Michael Meyer-Hermann
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany. .,Institute for Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig, Germany
| |
Collapse
|
35
|
Yang YW, Zhang CM, Huang XJ, Zhang XX, Zhang LK, Li JH, Hua ZC. Tumor-targeted delivery of a C-terminally truncated FADD (N-FADD) significantly suppresses the B16F10 melanoma via enhancing apoptosis. Sci Rep 2016; 6:34178. [PMID: 27767039 PMCID: PMC5073321 DOI: 10.1038/srep34178] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 09/08/2016] [Indexed: 01/21/2023] Open
Abstract
Fas-associated protein with death domain (FADD), a pivotal adaptor protein transmitting apoptotic signals, is indispensable for the induction of extrinsic apoptosis. However, overexpression of FADD can form large, filamentous aggregates, termed death effector filaments (DEFs) by self-association and initiate apoptosis independent of receptor cross-linking. A mutant of FADD, which is truncated of the C-terminal tail (m-FADD, 182–205 aa) named N-FADD (m-FADD, 1–181 aa), can dramatically up-regulate the strength of FADD self-association and increase apoptosis. In this study, it was found that over-expression of FADD or N-FADD caused apoptosis of B16F10 cells in vitro, even more, N-FADD showed a more potent apoptotic effect than FADD. Meanwhile, Attenuated Salmonella Typhimurium strain VNP20009 was engineered to express FADD or N-FADD under the control of a hypoxia-induced NirB promoter and each named VNP-pN-FADD and VNP-pN-N-FADD. The results showed both VNP-pN-FADD and VNP-pN-N-FADD delayed tumor growth in B16F10 mice model, while VNP-pN-N-FADD suppressed melanoma growth more significantly than VNP-pN-FADD. Additionally, VNP-pN-FADD and VNP-pN-N-FADD induced apoptosis of tumor cells by activating caspase-dependent apoptotic pathway. Our results show that N-FADD is a more potent apoptotic inducer and VNP20009-mediated targeted expression of N-FADD provides a possible cancer gene therapeutic approach for the treatment of melanoma.
Collapse
Affiliation(s)
- Yun-Wen Yang
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, Jiangsu, China
| | - Chun-Mei Zhang
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, Jiangsu, China
| | - Xian-Jie Huang
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, Jiangsu, China
| | - Xiao-Xin Zhang
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, Jiangsu, China
| | - Lin-Kai Zhang
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, Jiangsu, China
| | - Jia-Huang Li
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, Jiangsu, China.,Changzhou High-Tech Research Institute of Nanjing University and Jiangsu TargetPharma Laboratories Inc., Changzhou, 213164, Jiangsu, China
| | - Zi-Chun Hua
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, Jiangsu, China.,Changzhou High-Tech Research Institute of Nanjing University and Jiangsu TargetPharma Laboratories Inc., Changzhou, 213164, Jiangsu, China
| |
Collapse
|
36
|
Kazmierczak RA, Gentry B, Mumm T, Schatten H, Eisenstark A. Salmonella Bacterial Monotherapy Reduces Autochthonous Prostate Tumor Burden in the TRAMP Mouse Model. PLoS One 2016; 11:e0160926. [PMID: 27504973 PMCID: PMC4978392 DOI: 10.1371/journal.pone.0160926] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 07/26/2016] [Indexed: 12/31/2022] Open
Abstract
Attenuated Salmonella typhimurium injected in the circulatory system of mammals selectively targets tumors. Using weekly intraperitoneal injections of attenuated Salmonella strain CRC2631, we tested for regression and/or inhibition of tumor development in the TRAMP prostate tumor mouse model, which utilizes SV40 early region expression for autochthonous formation of prostate tumors that progress into metastatic, poorly differentiated prostatic carcinomas in an immunocompetent murine model. Thirteen weekly intraperitoneal administrations of 105–107 CFU CRC2631 into 10 week old mice were well tolerated by the TRAMP model. Sacrifice and histological analysis of TRAMP prostates at 22 weeks indicated that Salmonella monotherapy at administrated levels decrease visible tumor size (>29%) but did not significantly inhibit previously described SV40 expression-driven TRAMP tumor progression to undifferentiated carcinomas when histologically examined. In conclusion, this work demonstrates baseline results for CRC2631 Salmonella monotherapy using the immunocompetent TRAMP prostate tumor model in preparation for study of combination therapies that resolve autochthonously generated TRAMP prostate tumors, further reduce tumor size, or inhibit prostate tumor progression.
Collapse
Affiliation(s)
- Robert A. Kazmierczak
- Cancer Research Center, Columbia, Missouri, United States of America
- Division of Biological Sciences, University of Missouri, Columbia, Missouri, United States of America
- * E-mail:
| | - Bettina Gentry
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, United States of America
| | - Tyler Mumm
- Cancer Research Center, Columbia, Missouri, United States of America
- Division of Biological Sciences, University of Missouri, Columbia, Missouri, United States of America
| | - Heide Schatten
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, United States of America
| | - Abraham Eisenstark
- Cancer Research Center, Columbia, Missouri, United States of America
- Division of Biological Sciences, University of Missouri, Columbia, Missouri, United States of America
| |
Collapse
|
37
|
Nguyen VH, Min JJ. Salmonella-Mediated Cancer Therapy: Roles and Potential. Nucl Med Mol Imaging 2016; 51:118-126. [PMID: 28559936 DOI: 10.1007/s13139-016-0415-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 02/29/2016] [Accepted: 03/28/2016] [Indexed: 01/21/2023] Open
Abstract
The use of bacteria for cancer therapy, which was proposed many years ago, was not recognized as a potential therapeutic strategy until recently. Technological advances and updated knowledge have enabled the genetic engineering of bacteria for their safe and effective application in the treatment of cancer. The efficacy of radiotherapy depends mainly on tissue oxygen levels, and low oxygen concentrations in necrotic and hypoxic regions are a common cause of treatment failure. In addition, the distribution of a drug is important for the therapeutic effect of chemotherapy, and the poor vasculature in tumors impairs drug delivery, limiting the efficacy of a drug, especially in necrotic and hypoxic regions. Bacteria-mediated cancer therapy (BMCT) relies on facultative anaerobes that can survive in well or poorly oxygenated regions, and it therefore improves the therapeutic efficacy drug distribution throughout the tumor mass. Since the mid-1990s, the number of published bacterial therapy papers has increased rapidly, with a doubling time of 2.5 years in which the use of Salmonella increased significantly. BMCT is being reevaluated to overcome some of the drawbacks of conventional therapies. This review focuses on Salmonella-mediated cancer therapy as the most widely used type of BMCT.2.
Collapse
Affiliation(s)
- Vu Hong Nguyen
- Department of Experimental Therapeutics, Beckman Research Institute of City of Hope, Duarte, California, 1500 East Duarte Road, Duarte, CA 91010 USA
| | - Jung-Joon Min
- Department of Nuclear Medicine, Chonnam National University Medical School, 5 Hak1 dong, Dong-gu, Gwangju, 501-746 Republic of Korea
| |
Collapse
|
38
|
Deyneko IV, Kasnitz N, Leschner S, Weiss S. Composing a Tumor Specific Bacterial Promoter. PLoS One 2016; 11:e0155338. [PMID: 27171245 PMCID: PMC4865170 DOI: 10.1371/journal.pone.0155338] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 04/27/2016] [Indexed: 12/12/2022] Open
Abstract
Systemically applied Salmonella enterica spp. have been shown to invade and colonize neoplastic tissues where it retards the growth of many tumors. This offers the possibility to use the bacteria as a vehicle for the tumor specific delivery of therapeutic molecules. Specificity of such delivery is solely depending on promoter sequences that control the production of a target molecule. We have established the functional structure of bacterial promoters that are transcriptionally active exclusively in tumor tissues after systemic application. We observed that the specific transcriptional activation is accomplished by a combination of a weak basal promoter and a strong FNR binding site. This represents a minimal set of control elements required for such activation. In natural promoters, additional DNA remodeling elements are found that alter the level of transcription quantitatively. Inefficiency of the basal promoter ensures the absence of transcription outside tumors. As a proof of concept, we compiled an artificial promoter sequence from individual motifs representing FNR and basal promoter and showed specific activation in a tumor microenvironment. Our results open possibilities for the generation of promoters with an adjusted level of expression of target proteins in particular for applications in bacterial tumor therapy.
Collapse
Affiliation(s)
- Igor V. Deyneko
- Molecular Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- * E-mail:
| | - Nadine Kasnitz
- Molecular Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Sara Leschner
- Molecular Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Siegfried Weiss
- Molecular Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Institute of Immunology, Medical School Hannover, Hannover, Germany
| |
Collapse
|
39
|
Strains, Mechanism, and Perspective: Salmonella-Based Cancer Therapy. Int J Microbiol 2016; 2016:5678702. [PMID: 27190519 PMCID: PMC4848419 DOI: 10.1155/2016/5678702] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 03/11/2016] [Accepted: 03/20/2016] [Indexed: 01/21/2023] Open
Abstract
Recently, investigation of bacterial-based tumor therapy has regained focus due to progress in molecular, cellular, and microbial biology. Many bacteria such as Salmonella, Listeria, Escherichia, and Clostridium have proved to have tumor targeting and in some cases even tumor-destroying phenotypes. Furthermore, bacterial clinical treatments for cancer have been improved by combination with other therapeutic methods such as chemotherapeutic drugs and radioactive agents. Synthetic biology techniques have also driven the development of new bacterial-based cancer therapies. However, basic questions about the mechanisms of bacterial-mediated tumor targeting and destruction are still being elucidated. In this review, we focus on three tumor-therapeutic Salmonella models, the most intensively studied bacterial genus in this field. One of these Salmonella models is our Salmonella enterica serovar Typhimurium LT2 derived strain CRC2631, engineered to minimize toxicity but maximize tumor-targeting and destruction effects. The other two are VNP20009 and A1-R. We compare the means by which these therapeutic candidate strain models were selected for study, their tumor targeting and tumor destruction phenotypes in vitro and in vivo, and what is currently known about the mechanisms by which they target and destroy tumors.
Collapse
|
40
|
Felgner S, Kocijancic D, Frahm M, Weiss S. Bacteria in Cancer Therapy: Renaissance of an Old Concept. Int J Microbiol 2016; 2016:8451728. [PMID: 27051423 PMCID: PMC4802035 DOI: 10.1155/2016/8451728] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 02/03/2016] [Accepted: 02/11/2016] [Indexed: 01/20/2023] Open
Abstract
The rising incidence of cancer cases worldwide generates an urgent need of novel treatment options. Applying bacteria may represent a valuable therapeutic variant that is intensively investigated nowadays. Interestingly, the idea to apply bacteria wittingly or unwittingly dates back to ancient times and was revived in the 19th century mainly by the pioneer William Coley. This review summarizes and compares the results of the past 150 years in bacteria mediated tumor therapy from preclinical to clinical studies. Lessons we have learned from the past provide a solid foundation on which to base future efforts. In this regard, several perspectives are discussed by which bacteria in addition to their intrinsic antitumor effect can be used as vector systems that shuttle therapeutic compounds into the tumor. Strategic solutions like these provide a sound and more apt exploitation of bacteria that may overcome limitations of conventional therapies.
Collapse
Affiliation(s)
- Sebastian Felgner
- Department of Molecular Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Dino Kocijancic
- Department of Molecular Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Michael Frahm
- Department of Molecular Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Siegfried Weiss
- Department of Molecular Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
41
|
Lei L, Wang W, Xia C, Liu F. Salmonella Virulence Factor SsrAB Regulated Factor Modulates Inflammatory Responses by Enhancing the Activation of NF-κB Signaling Pathway. THE JOURNAL OF IMMUNOLOGY 2015; 196:792-802. [PMID: 26673132 DOI: 10.4049/jimmunol.1500679] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 11/14/2015] [Indexed: 12/20/2022]
Abstract
Effector proteins encoded by Salmonella pathogenicity islands play a key role in promoting bacterial intracellular survival, colonization, and pathogenesis. In this study, we investigated the function of the virulence-associated effector SrfA (SsrAB regulated factor) both in macrophages in vitro and in infected mice in vivo. SrfA was secreted into the cytoplasm during S. Typhimurium infection and disassociated IL-1R-associated kinase-1 (IRAK-1) from the IRAK-1-Toll interacting protein (Tollip) complex by interacting with Tollip. The released IRAK-1 was phosphorylated and subsequently activated the NF-κB signaling pathway, which enhanced the LPS-induced expression of inflammatory cytokines, such as IL-8, IL-1β, and TNF-α. The coupling of ubiquitin to endoplasmic reticulum degradation aa 183-219 domain of Tollip is the binding region for SrfA, and both the MDaa207-226 and CTaa357-377 regions of SrfA mediate binding to Tollip and NF-κB signaling activation. Deletion of SrfA in S. Typhimurium had no notable effects on its replication but impaired the induction of NF-κB activation in infected macrophages. The mice infected with srfA-deficient bacteria exhibited a decreased inflammatory response and an increased survival rate compared with those infected with wild-type S. Typhimurium. We conclude that SrfA is a novel Salmonella virulence effector that helps modulate host inflammatory responses by promoting NF-κB signaling activation.
Collapse
Affiliation(s)
- Lei Lei
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China; and
| | - Wenbiao Wang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China; and
| | - Chuan Xia
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China; and
| | - Fenyong Liu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China; and Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA 94720
| |
Collapse
|
42
|
Osswald A, Sun Z, Grimm V, Ampem G, Riegel K, Westendorf AM, Sommergruber W, Otte K, Dürre P, Riedel CU. Three-dimensional tumor spheroids for in vitro analysis of bacteria as gene delivery vectors in tumor therapy. Microb Cell Fact 2015; 14:199. [PMID: 26655167 PMCID: PMC4676896 DOI: 10.1186/s12934-015-0383-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 11/11/2015] [Indexed: 12/31/2022] Open
Abstract
Background Several studies in animal models demonstrated that obligate and facultative anaerobic bacteria of the genera Bifidobacterium, Salmonella, or Clostridium specifically colonize solid tumors. Consequently, these and other bacteria are discussed as live vectors to deliver therapeutic genes to inhibit tumor growth. Therapeutic approaches for cancer treatment using anaerobic bacteria have been investigated in different mouse models. In the present study, solid three-dimensional (3D) multicellular tumor spheroids (MCTS) of the colorectal adenocarcinoma cell line HT-29 were generated and tested for their potential to study prodrug-converting enzyme therapies using bacterial vectors in vitro. Results HT-29 MCTS resembled solid tumors displaying all relevant features with an outer zone of proliferating cells and hypoxic and apoptotic regions in the core. Upon incubation with HT-29 MCTS, Bifidobacterium bifidum S17 and Salmonella typhimurium YB1 selectively localized, survived and replicated in hypoxic areas inside MCTS. Furthermore, spores of the obligate anaerobe Clostridium sporogenes germinated in these hypoxic areas. To further evaluate the potential of MCTS to investigate therapeutic approaches using bacteria as gene delivery vectors, recombinant bifidobacteria expressing prodrug-converting enzymes were used. Expression of a secreted cytosine deaminase in combination with 5-fluorocytosine had no effect on growth of MCTS due to an intrinsic resistance of HT-29 cells to 5-fluorouracil, i.e. the converted drug. However, a combination of the prodrug CB1954 and a strain expressing a secreted chromate reductase effectively inhibited MCTS growth. Conclusions Collectively, the presented results indicate that MCTS are a suitable and reliable model to investigate live bacteria as gene delivery vectors for cancer therapy in vitro. Electronic supplementary material The online version of this article (doi:10.1186/s12934-015-0383-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Annika Osswald
- Institute of Microbiology and Biotechnology, University of Ulm, 89069, Ulm, Germany. .,Department of Lead Discovery, Boehringer Ingelheim RCV GmbH & Co KG, 1121, Vienna, Austria.
| | - Zhongke Sun
- Institute of Microbiology and Biotechnology, University of Ulm, 89069, Ulm, Germany. .,College of Life Sciences and Agriculture, Zhoukou Normal University, Chuanhui District, Zhoukou, 466001, People's Republic of China.
| | - Verena Grimm
- Institute of Microbiology and Biotechnology, University of Ulm, 89069, Ulm, Germany.
| | - Grace Ampem
- Institute of Microbiology and Biotechnology, University of Ulm, 89069, Ulm, Germany.
| | - Karin Riegel
- Institute of Microbiology and Biotechnology, University of Ulm, 89069, Ulm, Germany.
| | - Astrid M Westendorf
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany.
| | - Wolfgang Sommergruber
- Department of Lead Discovery, Boehringer Ingelheim RCV GmbH & Co KG, 1121, Vienna, Austria.
| | - Kerstin Otte
- Institute of Applied Biotechnology, University of Applied Sciences Biberach, 88400, Biberach, Germany.
| | - Peter Dürre
- Institute of Microbiology and Biotechnology, University of Ulm, 89069, Ulm, Germany.
| | - Christian U Riedel
- Institute of Microbiology and Biotechnology, University of Ulm, 89069, Ulm, Germany.
| |
Collapse
|
43
|
Lin IYC, Van TTH, Smooker PM. Live-Attenuated Bacterial Vectors: Tools for Vaccine and Therapeutic Agent Delivery. Vaccines (Basel) 2015; 3:940-72. [PMID: 26569321 PMCID: PMC4693226 DOI: 10.3390/vaccines3040940] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 10/30/2015] [Accepted: 10/30/2015] [Indexed: 12/14/2022] Open
Abstract
Genetically attenuated microorganisms, including pathogenic and commensal bacteria, can be engineered to carry and deliver heterologous antigens to elicit host immunity against both the vector as well as the pathogen from which the donor gene is derived. These live attenuated bacterial vectors have been given much attention due to their capacity to induce a broad range of immune responses including localized mucosal, as well as systemic humoral and/or cell-mediated immunity. In addition, the unique tumor-homing characteristics of these bacterial vectors has also been exploited for alternative anti-tumor vaccines and therapies. In such approach, tumor-associated antigen, immunostimulatory molecules, anti-tumor drugs, or nucleotides (DNA or RNA) are delivered. Different potential vectors are appropriate for specific applications, depending on their pathogenic routes. In this review, we survey and summarize the main features of the different types of live bacterial vectors and discussed the clinical applications in the field of vaccinology. In addition, different approaches for using live attenuated bacterial vectors for anti-cancer therapy is discussed, and some promising pre-clinical and clinical studies in this field are outlined.
Collapse
Affiliation(s)
- Ivan Y C Lin
- School of Applied Sciences, RMIT University, Plenty Road, Bundoora VIC-3083, Australia.
| | - Thi Thu Hao Van
- School of Applied Sciences, RMIT University, Plenty Road, Bundoora VIC-3083, Australia.
| | - Peter M Smooker
- School of Applied Sciences, RMIT University, Plenty Road, Bundoora VIC-3083, Australia.
| |
Collapse
|
44
|
Kempf H, Bleicher M, Meyer-Hermann M. Spatio-Temporal Dynamics of Hypoxia during Radiotherapy. PLoS One 2015; 10:e0133357. [PMID: 26273841 PMCID: PMC4537194 DOI: 10.1371/journal.pone.0133357] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2015] [Accepted: 06/26/2015] [Indexed: 12/27/2022] Open
Abstract
Tumour hypoxia plays a pivotal role in cancer therapy for most therapeutic approaches from radiotherapy to immunotherapy. The detailed and accurate knowledge of the oxygen distribution in a tumour is necessary in order to determine the right treatment strategy. Still, due to the limited spatial and temporal resolution of imaging methods as well as lacking fundamental understanding of internal oxygenation dynamics in tumours, the precise oxygen distribution map is rarely available for treatment planing. We employ an agent-based in silico tumour spheroid model in order to study the complex, localized and fast oxygen dynamics in tumour micro-regions which are induced by radiotherapy. A lattice-free, 3D, agent-based approach for cell representation is coupled with a high-resolution diffusion solver that includes a tissue density-dependent diffusion coefficient. This allows us to assess the space- and time-resolved reoxygenation response of a small subvolume of tumour tissue in response to radiotherapy. In response to irradiation the tumour nodule exhibits characteristic reoxygenation and re-depletion dynamics which we resolve with high spatio-temporal resolution. The reoxygenation follows specific timings, which should be respected in treatment in order to maximise the use of the oxygen enhancement effects. Oxygen dynamics within the tumour create windows of opportunity for the use of adjuvant chemotherapeutica and hypoxia-activated drugs. Overall, we show that by using modelling it is possible to follow the oxygenation dynamics beyond common resolution limits and predict beneficial strategies for therapy and in vitro verification. Models of cell cycle and oxygen dynamics in tumours should in the future be combined with imaging techniques, to allow for a systematic experimental study of possible improved schedules and to ultimately extend the reach of oxygenation monitoring available in clinical treatment.
Collapse
Affiliation(s)
- Harald Kempf
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Frankfurt Institute for Advanced Studies, Frankfurt, Germany
| | - Marcus Bleicher
- Frankfurt Institute for Advanced Studies, Frankfurt, Germany
| | - Michael Meyer-Hermann
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Institute for Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig, Germany
| |
Collapse
|
45
|
Drees JJ, Mertensotto MJ, Augustin LB, Schottel JL, Saltzman DA. Vasculature Disruption Enhances Bacterial Targeting of Autochthonous Tumors. J Cancer 2015; 6:843-8. [PMID: 26284135 PMCID: PMC4532981 DOI: 10.7150/jca.12491] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 06/23/2015] [Indexed: 12/13/2022] Open
Abstract
Attenuated Salmonella enterica serovar Typhimurium (S. Typhimurium) has been developed as a vector to deliver therapeutic agents to tumors. The potential of S. Typhimurium in cancer therapy is largely due to its reported propensity to accumulate at greater than 1,000-fold higher concentrations in tumors relative to healthy tissues. In this study, we compared bacterial colonization of tumors in a subcutaneous transplantation model with a more clinically relevant autochthonous tumor model. Following intravenous administration of attenuated S. Typhimurium strain SL3261, we observed approximately 10,000-fold less bacteria in autochthonous tumors that sporadically develop in transgenic BALB-neuT mice compared to tumors developed from subcutaneous transplantation of 4T1 murine breast cancer cells in BALB/c mice. Treatment of BALB-neuT mice with a vasculature-disrupting agent (VDA) prior to bacterial treatment caused necrosis of tumor tissue and significantly increased the bacterial targeting of autochthonous tumors by approximately 1,000-fold. These observations emphasize the importance of appropriate model selection in developing bacteria-based cancer therapies and demonstrate the potential of combining VDA pre-treatment with bacteria to facilitate targeting of clinically relevant tumors.
Collapse
Affiliation(s)
- Jeremy J Drees
- 1. Department of Surgery, University of Minnesota, 195 MMC, 420 Delaware St SE, Minneapolis, Minnesota 55455, USA
| | - Michael J Mertensotto
- 1. Department of Surgery, University of Minnesota, 195 MMC, 420 Delaware St SE, Minneapolis, Minnesota 55455, USA
| | - Lance B Augustin
- 1. Department of Surgery, University of Minnesota, 195 MMC, 420 Delaware St SE, Minneapolis, Minnesota 55455, USA
| | - Janet L Schottel
- 2. Department of Biochemistry, Molecular Biology & Biophysics, University of Minnesota, 1479 Gortner Ave., St. Paul, MN 55108, USA
| | - Daniel A Saltzman
- 1. Department of Surgery, University of Minnesota, 195 MMC, 420 Delaware St SE, Minneapolis, Minnesota 55455, USA
| |
Collapse
|
46
|
Wong S, Slavcev R. Treating cancer with infection: a review on bacterial cancer therapy. Lett Appl Microbiol 2015; 61:107-12. [DOI: 10.1111/lam.12436] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 04/08/2015] [Accepted: 04/27/2015] [Indexed: 12/13/2022]
Affiliation(s)
- S. Wong
- School of Pharmacy; University of Waterloo; Waterloo ON Canada
| | - R.A. Slavcev
- School of Pharmacy; University of Waterloo; Waterloo ON Canada
| |
Collapse
|
47
|
Sahari A, Traore MA, Scharf BE, Behkam B. Directed transport of bacteria-based drug delivery vehicles: bacterial chemotaxis dominates particle shape. Biomed Microdevices 2015; 16:717-25. [PMID: 24907051 DOI: 10.1007/s10544-014-9876-y] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Several attenuated and non-pathogenic bacterial species have been demonstrated to actively target diseased sites and successfully deliver plasmid DNA, proteins and other therapeutic agents into mammalian cells. These disease-targeting bacteria can be employed for targeted delivery of therapeutic and imaging cargos in the form of a bio-hybrid system. The bio-hybrid drug delivery system constructed here is comprised of motile Escherichia coli MG1655 bacteria and elliptical disk-shaped polymeric microparticles. The transport direction for these vehicles can be controlled through biased random walk of the attached bacteria in presence of chemoattractant gradients in a process known as chemotaxis. In this work, we utilize a diffusion-based microfluidic platform to establish steady linear concentration gradients of a chemoattractant and investigate the roles of chemotaxis and geometry in transport of bio-hybrid drug delivery vehicles. Our experimental results demonstrate for the first time that bacterial chemotactic response dominates the effect of body shape in extravascular transport; thus, the non-spherical system could be more favorable for drug delivery applications owing to the known benefits of using non-spherical particles for vascular transport (e.g. relatively long circulation time).
Collapse
Affiliation(s)
- Ali Sahari
- School of Biomedical Engineering and Sciences, Virginia Tech-Wake Forest University, Blacksburg, VA, 24061, USA
| | | | | | | |
Collapse
|
48
|
Stern C, Kasnitz N, Kocijancic D, Trittel S, Riese P, Guzman CA, Leschner S, Weiss S. Induction of CD4(+) and CD8(+) anti-tumor effector T cell responses by bacteria mediated tumor therapy. Int J Cancer 2015; 137:2019-28. [PMID: 25868911 DOI: 10.1002/ijc.29567] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 02/05/2015] [Accepted: 03/11/2015] [Indexed: 01/30/2023]
Abstract
Facultative anaerobic bacteria like E. coli can colonize solid tumors often resulting in tumor growth retardation or even clearance. Little mechanistic knowledge is available for this phenomenon which is however crucial for optimization and further implementation in the clinic. Here, we show that intravenous injections with E. coli TOP10 can induce clearance of CT26 tumors in BALB/c mice. Importantly, re-challenging mice which had cleared tumors showed that clearance was due to a specific immune reaction. Accordingly, lymphopenic mice never showed tumor clearance after infection. Depletion experiments revealed that during induction phase, CD8(+) T cells are the sole effectors responsible for tumor clearance while in the memory phase CD8(+) and CD4(+) T cells were involved. This was confirmed by adoptive transfer. CD4(+) and CD8(+) T cells could reject newly set tumors while CD8(+) T cells could even reject established tumors. Detailed analysis of adoptively transferred CD4(+) T cells during tumor challenge revealed expression of granzyme B, FasL, TNF-α and IFN-γ in such T cells that might be involved in the anti-tumor activity. Our findings should pave the way for further optimization steps of this promising therapy.
Collapse
Affiliation(s)
- Christian Stern
- Department of Molecular Immunology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Nadine Kasnitz
- Department of Molecular Immunology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Dino Kocijancic
- Department of Molecular Immunology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Stephanie Trittel
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Peggy Riese
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Carlos A Guzman
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Sara Leschner
- Department of Molecular Immunology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Siegfried Weiss
- Department of Molecular Immunology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| |
Collapse
|
49
|
Frahm M, Felgner S, Kocijancic D, Rohde M, Hensel M, Curtiss R, Erhardt M, Weiss S. Efficiency of conditionally attenuated Salmonella enterica serovar Typhimurium in bacterium-mediated tumor therapy. mBio 2015; 6:e00254-15. [PMID: 25873375 PMCID: PMC4453544 DOI: 10.1128/mbio.00254-15] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 03/23/2015] [Indexed: 01/05/2023] Open
Abstract
UNLABELLED Increasing numbers of cancer cases generate a great urge for new treatment options. Applying bacteria like Salmonella enterica serovar Typhimurium for cancer therapy represents an intensively explored option. These bacteria have been shown not only to colonize solid tumors but also to exhibit an intrinsic antitumor effect. In addition, they could serve as tumor-targeting vectors for therapeutic molecules. However, the pathogenic S. Typhimurium strains used for tumor therapy need to be attenuated for safe application. Here, lipopolysaccharide (LPS) deletion mutants (ΔrfaL, ΔrfaG, ΔrfaH, ΔrfaD, ΔrfaP, and ΔmsbB mutants) of Salmonella were investigated for efficiency in tumor therapy. Of such variants, the ΔrfaD and ΔrfaG deep rough mutants exhibited the best tumor specificity and lowest pathogenicity. However, the intrinsic antitumor effect was found to be weak. To overcome this limitation, conditional attenuation was tested by complementing the mutants with an inducible arabinose promoter. The chromosomal integration of the respective LPS biosynthesis genes into the araBAD locus exhibited the best balance of attenuation and therapeutic benefit. Thus, the present study establishes a basis for the development of an applicably cancer therapeutic bacterium. IMPORTANCE Cancer has become the second most frequent cause of death in industrialized countries. This and the drawbacks of routine therapies generate an urgent need for novel treatment options. Applying appropriately modified S. Typhimurium for therapy represents the major challenge of bacterium-mediated tumor therapy. In the present study, we demonstrated that Salmonella bacteria conditionally modified in their LPS phenotype exhibit a safe tumor-targeting phenotype. Moreover, they could represent a suitable vehicle to shuttle therapeutic compounds directly into cancerous tissue without harming the host.
Collapse
Affiliation(s)
- Michael Frahm
- Department of Molecular Immunology, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Sebastian Felgner
- Department of Molecular Immunology, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Dino Kocijancic
- Department of Molecular Immunology, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Manfred Rohde
- Central Facility for Microscopy, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Michael Hensel
- Division of Microbiology, University of Osnabrück, Osnabrück, Germany
| | - Roy Curtiss
- Biodesign Institute, Center for Infectious Diseases and Vaccinology, Tempe, Arizona, USA
| | - Marc Erhardt
- Junior Research Group Infection Biology of Salmonella, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Siegfried Weiss
- Department of Molecular Immunology, Helmholtz Center for Infection Research, Braunschweig, Germany
| |
Collapse
|
50
|
Zhang M, Forbes NS. Trg-deficient Salmonella colonize quiescent tumor regions by exclusively penetrating or proliferating. J Control Release 2014; 199:180-9. [PMID: 25523033 DOI: 10.1016/j.jconrel.2014.12.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 12/04/2014] [Accepted: 12/13/2014] [Indexed: 01/15/2023]
Abstract
Chemotherapeutics fail to effectively treat tumors because they cannot reach quiescent regions far from blood vessels. Motile Salmonella are an attractive delivery system that could break this therapeutic barrier. However, little is known about the dissemination and tissue penetration of individual bacteria in tumors after intravenous administration. We hypothesized that eliminating the Trg receptor would improve accumulation in tumor quiescence. To test this hypothesis, we deleted the trg gene from nonpathogenic Salmonella. To quantify individual bacterial behavior, we measured tissue penetration in a tumor-on-a-chip device and measured colony localization in mouse tumors using immunofluorescence. In tumors in vitro and in mice, trg(-) Salmonella penetrated farther into tissue than control bacteria. This difference in localization was caused by the inability to sense sugars in well perfused tissue. Three distinct bacterial phenotypes were observed: proliferating, penetrating, and inactive. Large proliferating colonies, containing more than 40% of individual bacteria, only formed less than 60μm from blood vessels. Small colonies, in comparison, were present both near (inactive) and far (penetrating) from vessels. The farthest was 361.2μm from a vessel, demonstrating the ability to target avascular regions. In addition, colonization was most pronounced in poorly vascularized tumor regions. We show that deletion of trg amplifies Salmonella accumulation in quiescent tumor regions, and, for the first time, identify biological processes that control bacterial distribution in tumors. Understanding how Salmonella penetrate tissue, target quiescence and specifically replicate in tumors are essential steps toward creating a tightly controlled, tunable bacterial therapy.
Collapse
Affiliation(s)
- Miaomin Zhang
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, USA; Pioneer Valley Life Sciences Institute, Springfield, MA, USA
| | - Neil S Forbes
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, USA; Pioneer Valley Life Sciences Institute, Springfield, MA, USA.
| |
Collapse
|