1
|
Ooi VY, Yeh TY. Recent Advances and Mechanisms of Phage-Based Therapies in Cancer Treatment. Int J Mol Sci 2024; 25:9938. [PMID: 39337427 PMCID: PMC11432602 DOI: 10.3390/ijms25189938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/07/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
The increasing interest in bacteriophage technology has prompted its novel applications to treat different medical conditions, most interestingly cancer. Due to their high specificity, manipulability, nontoxicity, and nanosize nature, phages are promising carriers in targeted therapy and cancer immunotherapy. This approach is particularly timely, as current challenges in cancer research include damage to healthy cells, inefficiency in targeting, obstruction by biological barriers, and drug resistance. Some cancers are being kept at the forefront of phage research, such as colorectal cancer and HCC, while others like lymphoma, cervical cancer, and myeloma have not been retouched in a decade. Common mechanisms are immunogenic antigen display on phage coats and the use of phage as transporters to carry drugs, genes, and other molecules. To date, popular phage treatments being tested are gene therapy and phage-based vaccines using M13 and λ phage, with some vaccines having advanced to human clinical trials. The results from most of these studies have been promising, but limitations in phage-based therapies such as reticuloendothelial system clearance or diffusion inefficiency must be addressed. Before phage-based therapies for cancer can be successfully used in oncology practice, more in-depth research and support from local governments are required.
Collapse
Affiliation(s)
| | - Ting-Yu Yeh
- Agricultural Biotechnology Laboratory, Auxergen Inc., Riti Rossi Colwell Center, 701 E Pratt Street, Baltimore, MD 21202, USA
| |
Collapse
|
2
|
Vernhes E, Larbi Chérif L, Ducrot N, Vanbergue C, Ouldali M, Zig L, Sidibe N, Hoos S, Ramirez-Chamorro L, Renouard M, Rossier O, England P, Schoehn G, Boulanger P, Benihoud K. Antigen self-anchoring onto bacteriophage T5 capsid-like particles for vaccine design. NPJ Vaccines 2024; 9:6. [PMID: 38177231 PMCID: PMC10766600 DOI: 10.1038/s41541-023-00798-5] [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: 10/19/2022] [Accepted: 12/12/2023] [Indexed: 01/06/2024] Open
Abstract
The promises of vaccines based on virus-like particles stimulate demand for universal non-infectious virus-like platforms that can be efficiently grafted with large antigens. Here, we harnessed the modularity and extreme affinity of the decoration protein pb10 for the capsid of bacteriophage T5. SPR experiments demonstrated that pb10 fused to mCherry or to the model antigen ovalbumin (Ova) retained picomolar affinity for DNA-free T5 capsid-like particles (T5-CLPs), while cryo-EM studies attested to the full occupancy of the 120 capsid binding sites. Mice immunization with CLP-bound pb10-Ova chimeras elicited strong long-lasting anti-Ova humoral responses involving a large panel of isotypes, as well as CD8+ T cell responses, without any extrinsic adjuvant. Therefore, T5-CLP constitutes a unique DNA-free bacteriophage capsid able to display a regular array of large antigens through highly efficient chemical-free anchoring. Its ability to elicit robust immune responses paves the way for further development of this novel vaccination platform.
Collapse
Affiliation(s)
- Emeline Vernhes
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198, Gif-sur-Yvette, France
| | - Linda Larbi Chérif
- Université Paris-Saclay, Gustave Roussy, CNRS, Metabolic and systemic aspects of oncogenesis for new therapeutic approaches (METSY), 94805, Villejuif, France
| | - Nicolas Ducrot
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198, Gif-sur-Yvette, France
| | - Clément Vanbergue
- Université Paris-Saclay, Gustave Roussy, CNRS, Metabolic and systemic aspects of oncogenesis for new therapeutic approaches (METSY), 94805, Villejuif, France
| | - Malika Ouldali
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198, Gif-sur-Yvette, France
| | - Lena Zig
- Université Paris-Saclay, Gustave Roussy, CNRS, Metabolic and systemic aspects of oncogenesis for new therapeutic approaches (METSY), 94805, Villejuif, France
| | - N'diaye Sidibe
- Université Paris-Saclay, Gustave Roussy, CNRS, Metabolic and systemic aspects of oncogenesis for new therapeutic approaches (METSY), 94805, Villejuif, France
| | - Sylviane Hoos
- Institut Pasteur, Biophysique Moléculaire, CNRS UMR 3528, Paris, France
| | - Luis Ramirez-Chamorro
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198, Gif-sur-Yvette, France
| | - Madalena Renouard
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198, Gif-sur-Yvette, France
| | - Ombeline Rossier
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198, Gif-sur-Yvette, France
| | - Patrick England
- Institut Pasteur, Biophysique Moléculaire, CNRS UMR 3528, Paris, France
| | - Guy Schoehn
- Univ. Grenoble Alpes, CNRS, CEA, IBS, F-38000, Grenoble, France
| | - Pascale Boulanger
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198, Gif-sur-Yvette, France.
| | - Karim Benihoud
- Université Paris-Saclay, Gustave Roussy, CNRS, Metabolic and systemic aspects of oncogenesis for new therapeutic approaches (METSY), 94805, Villejuif, France.
| |
Collapse
|
3
|
Yu T, Sun Z, Cao X, Pang Q, Deng H. Recent trends in T7 phage application in diagnosis and treatment of various diseases. Int Immunopharmacol 2022; 110:109071. [DOI: 10.1016/j.intimp.2022.109071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 06/25/2022] [Accepted: 07/14/2022] [Indexed: 11/05/2022]
|
4
|
Abstract
Bacteriophages have been explored for their uses in vaccine development, due to the ease of propagation while displaying epitopes in high density. Bacteriophage T7 has been demonstrated to be useful in the production of potential vaccine candidates for various diseases, including influenza A, foot-and mouth disease (FMD), and cancers. In this chapter, we described the use of phage T7 to display potential foot-and-mouth disease virus (FMDV) epitope, from cloning to expression, purification, and immunization in a mouse model.
Collapse
Affiliation(s)
- Chuan Loo Wong
- Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Chean Yeah Yong
- Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Khai Wooi Lee
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Selangor, Malaysia.
| |
Collapse
|
5
|
Bacteriophages as Therapeutic and Diagnostic Vehicles in Cancer. Pharmaceuticals (Basel) 2021; 14:ph14020161. [PMID: 33671476 PMCID: PMC7923149 DOI: 10.3390/ph14020161] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/15/2021] [Accepted: 02/16/2021] [Indexed: 12/11/2022] Open
Abstract
Evolution of nanomedicine is the re-design of synthetic and biological carriers to implement novel theranostic platforms. In recent years, bacteriophage research favors this process, which has opened up new roads in drug and gene delivery studies. By displaying antibodies, peptides, or proteins on the surface of different bacteriophages through the phage display technique, it is now possible to unravel specific molecular determinants of both cancer cells and tumor-associated microenvironmental molecules. Downstream applications are manifold, with peptides being employed most of the times to functionalize drug carriers and improve their therapeutic index. Bacteriophages themselves were proven, in this scenario, to be good carriers for imaging molecules and therapeutics as well. Moreover, manipulation of their genetic material to stably vehiculate suicide genes within cancer cells substantially changed perspectives in gene therapy. In this review, we provide examples of how amenable phages can be used as anticancer agents, especially because their systemic administration is possible. We also provide some insights into how their immunogenic profile can be modulated and exploited in immuno-oncology for vaccine production.
Collapse
|
6
|
Hess KL, Jewell CM. Phage display as a tool for vaccine and immunotherapy development. Bioeng Transl Med 2020; 5:e10142. [PMID: 31989033 PMCID: PMC6971447 DOI: 10.1002/btm2.10142] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 07/15/2019] [Accepted: 08/22/2019] [Indexed: 12/11/2022] Open
Abstract
Bacteriophages, or phages, are viruses that specifically infect bacteria and coopt the cellular machinery to create more phage proteins, eventually resulting in the release of new phage particles. Phages are heavily utilized in bioengineering for applications ranging from tissue engineering scaffolds to immune signal delivery. Of specific interest to vaccines and immunotherapies, phages have demonstrated an ability to activate both the innate and adaptive immune systems. The genome of these viral particles can be harnessed for DNA vaccination, or the surface proteins can be exploited for antigen display. More specifically, genes that encode an antigen of interest can be spliced into the phage genome, allowing antigenic proteins or peptides to be displayed by fusion to phage capsid proteins. Phages therefore present antigens to immune cells in a highly ordered and repetitive manner. This review discusses the use of phage with adjuvanting activity as antigen delivery vehicles for vaccination against infectious disease and cancer.
Collapse
Affiliation(s)
- Krystina L. Hess
- U.S. Army Combat Capabilities Development Command Chemical Biological CenterAberdeen Proving GroundMaryland
| | - Christopher M. Jewell
- Fischell Department of BioengineeringUniversity of MarylandCollege ParkMaryland
- Robert E. Fischell Institute for Biomedical DevicesCollege ParkMaryland
- Department of Microbiology and ImmunologyUniversity of Maryland Medical SchoolBaltimoreMaryland
- Marlene and Stewart Greenebaum Cancer CenterBaltimoreMaryland
- U.S. Department of Veterans AffairsBaltimoreMaryland
| |
Collapse
|
7
|
Kodumudi KN, Ramamoorthi G, Snyder C, Basu A, Jia Y, Awshah S, Beyer AP, Wiener D, Lam L, Zhang H, Greene MI, Costa RLB, Czerniecki BJ. Sequential Anti-PD1 Therapy Following Dendritic Cell Vaccination Improves Survival in a HER2 Mammary Carcinoma Model and Identifies a Critical Role for CD4 T Cells in Mediating the Response. Front Immunol 2019; 10:1939. [PMID: 31475002 PMCID: PMC6702967 DOI: 10.3389/fimmu.2019.01939] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 07/31/2019] [Indexed: 12/27/2022] Open
Abstract
Patients with metastatic HER2 breast cancer (MBC) often become resistant to HER 2 targeted therapy and have recurrence of disease. The Panacea trial suggested that HER2 MBC patients were more likely to respond to checkpoint therapy if TIL were present or if tumor expressed PD-L1. We assessed whether type I polarized dendritic cells (DC1) could improve checkpoint therapy in a preclinical model of HER2+ breast cancer. TUBO bearing mice were vaccinated with either MHC class I or class II HER2 peptide pulsed DC1 (class I or class II HER2-DC1) concurrently or sequentially with administration of anti-PD-1 or anti-PDL1. Infiltration of tumors by immune cells, induction of anti-HER2 immunity and response to therapy was evaluated. Class I or class II HER2-DC1 vaccinated mice generated anti-HER2 CD8 or CD4+ T cell immune responses and demonstrated delayed tumor growth. Combining both MHC class I and II HER2-pulsed DC1 did not further result in inhibition of tumor growth or enhanced survival compared to individual administration. Interestingly class II HER2-DC1 led to both increased CD4 and CD8 T cells in the tumor microenvironment while class I peptides typically resulted in only increased CD8 T cells. Anti-PD-1 but not anti-PD-L1 administered sequentially with class I or class II HER2-DC1 vaccine could improve the efficacy of HER2-DC1 vaccine as measured by tumor growth, survival, infiltration of tumors by T cells and increase in systemic anti-HER2 immune responses. Depletion of CD4+ T cells abrogated the anti-tumor efficacy of combination therapy with class II HER2-DC1 and anti-PD-1, suggesting that tumor regression was CD4 dependent. Since class II HER2-DC1 was as effective as class I, we combined class II HER2-DC1 vaccine with anti-rat neu antibodies and anti-PD-1 therapy. Combination therapy demonstrated further delay in tumor growth, and enhanced survival compared to control mice. In summary, Class II HER2-DC1 drives both a CD4 and CD8 T cell tumor infiltration that leads to increased survival, and in combination with anti-HER2 therapy and checkpoint blockade can improve survival in preclinical models of HER2 positive breast cancer and warrants exploration in patients with HER2 MBC.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal, Humanized/immunology
- Antibodies, Monoclonal, Humanized/pharmacology
- B7-H1 Antigen/antagonists & inhibitors
- B7-H1 Antigen/immunology
- B7-H1 Antigen/metabolism
- Breast Neoplasms/immunology
- Breast Neoplasms/metabolism
- Breast Neoplasms/therapy
- CD4-Positive T-Lymphocytes/drug effects
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/immunology
- Cell Line, Tumor
- Combined Modality Therapy
- Dendritic Cells/immunology
- Female
- Humans
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/therapy
- Mice, Inbred BALB C
- Mice, Transgenic
- Programmed Cell Death 1 Receptor/antagonists & inhibitors
- Programmed Cell Death 1 Receptor/immunology
- Programmed Cell Death 1 Receptor/metabolism
- Rats
- Receptor, ErbB-2/immunology
- Receptor, ErbB-2/metabolism
- Survival Analysis
- Treatment Outcome
- Tumor Burden/drug effects
- Tumor Burden/immunology
Collapse
Affiliation(s)
- Krithika N. Kodumudi
- Clinical Science & Immunology Program, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Ganesan Ramamoorthi
- Clinical Science & Immunology Program, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Colin Snyder
- Clinical Science & Immunology Program, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Amrita Basu
- Clinical Science & Immunology Program, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Yongsheng Jia
- Clinical Science & Immunology Program, H. Lee Moffitt Cancer Center, Tampa, FL, United States
- Department of Breast Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Sabrina Awshah
- Clinical Science & Immunology Program, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Amber P. Beyer
- Clinical Science & Immunology Program, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Doris Wiener
- Clinical Science & Immunology Program, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Lian Lam
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Hongtao Zhang
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Mark I. Greene
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Ricardo L. B. Costa
- Clinical Science & Immunology Program, H. Lee Moffitt Cancer Center, Tampa, FL, United States
- Department of Breast Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Brian J. Czerniecki
- Clinical Science & Immunology Program, H. Lee Moffitt Cancer Center, Tampa, FL, United States
- Department of Breast Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| |
Collapse
|
8
|
Liu T, Li B, Jiang Y, Zheng C, Zhang L, Wang Y. Screening and identification of novel specific markers of breast cancer stem cells. Oncol Lett 2019; 18:2262-2269. [PMID: 31452727 PMCID: PMC6676669 DOI: 10.3892/ol.2019.10535] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 05/08/2019] [Indexed: 01/11/2023] Open
Abstract
Breast cancer is the leading cause of death among women worldwide. Until recent years, triple negative breast cancer could be divided into 6 types according to different biomarkers with the development of sequence and microarray technology. However, these results rarely have therapeutic impact and still lack validation with the string criteria of clinical studies. Therefore, the present study aimed to screen novel markers of breast cancer stem cells and to verify the specificity in vitro and in vivo. In the present study, screening for phages specifically binding to breast cancer stem cells was performed, positive phage DNAs were extracted, and polypeptides were synthesized and labeled with FITC. The specificity of the polypeptides was identified in vitro and in vivo. Breast cancer stem cells were cultured and identified by flow cytometry. A phage random-peptide library was amplified and screened by culturing with breast cancer cells and breast cancer stem cells. The positive phage was identified by ELISA, and positive phage DNA was extracted. The DNA pellet was isolated and sent for external sequencing with the primer −96 gIII. Based on the sequencing results, a polypeptide was synthesized and labeled with FITC. The specificity to breast cancer stem cells was identified in vivo and vitro. Following three rounds of screening, the phage was enriched ~200-fold. Immunofluorescence demonstrated that two randomly selected phage clones, B8 and A3, had specific affinity to breast cancer stem cells. The results of the present study indicated that phage polypeptides that specifically bind to breast cancer stem cells were successfully screened through stem cell enrichment and phage display technology, which may be beneficial for targeted therapy and further study of breast cancer stem cells.
Collapse
Affiliation(s)
- Tingting Liu
- Breast Cancer Center, Shandong Cancer Hospital and Institute Affiliated to Shandong University, Jinan, Shandong 250117, P.R. China.,Department of Breast Surgery, Tai'an Central Hospital, Tai'an, Shandong 271000, P.R. China
| | - Baojiang Li
- Department of Breast Surgery, Tai'an Central Hospital, Tai'an, Shandong 271000, P.R. China
| | - Yunyun Jiang
- Department of Rehabilitation Medicine, Tai'an Central Hospital, Tai'an, Shandong 271000, P.R. China
| | - Chunhui Zheng
- Department of Oncology Surgery, Weifang People's Hospital, Weifang, Shandong 261000, P.R. China
| | - Li Zhang
- Department of Breast Oncology, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital/Ministry of Education, Tianjin 300060, P.R. China
| | - Yongsheng Wang
- Breast Cancer Center, Shandong Cancer Hospital and Institute Affiliated to Shandong University, Jinan, Shandong 250117, P.R. China
| |
Collapse
|
9
|
Arab A, Behravan N, Razazn A, Barati N, Mosaffa F, Nicastro J, Slavcev R, Behravan J. The viral approach to breast cancer immunotherapy. J Cell Physiol 2018; 234:1257-1267. [PMID: 30146692 DOI: 10.1002/jcp.27150] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 07/05/2018] [Indexed: 01/03/2023]
Abstract
Despite years of intensive research, breast cancer remains the leading cause of death in women worldwide. New technologies including oncolytic virus therapies, virus, and phage display are among the most powerful and advanced methods that have emerged in recent years with potential applications in cancer prevention and treatment. Oncolytic virus therapy is an interesting strategy for cancer treatment. Presently, a number of viruses from different virus families are under laboratory and clinical investigation as oncolytic therapeutics. Oncolytic viruses (OVs) have been shown to be able to induce and initiate a systemic antitumor immune response. The possibility of application of a multimodal therapy using a combination of the OV therapy with immune checkpoint inhibitors and cancer antigen vaccination holds a great promise in the future of cancer immunotherapy. Display of immunologic peptides on bacterial viruses (bacteriophages) is also increasingly being considered as a new and strong cancer vaccine delivery strategy. In phage display immunotherapy, a peptide or protein antigen is presented by genetic fusions to the phage coat proteins, and the phage construct formulation acts as a protective or preventive vaccine against cancer. In our laboratory, we have recently tested a few peptides (E75, AE37, and GP2) derived from HER2/neu proto-oncogene as vaccine delivery modalities for the treatment of TUBO breast cancer xenograft tumors of BALB/c mice. Here, in this paper, we discuss the latest advancements in the applications of OVs and bacterial viruses display systems as new and advanced modalities in cancer immune therapeutics.
Collapse
Affiliation(s)
- Atefeh Arab
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Atefeh Razazn
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Nastaran Barati
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Mosaffa
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Jessica Nicastro
- School of Pharmacy, University of Waterloo, Waterloo, ON, Canada.,Waterloo Institute of Nanotechnology, University of Waterloo, Waterloo, ON, Canada
| | - Roderick Slavcev
- School of Pharmacy, University of Waterloo, Waterloo, ON, Canada.,Waterloo Institute of Nanotechnology, University of Waterloo, Waterloo, ON, Canada.,Mediphage Bioceuticals, Inc., MaRS Centre, Toronto, ON, Canada
| | - Javad Behravan
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Mediphage Bioceuticals, Inc., MaRS Centre, Toronto, ON, Canada
| |
Collapse
|
10
|
Arab A, Nicastro J, Slavcev R, Razazan A, Barati N, Nikpoor AR, Brojeni AAM, Mosaffa F, Badiee A, Jaafari MR, Behravan J. Lambda phage nanoparticles displaying HER2-derived E75 peptide induce effective E75-CD8+ T response. Immunol Res 2017; 66:200-206. [DOI: 10.1007/s12026-017-8969-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
11
|
Deng X, Wang L, You X, Dai P, Zeng Y. Advances in the T7 phage display system (Review). Mol Med Rep 2017; 17:714-720. [PMID: 29115536 DOI: 10.3892/mmr.2017.7994] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Accepted: 08/10/2017] [Indexed: 11/06/2022] Open
Abstract
The present review describes the advantages and updated applications of the T7 phage display system in bioscience and medical science. Current phage display systems are based on various bacteriophage vectors, including M13, T7, T4 and f1. Of these, the M13 phage display is the most frequently used, however, the present review highlights the advantages of the T7 system. As a phage display platform, M13 contains single‑stranded DNA, while the T7 phage consists of double‑stranded DNA, which exhibits increased stability and is less prone to mutation during replication. Additional characteristics of the T7 phage include the following: The T7 phage does not depend on a protein secretion pathway in the lytic cycle; expressed peptides and proteins are usually located on the C‑terminal region of capsid protein gp10B, which avoids problems associated with steric hindrance; and T7 phage particles exhibit high stability under various extreme conditions, including high temperature and low pH, which facilitates effective high‑throughput affinity elutriation. Recent applications of the T7 phage display system have been instrumental in uncovering mechanisms of molecular interaction, particularly in the fields of antigen discovery, vaccine development, protein interaction, and cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Xiangying Deng
- Institute of Pathogenic Biology, Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, Hunan 421001, P.R. China
| | - Li Wang
- Institute of Pathogenic Biology, Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, Hunan 421001, P.R. China
| | - Xiaolong You
- Institute of Pathogenic Biology, Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, Hunan 421001, P.R. China
| | - Pei Dai
- Institute of Pathogenic Biology, Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, Hunan 421001, P.R. China
| | - Yanhua Zeng
- Institute of Pathogenic Biology, Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, Hunan 421001, P.R. China
| |
Collapse
|
12
|
Lee KL, Twyman RM, Fiering S, Steinmetz N. Virus-based nanoparticles as platform technologies for modern vaccines. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2016; 8:554-78. [PMID: 26782096 PMCID: PMC5638654 DOI: 10.1002/wnan.1383] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 11/11/2015] [Indexed: 12/25/2022]
Abstract
Nanoscale engineering is revolutionizing the development of vaccines and immunotherapies. Viruses have played a key role in this field because they can function as prefabricated nanoscaffolds with unique properties that are easy to modify. Viruses are immunogenic via multiple pathways, and antigens displayed naturally or by engineering on the surface can be used to create vaccines against the cognate virus, other pathogens, specific molecules or cellular targets such as tumors. This review focuses on the development of virus-based nanoparticle systems as vaccines indicated for the prevention or treatment of infectious diseases, chronic diseases, cancer, and addiction. WIREs Nanomed Nanobiotechnol 2016, 8:554-578. doi: 10.1002/wnan.1383 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Karin L. Lee
- Department of Biomedical Engineering, Case Western Reserve University Schools of Engineering and Medicine, Cleveland, OH 44106
| | | | - Steven Fiering
- Department of Microbiology and Immunology and Norris Cotton Cancer Center, The Geisel School of Medicine at Dartmouth, Lebanon, NH 03756
| | - Nicole Steinmetz
- Departments of Biomedical Engineering, Radiology, Materials Science and Engineering, and Macromolecular Science and Engineering, Case Western Reserve University and Medicine, Cleveland, OH 44106;
| |
Collapse
|
13
|
Costa LE, Goulart LR, Pereira NCDJ, Lima MIS, Duarte MC, Martins VT, Lage PS, Menezes-Souza D, Ribeiro TG, Melo MN, Fernandes AP, Soto M, Tavares CAP, Chávez-Fumagalli MA, Coelho EAF. Mimotope-based vaccines of Leishmania infantum antigens and their protective efficacy against visceral leishmaniasis. PLoS One 2014; 9:e110014. [PMID: 25333662 PMCID: PMC4198211 DOI: 10.1371/journal.pone.0110014] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 09/05/2014] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND The development of cost-effective prophylactic strategies to prevent leishmaniasis has become a high-priority. The present study has used the phage display technology to identify new immunogens, which were evaluated as vaccines in the murine model of visceral leishmaniasis (VL). Epitope-based immunogens, represented by phage-fused peptides that mimic Leishmania infantum antigens, were selected according to their affinity to antibodies from asymptomatic and symptomatic VL dogs' sera. METHODOLOGY/MAIN FINDINGS Twenty phage clones were selected after three selection cycles, and were evaluated by means of in vitro assays of the immune stimulation of spleen cells derived from naive and chronically infected with L. infantum BALB/c mice. Clones that were able to induce specific Th1 immune response, represented by high levels of IFN-γ and low levels of IL-4 were selected, and based on their selectivity and specificity, two clones, namely B10 and C01, were further employed in the vaccination protocols. BALB/c mice vaccinated with clones plus saponin showed both a high and specific production of IFN-γ, IL-12, and GM-CSF after in vitro stimulation with individual clones or L. infantum extracts. Additionally, these animals, when compared to control groups (saline, saponin, wild-type phage plus saponin, or non-relevant phage clone plus saponin), showed significant reductions in the parasite burden in the liver, spleen, bone marrow, and paws' draining lymph nodes. Protection was associated with an IL-12-dependent production of IFN-γ, mainly by CD8+ T cells, against parasite proteins. These animals also presented decreased parasite-mediated IL-4 and IL-10 responses, and increased levels of parasite-specific IgG2a antibodies. CONCLUSIONS/SIGNIFICANCE This study describes two phage clones that mimic L. infantum antigens, which were directly used as immunogens in vaccines and presented Th1-type immune responses, and that significantly reduced the parasite burden. This is the first study that describes phage-displayed peptides as successful immunogens in vaccine formulations against VL.
Collapse
Affiliation(s)
- Lourena Emanuele Costa
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Luiz Ricardo Goulart
- Instituto de Genética e Bioquímica, Universidade Federal de Uberlândia, Uberlândia, Minas Gerais, Brazil
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, United States of America
| | - Nathália Cristina de Jesus Pereira
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Mayara Ingrid Sousa Lima
- Instituto de Genética e Bioquímica, Universidade Federal de Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Mariana Costa Duarte
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Vivian Tamietti Martins
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Paula Sousa Lage
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Daniel Menezes-Souza
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Tatiana Gomes Ribeiro
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Maria Norma Melo
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Ana Paula Fernandes
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Manuel Soto
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Departamento de Biología Molecular, Universidad Autónoma de Madrid, Madrid, Spain
| | - Carlos Alberto Pereira Tavares
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Miguel Angel Chávez-Fumagalli
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Eduardo Antonio Ferraz Coelho
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Departamento de Patologia Clínica, COLTEC, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- * E-mail:
| |
Collapse
|
14
|
Milani A, Sangiolo D, Aglietta M, Valabrega G. Recent advances in the development of breast cancer vaccines. BREAST CANCER-TARGETS AND THERAPY 2014; 6:159-68. [PMID: 25339848 PMCID: PMC4204811 DOI: 10.2147/bctt.s38428] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The manipulation of the immune system through the administration of a vaccine to direct an effective and long-lasting immune response against breast cancer (BC) cells is an attractive strategy. Vaccines would have several theoretical advantages over standard therapies, including low toxicities, high specificity, and long-lasting efficacy due to the establishment of immunological memory. However, BC vaccines have failed to demonstrate meaningful results in clinical trials so far. This reflects the intrinsic difficulty in breaking the complex immune-escaping mechanisms developed by cancer cells. New vaccines should be able to elicit complex immunologic response involving multiple immune effectors such as cytotoxic and antibody-secreting B cells, innate immunity effectors, and memory cells. Moreover, especially in patients with large tumor burdens and metastatic disease, combining vaccines with other strategies, such as systemic BC therapies, passive immunotherapy, or immunomodulatory agents, could increase the effectiveness of each approach. Here, we review recent advances in BC vaccines, focusing on suitable targets and innovative strategies. We report results of most recent trials investigating active immunotherapy in BC and provide possible future perspectives in this field of research.
Collapse
Affiliation(s)
- Andrea Milani
- Department of Oncology, University of Torino, Torino, Italy
| | - Dario Sangiolo
- Department of Oncology, University of Torino, Torino, Italy
| | - Massimo Aglietta
- Department of Oncology, University of Torino, Torino, Italy ; FPO, Candiolo Cancer Institute, IRCCS, Torino, Italy
| | - Giorgio Valabrega
- Department of Oncology, University of Torino, Torino, Italy ; FPO, Candiolo Cancer Institute, IRCCS, Torino, Italy
| |
Collapse
|
15
|
Shukla S, Wen AM, Commandeur U, Steinmetz NF. Presentation of HER2 epitopes using a filamentous plant virus-based vaccination platform. J Mater Chem B 2014; 2:6249-6258. [DOI: 10.1039/c4tb00749b] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
16
|
Guo L, Yan DD, Yang D, Li Y, Wang X, Zalewski O, Yan B, Lu W. Combinatorial photothermal and immuno cancer therapy using chitosan-coated hollow copper sulfide nanoparticles. ACS NANO 2014; 8:5670-81. [PMID: 24801008 PMCID: PMC4072412 DOI: 10.1021/nn5002112] [Citation(s) in RCA: 370] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 05/06/2014] [Indexed: 05/19/2023]
Abstract
Near-infrared light-responsive inorganic nanoparticles have been shown to enhance the efficacy of cancer photothermal ablation therapy. However, current nanoparticle-mediated photothermal ablation is more effective in treating local cancer at the primary site than metastatic cancer. Here, we report the design of a near-infrared light-induced transformative nanoparticle platform that combines photothermal ablation with immunotherapy. The design is based on chitosan-coated hollow CuS nanoparticles that assemble the immunoadjuvants oligodeoxynucleotides containing the cytosine-guanine (CpG) motifs. Interestingly, these structures break down after laser excitation, reassemble, and transform into polymer complexes that improve tumor retention of the immunotherapy. In this "photothermal immunotherapy" approach, photothermal ablation-induced tumor cell death reduces tumor growth and releases tumor antigens into the surrounding milieu, while the immunoadjuvants potentiate host antitumor immunity. Our results indicated that combined photothermal immunotherapy is more effective than either immunotherapy or photothermal therapy alone against primary treated and distant untreated tumors in a mouse breast cancer model. These hollow CuS nanoparticles are biodegradable and can be eliminated from the body after laser excitation.
Collapse
Affiliation(s)
- Liangran Guo
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, The University of Rhode Island, Kingston, Rhode Island 02881, United States
| | - Daisy D. Yan
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, The University of Rhode Island, Kingston, Rhode Island 02881, United States
| | - Dongfang Yang
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, The University of Rhode Island, Kingston, Rhode Island 02881, United States
| | - Yajuan Li
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, The University of Rhode Island, Kingston, Rhode Island 02881, United States
| | - Xiaodong Wang
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, The University of Rhode Island, Kingston, Rhode Island 02881, United States
| | - Olivia Zalewski
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, The University of Rhode Island, Kingston, Rhode Island 02881, United States
| | - Bingfang Yan
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, The University of Rhode Island, Kingston, Rhode Island 02881, United States
| | - Wei Lu
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, The University of Rhode Island, Kingston, Rhode Island 02881, United States
- School of Pharmacy, Fudan University, Shanghai 201203, China
- Address correspondence to
| |
Collapse
|