1
|
Gay L, Suwan K, Hajitou A. Construction and utilization of a new generation of bacteriophage-based particles, or TPA, for guided systemic delivery of nucleic acids to tumors. Nat Protoc 2024:10.1038/s41596-024-01040-9. [PMID: 39237829 DOI: 10.1038/s41596-024-01040-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 06/12/2024] [Indexed: 09/07/2024]
Abstract
Successful delivery of nucleic acid therapeutics to diseased sites would present a pivotal advancement in cancer treatment. However, progress has been hindered by the lack of efficient tumor-selective vectors via clinical systemic routes, the blood-brain barrier for brain tumors and problems with repeated administrations. We present a new generation of M13 phage-based vectors termed transmorphic phage/adeno-associated virus (AAV) (TPA), wherein the phage genome has been excised to facilitate exclusive packaging of human AAV DNA by phage coat proteins. Here we provide a detailed protocol for the molecular cloning of DNA into the TPA construct, display of disease-specific ligands on the helper phage capsid for cell targeting and entry, and packaging of TPA DNA by helper phage coat proteins in a bacterial host. Furthermore, we provide methods for mammalian cell transduction and assessment of transgene expression in vitro as well as in vivo application of TPA particles in tumor-bearing mice. Unlike other similar methods, our protocol enables high-yield production and control of helper phage quantity in TPA preparations. Moreover, compared with existing M13 phage vectors, TPA particles can accommodate large size transgene inserts, despite being considerably more compact, providing superior gene delivery through enhanced diffusion across the extracellular matrix, improved cellular binding and entry and increased vector DNA accumulation in the nucleus. The protocol encompasses a timeline of 4-5 months, including construction and production of TPA particles with transgene and targeted ligand and in vitro/in vivo testing. This protocol can be conducted by researchers trained in basic molecular biology/bacteriology research techniques.
Collapse
Affiliation(s)
- Lauren Gay
- Cancer Phage Therapy, Department of Brain Sciences, Imperial College London, London, UK
| | - Keittisak Suwan
- Cancer Phage Therapy, Department of Brain Sciences, Imperial College London, London, UK
| | - Amin Hajitou
- Cancer Phage Therapy, Department of Brain Sciences, Imperial College London, London, UK.
| |
Collapse
|
2
|
Chongchai A, Bentayebi K, Chu G, Yan W, Waramit S, Phitak T, Kongtawelert P, Pothacharoen P, Suwan K, Hajitou A. Targeted treatment of chondrosarcoma with a bacteriophage-based particle delivering a secreted tumor necrosis factor-related apoptosis-inducing ligand. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200805. [PMID: 38745750 PMCID: PMC11090904 DOI: 10.1016/j.omton.2024.200805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 02/20/2024] [Accepted: 04/18/2024] [Indexed: 05/16/2024]
Abstract
Chondrosarcoma (CS) is a malignant cartilage-forming bone tumor that is inherently resistant to chemotherapy and radiotherapy, leaving surgery as the only treatment option. We have designed a tumor-targeted bacteriophage (phage)-derived particle (PDP), for targeted systemic delivery of cytokine-encoding transgenes to solid tumors. Phage has no intrinsic tropism for mammalian cells; therefore, it was engineered to display a double cyclic RGD4C ligand on the capsid to target tumors. To induce cancer cell death, we constructed a transgene cassette expressing a secreted form of the cytokine tumor necrosis factor-related apoptosis-inducing ligand (sTRAIL). We detected high expression of αvβ3 and αvβ5 integrin receptors of the RGD4C ligand, and of the TRAIL receptor-2 in human CS cells (SW1353), but not in primary normal chondrocytes. The RGD4C.PDP-Luc particle carrying a luciferase reporter gene, Luc, effectively and selectively mediated gene delivery to SW1353 cells, but not primary chondrocytes. Transduction of SW1353 cells with RGD4C.PDP-sTRAIL encoding a human sTRAIL, resulted in the expression of TRAIL and subsequent cell death without harming the normal chondrocytes. Intravenous administration of RGD4C.PDP-sTRAIL to mice with established human CS resulted in a decrease in tumor size and tumor viability. Altogether, RGD4C.PDP-sTRAIL can be used to target systemic treatment of CS with the sTRAIL.
Collapse
Affiliation(s)
- Aitthiphon Chongchai
- Cancer Phage Therapy Laboratory, Department of Brain Sciences, Imperial College London, W12 0NN London, UK
- Thailand Excellence Centre for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Kaoutar Bentayebi
- Cancer Phage Therapy Laboratory, Department of Brain Sciences, Imperial College London, W12 0NN London, UK
- Medical Biotechnology Laboratory, Rabat Medical and Pharmacy School, Mohammed V University, Rabat 10100, Morocco
| | - Grace Chu
- Cancer Phage Therapy Laboratory, Department of Brain Sciences, Imperial College London, W12 0NN London, UK
| | - Wenqing Yan
- Cancer Phage Therapy Laboratory, Department of Brain Sciences, Imperial College London, W12 0NN London, UK
| | - Sajee Waramit
- Cancer Phage Therapy Laboratory, Department of Brain Sciences, Imperial College London, W12 0NN London, UK
| | - Thanyaluck Phitak
- Thailand Excellence Centre for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Prachya Kongtawelert
- Thailand Excellence Centre for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Peraphan Pothacharoen
- Thailand Excellence Centre for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Keittisak Suwan
- Cancer Phage Therapy Laboratory, Department of Brain Sciences, Imperial College London, W12 0NN London, UK
| | - Amin Hajitou
- Cancer Phage Therapy Laboratory, Department of Brain Sciences, Imperial College London, W12 0NN London, UK
| |
Collapse
|
3
|
Silver KI, Patkar S, Mazcko C, Berger EP, Beck JA, LeBlanc AK. Patterns of metastatic progression and association with clinical outcomes in canine osteosarcoma: A necropsy study of 83 dogs. Vet Comp Oncol 2023; 21:646-655. [PMID: 37592810 PMCID: PMC10842475 DOI: 10.1111/vco.12927] [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: 03/13/2023] [Revised: 07/17/2023] [Accepted: 07/18/2023] [Indexed: 08/19/2023]
Abstract
Osteosarcoma is a highly metastatic primary bone tumour that occurs spontaneously in both pet dogs and humans. Patterns of metastasis to organs beyond the most common site (lung) are poorly characterised and it is unknown whether specific associations between patterns of metastatic progression and patient features exist. This retrospective study characterised the necropsy findings of 83 dogs receiving standardised therapy and clinical monitoring in a prospective clinical trial setting to document patterns of metastasis and correlate outcomes with these patterns and other patient and tumour-specific factors. A total of 20 different sites of metastasis were documented, with lung as the most common site, followed by bone, kidney, liver, and heart. Two distinct clusters of dogs were identified based on patterns of metastasis. There was no significant association between site of enrollment, trial arm, sex, serum alkaline phosphatase (ALP) activity, or tumour location and clinical outcomes. A second cancer type was identified at necropsy in 10 dogs (10/83; 12%). These data showcase the extensive nature of osteosarcoma metastasis beyond the lung and provide a benchmark for clinical monitoring of the disease. Further, this study provides insight into transcriptional features of primary tumours that may relate to a propensity for osteosarcoma metastasis to specific organs and tissues.
Collapse
Affiliation(s)
- K I Silver
- Comparative Oncology Program, Molecular Imaging Branch, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - S Patkar
- Artificial Intelligence Resource, Molecular Imaging Branch, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - C Mazcko
- Comparative Oncology Program, Molecular Imaging Branch, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - E P Berger
- Frederick National Laboratory for Cancer Research in the Comparative Oncology Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - J A Beck
- Comparative Oncology Program, Molecular Imaging Branch, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - A K LeBlanc
- Comparative Oncology Program, Molecular Imaging Branch, National Cancer Institute, NIH, Bethesda, Maryland, USA
| |
Collapse
|
4
|
Al‐Bahrani M, Asavarut P, Waramit S, Suwan K, Hajitou A. Transmorphic phage-guided systemic delivery of TNFα gene for the treatment of human pediatric medulloblastoma. FASEB J 2023; 37:e23038. [PMID: 37331004 PMCID: PMC10947044 DOI: 10.1096/fj.202300045r] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 05/12/2023] [Accepted: 06/05/2023] [Indexed: 06/20/2023]
Abstract
Medulloblastoma is the most common childhood brain tumor with an unfavorable prognosis and limited options of harmful treatments that are associated with devastating long-term side effects. Therefore, the development of safe, noninvasive, and effective therapeutic approaches is required to save the quality of life of young medulloblastoma survivors. We postulated that therapeutic targeting is a solution. Thus, we used a recently designed tumor-targeted bacteriophage (phage)-derived particle, named transmorphic phage/AAV, TPA, to deliver a transgene expressing the tumor necrosis factor-alpha (TNFα) for targeted systemic therapy of medulloblastoma. This vector was engineered to display the double-cyclic RGD4C ligand to selectively target tumors after intravenous administration. Furthermore, the lack of native phage tropism in mammalian cells warrants safe and selective systemic delivery to the tumor microenvironment. In vitro RGD4C.TPA.TNFα treatment of human medulloblastoma cells generated efficient and selective TNFα expression, subsequently triggering cell death. Combination with the chemotherapeutic drug cisplatin used clinically against medulloblastoma resulted in augmented effect through the enhancement of TNFα gene expression. Systemic administration of RGD4C.TPA.TNFα to mice-bearing subcutaneous medulloblastoma xenografts resulted in selective tumor homing of these particles and consequently, targeted tumor expression of TNFα, apoptosis, and destruction of the tumor vasculature. Thus, our RGD4C.TPA.TNFα particle provides selective and efficient systemic delivery of TNFα to medulloblastoma, yielding a potential TNFα anti-medulloblastoma therapy while sparing healthy tissues from the systemic toxicity of this cytokine.
Collapse
Affiliation(s)
- Mariam Al‐Bahrani
- Phage Therapy Group, Department of Brain SciencesImperial College LondonLondonUK
- Present address:
Department of Medical Laboratory Sciences, Faculty of Allied Health SciencesKuwait UniversityKuwait CityKuwait
| | - Paladd Asavarut
- Phage Therapy Group, Department of Brain SciencesImperial College LondonLondonUK
| | - Sajee Waramit
- Phage Therapy Group, Department of Brain SciencesImperial College LondonLondonUK
| | - Keittisak Suwan
- Phage Therapy Group, Department of Brain SciencesImperial College LondonLondonUK
| | - Amin Hajitou
- Phage Therapy Group, Department of Brain SciencesImperial College LondonLondonUK
| |
Collapse
|
5
|
Bacteriophage-Mediated Cancer Gene Therapy. Int J Mol Sci 2022; 23:ijms232214245. [PMID: 36430720 PMCID: PMC9697857 DOI: 10.3390/ijms232214245] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/12/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022] Open
Abstract
Bacteriophages have long been considered only as infectious agents that affect bacterial hosts. However, recent studies provide compelling evidence that these viruses are able to successfully interact with eukaryotic cells at the levels of the binding, entry and expression of their own genes. Currently, bacteriophages are widely used in various areas of biotechnology and medicine, but the most intriguing of them is cancer therapy. There are increasing studies confirming the efficacy and safety of using phage-based vectors as a systemic delivery vehicle of therapeutic genes and drugs in cancer therapy. Engineered bacteriophages, as well as eukaryotic viruses, demonstrate a much greater efficiency of transgene delivery and expression in cancer cells compared to non-viral gene transfer methods. At the same time, phage-based vectors, in contrast to eukaryotic viruses-based vectors, have no natural tropism to mammalian cells and, as a result, provide more selective delivery of therapeutic cargos to target cells. Moreover, numerous data indicate the presence of more complex molecular mechanisms of interaction between bacteriophages and eukaryotic cells, the further study of which is necessary both for the development of gene therapy methods and for understanding the cancer nature. In this review, we summarize the key results of research into aspects of phage-eukaryotic cell interaction and, in particular, the use of phage-based vectors for highly selective and effective systemic cancer gene therapy.
Collapse
|
6
|
Tsedev U, Lin CW, Hess GT, Sarkaria JN, Lam FC, Belcher AM. Phage Particles of Controlled Length and Genome for In Vivo Targeted Glioblastoma Imaging and Therapeutic Delivery. ACS NANO 2022; 16:11676-11691. [PMID: 35830573 DOI: 10.1021/acsnano.1c08720] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
M13 bacteriophage (phage) are versatile, genetically tunable nanocarriers that have been recently adapted for use as diagnostic and therapeutic platforms. Applying p3 capsid chlorotoxin fusion with the "inho" circular single-stranded DNA (cssDNA) gene packaging system, we produced miniature chlorotoxin inho (CTX-inho) phage particles with a minimum length of 50 nm that can target intracranial orthotopic patient-derived GBM22 glioblastoma tumors in the brains of mice. Systemically administered indocyanine green conjugated CTX-inho phage accumulated in brain tumors, facilitating shortwave infrared detection. Furthermore, we show that our inho phage can carry cssDNA that are transcriptionally active when delivered to GBM22 glioma cells in vitro. The ability to modulate the capsid display, surface loading, phage length, and cssDNA gene content makes the recombinant M13 phage particle an ideal delivery platform.
Collapse
Affiliation(s)
- Uyanga Tsedev
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Ching-Wei Lin
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei 10617, Taiwan
| | - Gaelen T Hess
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53705, Unites States
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota 55902, United States
| | - Fred C Lam
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Division of Neurosurgery, Saint Elizabeth's Medical Center, Brighton, Massachusetts 02135, United States
| | - Angela M Belcher
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
7
|
Asavarut P, Waramit S, Suwan K, Marais GJK, Chongchai A, Benjathummarak S, Al‐Bahrani M, Vila‐Gomez P, Williams M, Kongtawelert P, Yata T, Hajitou A. Systemically targeted cancer immunotherapy and gene delivery using transmorphic particles. EMBO Mol Med 2022; 14:e15418. [PMID: 35758207 PMCID: PMC9358398 DOI: 10.15252/emmm.202115418] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 05/23/2022] [Accepted: 05/24/2022] [Indexed: 01/21/2023] Open
Abstract
Immunotherapy is a powerful tool for cancer treatment, but the pleiotropic nature of cytokines and immunological agents strongly limits clinical translation and safety. To address this unmet need, we designed and characterised a systemically targeted cytokine gene delivery system through transmorphic encapsidation of human recombinant adeno-associated virus DNA using coat proteins from a tumour-targeted bacteriophage (phage). We show that Transmorphic Phage/AAV (TPA) particles provide superior delivery of transgenes over current phage-derived vectors through greater diffusion across the extracellular space and improved intracellular trafficking. We used TPA to target the delivery of cytokine-encoding transgenes for interleukin-12 (IL12), and novel isoforms of IL15 and tumour necrosis factor alpha (TNF α ) for tumour immunotherapy. Our results demonstrate selective and efficient gene delivery and immunotherapy against solid tumours in vivo, without harming healthy organs. Our transmorphic particle system provides a promising modality for safe and effective gene delivery, and cancer immunotherapies through cross-species complementation of two commonly used viruses.
Collapse
Affiliation(s)
- Paladd Asavarut
- Cancer Phagotherapy, Department of Brain SciencesImperial College LondonLondonUK
| | - Sajee Waramit
- Cancer Phagotherapy, Department of Brain SciencesImperial College LondonLondonUK
| | - Keittisak Suwan
- Cancer Phagotherapy, Department of Brain SciencesImperial College LondonLondonUK
| | - Gert J K Marais
- Cancer Phagotherapy, Department of Brain SciencesImperial College LondonLondonUK
| | - Aitthiphon Chongchai
- Cancer Phagotherapy, Department of Brain SciencesImperial College LondonLondonUK
- Thailand Excellence Centre for Tissue Engineering and Stem Cells, Faculty of MedicineChiang Mai UniversityChiang MaiThailand
| | - Surachet Benjathummarak
- Cancer Phagotherapy, Department of Brain SciencesImperial College LondonLondonUK
- Center of Excellence for Antibody Research, Faculty of Tropical MedicineMahidol UniversityBangkokThailand
| | - Mariam Al‐Bahrani
- Cancer Phagotherapy, Department of Brain SciencesImperial College LondonLondonUK
| | - Paula Vila‐Gomez
- Cancer Phagotherapy, Department of Brain SciencesImperial College LondonLondonUK
| | | | - Prachya Kongtawelert
- Thailand Excellence Centre for Tissue Engineering and Stem Cells, Faculty of MedicineChiang Mai UniversityChiang MaiThailand
| | - Teerapong Yata
- Cancer Phagotherapy, Department of Brain SciencesImperial College LondonLondonUK
- Present address:
Department of PhysiologyChulalongkorn UniversityBangkokThailand
| | - Amin Hajitou
- Cancer Phagotherapy, Department of Brain SciencesImperial College LondonLondonUK
| |
Collapse
|
8
|
Hendricks-Wenger A, Arnold L, Gannon J, Simon A, Singh N, Sheppard H, Nagai-Singer MA, Imran KM, Lee K, Clark-Deener S, Byron C, Edwards MR, Larson MM, Rossmeisl JH, Coutermarsh-Ott SL, Eden K, Dervisis N, Klahn S, Tuohy J, Allen IC, Vlaisavljevich E. Histotripsy Ablation in Preclinical Animal Models of Cancer and Spontaneous Tumors in Veterinary Patients: A Review. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2022; 69:5-26. [PMID: 34478363 PMCID: PMC9284566 DOI: 10.1109/tuffc.2021.3110083] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
New therapeutic strategies are direly needed in the fight against cancer. Over the last decade, several tumor ablation strategies have emerged as stand-alone or combination therapies. Histotripsy is the first completely noninvasive, nonthermal, and nonionizing tumor ablation method. Histotripsy can produce consistent and rapid ablations, even near critical structures. Additional benefits include real-time image guidance, high precision, and the ability to treat tumors of any predetermined size and shape. Unfortunately, the lack of clinically and physiologically relevant preclinical cancer models is often a significant limitation with all focal tumor ablation strategies. The majority of studies testing histotripsy for cancer treatment have focused on small animal models, which have been critical in moving this field forward and will continue to be essential for providing mechanistic insight. While these small animal models have notable translational value, there are significant limitations in terms of scale and anatomical relevance. To address these limitations, a diverse range of large animal models and spontaneous tumor studies in veterinary patients have emerged to complement existing rodent models. These models and veterinary patients are excellent at providing realistic avenues for developing and testing histotripsy devices and techniques designed for future use in human patients. Here, we provide a review of animal models used in preclinical histotripsy studies and compare histotripsy ablation in these models using a series of original case reports across a broad spectrum of preclinical animal models and spontaneous tumors in veterinary patients.
Collapse
|
9
|
Kabwe M, Dashper S, Bachrach G, Tucci J. Bacteriophage manipulation of the microbiome associated with tumour microenvironments-can this improve cancer therapeutic response? FEMS Microbiol Rev 2021; 45:6188389. [PMID: 33765142 DOI: 10.1093/femsre/fuab017] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 03/21/2021] [Indexed: 12/11/2022] Open
Abstract
Some cancer treatment failures have been attributed to the tumour microbiota, with implications that microbiota manipulation may improve treatment efficacy. While antibiotics have been used to control bacterial growth, their dysbiotic effects on the microbiome, failure to penetrate biofilms and decreased efficacy due to increasing antimicrobial resistance by bacteria, suggest alternatives are needed. Bacteriophages may provide a precise means for targeting oncobacteria whose relative abundance is increased in tumour tissue microbiomes. Fusobacterium, Streptococcus, Peptostreptococcus, Prevotella, Parvimonas, and Treponema species are prevalent in tumour tissue microbiomes of some cancers. They may promote cancer growth by dampening immunity, stimulating release of proinflammatory cytokines, and directly interacting with cancer cells to stimulate proliferation. Lytic bacteriophages against some of these oncobacteria have been isolated and characterised. The search continues for others. The possibility exists for their testing as adjuncts to complement existing therapies. In this review, we highlight the role of oncobacteria, specifically those whose relative abundance in the intra-tumour microbiome is increased, and discuss the potential for bacteriophages against these micro-organisms to augment existing cancer therapies. The capacity for bacteriophages to modulate immunity and kill specific bacteria makes them suitable candidates to manipulate the tumour microbiome and negate the effects of these oncobacteria.
Collapse
Affiliation(s)
- Mwila Kabwe
- Department of Pharmacy and Biomedical Sciences, La Trobe Institute for Molecular Science, La Trobe University, Sharon St. Bendigo, Victoria 3550, Australia
| | - Stuart Dashper
- Melbourne Dental School, University of Melbourne, 720 Swanston St, Parkville, Victoria 3010, Australia
| | - Gilad Bachrach
- The Institute of Dental Sciences, The Hebrew University-Hadassah School of Dental Medicine, PO Box 12272, Jerusalem 9112102, Israel
| | - Joseph Tucci
- Department of Pharmacy and Biomedical Sciences, La Trobe Institute for Molecular Science, La Trobe University, Sharon St. Bendigo, Victoria 3550, Australia
| |
Collapse
|
10
|
Staquicini DI, Tang FHF, Markosian C, Yao VJ, Staquicini FI, Dodero-Rojas E, Contessoto VG, Davis D, O'Brien P, Habib N, Smith TL, Bruiners N, Sidman RL, Gennaro ML, Lattime EC, Libutti SK, Whitford PC, Burley SK, Onuchic JN, Arap W, Pasqualini R. Design and proof of concept for targeted phage-based COVID-19 vaccination strategies with a streamlined cold-free supply chain. Proc Natl Acad Sci U S A 2021; 118:e2105739118. [PMID: 34234013 PMCID: PMC8325333 DOI: 10.1073/pnas.2105739118] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Development of effective vaccines against coronavirus disease 2019 (COVID-19) is a global imperative. Rapid immunization of the entire human population against a widespread, continually evolving, and highly pathogenic virus is an unprecedented challenge, and different vaccine approaches are being pursued. Engineered filamentous bacteriophage (phage) particles have unique potential in vaccine development due to their inherent immunogenicity, genetic plasticity, stability, cost-effectiveness for large-scale production, and proven safety profile in humans. Herein we report the development and initial evaluation of two targeted phage-based vaccination approaches against SARS-CoV-2: dual ligand peptide-targeted phage and adeno-associated virus/phage (AAVP) particles. For peptide-targeted phage, we performed structure-guided antigen design to select six solvent-exposed epitopes of the SARS-CoV-2 spike (S) protein. One of these epitopes displayed on the major capsid protein pVIII of phage induced a specific and sustained humoral response when injected in mice. These phage were further engineered to simultaneously display the peptide CAKSMGDIVC on the minor capsid protein pIII to enable their transport from the lung epithelium into the systemic circulation. Aerosolization of these "dual-display" phage into the lungs of mice generated a systemic and specific antibody response. In the second approach, targeted AAVP particles were engineered to deliver the entire S protein gene under the control of a constitutive CMV promoter. This induced tissue-specific transgene expression, stimulating a systemic S protein-specific antibody response in mice. With these proof-of-concept preclinical experiments, we show that both targeted phage- and AAVP-based particles serve as robust yet versatile platforms that can promptly yield COVID-19 vaccine prototypes for translational development.
Collapse
Affiliation(s)
- Daniela I Staquicini
- Rutgers Cancer Institute of New Jersey, Newark, NJ 07101
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103
| | - Fenny H F Tang
- Rutgers Cancer Institute of New Jersey, Newark, NJ 07101
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103
| | - Christopher Markosian
- Rutgers Cancer Institute of New Jersey, Newark, NJ 07101
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103
| | - Virginia J Yao
- Rutgers Cancer Institute of New Jersey, Newark, NJ 07101
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103
| | - Fernanda I Staquicini
- Rutgers Cancer Institute of New Jersey, Newark, NJ 07101
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103
| | | | - Vinícius G Contessoto
- Center for Theoretical Biological Physics, Rice University, Houston, TX 77005
- Department of Physics, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University, São José do Rio Preto, SP 15054, Brazil
| | - Deodate Davis
- Rutgers Cancer Institute of New Jersey, Newark, NJ 07101
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103
| | - Paul O'Brien
- Rutgers Cancer Institute of New Jersey, Newark, NJ 07101
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103
| | - Nazia Habib
- Rutgers Cancer Institute of New Jersey, Newark, NJ 07101
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103
| | - Tracey L Smith
- Rutgers Cancer Institute of New Jersey, Newark, NJ 07101
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103
| | - Natalie Bruiners
- Public Health Research Institute, Rutgers New Jersey Medical School, Newark, NJ 07103
| | - Richard L Sidman
- Department of Neurology, Harvard Medical School, Boston, MA 02115
| | - Maria L Gennaro
- Public Health Research Institute, Rutgers New Jersey Medical School, Newark, NJ 07103
| | - Edmund C Lattime
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901
| | - Steven K Libutti
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901
| | - Paul C Whitford
- Department of Physics and Center for Theoretical Biological Physics, Northeastern University, Boston, MA 02115
| | - Stephen K Burley
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901
- RCSB Protein Data Bank and Institute for Quantitative Biomedicine, Rutgers, The State University of New Jersey, Piscataway, NJ 08854
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854
- RCSB Protein Data Bank, San Diego Supercomputer Center and Skaggs School of Pharmacy & Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92067
| | - José N Onuchic
- Center for Theoretical Biological Physics, Rice University, Houston, TX 77005;
- Department of Biosciences, Rice University, Houston, TX 77005
- Department of Chemistry, Rice University, Houston, TX 77005
- Department of Physics and Astronomy, Rice University, Houston, TX 77005
| | - Wadih Arap
- Rutgers Cancer Institute of New Jersey, Newark, NJ 07101;
- Division of Hematology/Oncology, Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103
| | - Renata Pasqualini
- Rutgers Cancer Institute of New Jersey, Newark, NJ 07101;
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103
| |
Collapse
|
11
|
Chongchai A, Waramit S, Suwan K, Al-Bahrani M, Udomruk S, Phitak T, Kongtawelert P, Pothacharoen P, Hajitou A. Bacteriophage-mediated therapy of chondrosarcoma by selective delivery of the tumor necrosis factor alpha (TNFα) gene. FASEB J 2021; 35:e21487. [PMID: 33811705 DOI: 10.1096/fj.202002539r] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 02/03/2021] [Accepted: 02/15/2021] [Indexed: 12/31/2022]
Abstract
Chondrosarcoma is a cartilage-forming bone tumor, well known for intrinsic resistance to chemotherapy and radiotherapy. We have designed a targeted chondrosarcoma gene therapy using a bacteriophage (phage) particle to deliver therapeutic genes. Phage has no tropism for mammalian cells, allowing engineered phage to be targeted to specific cell surface receptors in cancer. We modified the phage capsid to display the RGD4C ligand on the pIII minor coat proteins to specifically bind to αvβ3 or αvβ5 integrin receptors. The endosomal escape peptide, H5WYG, was also displayed on recombinant pVIII major coat proteins to enhance gene delivery. Finally, a human tumor necrosis factor alpha (TNFα) therapeutic transgene expression cassette was incorporated into the phage genome. First, we found that human chondrosarcoma cells (SW1353) have high expression of αvβ3, αvβ5 integrin receptors, and both TNFα receptors. Targeted particle encoding a luciferase reporter gene efficiently and selectively mediated gene delivery to these cells. When SW1353 cells were treated with the targeted particle encoding a TNFα transgene, significant cell killing was evident and was associated with high expression of TNFα and apoptosis-related genes. In vivo, mice with established human chondrosarcoma showed suppression of tumors upon repetitive intravenous administrations of the targeted phage. These data show that our phage-based particle is a promising, selective, and efficient tool for targeted chondrosarcoma therapy.
Collapse
Affiliation(s)
- Aitthiphon Chongchai
- Thailand Excellence Centre for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Cancer Phage Therapy Group, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Sajee Waramit
- Cancer Phage Therapy Group, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Keittisak Suwan
- Cancer Phage Therapy Group, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Mariam Al-Bahrani
- Cancer Phage Therapy Group, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Sasimol Udomruk
- Thailand Excellence Centre for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Thanyaluck Phitak
- Thailand Excellence Centre for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Prachya Kongtawelert
- Thailand Excellence Centre for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Peraphan Pothacharoen
- Thailand Excellence Centre for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Amin Hajitou
- Cancer Phage Therapy Group, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
| |
Collapse
|
12
|
Mestrinho LA, Santos RR. Translational oncotargets for immunotherapy: From pet dogs to humans. Adv Drug Deliv Rev 2021; 172:296-313. [PMID: 33705879 DOI: 10.1016/j.addr.2021.02.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 02/10/2021] [Accepted: 02/27/2021] [Indexed: 12/21/2022]
Abstract
Preclinical studies in rodent models have been a pivotal role in human clinical research, but many of them fail in the translational process. Spontaneous tumors in pet dogs have the potential to bridge the gap between preclinical models and human clinical trials. Their natural occurrence in an immunocompetent system overcome the limitations of preclinical rodent models. Due to its reasonable cellular, molecular, and genetic homology to humans, the pet dog represents a valuable model to accelerate the translation of preclinical studies to clinical trials in humans, actually with benefits for both species. Moreover, their unique genetic features of breeding and breed-related mutations have contributed to assess and optimize therapeutics in individuals with different genetic backgrounds. This review aims to outline four main immunotherapy approaches - cancer vaccines, adaptive T-cell transfer, antibodies, and cytokines -, under research in veterinary medicine and how they can serve the clinical application crosstalk with humans.
Collapse
|
13
|
Staquicini DI, Tang FHF, Markosian C, Yao VJ, Staquicini FI, Dodero-Rojas E, Contessoto VG, Davis D, O’Brien P, Habib N, Smith TL, Bruiners N, Sidman RL, Gennaro ML, Lattime EC, Libutti SK, Whitford PC, Burley SK, Onuchic JN, Arap W, Pasqualini R. Design and proof-of-concept for targeted phage-based COVID-19 vaccination strategies with a streamlined cold-free supply chain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.03.15.435496. [PMID: 33758865 PMCID: PMC7987025 DOI: 10.1101/2021.03.15.435496] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Development of effective vaccines against Coronavirus Disease 2019 (COVID-19) is a global imperative. Rapid immunization of the world human population against a widespread, continually evolving, and highly pathogenic virus is an unprecedented challenge, and many different vaccine approaches are being pursued to meet this task. Engineered filamentous bacteriophage (phage) have unique potential in vaccine development due to their inherent immunogenicity, genetic plasticity, stability, cost-effectiveness for large-scale production, and proven safety profile in humans. Herein we report the design, development, and initial evaluation of targeted phage-based vaccination approaches against Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) by using dual ligand peptide-targeted phage and adeno-associated virus/phage (AAVP) particles. Towards a unique phage- and AAVP-based dual-display candidate approach, we first performed structure-guided antigen design to select six solvent-exposed epitopes of the SARS-CoV-2 spike (S) protein for display on the recombinant major capsid coat protein pVIII. Targeted phage particles carrying one of these epitopes induced a strong and specific humoral response. In an initial experimental approach, when these targeted phage particles were further genetically engineered to simultaneously display a ligand peptide (CAKSMGDIVC) on the minor capsid protein pIII, which enables receptor-mediated transport of phage particles from the lung epithelium into the systemic circulation (termed "dual-display"), they enhanced a systemic and specific spike (S) protein-specific antibody response upon aerosolization into the lungs of mice. In a second line of investigation, we engineered targeted AAVP particles to deliver the entire S protein gene under the control of a constitutive cytomegalovirus (CMV) promoter, which induced tissue-specific transgene expression stimulating a systemic S protein-specific antibody response. As proof-of-concept preclinical experiments, we show that targeted phage- and AAVP-based particles serve as robust yet versatile enabling platforms for ligand-directed immunization and promptly yield COVID-19 vaccine prototypes for further translational development. SIGNIFICANCE The ongoing COVID-19 global pandemic has accounted for over 2.5 million deaths and an unprecedented impact on the health of mankind worldwide. Over the past several months, while a few COVID-19 vaccines have received Emergency Use Authorization and are currently being administered to the entire human population, the demand for prompt global immunization has created enormous logistical challenges--including but not limited to supply, access, and distribution--that justify and reinforce the research for additional strategic alternatives. Phage are viruses that only infect bacteria and have been safely administered to humans as antibiotics for decades. As experimental proof-of-concept, we demonstrated that aerosol pulmonary vaccination with lung-targeted phage particles that display short epitopes of the S protein on the capsid as well as preclinical vaccination with targeted AAVP particles carrying the S protein gene elicit a systemic and specific immune response against SARS-CoV-2 in immunocompetent mice. Given that targeted phage- and AAVP-based viral particles are sturdy yet simple to genetically engineer, cost-effective for rapid large-scale production in clinical grade, and relatively stable at room temperature, such unique attributes might perhaps become additional tools towards COVID-19 vaccine design and development for immediate and future unmet needs.
Collapse
Affiliation(s)
- Daniela I. Staquicini
- Rutgers Cancer Institute of New Jersey, Newark, NJ 07101
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103
| | - Fenny H. F. Tang
- Rutgers Cancer Institute of New Jersey, Newark, NJ 07101
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103
| | - Christopher Markosian
- Rutgers Cancer Institute of New Jersey, Newark, NJ 07101
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103
| | - Virginia J. Yao
- Rutgers Cancer Institute of New Jersey, Newark, NJ 07101
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103
| | - Fernanda I. Staquicini
- Rutgers Cancer Institute of New Jersey, Newark, NJ 07101
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103
| | | | - Vinícius G. Contessoto
- Center for Theoretical Biological Physics, Rice University, Houston, TX 77005
- Department of Physics, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University, São José do Rio Preto, SP 15054, Brazil. Institute, Rutgers New Jersey Medical School, Newark, NJ 07103
| | - Deodate Davis
- Rutgers Cancer Institute of New Jersey, Newark, NJ 07101
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103
| | - Paul O’Brien
- Rutgers Cancer Institute of New Jersey, Newark, NJ 07101
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103
| | - Nazia Habib
- Rutgers Cancer Institute of New Jersey, Newark, NJ 07101
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103
| | - Tracey L. Smith
- Rutgers Cancer Institute of New Jersey, Newark, NJ 07101
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103
| | - Natalie Bruiners
- Public Health Research Institute, Rutgers New Jersey Medical School, Newark, NJ 07103
| | | | - Maria L. Gennaro
- Public Health Research Institute, Rutgers New Jersey Medical School, Newark, NJ 07103
| | - Edmund C. Lattime
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901
| | - Steven K. Libutti
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901
| | - Paul C. Whitford
- Department of Physics and Center for Theoretical Biological Physics, Northeastern University, Boston, MA 02115
| | - Stephen K. Burley
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901
- RCSB Protein Data Bank and Institute for Quantitative Biomedicine, Rutgers, The State University of New Jersey, Piscataway, NJ 08854
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854
- RCSB Protein Data Bank, San Diego Supercomputer Center and Skaggs School of Pharmacy & Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92067
| | - José N. Onuchic
- Center for Theoretical Biological Physics, Rice University, Houston, TX 77005
- Department of Biosciences, Rice University, Houston, TX 77005
- Department of Chemistry, Rice University, Houston, TX 77005
- Department of Physics and Astronomy, Rice University, Houston, TX 77005
| | - Wadih Arap
- Rutgers Cancer Institute of New Jersey, Newark, NJ 07101
- Division of Hematology/Oncology, Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103
| | - Renata Pasqualini
- Rutgers Cancer Institute of New Jersey, Newark, NJ 07101
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103
| |
Collapse
|
14
|
Abstract
Comparative oncology clinical trials play an important and growing role in cancer research and drug development efforts. These trials, typically conducted in companion (pet) dogs, allow assessment of novel anticancer agents and combination therapies in a veterinary clinical setting that supports serial biologic sample collections and exploration of dose, schedule and corresponding pharmacokinetic/pharmacodynamic relationships. Further, an intact immune system and natural co-evolution of tumour and microenvironment support exploration of novel immunotherapeutic strategies. Substantial improvements in our collective understanding of the molecular landscape of canine cancers have occurred in the past 10 years, facilitating translational research and supporting the inclusion of comparative studies in drug development. The value of the approach is demonstrated in various clinical trial settings, including single-agent or combination response rates, inhibition of metastatic progression and randomized comparison of multiple agents in a head-to-head fashion. Such comparative oncology studies have been purposefully included in the developmental plan for several US FDA-approved and up-and-coming anticancer drugs. Challenges for this field include keeping pace with technology and data dissemination/harmonization, improving annotation of the canine genome and immune system, and generation of canine-specific validated reagents to support integration of correlative biology within clinical trial efforts.
Collapse
Affiliation(s)
- Amy K LeBlanc
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Christina N Mazcko
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
15
|
Tsafa E, Bentayebi K, Topanurak S, Yata T, Przystal J, Fongmoon D, Hajji N, Waramit S, Suwan K, Hajitou A. Doxorubicin Improves Cancer Cell Targeting by Filamentous Phage Gene Delivery Vectors. Int J Mol Sci 2020; 21:E7867. [PMID: 33114050 PMCID: PMC7660303 DOI: 10.3390/ijms21217867] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/18/2020] [Accepted: 10/19/2020] [Indexed: 12/14/2022] Open
Abstract
Merging targeted systemic gene delivery and systemic chemotherapy against cancer, chemovirotherapy, has the potential to improve chemotherapy and gene therapy treatments and overcome cancer resistance. We introduced a bacteriophage (phage) vector, named human adeno-associated virus (AAV)/phage or AAVP, for the systemic targeting of therapeutic genes to cancer. The vector was designed as a hybrid between a recombinant adeno-associated virus genome (rAAV) and a filamentous phage capsid. To achieve tumor targeting, we displayed on the phage capsid the double-cyclic CDCRGDCFC (RGD4C) ligand that binds the alpha-V/beta-3 (αvβ3) integrin receptor. Here, we investigated a combination of doxorubicin chemotherapeutic drug and targeted gene delivery by the RGD4C/AAVP vector. Firstly, we showed that doxorubicin boosts transgene expression from the RGD4C/AAVP in two-dimensional (2D) cell cultures and three-dimensional (3D) tumor spheres established from human and murine cancer cells, while preserving selective gene delivery by RGD4C/AAVP. Next, we confirmed that doxorubicin does not increase vector attachment to cancer cells nor vector cell entry. In contrast, doxorubicin may alter the intracellular trafficking of the vector by facilitating nuclear accumulation of the RGD4C/AAVP genome through destabilization of the nuclear membrane. Finally, a combination of doxorubicin and RGD4C/AAVP-targeted suicide gene therapy exerts a synergistic effect to destroy human and murine tumor cells in 2D and 3D tumor sphere settings.
Collapse
Affiliation(s)
- Effrosyni Tsafa
- Phage Therapy Group, Department of Brain Sciences, Imperial College London, London W12 0NN, UK; (E.T.); (K.B.); (T.Y.); (J.P.); (S.W.)
| | - Kaoutar Bentayebi
- Phage Therapy Group, Department of Brain Sciences, Imperial College London, London W12 0NN, UK; (E.T.); (K.B.); (T.Y.); (J.P.); (S.W.)
| | - Supachai Topanurak
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand;
| | - Teerapong Yata
- Phage Therapy Group, Department of Brain Sciences, Imperial College London, London W12 0NN, UK; (E.T.); (K.B.); (T.Y.); (J.P.); (S.W.)
| | - Justyna Przystal
- Phage Therapy Group, Department of Brain Sciences, Imperial College London, London W12 0NN, UK; (E.T.); (K.B.); (T.Y.); (J.P.); (S.W.)
| | - Duriya Fongmoon
- Department of Medical Services, Lampang Cancer Hospital, Ministry of Public Health, Lampang 52000, Thailand;
| | - Nabil Hajji
- John Fulcher Neuro-Oncology Laboratory, Department of Brain Sciences, Imperial College London, London W12 0NN, UK;
| | - Sajee Waramit
- Phage Therapy Group, Department of Brain Sciences, Imperial College London, London W12 0NN, UK; (E.T.); (K.B.); (T.Y.); (J.P.); (S.W.)
| | - Keittisak Suwan
- Phage Therapy Group, Department of Brain Sciences, Imperial College London, London W12 0NN, UK; (E.T.); (K.B.); (T.Y.); (J.P.); (S.W.)
| | - Amin Hajitou
- Phage Therapy Group, Department of Brain Sciences, Imperial College London, London W12 0NN, UK; (E.T.); (K.B.); (T.Y.); (J.P.); (S.W.)
| |
Collapse
|
16
|
Yang Zhou J, Suwan K, Hajitou A. Initial Steps for the Development of a Phage-Mediated Gene Replacement Therapy Using CRISPR-Cas9 Technology. J Clin Med 2020; 9:E1498. [PMID: 32429407 PMCID: PMC7290871 DOI: 10.3390/jcm9051498] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 05/04/2020] [Accepted: 05/14/2020] [Indexed: 12/26/2022] Open
Abstract
p53 gene (TP53) replacement therapy has shown promising results in cancer gene therapy. However, it has been hampered, mostly because of the gene delivery vector of choice. CRISPR-Cas9 technology (clustered regularly interspaced short palindromic repeats/CRISPR-associated protein 9) can knock out the mutated TP53 (mutTP53), but due to its large size, many viral vectors are not suitable or require implemented strategies that lower the therapeutic efficiency. Here, we introduced a bacteriophage or phage-based vector with the ability to target cancer cells and aimed to investigate the feasibility of using this vector to deliver CRISPR-Cas9 transgene in human lung adenocarcinoma cells. First, we produced a tumour-targeted bacteriophage carrying a CRISPR-Cas9 transgene cassette. Next, we investigated any negative impact on vector titers via quantitative polymerase chain reaction (qPCR) and colony-forming agar plate. Last, we combined Western blot analysis and immunofluorescence staining to prove cell transduction in vitro. We showed that the tumour-targeted bacteriophage can package a large-size vector genome, ~10 kb, containing the CRISPR-Cas9 sequence without any negative impact on the active or total number of bacteriophage particles. Then, we detected expression of the Cas9 in human lung adenocarcinoma cells in a targeted and efficient manner. Finally, we proved loss of p53 protein expression when a p53 gRNA was incorporated into the CRISPR-Cas9 phage DNA construct. These proof-of-concept findings support the use of engineered bacteriophage for TP53 replacement therapy in lung cancer.
Collapse
Affiliation(s)
| | - Keittisak Suwan
- Phage Therapy Group, Department of Brain Sciences, Imperial College London, London W12 0NN, UK;
| | - Amin Hajitou
- Phage Therapy Group, Department of Brain Sciences, Imperial College London, London W12 0NN, UK;
| |
Collapse
|
17
|
Vail DM, LeBlanc AK, Jeraj R. Advanced Cancer Imaging Applied in the Comparative Setting. Front Oncol 2020; 10:84. [PMID: 32117739 PMCID: PMC7019008 DOI: 10.3389/fonc.2020.00084] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 01/16/2020] [Indexed: 11/13/2022] Open
Abstract
The potential for companion (pet) species with spontaneously arising tumors to act as surrogates for preclinical development of advanced cancer imaging technologies has become more apparent in the last decade. The utility of the companion model specifically centers around issues related to body size (including spatial target/normal anatomic characteristics), physical size and spatial distribution of metastasis, tumor heterogeneity, the presence of an intact syngeneic immune system and a syngeneic tumor microenvironment shaped by the natural evolution of the cancer. Companion species size allows the use of similar equipment, hardware setup, software, and scan protocols which provide the opportunity for standardization and harmonization of imaging operating procedures and quality assurance across imaging protocols, imaging hardware, and the imaged species. Murine models generally do not replicate the size and spatial distribution of human metastatic cancer and these factors strongly influence image resolution and dosimetry. The following review will discuss several aspects of comparative cancer imaging in more detail while providing several illustrative examples of investigational approaches performed or currently under exploration at our institutions. Topics addressed include a discussion on interested consortia; image quality assurance and harmonization; image-based biomarker development and validation; contrast agent and radionuclide tracer development; advanced imaging to assess and predict response to cytotoxic and immunomodulatory anticancer agents; imaging of the tumor microenvironment; development of novel theranostic approaches; cell trafficking assessment via non-invasive imaging; and intraoperative imaging to inform surgical oncology decision making. Taken in totality, these comparative opportunities predict that safety, diagnostic and efficacy data generated in companion species with naturally developing and progressing cancers would better recapitulate the human cancer condition than that of artificial models in small rodent systems and ultimately accelerate the integration of novel imaging technologies into clinical practice. It is our hope that the examples presented should serve to provide those involved in cancer investigations who are unfamiliar with available comparative methodologies an understanding of the potential utility of this approach.
Collapse
Affiliation(s)
- David M Vail
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, United States.,Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Amy K LeBlanc
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| | - Robert Jeraj
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, United States.,Department of Medical Physics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
18
|
Dow S. A Role for Dogs in Advancing Cancer Immunotherapy Research. Front Immunol 2020; 10:2935. [PMID: 32010120 PMCID: PMC6979257 DOI: 10.3389/fimmu.2019.02935] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 11/29/2019] [Indexed: 12/27/2022] Open
Abstract
While rodent cancer models are essential for early proof-of-concept and mechanistic studies for immune therapies, these models have limitations with regards to predicting the ultimate effectiveness of new immunotherapies in humans. As a unique spontaneous, large animal model of cancer, the value of conducting studies in pet dogs with cancer has been increasingly recognized by the research community. This review will therefore summarize key aspects of the dog cancer immunotherapy model and the role that these studies may play in the overall immunotherapy drug research effort. We will focus on cancer types and settings in which the dog model is most likely to impact clinical immuno-oncology research and drug development. Immunological reagent availability is discussed, along with some unique opportunities and challenges associated with the dog immunotherapy model. Overall it is hoped that this review will increase awareness of the dog cancer immunotherapy model and stimulate additional collaborative studies to benefit both man and man's best friend.
Collapse
Affiliation(s)
- Steven Dow
- Flint Animal Cancer Center, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| |
Collapse
|
19
|
Thamm DH. Canine Cancer: Strategies in Experimental Therapeutics. Front Oncol 2019; 9:1257. [PMID: 31803625 PMCID: PMC6873901 DOI: 10.3389/fonc.2019.01257] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 10/31/2019] [Indexed: 12/13/2022] Open
Abstract
Cancer is the most common cause of death in adult dogs. Many features of spontaneously developing tumors in pet dogs contribute to their potential utility as a human disease model. These include similar environmental exposures, similar clonal evolution as it applies to important factors such as immune avoidance, a favorable body size for imaging and serial biopsy, and a relatively contracted time course of disease progression, which makes evaluation of temporal endpoints such as progression free or overall survival feasible in a comparatively short time frame. These criteria have been leveraged to evaluate novel local therapies, demonstrate proof of tumor target inhibition or tumor localization, evaluate potential antimetastatic approaches, and assess the efficacy, safety and immune effects of a variety of immune-based therapeutics. Some of these canine proof of concept studies have been instrumental in informing subsequent human clinical trials. This review will cover key aspects of clinical trials in dogs with spontaneous neoplasia, with examples of how these studies have contributed to human cancer therapeutic development.
Collapse
Affiliation(s)
- Douglas H Thamm
- Flint Animal Cancer Center, Colorado State University, Fort Collins, CO, United States.,Cell and Molecular Biology Graduate Program, Colorado State University, Fort Collins, CO, United States.,University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
20
|
Regan D, Garcia K, Thamm D. Clinical, Pathological, and Ethical Considerations for the Conduct of Clinical Trials in Dogs with Naturally Occurring Cancer: A Comparative Approach to Accelerate Translational Drug Development. ILAR J 2019; 59:99-110. [PMID: 30668709 DOI: 10.1093/ilar/ily019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 11/26/2018] [Indexed: 01/21/2023] Open
Abstract
The role of comparative oncology in translational research is receiving increasing attention from drug developers and the greater biomedical research community. Pet dogs with spontaneous cancer are important and underutilized translational models, owing to dogs' large size and relative outbreeding, combined with their high incidence of certain tumor histotypes with significant biological, genetic, and histological similarities to their human tumor counterparts. Dogs with spontaneous tumors naturally develop therapy resistance and spontaneous metastasis, all in the context of an intact immune system. These fundamental features of cancer biology are often lacking in induced or genetically engineered preclinical tumor models and likely contribute to their poor predictive value and the associated overall high failure rate in oncology drug development. Thus, the conduct of clinical trials in pet dogs with naturally occurring cancer represents a viable surrogate and valuable intermediary step that should be increasingly incorporated into the cancer drug discovery and development pipeline. The development of molecular-targeted therapies has resulted in an expanded role of the pathologist in human oncology trials, and similarly the expertise of veterinary pathologists will be increasingly valuable to all phases of comparative oncology trial design and conduct. In this review, we provide a framework of clinical, ethical, and pathology-focused considerations for the increasing integration of translational research investigations in dogs with spontaneous cancer as a means to accelerate clinical cancer discovery and drug development.
Collapse
Affiliation(s)
- Daniel Regan
- Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Kelly Garcia
- Biologic Resources Laboratory, University of Illinois, Chicago, Illinois
| | - Douglas Thamm
- Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado
| |
Collapse
|
21
|
Suwan K, Yata T, Waramit S, Przystal JM, Stoneham CA, Bentayebi K, Asavarut P, Chongchai A, Pothachareon P, Lee KY, Topanurak S, Smith TL, Gelovani JG, Sidman RL, Pasqualini R, Arap W, Hajitou A. Next-generation of targeted AAVP vectors for systemic transgene delivery against cancer. Proc Natl Acad Sci U S A 2019; 116:18571-18577. [PMID: 31375630 PMCID: PMC6744886 DOI: 10.1073/pnas.1906653116] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Bacteriophage (phage) have attractive advantages as delivery systems compared with mammalian viruses, but have been considered poor vectors because they lack evolved strategies to confront and overcome mammalian cell barriers to infective agents. We reasoned that improved efficacy of delivery might be achieved through structural modification of the viral capsid to avoid pre- and postinternalization barriers to mammalian cell transduction. We generated multifunctional hybrid adeno-associated virus/phage (AAVP) particles to enable simultaneous display of targeting ligands on the phage's minor pIII proteins and also degradation-resistance motifs on the very numerous pVIII coat proteins. This genetic strategy of directed evolution bestows a next-generation of AAVP particles that feature resistance to fibrinogen adsorption or neutralizing antibodies and ability to escape endolysosomal degradation. This results in superior gene transfer efficacy in vitro and also in preclinical mouse models of rodent and human solid tumors. Thus, the unique functions of our next-generation AAVP particles enable improved targeted gene delivery to tumor cells.
Collapse
Affiliation(s)
- Keittisak Suwan
- Phage Therapy Group, Department of Brain Sciences, Imperial College London, W12 0NN London, United Kingdom
| | - Teerapong Yata
- Phage Therapy Group, Department of Brain Sciences, Imperial College London, W12 0NN London, United Kingdom
| | - Sajee Waramit
- Phage Therapy Group, Department of Brain Sciences, Imperial College London, W12 0NN London, United Kingdom
| | - Justyna M Przystal
- Phage Therapy Group, Department of Brain Sciences, Imperial College London, W12 0NN London, United Kingdom
| | - Charlotte A Stoneham
- Phage Therapy Group, Department of Brain Sciences, Imperial College London, W12 0NN London, United Kingdom
| | - Kaoutar Bentayebi
- Phage Therapy Group, Department of Brain Sciences, Imperial College London, W12 0NN London, United Kingdom
| | - Paladd Asavarut
- Phage Therapy Group, Department of Brain Sciences, Imperial College London, W12 0NN London, United Kingdom
| | - Aitthiphon Chongchai
- Thailand Excellence Center for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, 50200 Chiang Mai, Thailand
| | - Peraphan Pothachareon
- Thailand Excellence Center for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, 50200 Chiang Mai, Thailand
| | - Koon-Yang Lee
- Department of Aeronautics, Imperial College London, SW7 2AZ London, United Kingdom
| | - Supachai Topanurak
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, 10400 Bangkok, Thailand
| | - Tracey L Smith
- Rutgers Cancer Institute of New Jersey, Newark, NJ 07103
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103
| | - Juri G Gelovani
- Karmanos Cancer Institute, School of Medicine, Wayne State University, Detroit, MI 48201
- Department of Biomedical Engineering, College of Engineering, Wayne State University, Detroit, MI 48201
| | - Richard L Sidman
- Department of Neurology, Harvard Medical School, Boston, MA 02115;
| | - Renata Pasqualini
- Rutgers Cancer Institute of New Jersey, Newark, NJ 07103;
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103
| | - Wadih Arap
- Rutgers Cancer Institute of New Jersey, Newark, NJ 07103;
- Division of Hematology/Oncology, Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103
| | - Amin Hajitou
- Phage Therapy Group, Department of Brain Sciences, Imperial College London, W12 0NN London, United Kingdom;
| |
Collapse
|
22
|
Staquicini FI, Smith TL, Tang FHF, Gelovani JG, Giordano RJ, Libutti SK, Sidman RL, Cavenee WK, Arap W, Pasqualini R. Targeted AAVP-based therapy in a mouse model of human glioblastoma: a comparison of cytotoxic versus suicide gene delivery strategies. Cancer Gene Ther 2019; 27:301-310. [PMID: 31130731 PMCID: PMC6879804 DOI: 10.1038/s41417-019-0101-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 04/09/2019] [Accepted: 04/27/2019] [Indexed: 11/29/2022]
Abstract
Glioblastoma persists as a uniformly deadly diagnosis for patients and effective therapeutic options are gravely needed. Recently, targeted gene therapy approaches are reemerging as attractive experimental clinical agents. Our ligand-directed hybrid virus of adeno-associated virus and phage (AAVP) is a targeted gene delivery vector that has been used in several formulations displaying targeting ligand peptides to deliver clinically applicable transgenes. Here we compared different constructs side-by-side in a tumor model, an orthotopic model of xenograft human glioblastoma cells stereotactically implanted in immunodeficient mice. We have used divergent therapeutic strategies for two AAVP constructs, both displaying a double-cyclic RGD4C motif ligand specific for alpha V integrins expressed in tumor vascular endothelium, but carrying different genes of interest for the treatment of intracranial xenografted tumors. One construct delivered tumor necrosis factor (TNF), a purely cytotoxic gene for antitumor activity (RGD4C-AAVP-TNF); in the other construct, we delivered Herpes simplex virus thymidine kinase (HSVtk) for in tandem molecular-genetic imaging and targeted therapy (RGD4C-AAVP-HSVtk) utilizing ganciclovir (GCV) for a suicide gene therapy. Both AAVP constructs demonstrated antitumor activity, with damage to the tumor-associated neovasculature and induction of cell death evident after treatment. In addition, the ability to monitor transgene expression with a radiolabeled HSVtk substrate pre and post GCV treatment demonstrated the theranostic potential of RGD4C-AAVP-HSVtk. We conclude that targeted AAVP constructs delivering either cytotoxic TNF or theranostic HSVtk followed by suicide gene therapy with GCV have comparable preclinical efficacy, at least in this standard experimental model. The results presented here provide a blueprint for future studies of targeted gene delivery against human glioblastomas and other brain tumors.
Collapse
Affiliation(s)
- Fernanda I Staquicini
- Rutgers Cancer Institute of New Jersey and Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Tracey L Smith
- Rutgers Cancer Institute of New Jersey and Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Fenny H F Tang
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Juri G Gelovani
- Karmanos Cancer Institute, School of Medicine and Department of Biomedical Engineering, College of Engineering, Wayne State University, Detroit, MI, USA
| | - Ricardo J Giordano
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Steven K Libutti
- Rutgers Cancer Institute of New Jersey and Department of Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Richard L Sidman
- Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Webster K Cavenee
- Ludwig Institute for Cancer Research, University of California-San Diego, La Jolla, CA, USA
| | - Wadih Arap
- Rutgers Cancer Institute of New Jersey and Division of Hematology/Oncology, Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA.
| | - Renata Pasqualini
- Rutgers Cancer Institute of New Jersey and Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ, USA.
| |
Collapse
|
23
|
Huh H, Wong S, St Jean J, Slavcev R. Bacteriophage interactions with mammalian tissue: Therapeutic applications. Adv Drug Deliv Rev 2019; 145:4-17. [PMID: 30659855 DOI: 10.1016/j.addr.2019.01.003] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 11/30/2018] [Accepted: 01/03/2019] [Indexed: 12/12/2022]
Abstract
The human body is a large reservoir for bacterial viruses known as bacteriophages (phages), which participate in dynamic interactions with their bacterial and human hosts that ultimately affect human health. The current growing interest in human resident phages is paralleled by new uses of phages, including the design of engineered phages for therapeutic applications. Despite the increasing number of clinical trials being conducted, the understanding of the interaction of phages and mammalian cells and tissues is still largely unknown. The presence of phages in compartments within the body previously considered purely sterile, suggests that phages possess a unique capability of bypassing anatomical and physiological barriers characterized by varying degrees of selectivity and permeability. This review will discuss the direct evidence of the accumulation of bacteriophages in various tissues, focusing on the unique capability of phages to traverse relatively impermeable barriers in mammals and its relevance to its current applications in therapy.
Collapse
Affiliation(s)
- Haein Huh
- School of Pharmacy, University of Waterloo, 10A Victoria St S, Kitchener N2G 1C5, Canada
| | - Shirley Wong
- School of Pharmacy, University of Waterloo, 10A Victoria St S, Kitchener N2G 1C5, Canada
| | - Jesse St Jean
- School of Pharmacy, University of Waterloo, 10A Victoria St S, Kitchener N2G 1C5, Canada
| | - Roderick Slavcev
- School of Pharmacy, University of Waterloo, 10A Victoria St S, Kitchener N2G 1C5, Canada.
| |
Collapse
|
24
|
Mochel JP, Ekker SC, Johannes CM, Jergens AE, Allenspach K, Bourgois-Mochel A, Knouse M, Benzekry S, Wierson W, LeBlanc AK, Kenderian SS. CAR T Cell Immunotherapy in Human and Veterinary Oncology: Changing the Odds Against Hematological Malignancies. AAPS JOURNAL 2019; 21:50. [PMID: 30963322 DOI: 10.1208/s12248-019-0322-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 03/17/2019] [Indexed: 01/14/2023]
Abstract
The advent of the genome editing era brings forth the promise of adoptive cell transfer using engineered chimeric antigen receptor (CAR) T cells for targeted cancer therapy. CAR T cell immunotherapy is probably one of the most encouraging developments for the treatment of hematological malignancies. In 2017, two CAR T cell therapies were approved by the US Food and Drug Administration: one for the treatment of pediatric acute lymphoblastic leukemia (ALL) and the other for adult patients with advanced lymphomas. However, despite significant progress in the area, CAR T cell therapy is still in its early days and faces significant challenges, including the complexity and costs associated with the technology. B cell lymphoma is the most common hematopoietic cancer in dogs, with an incidence approaching 0.1% and a total of 20-100 cases per 100,000 individuals. It is a widely accepted naturally occurring model for human non-Hodgkin's lymphoma. Current treatment is with combination chemotherapy protocols, which prolong life for less than a year in canines and are associated with severe dose-limiting side effects, such as gastrointestinal and bone marrow toxicity. To date, one canine study generated CAR T cells by transfection of mRNA for CAR domain expression. While this was shown to provide a transient anti-tumor activity, results were modest, indicating that stable, genomic integration of CAR modules is required in order to achieve lasting therapeutic benefit. This commentary summarizes the current state of knowledge on CAR T cell immunotherapy in human medicine and its potential applications in animal health, while discussing the potential of the canine model as a translational system for immuno-oncology research.
Collapse
Affiliation(s)
- Jonathan P Mochel
- Department of Biomedical Sciences, Iowa State University, Ames, Iowa, 50011, USA. .,Iowa State University College of Vet. Medicine, 2448 Lloyd, 1809 S Riverside Dr., Ames, Iowa, 50011-1250, USA.
| | - Stephen C Ekker
- Mayo Clinic Cancer Center Department of Biochemistry and Molecular Biology, Rochester, Minnesota, 55905, USA
| | - Chad M Johannes
- Department of Veterinary Clinical Sciences, Iowa State University, Ames, Iowa, 50011, USA
| | - Albert E Jergens
- Department of Veterinary Clinical Sciences, Iowa State University, Ames, Iowa, 50011, USA
| | - Karin Allenspach
- Department of Veterinary Clinical Sciences, Iowa State University, Ames, Iowa, 50011, USA
| | - Agnes Bourgois-Mochel
- Department of Veterinary Clinical Sciences, Iowa State University, Ames, Iowa, 50011, USA
| | - Michael Knouse
- Department of Biomedical Sciences, Iowa State University, Ames, Iowa, 50011, USA
| | - Sebastien Benzekry
- Team MONC, Institut National de Recherche en Informatique et en Automatique, Bordeaux, France
| | - Wesley Wierson
- Department of Genetics, Development, and Cell Biology, Iowa State University, Ames, Iowa, 50011, USA
| | - Amy K LeBlanc
- Comparative Oncology Program, Center for Cancer Research National Cancer Institute, Bethesda, Maryland, 20892, USA
| | - Saad S Kenderian
- Department of Medicine, Mayo Clinic Division of Hematology, Rochester, Minnesota, 55905, USA.,Department of Immunology, Mayo Clinic, Rochester, Minnesota, 55905, USA
| |
Collapse
|
25
|
Przystal JM, Waramit S, Pranjol MZI, Yan W, Chu G, Chongchai A, Samarth G, Olaciregui NG, Tabatabai G, Carcaboso AM, Aboagye EO, Suwan K, Hajitou A. Efficacy of systemic temozolomide-activated phage-targeted gene therapy in human glioblastoma. EMBO Mol Med 2019; 11:e8492. [PMID: 30808679 PMCID: PMC6460351 DOI: 10.15252/emmm.201708492] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 02/01/2019] [Accepted: 02/04/2019] [Indexed: 12/23/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most lethal primary intracranial malignant neoplasm in adults and most resistant to treatment. Integration of gene therapy and chemotherapy, chemovirotherapy, has the potential to improve treatment. We have introduced an intravenous bacteriophage (phage) vector for dual targeting of therapeutic genes to glioblastoma. It is a hybrid AAV/phage, AAVP, designed to deliver a recombinant adeno-associated virus genome (rAAV) by the capsid of M13 phage. In this vector, dual tumor targeting is first achieved by phage capsid display of the RGD4C ligand that binds the αvβ3 integrin receptor. Second, genes are expressed from a tumor-activated and temozolomide (TMZ)-induced promoter of the glucose-regulated protein, Grp78 Here, we investigated systemic combination therapy using TMZ and targeted suicide gene therapy by the RGD4C/AAVP-Grp78 Firstly, in vitro we showed that TMZ increases endogenous Grp78 gene expression and boosts transgene expression from the RGD4C/AAVP-Grp78 in human GBM cells. Next, RGD4C/AAVP-Grp78 targets intracranial tumors in mice following intravenous administration. Finally, combination of TMZ and RGD4C/AAVP-Grp78 targeted gene therapy exerts a synergistic effect to suppress growth of orthotopic glioblastoma.
Collapse
Affiliation(s)
- Justyna Magdalena Przystal
- Phage Therapy Group, Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK
| | - Sajee Waramit
- Phage Therapy Group, Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK
| | - Md Zahidul Islam Pranjol
- Phage Therapy Group, Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK
| | - Wenqing Yan
- Phage Therapy Group, Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK
| | - Grace Chu
- Phage Therapy Group, Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK
| | - Aitthiphon Chongchai
- Thailand Excellence Centre for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine Chiang Mai University, Chiang Mai, Thailand
| | - Gargi Samarth
- Phage Therapy Group, Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK
| | - Nagore Gene Olaciregui
- Institute de Recerca Sant Joan de Deu, Barcelona, Spain
- Department of Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Barcelona, Spain
| | - Ghazaleh Tabatabai
- Interdisciplinary Division of Neuro-Oncology, Hertie Institute for Clinical Brain Research, Center for CNS Tumors, Comprehensive Cancer Center, University Hospital Tübingen, Eberhard Karls University, Tübingen, Germany
- German Cancer Consortium (DKTK), DKFZ Partner Site Tübingen, Tübingen, Germany
| | - Angel Montero Carcaboso
- Institute de Recerca Sant Joan de Deu, Barcelona, Spain
- Department of Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Barcelona, Spain
| | - Eric Ofori Aboagye
- Comprehensive Cancer Imaging Centre, Imperial College London, Faculty of Medicine, London, UK
| | - Keittisak Suwan
- Phage Therapy Group, Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK
| | - Amin Hajitou
- Phage Therapy Group, Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK
| |
Collapse
|
26
|
Fan TM, Selting KA. Exploring the Potential Utility of Pet Dogs With Cancer for Studying Radiation-Induced Immunogenic Cell Death Strategies. Front Oncol 2019; 8:680. [PMID: 30697532 PMCID: PMC6340932 DOI: 10.3389/fonc.2018.00680] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 12/27/2018] [Indexed: 01/21/2023] Open
Abstract
Radiotherapy serves as a foundational pillar for the therapeutic management of diverse solid tumors through the generation of lethal DNA damage and induction of cell death. While the direct cytotoxic effects of radiation therapy remain a cornerstone for cancer management, in the era of immunooncology there is renewed and focused interest in exploiting the indirect bystander activities of radiation, termed abscopal effects. In radioimmunobiologic terms, abscopal effects describe the radiotherapy-induced regression of cancerous lesions distant from the primary site of radiation delivery and rely upon the induction of immunogenic cell death and consequent systemic anticancer immune activation. Despite the promise of radiation therapy for awaking potent anticancer immune responses, the purposeful harnessing of abscopal effects with radiotherapy remain clinically elusive. In part, failure to fully leverage and clinically implement the promise of radiation-induced abscopal effects stems from limitations associated with existing conventional tumor models which inadequately recapitulate the complexity of malignant transformation and the dynamic nature of tumor immune surveillance. To supplement this existing gap in modeling systems, pet dogs diagnosed with solid tumors including melanoma and osteosarcoma, which are both metastatic and immunogenic in nature, could potentially serve as unique resources for exploring the fundamental underpinnings required for maximizing radiation-induced abscopal effects. Given the spontaneous course of cancer development in the context of operative immune mechanisms, pet dogs treated with radiotherapy for metastatic solid tumors might be leveraged as valuable model systems for realizing the science and best clinical practices necessary to generate potent abscopal effects with anti-metastatic immune activities.
Collapse
Affiliation(s)
- Timothy M Fan
- Comparative Oncology Research Laboratory, Department of Veterinary Clinical Medicine, College of Veterinary Medicine, University of Illinois at Urbana-Champaign Urbana, IL, United States
| | - Kimberly A Selting
- Comparative Oncology Research Laboratory, Department of Veterinary Clinical Medicine, College of Veterinary Medicine, University of Illinois at Urbana-Champaign Urbana, IL, United States
| |
Collapse
|
27
|
Burton JH, Mazcko C, LeBlanc A, Covey JM, Ji J, Kinders RJ, Parchment RE, Khanna C, Paoloni M, Lana S, Weishaar K, London C, Kisseberth W, Krick E, Vail D, Childress M, Bryan JN, Barber L, Ehrhart EJ, Kent M, Fan T, Kow K, Northup N, Wilson-Robles H, Tomaszewski J, Holleran JL, Muzzio M, Eiseman J, Beumer JH, Doroshow JH, Pommier Y. NCI Comparative Oncology Program Testing of Non-Camptothecin Indenoisoquinoline Topoisomerase I Inhibitors in Naturally Occurring Canine Lymphoma. Clin Cancer Res 2018; 24:5830-5840. [PMID: 30061364 PMCID: PMC6312717 DOI: 10.1158/1078-0432.ccr-18-1498] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Revised: 06/19/2018] [Accepted: 07/23/2018] [Indexed: 12/16/2022]
Abstract
PURPOSE Only one chemical class of topoisomerase I (TOP1) inhibitors is FDA approved, the camptothecins with irinotecan and topotecan widely used. Because of their limitations (chemical instability, drug efflux-mediated resistance, and diarrhea), novel TOP1 inhibitors are warranted. Indenoisoquinoline non-camptothecin topoisomerase I (TOP1) inhibitors overcome chemical instability and drug resistance that limit camptothecin use. Three indenoisoquinolines, LMP400 (indotecan), LMP776 (indimitecan), and LMP744, were examined in a phase I study for lymphoma-bearing dogs to evaluate differential efficacy, pharmacodynamics, toxicology, and pharmacokinetics. EXPERIMENTAL DESIGN Eighty-four client-owned dogs with lymphomas were enrolled in dose-escalation cohorts for each indenoisoquinoline, with an expansion phase for LMP744. Efficacy, tolerability, pharmacokinetics, and target engagement were determined. RESULTS The MTDs were 17.5 mg/m2 for LMP 776 and 100 mg/m2 for LMP744; bone marrow toxicity was dose-limiting; up to 65 mg/m2 LMP400 was well-tolerated and MTD was not reached. None of the drugs induced notable diarrhea. Sustained tumor accumulation was observed for LMP744; γH2AX induction was demonstrated in tumors 2 and 6 hours after treatment; a decrease in TOP1 protein was observed in most lymphoma samples across all compounds and dose levels, which is consistent with the fact that tumor response was also observed at low doses LMP744. Objective responses were documented for all indenoisoquinolines; efficacy (13/19 dogs) was greatest for LMP744. CONCLUSIONS These results demonstrate proof-of-mechanism for indenoisoquinoline TOP1 inhibitors supporting their further clinical development. They also highlight the value of the NCI Comparative Oncology Program (https://ccr.cancer.gov/Comparative-Oncology-Program) for evaluating novel therapies in immunocompetent pets with cancers.
Collapse
Affiliation(s)
- Jenna H Burton
- School of Veterinary Medicine, University of California Davis, Davis, California
| | - Christina Mazcko
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Amy LeBlanc
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Joseph M Covey
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland
| | - Jiuping Ji
- Clinical Pharmacodynamic Biomarker Program, Applied/Developmental Research Directorate, Leidos Biomedical Research, Inc., NCI Campus at Frederick, Frederick, Maryland
| | - Robert J Kinders
- Clinical Pharmacodynamic Biomarker Program, Applied/Developmental Research Directorate, Leidos Biomedical Research, Inc., NCI Campus at Frederick, Frederick, Maryland
| | - Ralph E Parchment
- Clinical Pharmacodynamic Biomarker Program, Applied/Developmental Research Directorate, Leidos Biomedical Research, Inc., NCI Campus at Frederick, Frederick, Maryland
| | - Chand Khanna
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Melissa Paoloni
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Sue Lana
- College of Veterinary Medicine, The Ohio State University, Columbus, Ohio
| | - Kristen Weishaar
- College of Veterinary Medicine, The Ohio State University, Columbus, Ohio
| | - Cheryl London
- School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - William Kisseberth
- School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Erika Krick
- School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - David Vail
- College of Veterinary Medicine, Purdue University, West Lafayette, Indiana
| | - Michael Childress
- College of Veterinary Medicine, University of Missouri-Columbia, Columbia, Missouri
| | - Jeffrey N Bryan
- School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts
| | - Lisa Barber
- College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - E J Ehrhart
- College of Veterinary Medicine, The Ohio State University, Columbus, Ohio
| | - Michael Kent
- School of Veterinary Medicine, University of California Davis, Davis, California
| | - Timothy Fan
- College of Veterinary Medicine, University of Florida, Gainesville, Florida
| | - Kelvin Kow
- College of Veterinary Medicine, University of Georgia, Athens, Georgia
| | - Nicole Northup
- College of Veterinary Medicine, Texas A&M University, College Station, Texas
| | - Heather Wilson-Robles
- Cancer Therapeutics Program, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania
| | - Joseph Tomaszewski
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland
| | | | - Miguel Muzzio
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda
| | - Julie Eiseman
- Life Science Group, IIT Research Institute, Chicago, Illinois
| | - Jan H Beumer
- Life Science Group, IIT Research Institute, Chicago, Illinois
| | - James H Doroshow
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda
| | - Yves Pommier
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda.
| |
Collapse
|
28
|
Abstract
Pet dogs are becoming increasingly recognized as a population with the potential to inform medical research through their treatment for a variety of maladies by veterinary health professionals. This is the basis of the One Health initiative, supporting the idea of collaboration between human and animal health researchers and clinicians to study spontaneous disease processes and treatment in animals to inform human health. Cancer is a major health burden in pet dogs, accounting for approximately 30% of deaths across breeds. As such, pet dogs with cancer are becoming increasingly recognized as a resource for studying the pharmacology and therapeutic potential of anticancer drugs and therapies under development. This was recently highlighted by a National Academy of Medicine Workshop on Comparative Oncology that took place in mid-2015 (http://www.nap.edu/21830). One component of cancer burden in dogs is their significantly higher incidence of sarcomas as compared to humans. This increased incidence led to canine osteosarcoma being an important component in the development of surgical approaches for osteosarcoma in children. Included in this review of sarcomas in dogs is a description of the incidence, pathology, molecular characteristics and previous translational therapeutic studies associated with these tumors. An understanding of the patho-physiological and molecular characteristics of these naturally occurring canine sarcomas holds great promise for effective incorporation into drug development schemas, for evaluation of target modulation or other pharmacodynamic measures associated with therapeutic response. These data could serve to supplement other preclinical data and bolster clinical investigations in tumor types for which there is a paucity of human patients for clinical trials.
Collapse
Affiliation(s)
- Daniel L Gustafson
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA; Flint Animal Cancer Center, Colorado State University, Fort Collins, CO 80523, USA; University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, CO 80045, USA.
| | - Dawn L Duval
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA; Flint Animal Cancer Center, Colorado State University, Fort Collins, CO 80523, USA; University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Daniel P Regan
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA; Flint Animal Cancer Center, Colorado State University, Fort Collins, CO 80523, USA; University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Douglas H Thamm
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA; Flint Animal Cancer Center, Colorado State University, Fort Collins, CO 80523, USA; University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
29
|
Chira S, Gulei D, Hajitou A, Berindan-Neagoe I. Restoring the p53 'Guardian' Phenotype in p53-Deficient Tumor Cells with CRISPR/Cas9. Trends Biotechnol 2018; 36:653-660. [PMID: 29478674 DOI: 10.1016/j.tibtech.2018.01.014] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Revised: 01/26/2018] [Accepted: 01/30/2018] [Indexed: 12/26/2022]
Abstract
With an increasing prevalence in the human population, cancer has become one of the most investigated fields of medicine. Among the potential targets for cancer therapy is the tumor suppressor gene TP53, which is found in a mutated state in approximately 50% of human cancers and is often associated with poor prognosis. We propose a novel, highly tumor-specific delivery system for TP53, based on the CRISPR/Cas9 genome editing technology. This system will restore the normal p53 phenotype in tumor cells by replacing the mutant TP53 gene with a functional copy, leading to sustained expression of p53 protein and tumor regression.
Collapse
Affiliation(s)
- Sergiu Chira
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania.
| | - Diana Gulei
- MedFuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Amin Hajitou
- Cancer Phage Therapy Group, Division of Brain Sciences, Imperial College London, W12 0NN London, UK
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; MedFuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; Department of Functional Genomics and Experimental Pathology, Oncology Institute Prof. Dr. Ion Chiricuta, 400015 Cluj-Napoca, Romania
| |
Collapse
|
30
|
Tsafa E, Al-Bahrani M, Bentayebi K, Przystal J, Suwan K, Hajitou A. The natural dietary genistein boosts bacteriophage-mediated cancer cell killing by improving phage-targeted tumor cell transduction. Oncotarget 2018; 7:52135-52149. [PMID: 27437775 PMCID: PMC5239540 DOI: 10.18632/oncotarget.10662] [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: 11/24/2015] [Accepted: 06/16/2016] [Indexed: 12/25/2022] Open
Abstract
Gene therapy has long been regarded as a promising treatment for cancer. However, cancer gene therapy is still facing the challenge of targeting gene delivery vectors specifically to tumors when administered via clinically acceptable non-invasive systemic routes (i.e. intravenous). The bacteria virus, bacteriophage (phage), represents a new generation of promising vectors in systemic gene delivery since their targeting can be achieved through phage capsid display ligands, which enable them to home to specific tumor receptors without the need to ablate any native eukaryotic tropism. We have previously reported a tumor specific bacteriophage vector named adeno-associated virus/phage, or AAVP, in which gene expression is under a recombinant human rAAV2 virus genome targeted to tumors via a ligand-directed phage capsid. However, cancer gene therapy with this tumor-targeted vector achieved variable outcomes ranging from tumor regression to no effect in both experimental and natural preclinical models. Herein, we hypothesized that combining the natural dietary genistein, with proven anticancer activity, would improve bacteriophage anticancer safe therapy. We show that combination treatment with genistein and AAVP increased targeted cancer cell killing by AAVP carrying the gene for Herpes simplex virus thymidine kinase (HSVtk) in 2D tissue cultures and 3D tumor spheroids. We found this increased tumor cell killing was associated with enhanced AAVP-mediated gene expression. Next, we established that genistein protects AAVP against proteasome degradation and enhances vector genome accumulation in the nucleus. Combination of genistein and phage-guided virotherapy is a safe and promising strategy that should be considered in anticancer therapy with AAVP.
Collapse
Affiliation(s)
- Effrosyni Tsafa
- Phage Therapy Group, Department of Medicine, Imperial College London, Hammersmith Hospital Campus, London, United Kingdom
| | - Mariam Al-Bahrani
- Phage Therapy Group, Department of Medicine, Imperial College London, Hammersmith Hospital Campus, London, United Kingdom
| | - Kaoutar Bentayebi
- Biotechnology Laboratory (Medbiotech), Medical and Pharmacy School, University Mohammed V de Rabat, Rabat, Morocco
| | - Justyna Przystal
- Phage Therapy Group, Department of Medicine, Imperial College London, Hammersmith Hospital Campus, London, United Kingdom
| | - Keittisak Suwan
- Phage Therapy Group, Department of Medicine, Imperial College London, Hammersmith Hospital Campus, London, United Kingdom
| | - Amin Hajitou
- Phage Therapy Group, Department of Medicine, Imperial College London, Hammersmith Hospital Campus, London, United Kingdom
| |
Collapse
|
31
|
Gulei D, Berindan-Neagoe I. CRISPR/Cas9: A Potential Life-Saving Tool. What's next? MOLECULAR THERAPY. NUCLEIC ACIDS 2017; 9:333-336. [PMID: 29246311 PMCID: PMC5684489 DOI: 10.1016/j.omtn.2017.10.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 10/17/2017] [Accepted: 10/18/2017] [Indexed: 12/11/2022]
Affiliation(s)
- Diana Gulei
- MEDFUTURE-Research Center for Advanced Medicine, "Iuliu-Hatieganu" University of Medicine and Pharmacy, Marinescu 23 Street, 400337 Cluj-Napoca, Romania
| | - Ioana Berindan-Neagoe
- MEDFUTURE-Research Center for Advanced Medicine, "Iuliu-Hatieganu" University of Medicine and Pharmacy, Marinescu 23 Street, 400337 Cluj-Napoca, Romania; Research Center for Functional Genomics, Biomedicine and Translational Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, Marinescu 23 Street, 400337 Cluj-Napoca, Romania; Department of Functional Genomics and Experimental Pathology, The Oncology Institute "Prof. Dr. Ion Chiricuta," Republicii 34-36 Street, Cluj-Napoca, Romania.
| |
Collapse
|
32
|
The first report of cases of pet dogs with naturally occurring cancer treated with the antitumor peptide CIGB-552. Res Vet Sci 2017; 114:502-510. [DOI: 10.1016/j.rvsc.2017.09.029] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 08/07/2017] [Accepted: 09/27/2017] [Indexed: 01/13/2023]
|
33
|
Panek WK, Kane JR, Young JS, Rashidi A, Kim JW, Kanojia D, Lesniak MS. Hitting the nail on the head: combining oncolytic adenovirus-mediated virotherapy and immunomodulation for the treatment of glioma. Oncotarget 2017; 8:89391-89405. [PMID: 29179527 PMCID: PMC5687697 DOI: 10.18632/oncotarget.20810] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Accepted: 08/26/2017] [Indexed: 12/31/2022] Open
Abstract
Glioblastoma is a highly aggressive malignant brain tumor with a poor prognosis and the median survival 14.6 months. Immunomodulatory proteins and oncolytic viruses represent two treatment approaches that have recently been developed for patients with glioblastoma that could extend patient survival and result in better treatment outcomes for patients with this disease. Together, these approaches could potentially augment the treatment efficacy and strength of these anti-tumor therapies. In addition to oncolytic activities, this combinatory approach introduces immunomodulation locally only where cancerous cells are present. This thereby results in the change of the tumor microenvironment from immune-suppressive to immune-vulnerable via activation of cytotoxic T cells or through the removal of glioma cells immune-suppressive capability. This review discusses the strengths and weaknesses of adenoviral oncolytic therapy, and highlights the genetic modifications that result in more effective and targeted viral agents. Additionally, the mechanism of action of immune-activating agents is described and the results of previous clinical trials utilizing these treatments in other solid tumors are reviewed. The feasibility, synergy, and limitations for treatments that combine these two approaches are outlined and areas for which more work is needed are considered.
Collapse
Affiliation(s)
- Wojciech K Panek
- Department of Neurological Surgery, Northwestern University, Chicago, IL, 60611, USA
| | - J Robert Kane
- Department of Neurological Surgery, Northwestern University, Chicago, IL, 60611, USA
| | - Jacob S Young
- Pritzker School of Medicine, University of Chicago, Chicago, IL, 60637, USA
| | - Aida Rashidi
- Department of Neurological Surgery, Northwestern University, Chicago, IL, 60611, USA
| | - Julius W Kim
- Department of Neurological Surgery, Northwestern University, Chicago, IL, 60611, USA
| | - Deepak Kanojia
- Department of Neurological Surgery, Northwestern University, Chicago, IL, 60611, USA
| | - Maciej S Lesniak
- Department of Neurological Surgery, Northwestern University, Chicago, IL, 60611, USA
| |
Collapse
|
34
|
Gianferante DM, Mirabello L, Savage SA. Germline and somatic genetics of osteosarcoma - connecting aetiology, biology and therapy. Nat Rev Endocrinol 2017; 13:480-491. [PMID: 28338660 DOI: 10.1038/nrendo.2017.16] [Citation(s) in RCA: 316] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Clinical outcomes and treatment modalities for osteosarcoma, the most common primary cancer of bone, have changed very little over the past 30 years. The peak incidence of osteosarcoma occurs during the adolescent growth spurt, which suggests that bone growth and pubertal hormones are important in the aetiology of the disease. Tall stature, high birth weight and certain inherited cancer predisposition syndromes are well-described risk factors for osteosarcoma. Common genetic variants are also associated with osteosarcoma. The somatic genome of osteosarcoma is highly aneuploid, exhibits extensive intratumoural heterogeneity and has a higher mutation rate than most other paediatric cancers. Complex pathways related to bone growth and development and tumorigenesis are also important in osteosarcoma biology. In this Review, we discuss the contributions of germline and somatic genetics, tumour biology and animal models in improving our understanding of osteosarcoma aetiology, and their potential to identify novel therapeutic targets and thus improve the lives of patients with osteosarcoma.
Collapse
Affiliation(s)
- D Matthew Gianferante
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, 9609 Medical Center Drive, Bethesda, Maryland 20892, USA
| | - Lisa Mirabello
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, 9609 Medical Center Drive, Bethesda, Maryland 20892, USA
| | - Sharon A Savage
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, 9609 Medical Center Drive, Bethesda, Maryland 20892, USA
| |
Collapse
|
35
|
An AAVP-based solid-phase transducing matrix for transgene delivery: potential for translational applications. Cancer Gene Ther 2017; 24:358-360. [PMID: 28548103 PMCID: PMC5605672 DOI: 10.1038/cgt.2017.19] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 04/14/2017] [Accepted: 04/19/2017] [Indexed: 11/25/2022]
Abstract
A hybrid vector of adeno-associated virus and phage (termed AAVP) has been introduced as a platform for systemic ligand-directed delivery of transgenes to tumors over the past decade. A series of studies have evaluated the AAVP platform for potential theranostic or purely therapeutic applications in several tumor models. Sufficient ligand-directed tumor targeting consistently resulted in specific molecular-genetic imaging and/or anti-tumor responses to ‘suicide’ transgene delivery. However, efforts to optimize transduction efficiency are still ongoing. Here, we set out to expand the translational utility of AAVP by combining it with gold (Au) nanoparticles in order to generate a ‘transducing matrix’ for improved targeted gene delivery in solid phase. Targeted AAVP-based solid-phase transduction is superior to conventional transduction in soluble (aqueous) environments. This transducing matrix is stable and can be further modified with additional attributes (for example, magnetization) for targeted imaging and therapeutic gene delivery. Notably, it spontaneously assembles around cells in vitro to markedly enhance transduction capabilities compared with AAVP alone. This versatile nanoplatform may enable new applications of AAVP for transgene delivery in translational settings including, for example, efforts toward complex tissue patterning.
Collapse
|
36
|
Finocchiaro LME, Glikin GC. Recent clinical trials of cancer immunogene therapy in companion animals. World J Exp Med 2017; 7:42-48. [PMID: 28589078 PMCID: PMC5439171 DOI: 10.5493/wjem.v7.i2.42] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Revised: 03/22/2017] [Accepted: 05/05/2017] [Indexed: 02/06/2023] Open
Abstract
This mini-review presents the results of veterinary clinical trials on immunogene therapy published from 2014 to 2016. A variety of tumors, among them melanoma (canine and equine), mastocytoma (canine), mammary adenocarcinoma (canine) and fibrosarcoma (feline) were treated by using diverse strategies. Non-viral vectors were usually employed to transfer genes of cytokines, suicide enzymes and/or tumor associated antigens. In general terms, minor or no adverse collateral effects were related to these procedures, and treated patients frequently improved their conditions (better quality of life, delayed or suppressed recurrence or metastatic spread, increased survival). Some of these new methodologies have a promising future if applied as adjuvant treatments of standard approaches. The auspicious results, derived from immunogene therapy studies carried out in companion animals, warrant their imperative usage in veterinary clinical oncology. Besides, they provide a strong preclinical basis (safety assays and proofs of concept) for analogous human clinical trials.
Collapse
|
37
|
Statz CM, Patterson SE, Mockus SM. Barriers preventing the adoption of comprehensive cancer genomic profiling in the clinic. Expert Rev Mol Diagn 2017; 17:549-555. [DOI: 10.1080/14737159.2017.1319280] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
38
|
Abstract
This article summarizes the relevant definitions related to biomarkers; reviews the general processes related to biomarker discovery and ultimate acceptance and use; and finally summarizes and reviews, to the extent possible, examples of the types of biomarkers used in animal species within veterinary clinical practice and human and veterinary drug development. We highlight opportunities for collaboration and coordination of research within the veterinary community and leveraging of resources from human medicine to support biomarker discovery and validation efforts for veterinary medicine.
Collapse
Affiliation(s)
- Michael J Myers
- Center for Veterinary Medicine, Food and Drug Administration, Rockville, Maryland 20855;
| | - Emily R Smith
- Center for Veterinary Medicine, Food and Drug Administration, Rockville, Maryland 20855;
| | - Phillip G Turfle
- Center for Veterinary Medicine, Food and Drug Administration, Rockville, Maryland 20855;
| |
Collapse
|
39
|
Yao VJ, D'Angelo S, Butler KS, Theron C, Smith TL, Marchiò S, Gelovani JG, Sidman RL, Dobroff AS, Brinker CJ, Bradbury ARM, Arap W, Pasqualini R. Ligand-targeted theranostic nanomedicines against cancer. J Control Release 2016; 240:267-286. [PMID: 26772878 PMCID: PMC5444905 DOI: 10.1016/j.jconrel.2016.01.002] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 12/17/2015] [Accepted: 01/02/2016] [Indexed: 02/06/2023]
Abstract
Nanomedicines have significant potential for cancer treatment. Although the majority of nanomedicines currently tested in clinical trials utilize simple, biocompatible liposome-based nanocarriers, their widespread use is limited by non-specificity and low target site concentration and thus, do not provide a substantial clinical advantage over conventional, systemic chemotherapy. In the past 20years, we have identified specific receptors expressed on the surfaces of tumor endothelial and perivascular cells, tumor cells, the extracellular matrix and stromal cells using combinatorial peptide libraries displayed on bacteriophage. These studies corroborate the notion that unique receptor proteins such as IL-11Rα, GRP78, EphA5, among others, are differentially overexpressed in tumors and present opportunities to deliver tumor-specific therapeutic drugs. By using peptides that bind to tumor-specific cell-surface receptors, therapeutic agents such as apoptotic peptides, suicide genes, imaging dyes or chemotherapeutics can be precisely and systemically delivered to reduce tumor growth in vivo, without harming healthy cells. Given the clinical applicability of peptide-based therapeutics, targeted delivery of nanocarriers loaded with therapeutic cargos seems plausible. We propose a modular design of a functionalized protocell in which a tumor-targeting moiety, such as a peptide or recombinant human antibody single chain variable fragment (scFv), is conjugated to a lipid bilayer surrounding a silica-based nanocarrier core containing a protected therapeutic cargo. The functionalized protocell can be tailored to a specific cancer subtype and treatment regimen by exchanging the tumor-targeting moiety and/or therapeutic cargo or used in combination to create unique, theranostic agents. In this review, we summarize the identification of tumor-specific receptors through combinatorial phage display technology and the use of antibody display selection to identify recombinant human scFvs against these tumor-specific receptors. We compare the characteristics of different types of simple and complex nanocarriers, and discuss potential types of therapeutic cargos and conjugation strategies. The modular design of functionalized protocells may improve the efficacy and safety of nanomedicines for future cancer therapy.
Collapse
Affiliation(s)
- Virginia J Yao
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131; Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM 87131
| | - Sara D'Angelo
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131; Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM 87131
| | - Kimberly S Butler
- Department of Chemical and Biological Engineering, University of New Mexico, Albuquerque, NM 87131
| | - Christophe Theron
- Department of Chemical and Biological Engineering, University of New Mexico, Albuquerque, NM 87131
| | - Tracey L Smith
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131; Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM 87131
| | - Serena Marchiò
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131; Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM 87131; Department of Oncology, University of Turin, Candiolo, 10060, Italy
| | - Juri G Gelovani
- Department of Biomedical Engineering, College of Engineering and School of Medicine, Wayne State University, Detroit, MI 48201
| | - Richard L Sidman
- Department of Neurology, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA 02215
| | - Andrey S Dobroff
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131; Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM 87131
| | - C Jeffrey Brinker
- Department of Chemical and Biological Engineering, University of New Mexico, Albuquerque, NM 87131; Center for Micro-Engineered Materials, University of New Mexico, Albuquerque, NM 87131; Cancer Research and Treatment Center, Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, University of New Mexico, Albuquerque, NM 87131; Self-Assembled Materials Department, Sandia National Laboratories, Albuquerque, NM 87185
| | - Andrew R M Bradbury
- Bioscience Division, Los Alamos National Laboratories, Los Alamos, NM, 87545
| | - Wadih Arap
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131; Division of Hematology/Oncology, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM 87131.
| | - Renata Pasqualini
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131; Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM 87131.
| |
Collapse
|
40
|
Towards a transcriptome-based theranostic platform for unfavorable breast cancer phenotypes. Proc Natl Acad Sci U S A 2016; 113:12780-12785. [PMID: 27791177 DOI: 10.1073/pnas.1615288113] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Inflammatory breast carcinoma (IBC) is one of the most lethal forms of human breast cancer, and effective treatment for IBC is an unmet clinical need in contemporary oncology. Tumor-targeted theranostic approaches are emerging in precision medicine, but only a few specific biomarkers are available. Here we report up-regulation of the 78-kDa glucose-regulated protein (GRP78) in two independent discovery and validation sets of specimens derived from IBC patients, suggesting translational promise for clinical applications. We show that a GRP78-binding motif displayed on either bacteriophage or adeno-associated virus/phage (AAVP) particles or loop-grafted onto a human antibody fragment specifically targets orthotopic IBC and other aggressive breast cancer models in vivo. To evaluate the theranostic value, we used GRP78-targeting AAVP particles to deliver the human Herpes simplex virus thymidine kinase type-1 (HSVtk) transgene, obtaining simultaneous in vivo diagnosis through PET imaging and tumor treatment by selective activation of the prodrug ganciclovir at tumor sites. Translation of this AAVP system is expected simultaneously to image, monitor, and treat the IBC phenotype and possibly other aggressive (e.g., invasive and/or metastatic) subtypes of breast cancer, based on the inducible cell-surface expression of the stress-response chaperone GRP78, and possibily other cell-surface receptors in human tumors.
Collapse
|
41
|
Targeted molecular-genetic imaging and ligand-directed therapy in aggressive variant prostate cancer. Proc Natl Acad Sci U S A 2016; 113:12786-12791. [PMID: 27791181 DOI: 10.1073/pnas.1615400113] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Aggressive variant prostate cancers (AVPC) are a clinically defined group of tumors of heterogeneous morphologies, characterized by poor patient survival and for which limited diagnostic and treatment options are currently available. We show that the cell surface 78-kDa glucose-regulated protein (GRP78), a receptor that binds to phage-display-selected ligands, such as the SNTRVAP motif, is a candidate target in AVPC. We report the presence and accessibility of this receptor in clinical specimens from index patients. We also demonstrate that human AVPC cells displaying GRP78 on their surface could be effectively targeted both in vitro and in vivo by SNTRVAP, which also enabled specific delivery of siRNA species to tumor xenografts in mice. Finally, we evaluated ligand-directed strategies based on SNTRVAP-displaying adeno-associated virus/phage (AAVP) particles in mice bearing MDA-PCa-118b, a patient-derived xenograft (PDX) of castration-resistant prostate cancer bone metastasis that we exploited as a model of AVPC. For theranostic (a merging of the terms therapeutic and diagnostic) studies, GRP78-targeting AAVP particles served to deliver the human Herpes simplex virus thymidine kinase type-1 (HSVtk) gene, which has a dual function as a molecular-genetic sensor/reporter and a cell suicide-inducing transgene. We observed specific and simultaneous PET imaging and treatment of tumors in this preclinical model of AVPC. Our findings demonstrate the feasibility of GPR78-targeting, ligand-directed theranostics for translational applications in AVPC.
Collapse
|
42
|
Barbu EM, Cady KC, Hubby B. Phage Therapy in the Era of Synthetic Biology. Cold Spring Harb Perspect Biol 2016; 8:cshperspect.a023879. [PMID: 27481531 DOI: 10.1101/cshperspect.a023879] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
For more than a century, bacteriophage (or phage) research has enabled some of the most important discoveries in biological sciences and has equipped scientists with many of the molecular biology tools that have advanced our understanding of replication, maintenance, and expression of genetic material. Phages have also been recognized and exploited as natural antimicrobial agents and nanovectors for gene therapy, but their potential as therapeutics has not been fully exploited in Western medicine because of challenges such as narrow host range, bacterial resistance, and unique pharmacokinetics. However, increasing concern related to the emergence of bacteria resistant to multiple antibiotics has heightened interest in phage therapy and the development of strategies to overcome hurdles associated with bacteriophage therapeutics. Recent progress in sequencing technologies, DNA manipulation, and synthetic biology allowed scientists to refactor the entire bacterial genome of Mycoplasma mycoides, thereby creating the first synthetic cell. These new strategies for engineering genomes may have the potential to accelerate the construction of designer phage genomes with superior therapeutic potential. Here, we discuss the use of phage as therapeutics, as well as how synthetic biology can create bacteriophage with desirable attributes.
Collapse
Affiliation(s)
| | - Kyle C Cady
- Synthetic Genomics, La Jolla, California 92037
| | - Bolyn Hubby
- Synthetic Genomics, La Jolla, California 92037
| |
Collapse
|
43
|
LeBlanc AK, Mazcko C, Brown DE, Koehler JW, Miller AD, Miller CR, Bentley RT, Packer RA, Breen M, Boudreau CE, Levine JM, Simpson RM, Halsey C, Kisseberth W, Rossmeisl JH, Dickinson PJ, Fan TM, Corps K, Aldape K, Puduvalli V, Pluhar GE, Gilbert MR. Creation of an NCI comparative brain tumor consortium: informing the translation of new knowledge from canine to human brain tumor patients. Neuro Oncol 2016; 18:1209-18. [PMID: 27179361 PMCID: PMC4999002 DOI: 10.1093/neuonc/now051] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 02/27/2016] [Indexed: 12/14/2022] Open
Abstract
On September 14-15, 2015, a meeting of clinicians and investigators in the fields of veterinary and human neuro-oncology, clinical trials, neuropathology, and drug development was convened at the National Institutes of Health campus in Bethesda, Maryland. This meeting served as the inaugural event launching a new consortium focused on improving the knowledge, development of, and access to naturally occurring canine brain cancer, specifically glioma, as a model for human disease. Within the meeting, a SWOT (strengths, weaknesses, opportunities, and threats) assessment was undertaken to critically evaluate the role that naturally occurring canine brain tumors could have in advancing this aspect of comparative oncology aimed at improving outcomes for dogs and human beings. A summary of this meeting and subsequent discussion are provided to inform the scientific and clinical community of the potential for this initiative. Canine and human comparisons represent an unprecedented opportunity to complement conventional brain tumor research paradigms, addressing a devastating disease for which innovative diagnostic and treatment strategies are clearly needed.
Collapse
Affiliation(s)
- Amy K LeBlanc
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - Christina Mazcko
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - Diane E Brown
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - Jennifer W Koehler
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - Andrew D Miller
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - C Ryan Miller
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - R Timothy Bentley
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - Rebecca A Packer
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - Matthew Breen
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - C Elizabeth Boudreau
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - Jonathan M Levine
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - R Mark Simpson
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - Charles Halsey
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - William Kisseberth
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - John H Rossmeisl
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - Peter J Dickinson
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - Timothy M Fan
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - Kara Corps
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - Kenneth Aldape
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - Vinay Puduvalli
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - G Elizabeth Pluhar
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - Mark R Gilbert
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| |
Collapse
|
44
|
Chimeric adeno-associated virus and bacteriophage: a potential targeted gene therapy vector for malignant glioma. Ther Deliv 2016; 5:975-90. [PMID: 25375341 DOI: 10.4155/tde.14.58] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The incipient development of gene therapy for cancer has fuelled its progression from bench to bedside in mere decades. Of all malignancies that exist, gliomas are the largest class of brain tumors, and are renowned for their aggressiveness and resistance to therapy. In order for gene therapy to achieve clinical success, a multitude of barriers ranging from glioma tumor physiology to vector biology must be overcome. Many viral gene delivery systems have been subjected to clinical investigation; however, with highly limited success. In this review, the current progress and challenges of gene therapy for malignant glioma are discussed. Moreover, we highlight the hybrid adeno-associated virus and bacteriophage vector as a potential candidate for targeted gene delivery to brain tumors.
Collapse
|
45
|
AAVP displaying octreotide for ligand-directed therapeutic transgene delivery in neuroendocrine tumors of the pancreas. Proc Natl Acad Sci U S A 2016; 113:2466-71. [PMID: 26884209 DOI: 10.1073/pnas.1525709113] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Patients with inoperable or unresectable pancreatic neuroendocrine tumors (NETs) have limited treatment options. These rare human tumors often express somatostatin receptors (SSTRs) and thus are clinically responsive to certain relatively stable somatostatin analogs, such as octreotide. Unfortunately, however, this tumor response is generally short-lived. Here we designed a hybrid adeno-associated virus and phage (AAVP) vector displaying biologically active octreotide on the viral surface for ligand-directed delivery, cell internalization, and transduction of an apoptosis-promoting tumor necrosis factor (TNF) transgene specifically to NETs. These functional attributes of AAVP-TNF particles displaying the octreotide peptide motif (termed Oct-AAVP-TNF) were confirmed in vitro, in SSTR type 2-expressing NET cells, and in vivo using cohorts of pancreatic NET-bearing Men1 tumor-suppressor gene KO mice, a transgenic model of functioning (i.e., insulin-secreting) tumors that genetically and clinically recapitulates the human disease. Finally, preclinical imaging and therapeutic experiments with pancreatic NET-bearing mice demonstrated that Oct-AAVP-TNF lowered tumor metabolism and insulin secretion, reduced tumor size, and improved mouse survival. Taken together, these proof-of-concept results establish Oct-AAVP-TNF as a strong therapeutic candidate for patients with NETs of the pancreas. More broadly, the demonstration that a known, short, biologically active motif can direct tumor targeting and receptor-mediated internalization of AAVP particles may streamline the potential utility of myriad other short peptide motifs and provide a blueprint for therapeutic applications in a variety of cancers and perhaps many nonmalignant diseases as well.
Collapse
|
46
|
Saba C, Paoloni M, Mazcko C, Kisseberth W, Burton JH, Smith A, Wilson-Robles H, Allstadt S, Vail D, Henry C, Lana S, Ehrhart EJ, Charles B, Kent M, Lawrence J, Burgess K, Borgatti A, Suter S, Woods P, Gordon I, Vrignaud P, Khanna C, LeBlanc AK. A Comparative Oncology Study of Iniparib Defines Its Pharmacokinetic Profile and Biological Activity in a Naturally-Occurring Canine Cancer Model. PLoS One 2016; 11:e0149194. [PMID: 26866698 PMCID: PMC4751284 DOI: 10.1371/journal.pone.0149194] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 01/28/2016] [Indexed: 12/25/2022] Open
Abstract
Development of iniparib as an anti-cancer agent was hindered in part by lingering questions regarding its mechanism of action, the activity of its metabolites, and their potential accumulation in tumors. Due to strong similarities in metabolism of iniparib between humans and dogs, a veterinary clinical trial in pet dogs with spontaneous cancers was designed to answer specific questions pertaining to pharmacokinetic exposures and tolerability of iniparib. Dogs were treated with iniparib alone and in combination with carboplatin chemotherapy. Iniparib doses ranged between 10-70 mg/kg intravenously (IV). Plasma, tumor and normal tissue samples were collected before and at various time points scheduled after exposure for pharmacokinetic and biologic analysis. The primary endpoints included characterization of dose-limiting toxicities (DLT) and determination of the drug exposures that could be achieved in both normal and tumor tissues. Nineteen dogs were treated. DLT included fever, anorexia, diarrhea, neutropenia, and thrombocytopenia; most effects were attributable to carboplatin based on the timing of adverse event onset. The maximum tolerated dose (MTD) of iniparib was not identified. Moderate to high variability in plasma exposure was noted for iniparib and all metabolites between animals. When quantifiable, iniparib and metabolite plasma:tumor ratios were < 0.088 and <1.7, respectively. In this study, iniparib was well tolerated as a single agent and in combination with carboplatin over a range of doses. However, clinically relevant concentrations of the parent drug and selected metabolites were not detectable in canine tumor tissues at any studied dose, thus eliminating expectations for clinical responses in dogs or humans. Negative clinical trials in humans, and the uncertainties of its mechanism of action, ultimately led to the decision to stop clinical development of the drug. Nevertheless, the questions that can be asked and answered within the comparative oncology approach are evident from this successfully executed comparative clinical trial and exemplify the value of such studies in drug development.
Collapse
Affiliation(s)
- Corey Saba
- College of Veterinary Medicine, University of Georgia, Athens, Georgia, United States of America
| | - Melissa Paoloni
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Christina Mazcko
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - William Kisseberth
- College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Jenna H. Burton
- College of Veterinary Medicine and Biological Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - Annette Smith
- College of Veterinary Medicine, Auburn University, Auburn, Alabama, United States of America
| | - Heather Wilson-Robles
- College of Veterinary Medicine, Texas A&M University, College Station, Texas, United States of America
| | - Sara Allstadt
- School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - David Vail
- School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Carolyn Henry
- College of Veterinary Medicine, University of Missouri-Columbia, Columbia, Missouri, United States of America
| | - Susan Lana
- College of Veterinary Medicine and Biological Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - E. J. Ehrhart
- College of Veterinary Medicine and Biological Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - Brad Charles
- College of Veterinary Medicine and Biological Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - Michael Kent
- School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - Jessica Lawrence
- College of Veterinary Medicine, University of Georgia, Athens, Georgia, United States of America
| | - Kristine Burgess
- School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, United States of America
| | - Antonella Borgatti
- College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Steve Suter
- College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Paul Woods
- Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Ira Gordon
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | | | - Chand Khanna
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Amy K. LeBlanc
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
47
|
Self-targeting of TNF-releasing cancer cells in preclinical models of primary and metastatic tumors. Proc Natl Acad Sci U S A 2016; 113:2223-8. [PMID: 26858439 DOI: 10.1073/pnas.1525697113] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Circulating cancer cells can putatively colonize distant organs to form metastases or to reinfiltrate primary tumors themselves through a process termed "tumor self-seeding." Here we exploit this biological attribute to deliver tumor necrosis factor alpha (TNF), a potent antitumor cytokine, directly to primary and metastatic tumors in a mechanism that we have defined as "tumor self-targeting." For this purpose, we genetically engineered mouse mammary adenocarcinoma (TSA), melanoma (B16-F10), and Lewis lung carcinoma cells to produce and release murine TNF. In a series of intervention trials, systemic administration of TNF-expressing tumor cells was associated with reduced growth of both primary tumors and metastatic colonies in immunocompetent mice. We show that these malignant cells home to tumors, locally release TNF, damage neovascular endothelium, and induce massive cancer cell apoptosis. We also demonstrate that such tumor-cell-mediated delivery avoids or minimizes common side effects often associated with TNF-based therapy, such as acute inflammation and weight loss. Our study provides proof of concept that genetically modified circulating tumor cells may serve as targeted vectors to deliver anticancer agents. In a clinical context, this unique paradigm represents a personalized approach to be translated into applications potentially using patient-derived circulating tumor cells as self-targeted vectors for drug delivery.
Collapse
|
48
|
Dervisis N, Klahn S. Therapeutic Innovations: Tyrosine Kinase Inhibitors in Cancer. Vet Sci 2016; 3:vetsci3010004. [PMID: 29056714 PMCID: PMC5644617 DOI: 10.3390/vetsci3010004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 01/12/2016] [Accepted: 01/18/2016] [Indexed: 01/06/2023] Open
Abstract
Conventional cytotoxic chemotherapy involving DNA-interacting agents and indiscriminate cell death is no longer the future of cancer management. While chemotherapy is not likely to completely disappear from the armamentarium; the use of targeted therapies in combination with conventional treatment is becoming the standard of care in human medicine. Tyrosine kinases are pivotal points of functional cellular pathways and have been implicated in malignancy, inflammatory, and immune-mediated diseases. Pharmaceutical interventions targeting aberrant tyrosine kinase signaling has exploded and is the second most important area of drug development. The “Valley of Death” between drug discovery and approval threatens to blunt the enormous strides in cancer management seen thus far. Kinase inhibitors, as targeted small molecules, hold promise in the treatment and diagnosis of cancer. However, there are still many unanswered questions regarding the use of kinase inhibitors in the interpretation and management of cancer. Comparative oncology has the potential to address restrictions and limitations in the advancement in kinase inhibitor therapy.
Collapse
Affiliation(s)
- Nikolaos Dervisis
- Virginia Maryland College of Veterinary Medicine, 245 Duck Pond Dr., Blacksburg, VA 24061, USA.
| | - Shawna Klahn
- Virginia Maryland College of Veterinary Medicine, 245 Duck Pond Dr., Blacksburg, VA 24061, USA.
| |
Collapse
|
49
|
LeBlanc AK, Mazcko CN, Khanna C. Defining the Value of a Comparative Approach to Cancer Drug Development. Clin Cancer Res 2015; 22:2133-8. [PMID: 26712689 DOI: 10.1158/1078-0432.ccr-15-2347] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 12/02/2015] [Indexed: 12/24/2022]
Abstract
Comparative oncology as a tool in drug development requires a deeper examination of the value of the approach and examples of where this approach can satisfy unmet needs. This review seeks to demonstrate types of drug development questions that are best answered by the comparative oncology approach. We believe common perceived risks of the comparative approach relate to uncertainty of how regulatory bodies will prioritize or react to data generated from these unique studies conducted in diseased animals, and how these new data will affect ongoing human clinical trials. We contend that it is reasonable to consider these data as potentially informative and valuable to cancer drug development, but as supplementary to conventional preclinical studies and human clinical trials particularly as they relate to the identification of drug-associated adverse events. Clin Cancer Res; 22(9); 2133-8. ©2015 AACR.
Collapse
Affiliation(s)
- Amy K LeBlanc
- Comparative Oncology Program, Center for Cancer Research, NCI, NIH, Bethesda, Maryland.
| | - Christina N Mazcko
- Comparative Oncology Program, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Chand Khanna
- Comparative Oncology Program, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| |
Collapse
|
50
|
Richards KL, Suter SE. Man's best friend: what can pet dogs teach us about non-Hodgkin's lymphoma? Immunol Rev 2015; 263:173-91. [PMID: 25510277 DOI: 10.1111/imr.12238] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Animal models are essential for understanding lymphoma biology and testing new treatments prior to human studies. Spontaneously arising lymphomas in pet dogs represent an underutilized resource that could be used to complement current mouse lymphoma models, which do not adequately represent all aspects of the human disease. Canine lymphoma resembles human lymphoma in many important ways, including characteristic translocations and molecular abnormalities and similar therapeutic responses to chemotherapy, radiation, and newer targeted therapies (e.g. ibrutinib). Given the large number of pet dogs and high incidence of lymphoma, particularly in susceptible breeds, dogs represent a largely untapped resource for advancing the understanding and treatment of human lymphoma. This review highlights similarities in molecular biology, diagnosis, treatment, and outcomes between human and canine lymphoma. It also describes resources that are currently available to study canine lymphoma, advantages to be gained by exploiting the genetic breed structure in dogs, and current and future challenges and opportunities to take full advantage of this resource for lymphoma studies.
Collapse
Affiliation(s)
- Kristy L Richards
- Center for Comparative Medicine and Translational Research, North Carolina State University, Raleigh, NC, USA; Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA; Division of Hematology/Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | |
Collapse
|