1
|
Dinda R, Garribba E, Sanna D, Crans DC, Costa Pessoa J. Hydrolysis, Ligand Exchange, and Redox Properties of Vanadium Compounds: Implications of Solution Transformation on Biological, Therapeutic, and Environmental Applications. Chem Rev 2025; 125:1468-1603. [PMID: 39818783 DOI: 10.1021/acs.chemrev.4c00475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Vanadium is a transition metal with important industrial, technological, biological, and biomedical applications widespread in the environment and in living beings. The different reactions that vanadium compounds (VCs) undergo in the presence of proteins, nucleic acids, lipids and metabolites under mild physiological conditions are reviewed. In the environment vanadium is present naturally or through anthropogenic sources, the latter having an environmental impact caused by the dispersion of VCs in the atmosphere and aquifers. Vanadium has a versatile chemistry with interconvertible oxidation states, variable coordination number and geometry, and ability to form polyoxidovanadates with various nuclearity and structures. If a VC is added to a water-containing environment it can undergo hydrolysis, ligand-exchange, redox, and other types of changes, determined by the conditions and speciation chemistry of vanadium. Importantly, the solution is likely to differ from the VC introduced into the system and varies with concentration. Here, vanadium redox, hydrolytic and ligand-exchange chemical reactions, the influence of pH, concentration, salt, specific solutes, biomolecules, and VCs on the speciation are described. One of our goals with this work is highlight the need for assessment of the VC speciation, so that beneficial or toxic species might be identified and mechanisms of action be elucidated.
Collapse
Affiliation(s)
- Rupam Dinda
- Department of Chemistry, National Institute of Technology, Rourkela, 769008 Odisha, India
| | - Eugenio Garribba
- Dipartimento di Medicina, Chirurgia e Farmacia, Università di Sassari, Viale San Pietro, I-07100 Sassari, Italy
| | - Daniele Sanna
- Istituto di Chimica Biomolecolare, Consiglio Nazionale delle Ricerche, Trav. La Crucca 3, I-07040 Sassari, Italy
| | - Debbie C Crans
- Department Chemistry and Cell and Molecular Biology, Colorado State University, Fort Collins, Colorado 80523, United States
| | - João Costa Pessoa
- Centro de Química Estrutural and Departamento de Engenharia Química, Institute of Molecular Sciences, Instituto Superior Técnico, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| |
Collapse
|
2
|
Alade A, Persad M, Bitar G, Dragan A, Fotiadis N, Shur J, Wong KH, Ng-Cheng-Hin B, Paleri V, Harrington K, Dafydd DA. A Review of Contemporary Image Guidance Techniques in Head and Neck Cancer. Head Neck 2025; 47:394-399. [PMID: 39503156 DOI: 10.1002/hed.27968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 10/04/2024] [Indexed: 12/13/2024] Open
Abstract
BACKGROUND Traditional head and neck cancer treatment involves open surgery, cytotoxic chemotherapy, and conventional radiotherapy planning. Emerging techniques aim to improve precision and reduce associated toxicity and functional impairment in current practice. This review article describes four such adaptations in image guidance, tailored to next generation therapies. METHODS This is a review of current literature, including feasibility studies from our cancer center, relating to: saline-aided intra-oral ultrasound-guided retropharyngeal biopsy; intra-oral ultrasound guided trans-oral robotic surgery (TORS); ultrasound-guided injection of "directly injected therapies"; and magnetic resonance imaging-guided radiotherapy. RESULTS Presented within the context of the wider literature, initial local experience and data indicate good technical outcomes and patient tolerance, and low technical complications in all four image guidance techniques. CONCLUSION Initial findings suggest a potentially important future role for these four image guidance techniques, on which next generation therapies are reliant. The broader implications on cross-disciplinary collaboration are also explored herein.
Collapse
Affiliation(s)
- Adebayo Alade
- Radiology Department, The Royal Marsden Hospital, London, UK
- Surgical Department, The Royal Marsden Hospital, London, UK
- Oncology Department, The Royal Marsden Hospital, London, UK
| | - Melissa Persad
- Radiology Department, The Royal Marsden Hospital, London, UK
- Surgical Department, The Royal Marsden Hospital, London, UK
- Oncology Department, The Royal Marsden Hospital, London, UK
| | - George Bitar
- Radiology Department, The Royal Marsden Hospital, London, UK
- Surgical Department, The Royal Marsden Hospital, London, UK
- Oncology Department, The Royal Marsden Hospital, London, UK
| | - Alina Dragan
- Radiology Department, The Royal Marsden Hospital, London, UK
- Surgical Department, The Royal Marsden Hospital, London, UK
- Oncology Department, The Royal Marsden Hospital, London, UK
| | - Nicos Fotiadis
- Radiology Department, The Royal Marsden Hospital, London, UK
- Surgical Department, The Royal Marsden Hospital, London, UK
- Oncology Department, The Royal Marsden Hospital, London, UK
| | - Joshua Shur
- Radiology Department, The Royal Marsden Hospital, London, UK
- Surgical Department, The Royal Marsden Hospital, London, UK
- Oncology Department, The Royal Marsden Hospital, London, UK
| | - Kee Howe Wong
- Radiology Department, The Royal Marsden Hospital, London, UK
- Surgical Department, The Royal Marsden Hospital, London, UK
- Oncology Department, The Royal Marsden Hospital, London, UK
| | - Brian Ng-Cheng-Hin
- Radiology Department, The Royal Marsden Hospital, London, UK
- Surgical Department, The Royal Marsden Hospital, London, UK
- Oncology Department, The Royal Marsden Hospital, London, UK
| | - Vinidh Paleri
- Radiology Department, The Royal Marsden Hospital, London, UK
- Surgical Department, The Royal Marsden Hospital, London, UK
- Oncology Department, The Royal Marsden Hospital, London, UK
| | - Kevin Harrington
- Radiology Department, The Royal Marsden Hospital, London, UK
- Surgical Department, The Royal Marsden Hospital, London, UK
- Oncology Department, The Royal Marsden Hospital, London, UK
| | - Derfel Ap Dafydd
- Radiology Department, The Royal Marsden Hospital, London, UK
- Surgical Department, The Royal Marsden Hospital, London, UK
- Oncology Department, The Royal Marsden Hospital, London, UK
| |
Collapse
|
3
|
Bitar GG, Persad M, Dragan A, Alade A, Jiménez-Labaig P, Johnston E, Withey SJ, Fotiadis N, Harrington KJ, Ap Dafydd D. Ultrasound-guided intra-tumoral administration of directly-injected therapies: a review of the technical and logistical considerations. Cancer Imaging 2024; 24:145. [PMID: 39456110 PMCID: PMC11515368 DOI: 10.1186/s40644-024-00763-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 08/18/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Directly-injected therapies (DIT) include a broad range of agents within a developing research field in cancer immunotherapy, with encouraging clinical trial results in various tumour subtypes. Currently, the majority of such therapies are only available within clinical trials; however, more recently, talimogene laherparepvec (T-VEC, Imlygic) has been approved as the first oncolytic virus therapy in the USA and Europe. Our institution contributes to multiple different trials exploring the efficacy of DIT, the majority of which are performed by oncologists in clinic. However, specific, challenging cases - mainly neck tumours - require image-guided administration. MAIN BODY This review article addresses the technical and logistical factors relevant to the incorporation of image-guided DIT into an established ultrasound service. Image-guidance (usually with ultrasound) is frequently needed for certain targets that cannot be palpated or are in high-risk locations, e.g. adjacent to blood vessels. A multi-disciplinary approach is essential to facilitate a safe and efficient service, including careful case-selection. Certain protocols and guidance need to be followed when incorporating such a service into an established ultrasound practice to enhance efficiency and optimise safety. Key learning points are drawn from the literature and from our early experience at a tertiary cancer centre following image guided DIT for an initial cohort of 22 patients (including 11 with a neck mass), addressing trial protocols, pre-procedure work-up, organisation, planning, consent, technical aspects, procedure tolerability, technical success, and post-procedure considerations. CONCLUSION With appropriate planning and coordination, and application of the learning points discussed herein, image-guided administration of DIT can be safely and efficiently incorporated into an established procedural ultrasound list. This has relevance to cancer centres, radiology departments, individual radiologists, and other team members with a future role in meeting the emerging need for these procedures. This paper provides advice on developing such an imaging service, and offers certain insights into the evolving remit of radiologists within cancer care in the near future.
Collapse
Affiliation(s)
| | | | - Alina Dragan
- The Royal Marsden NHS Foundation Trust, Fulham Road, London, UK
| | - Adebayo Alade
- The Royal Marsden NHS Foundation Trust, Fulham Road, London, UK
| | - Pablo Jiménez-Labaig
- The Royal Marsden NHS Foundation Trust, Fulham Road, London, UK
- The Institute of Cancer Research, Fulham Road, London, UK
| | - Edward Johnston
- The Royal Marsden NHS Foundation Trust, Fulham Road, London, UK
- The Institute of Cancer Research, Fulham Road, London, UK
| | - Samuel J Withey
- The Royal Marsden NHS Foundation Trust, Fulham Road, London, UK
| | - Nicos Fotiadis
- The Royal Marsden NHS Foundation Trust, Fulham Road, London, UK
- The Institute of Cancer Research, Fulham Road, London, UK
| | - Kevin J Harrington
- The Royal Marsden NHS Foundation Trust, Fulham Road, London, UK
- The Institute of Cancer Research, Fulham Road, London, UK
| | - Derfel Ap Dafydd
- The Royal Marsden NHS Foundation Trust, Fulham Road, London, UK.
| |
Collapse
|
4
|
Cetin B, Erendor F, Eksi YE, Sanlioglu AD, Sanlioglu S. Gene and cell therapy of human genetic diseases: Recent advances and future directions. J Cell Mol Med 2024; 28:e70056. [PMID: 39245805 PMCID: PMC11381193 DOI: 10.1111/jcmm.70056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 08/20/2024] [Indexed: 09/10/2024] Open
Abstract
Disruptions in normal development and the emergence of health conditions often result from the malfunction of vital genes in the human body. Decades of scientific research have focused on techniques to modify or substitute defective genes with healthy alternatives, marking a new era in disease treatment, prevention and cure. Recent strides in science and technology have reshaped our understanding of disorders, medication development and treatment recommendations, with human gene and cell therapy at the forefront of this transformative shift. Its primary objective is the modification of genes or adjustment of cell behaviour for therapeutic purposes. In this review, we focus on the latest advances in gene and cell therapy for treating human genetic diseases, with a particular emphasis on FDA and EMA-approved therapies and the evolving landscape of genome editing. We examine the current state of innovative gene editing technologies, particularly the CRISPR-Cas systems. As we explore the progress, ethical considerations and prospects of these innovations, we gain insight into their potential to revolutionize the treatment of genetic diseases, along with a discussion of the challenges associated with their regulatory pathways. This review traces the origins and evolution of these therapies, from conceptual ideas to practical clinical applications, marking a significant milestone in the field of medical science.
Collapse
Affiliation(s)
- Busra Cetin
- Department of Gene and Cell Therapy, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Fulya Erendor
- Department of Gene and Cell Therapy, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Yunus E Eksi
- Department of Gene and Cell Therapy, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Ahter D Sanlioglu
- Department of Gene and Cell Therapy, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Salih Sanlioglu
- Department of Gene and Cell Therapy, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| |
Collapse
|
5
|
Chen YS, Jin E, Day PJ. Use of Drug Sensitisers to Improve Therapeutic Index in Cancer. Pharmaceutics 2024; 16:928. [PMID: 39065625 PMCID: PMC11279903 DOI: 10.3390/pharmaceutics16070928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/04/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
The clinical management of malignant tumours is challenging, often leading to severe adverse effects and death. Drug resistance (DR) antagonises the effectiveness of treatments, and increasing drug dosage can worsen the therapeutic index (TI). Current efforts to overcome DR predominantly involve the use of drug combinations, including applying multiple anti-cancerous drugs, employing drug sensitisers, which are chemical agents that enhance pharmacokinetics (PK), including the targeting of cellular pathways and regulating pertinent membrane transporters. While combining multiple compounds may lead to drug-drug interactions (DDI) or polypharmacy effect, the use of drug sensitisers permits rapid attainment of effective treatment dosages at the disease site to prevent early DR and minimise side effects and will reduce the chance of DDI as lower drug doses are required. This review highlights the essential use of TI in evaluating drug dosage for cancer treatment and discusses the lack of a unified standard for TI within the field. Commonly used benefit-risk assessment criteria are summarised, and the critical exploration of the current use of TI in the pharmaceutical industrial sector is included. Specifically, this review leads to the discussion of drug sensitisers to facilitate improved ratios of effective dose to toxic dose directly in humans. The combination of drug and sensitiser molecules might see additional benefits to rekindle those drugs that failed late-stage clinical trials by the removal of detrimental off-target activities through the use of lower drug doses. Drug combinations and employing drug sensitisers are potential means to combat DR. The evolution of drug combinations and polypharmacy on TI are reviewed. Notably, the novel binary weapon approach is introduced as a new opportunity to improve TI. This review emphasises the urgent need for a criterion to systematically evaluate drug safety and efficiency for practical implementation in the field.
Collapse
Affiliation(s)
- Yu-Shan Chen
- Division of Evolution, Infection and Genomics, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, UK; (Y.-S.C.); (E.J.)
| | - Enhui Jin
- Division of Evolution, Infection and Genomics, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, UK; (Y.-S.C.); (E.J.)
| | - Philip J. Day
- Division of Evolution, Infection and Genomics, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, UK; (Y.-S.C.); (E.J.)
- Department of Medicine, University of Cape Town, Cape Town 7925, South Africa
| |
Collapse
|
6
|
Fu J, Feng Y, Sun Y, Yi R, Tian J, Zhao W, Sun D, Zhang C. A Multi-Drug Concentration Gradient Mixing Chip: A Novel Platform for High-Throughput Drug Combination Screening. BIOSENSORS 2024; 14:212. [PMID: 38785686 PMCID: PMC11117479 DOI: 10.3390/bios14050212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 04/21/2024] [Accepted: 04/22/2024] [Indexed: 05/25/2024]
Abstract
Combinatorial drug therapy has emerged as a critically important strategy in medical research and patient treatment and involves the use of multiple drugs in concert to achieve a synergistic effect. This approach can enhance therapeutic efficacy while simultaneously mitigating adverse side effects. However, the process of identifying optimal drug combinations, including their compositions and dosages, is often a complex, costly, and time-intensive endeavor. To surmount these hurdles, we propose a novel microfluidic device capable of simultaneously generating multiple drug concentration gradients across an interlinked array of culture chambers. This innovative setup allows for the real-time monitoring of live cell responses. With minimal effort, researchers can now explore the concentration-dependent effects of single-agent and combination drug therapies. Taking neural stem cells (NSCs) as a case study, we examined the impacts of various growth factors-epithelial growth factor (EGF), platelet-derived growth factor (PDGF), and fibroblast growth factor (FGF)-on the differentiation of NSCs. Our findings indicate that an overdose of any single growth factor leads to an upsurge in the proportion of differentiated NSCs. Interestingly, the regulatory effects of these growth factors can be modulated by the introduction of additional growth factors, whether singly or in combination. Notably, a reduced concentration of these additional factors resulted in a decreased number of differentiated NSCs. Our results affirm that the successful application of this microfluidic device for the generation of multi-drug concentration gradients has substantial potential to revolutionize drug combination screening. This advancement promises to streamline the process and accelerate the discovery of effective therapeutic drug combinations.
Collapse
Affiliation(s)
- Jiahao Fu
- State Key Laboratory of Photon-Technology in Western China Energy, Institute of Photonics and Photon-Technology, Northwest University, Xi’an 710127, China
| | - Yibo Feng
- State Key Laboratory of Photon-Technology in Western China Energy, Institute of Photonics and Photon-Technology, Northwest University, Xi’an 710127, China
| | - Yu Sun
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi’an 710127, China (R.Y.)
| | - Ruiya Yi
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi’an 710127, China (R.Y.)
| | - Jing Tian
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi’an 710127, China (R.Y.)
- Huaxin Microfish Biotechnology Co., Ltd., Taicang 215400, China
- Center for Automated and Innovative Drug Discovery, Northwest University, Xi’an 710127, China
| | - Wei Zhao
- State Key Laboratory of Photon-Technology in Western China Energy, Institute of Photonics and Photon-Technology, Northwest University, Xi’an 710127, China
| | - Dan Sun
- State Key Laboratory of Photon-Technology in Western China Energy, Institute of Photonics and Photon-Technology, Northwest University, Xi’an 710127, China
- Huaxin Microfish Biotechnology Co., Ltd., Taicang 215400, China
- Center for Automated and Innovative Drug Discovery, Northwest University, Xi’an 710127, China
| | - Ce Zhang
- State Key Laboratory of Photon-Technology in Western China Energy, Institute of Photonics and Photon-Technology, Northwest University, Xi’an 710127, China
- Huaxin Microfish Biotechnology Co., Ltd., Taicang 215400, China
| |
Collapse
|
7
|
Ghasemi Darestani N, Gilmanova AI, Al-Gazally ME, Zekiy AO, Ansari MJ, Zabibah RS, Jawad MA, Al-Shalah SAJ, Rizaev JA, Alnassar YS, Mohammed NM, Mustafa YF, Darvishi M, Akhavan-Sigari R. Mesenchymal stem cell-released oncolytic virus: an innovative strategy for cancer treatment. Cell Commun Signal 2023; 21:43. [PMID: 36829187 PMCID: PMC9960453 DOI: 10.1186/s12964-022-01012-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 12/10/2022] [Indexed: 02/26/2023] Open
Abstract
Oncolytic viruses (OVs) infect, multiply, and finally remove tumor cells selectively, causing no damage to normal cells in the process. Because of their specific features, such as, the ability to induce immunogenic cell death and to contain curative transgenes in their genomes, OVs have attracted attention as candidates to be utilized in cooperation with immunotherapies for cancer treatment. This treatment takes advantage of most tumor cells' inherent tendency to be infected by certain OVs and both innate and adaptive immune responses are elicited by OV infection and oncolysis. OVs can also modulate tumor microenvironment and boost anti-tumor immune responses. Mesenchymal stem cells (MSC) are gathering interest as promising anti-cancer treatments with the ability to address a wide range of cancers. MSCs exhibit tumor-trophic migration characteristics, allowing them to be used as delivery vehicles for successful, targeted treatment of isolated tumors and metastatic malignancies. Preclinical and clinical research were reviewed in this study to discuss using MSC-released OVs as a novel method for the treatment of cancer. Video Abstract.
Collapse
Affiliation(s)
| | - Anna I Gilmanova
- Department of Prosthetic Dentistry of the I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russian Federation
| | | | - Angelina O Zekiy
- Department of Prosthetic Dentistry of the I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russian Federation
| | - Mohammad Javed Ansari
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Rahman S Zabibah
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | | | - Saif A J Al-Shalah
- Medical Laboratories Techniques Department, Al-Mustaqbal University College, Babylon, Iraq
| | - Jasur Alimdjanovich Rizaev
- Department of Public Health and Healthcare Management, Rector, Samarkand State Medical University, Samarkand, Uzbekistan
| | | | | | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, 41001, Iraq
| | - Mohammad Darvishi
- Department of Aerospace and Subaquatic Medicine, Infectious Diseases and Tropical Medicine Research Center (IDTMRC), AJA University of Medical Sciences, Tehran, Iran.
| | - Reza Akhavan-Sigari
- Department of Neurosurgery, University Medical Center, Tuebingen, Germany.,Department of Health Care Management and Clinical Research, Collegium Humanum Warsaw Management University, Warsaw, Poland
| |
Collapse
|
8
|
Alluqmani N, Jirovec A, Taha Z, Varette O, Chen A, Serrano D, Maznyi G, Khan S, Forbes NE, Arulanandam R, Auer RC, Diallo JS. Vanadyl sulfate-enhanced oncolytic virus immunotherapy mediates the antitumor immune response by upregulating the secretion of pro-inflammatory cytokines and chemokines. Front Immunol 2022; 13:1032356. [PMID: 36532027 PMCID: PMC9749062 DOI: 10.3389/fimmu.2022.1032356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/09/2022] [Indexed: 11/29/2022] Open
Abstract
Oncolytic viruses (OVs) are promising anticancer treatments that specifically replicate in and kill cancer cells and have profound immunostimulatory effects. We previously reported the potential of vanadium-based compounds such as vanadyl sulfate (VS) as immunostimulatory enhancers of OV immunotherapy. These compounds, in conjunction with RNA-based OVs such as oncolytic vesicular stomatitis virus (VSVΔ51), improve viral spread and oncolysis, leading to long-term antitumor immunity and prolonged survival in resistant tumor models. This effect is associated with a virus-induced antiviral type I IFN response shifting towards a type II IFN response in the presence of vanadium. Here, we investigated the systemic impact of VS+VSVΔ51 combination therapy to understand the immunological mechanism of action leading to improved antitumor responses. VS+VSVΔ51 combination therapy significantly increased the levels of IFN-γ and IL-6, and improved tumor antigen-specific T-cell responses. Supported by immunological profiling and as a proof of concept for the design of more effective therapeutic regimens, we found that local delivery of IL-12 using VSVΔ51 in combination with VS further improved therapeutic outcomes in a syngeneic CT26WT colon cancer model.
Collapse
Affiliation(s)
- Nouf Alluqmani
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON, Canada,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, ON, Canada,Research Center, Molecular Oncology Department King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Anna Jirovec
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON, Canada,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, ON, Canada
| | - Zaid Taha
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON, Canada,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, ON, Canada
| | - Oliver Varette
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Andrew Chen
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Daniel Serrano
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Glib Maznyi
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Sarwat Khan
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Nicole E. Forbes
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Rozanne Arulanandam
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Rebecca C. Auer
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON, Canada,Department of Surgery, University of Ottawa, Ottawa, ON, Canada
| | - Jean-Simon Diallo
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON, Canada,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, ON, Canada,*Correspondence: Jean-Simon Diallo,
| |
Collapse
|
9
|
Abstract
Glioblastoma is the most aggressive primary brain tumor with a poor prognosis. The 2021 WHO CNS5 classification has further stressed the importance of molecular signatures in diagnosis although therapeutic breakthroughs are still lacking. In this review article, updates on the current and novel therapies in IDH-wildtype GBM will be discussed.
Collapse
Affiliation(s)
- Jawad M Melhem
- Division of Neurology, Department of Medicine, Faculty of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
| | - Jay Detsky
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, Canada
| | - Mary Jane Lim-Fat
- Division of Neurology, Department of Medicine, Faculty of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
| | - James R Perry
- Division of Neurology, Department of Medicine, Faculty of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada.
| |
Collapse
|
10
|
Cassidy T, Humphries AR. A mathematical model of viral oncology as an immuno-oncology instigator. MATHEMATICAL MEDICINE AND BIOLOGY-A JOURNAL OF THE IMA 2021; 37:117-151. [PMID: 31329873 DOI: 10.1093/imammb/dqz008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 02/15/2019] [Accepted: 03/26/2019] [Indexed: 12/14/2022]
Abstract
We develop and analyse a mathematical model of tumour-immune interaction that explicitly incorporates heterogeneity in tumour cell cycle duration by using a distributed delay differential equation. We derive a necessary and sufficient condition for local stability of the cancer-free equilibrium in which the amount of tumour-immune interaction completely characterizes disease progression. Consistent with the immunoediting hypothesis, we show that decreasing tumour-immune interaction leads to tumour expansion. Finally, by simulating the mathematical model, we show that the strength of tumour-immune interaction determines the long-term success or failure of viral therapy.
Collapse
Affiliation(s)
- Tyler Cassidy
- Department of Mathematics and Statistics, McGill University, Montreal, Canada
| | - Antony R Humphries
- Department of Mathematics and Statistics, McGill University, Montreal, Canada.,Department of Physiology, McGill University, Montreal, Canada
| |
Collapse
|
11
|
Piper K, DePledge L, Karsy M, Cobbs C. Glioma Stem Cells as Immunotherapeutic Targets: Advancements and Challenges. Front Oncol 2021; 11:615704. [PMID: 33718170 PMCID: PMC7945033 DOI: 10.3389/fonc.2021.615704] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 01/07/2021] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma is the most common and lethal primary brain malignancy. Despite major investments in research into glioblastoma biology and drug development, treatment remains limited and survival has not substantially improved beyond 1-2 years. Cancer stem cells (CSC) or glioma stem cells (GSC) refer to a population of tumor originating cells capable of self-renewal and differentiation. While controversial and challenging to study, evidence suggests that GCSs may result in glioblastoma tumor recurrence and resistance to treatment. Multiple treatment strategies have been suggested at targeting GCSs, including immunotherapy, posttranscriptional regulation, modulation of the tumor microenvironment, and epigenetic modulation. In this review, we discuss recent advances in glioblastoma treatment specifically focused on targeting of GCSs as well as their potential integration into current clinical pathways and trials.
Collapse
Affiliation(s)
- Keenan Piper
- Ben & Catherine Ivy Center for Advanced Brain Tumor Treatment, Swedish Neuroscience Institute, Seattle, WA, United States.,Sidney Kimmel Medical College, Philadelphia, PA, United States
| | - Lisa DePledge
- Ben & Catherine Ivy Center for Advanced Brain Tumor Treatment, Swedish Neuroscience Institute, Seattle, WA, United States.,University of Washington School of Medicine, Spokane, WA, United States
| | - Michael Karsy
- Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, PA, United States
| | - Charles Cobbs
- Ben & Catherine Ivy Center for Advanced Brain Tumor Treatment, Swedish Neuroscience Institute, Seattle, WA, United States
| |
Collapse
|
12
|
Rodríguez Stewart RM, Raghuram V, Berry JTL, Joshi GN, Mainou BA. Noncanonical Cell Death Induction by Reassortant Reovirus. J Virol 2020; 94:e01613-20. [PMID: 32847857 PMCID: PMC7592226 DOI: 10.1128/jvi.01613-20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 08/15/2020] [Indexed: 12/12/2022] Open
Abstract
Triple-negative breast cancer (TNBC) constitutes 10 to 15% of all breast cancer and is associated with worse prognosis than other subtypes of breast cancer. Current therapies are limited to cytotoxic chemotherapy, radiation, and surgery, leaving a need for targeted therapeutics to improve outcomes for TNBC patients. Mammalian orthoreovirus (reovirus) is a nonenveloped, segmented, double-stranded RNA virus in the Reoviridae family. Reovirus preferentially kills transformed cells and is in clinical trials to assess its efficacy against several types of cancer. We previously engineered a reassortant reovirus, r2Reovirus, that infects TNBC cells more efficiently and induces cell death with faster kinetics than parental reoviruses. In this study, we sought to understand the mechanisms by which r2Reovirus induces cell death in TNBC cells. We show that r2Reovirus infection of TNBC cells of a mesenchymal stem-like (MSL) lineage downregulates the mitogen-activated protein kinase/extracellular signal-related kinase pathway and induces nonconventional cell death that is caspase-dependent but caspase 3-independent. Infection of different MSL lineage TNBC cells with r2Reovirus results in caspase 3-dependent cell death. We map the enhanced oncolytic properties of r2Reovirus in TNBC to epistatic interactions between the type 3 Dearing M2 gene segment and type 1 Lang genes. These findings suggest that the genetic composition of the host cell impacts the mechanism of reovirus-induced cell death in TNBC. Together, our data show that understanding host and virus determinants of cell death can identify novel properties and interactions between host and viral gene products that can be exploited for the development of improved viral oncolytics.IMPORTANCE TNBC is unresponsive to hormone therapies, leaving patients afflicted with this disease with limited treatment options. We previously engineered an oncolytic reovirus (r2Reovirus) with enhanced infective and cytotoxic properties in TNBC cells. However, how r2Reovirus promotes TNBC cell death is not known. In this study, we show that reassortant r2Reovirus can promote nonconventional caspase-dependent but caspase 3-independent cell death and that the mechanism of cell death depends on the genetic composition of the host cell. We also map the enhanced oncolytic properties of r2Reovirus in TNBC to interactions between a type 3 M2 gene segment and type 1 genes. Our data show that understanding the interplay between the host cell environment and the genetic composition of oncolytic viruses is crucial for the development of efficacious viral oncolytics.
Collapse
Affiliation(s)
- Roxana M Rodríguez Stewart
- Emory University, Atlanta, Georgia, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | | | - Jameson T L Berry
- Emory University, Atlanta, Georgia, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | | | - Bernardo A Mainou
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
- Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| |
Collapse
|
13
|
Cai L, Liu Z. Novel recombinant coxsackievirus B3 with genetically inserted basic peptide elicits robust antitumor activity against lung cancer. Cancer Med 2020; 9:5210-5220. [PMID: 32459400 PMCID: PMC7367620 DOI: 10.1002/cam4.3143] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 04/01/2020] [Accepted: 04/24/2020] [Indexed: 12/19/2022] Open
Abstract
Cancer therapy that utilizes oncolytic virus may offer an exciting alternative, and coxsackievirus B3 (CVB3) is a potent oncolytic virus. This study was to assess the oncolytic activities of novel recombinant CVB3 with genetically inserted basic peptides in lung cancer. Recombinant CVB3 was produced in Vero cells, with or without genetically inserted basic peptides. In vitro and in vivo experiments with nude mouse models bearing human lung carcinoma xenografts were performed to examine the antitumor activities. Cytokines and immune responses to the recombinant CVB3 were determined in cynomolgus monkeys. Recombinant CVB3 with genetically inserted basic peptides was associated with significantly higher pH values within tumors. Mice treated with recombinant CVB3 showed significantly less tumor progression, and recombinant CVB3 with genetically inserted basic peptides appeared to enhance tumor suppression. Recombinant CVB3 was associated with significantly less proliferation of various lung cancer cells without affecting proliferation of normal lung fibroblasts. The cytokine profiles of the cynomolgus monkeys were comparable among control group (normal saline solution) and those given recombinant CVB3 with or without fused basic peptides, with no induction of excessive cytokine or immune responses. In conclusions, recombinant CVB3, especially those with fused basic peptides, possess strong antitumor activities without eliciting excessive immune responses.
Collapse
Affiliation(s)
- Ligang Cai
- Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhiyi Liu
- Wuhan Boweid Biotechnology Co., Ltd., Wuhan, Hubei, China
| |
Collapse
|
14
|
Ji W, Li L, Zhou S, Qiu L, Qian Z, Zhang H, Zhao P. Combination immunotherapy of oncolytic virus nanovesicles and PD-1 blockade effectively enhances therapeutic effects and boosts antitumour immune response. J Drug Target 2020; 28:982-990. [PMID: 32379004 DOI: 10.1080/1061186x.2020.1766473] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Immunotherapies are changing the landscape of melanoma treatment, but 70% of the melanoma patients have no response to immune checkpoint inhibitors or oncolytic virus therapy. Thus, novel formulations are needed to improve the population benefiting from immunotherapy. Here, we report a combined therapeutic modality based on oncolytic virus nanovesicles composed of CaCl2, oncolytic virus Ad5, lecithin and cholesterol (Lipo-Cap-Ad5) with immune checkpoint blockade (anti-PD-1 antibody). We investigated in vivo antitumour activity, systemic toxicity and mechanism of antitumour immune responses of Lipo-Cap-Ad5 + anti-PD-1 blockade, in a murine B16F10 tumour xenograft model. Through a series of in vivo studies, we found that Lipo-Cap-Ad5 in combination with anti-PD-1 blockade drastically reduced the tumour growth by 76.6%, and prolonged animals' survival with no obvious toxicity observed in heart, liver and kidney. The combination therapy facilitates tumour infiltration of effector CD4+, CD8+ T cells and increases secretion of TNF-α and IFN-γ. Therefore, Lipo-Cap-Ad5 in combination with anti-PD-1 blockade can potentiate and activate the immune system synergistically, ultimately creating a pro-inflammatory environment. These results suggest that combination immunotherapy of Lipo-Cap-Ad5 and anti-PD-1 blockade developed in this study has promising applications to enhance therapeutic efficacy with the potential of being translated into clinical practice.
Collapse
Affiliation(s)
- Wei Ji
- Public Laboratory, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Lanfang Li
- Department of Lymphoma, Key Laboratory of Cancer Prevention and Therapy, Sino-US Center for Lymphoma and Leukemia, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Shiyong Zhou
- Department of Lymphoma, Key Laboratory of Cancer Prevention and Therapy, Sino-US Center for Lymphoma and Leukemia, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Lihua Qiu
- Department of Lymphoma, Key Laboratory of Cancer Prevention and Therapy, Sino-US Center for Lymphoma and Leukemia, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Zhengzi Qian
- Department of Lymphoma, Key Laboratory of Cancer Prevention and Therapy, Sino-US Center for Lymphoma and Leukemia, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Huilai Zhang
- Department of Lymphoma, Key Laboratory of Cancer Prevention and Therapy, Sino-US Center for Lymphoma and Leukemia, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Peiqi Zhao
- Department of Lymphoma, Key Laboratory of Cancer Prevention and Therapy, Sino-US Center for Lymphoma and Leukemia, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
15
|
Alayo QA, Ito H, Passaro C, Zdioruk M, Mahmoud AB, Grauwet K, Zhang X, Lawler SE, Reardon DA, Goins WF, Fernandez S, Chiocca EA, Nakashima H. Glioblastoma infiltration of both tumor- and virus-antigen specific cytotoxic T cells correlates with experimental virotherapy responses. Sci Rep 2020; 10:5095. [PMID: 32198420 PMCID: PMC7083912 DOI: 10.1038/s41598-020-61736-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 02/20/2020] [Indexed: 02/05/2023] Open
Abstract
The mode of action for oncolytic viruses (OVs) in cancer treatment is thought to depend on a direct initial cytotoxic effect against infected tumor cells and subsequent activation of immune cell responses directed against the neoplasm. To study both of these effects in a mouse model of glioblastoma (GBM), we employed murine GBM cells engineered to constitutively express the type I Herpes Simplex Virus (HSV1) HSV-1 receptor, nectin-1, to allow for more efficient infection and replication by oncolytic HSV (oHSV). These cells were further engineered with a surrogate tumor antigen to facilitate assays of T cell activity. We utilized MRI-based volumetrics to measure GBM responses after injection with the oHSV and bioluminescent imaging (BLI) to determine oHSV replicative kinetics in the injected tumor mass. We found increased infiltration of both surrogate tumor antigen- and oHSV antigen-specific CD8+ T cells within 7 days after oHSV injection. There was no increase in tumor infiltrating CD8+ T cells expressing “exhaustion” markers, yet oHSV infection led to a reduction in PD-1+ CD8+ T cells in injected GBMs and an increase in IFNγ+ CD8+ T cells. There was a significant direct correlation between oHSV-mediated reduction in GBM volume and increased infiltration of both viral and tumor antigen-specific CD8+ T cells, as well as oHSV intratumoral gene activity. These findings imply that CD8+ T cell cytotoxicity against both tumor and viral antigens as well as intratumoral oHSV gene expression are important in oHSV-mediated GBM therapy.
Collapse
Affiliation(s)
- Quazim A Alayo
- Harvey W. Cushing Neuro-oncology Laboratories (HCNL), Department of Neurosurgery, Harvard Medical School and Brigham and Women's Hospital, 02115, Boston, MA, USA
| | - Hirotaka Ito
- Harvey W. Cushing Neuro-oncology Laboratories (HCNL), Department of Neurosurgery, Harvard Medical School and Brigham and Women's Hospital, 02115, Boston, MA, USA
| | - Carmela Passaro
- Harvey W. Cushing Neuro-oncology Laboratories (HCNL), Department of Neurosurgery, Harvard Medical School and Brigham and Women's Hospital, 02115, Boston, MA, USA
| | - Mykola Zdioruk
- Harvey W. Cushing Neuro-oncology Laboratories (HCNL), Department of Neurosurgery, Harvard Medical School and Brigham and Women's Hospital, 02115, Boston, MA, USA
| | - Ahmad Bakur Mahmoud
- Harvey W. Cushing Neuro-oncology Laboratories (HCNL), Department of Neurosurgery, Harvard Medical School and Brigham and Women's Hospital, 02115, Boston, MA, USA.,College of Applied Medical Sciences, Taibah University, 42353, Madinah, Saudi Arabia
| | - Korneel Grauwet
- Harvey W. Cushing Neuro-oncology Laboratories (HCNL), Department of Neurosurgery, Harvard Medical School and Brigham and Women's Hospital, 02115, Boston, MA, USA.,Cancer Center and Department of Medicine, Massachusetts General Hospital, Boston, 02114, MA, USA
| | - Xiaoli Zhang
- Center for Biostatistics, Department of Biomedical Informatics, The Ohio State University, 43210, Columbus, OH, USA
| | - Sean E Lawler
- Harvey W. Cushing Neuro-oncology Laboratories (HCNL), Department of Neurosurgery, Harvard Medical School and Brigham and Women's Hospital, 02115, Boston, MA, USA
| | - David A Reardon
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, 02115, Boston, MA, USA
| | - William F Goins
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, 15219, Pittsburgh, PA, USA
| | - Soledad Fernandez
- Center for Biostatistics, Department of Biomedical Informatics, The Ohio State University, 43210, Columbus, OH, USA
| | - E Antonio Chiocca
- Harvey W. Cushing Neuro-oncology Laboratories (HCNL), Department of Neurosurgery, Harvard Medical School and Brigham and Women's Hospital, 02115, Boston, MA, USA.
| | - Hiroshi Nakashima
- Harvey W. Cushing Neuro-oncology Laboratories (HCNL), Department of Neurosurgery, Harvard Medical School and Brigham and Women's Hospital, 02115, Boston, MA, USA.
| |
Collapse
|
16
|
Cassidy T, Craig M. Determinants of combination GM-CSF immunotherapy and oncolytic virotherapy success identified through in silico treatment personalization. PLoS Comput Biol 2019; 15:e1007495. [PMID: 31774808 PMCID: PMC6880985 DOI: 10.1371/journal.pcbi.1007495] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 10/20/2019] [Indexed: 12/15/2022] Open
Abstract
Oncolytic virotherapies, including the modified herpes simplex virus talimogene laherparepvec (T-VEC), have shown great promise as potent instigators of anti-tumour immune effects. The OPTiM trial, in particular, demonstrated the superior anti-cancer effects of T-VEC as compared to systemic immunotherapy treatment using exogenous administration of granulocyte-macrophage colony-stimulating factor (GM-CSF). Theoretically, a combined approach leveraging exogenous cytokine immunotherapy and oncolytic virotherapy would elicit an even greater immune response and improve patient outcomes. However, regimen scheduling of combination immunostimulation and T-VEC therapy has yet to be established. Here, we calibrate a computational biology model of sensitive and resistant tumour cells and immune interactions for implementation into an in silico clinical trial to test and individualize combination immuno- and virotherapy. By personalizing and optimizing combination oncolytic virotherapy and immunostimulatory therapy, we show improved simulated patient outcomes for individuals with late-stage melanoma. More crucially, through evaluation of individualized regimens, we identified determinants of combination GM-CSF and T-VEC therapy that can be translated into clinically-actionable dosing strategies without further personalization. Our results serve as a proof-of-concept for interdisciplinary approaches to determining combination therapy, and suggest promising avenues of investigation towards tailored combination immunotherapy/oncolytic virotherapy. The advent of biological therapies for anti-cancer treatment has had a significant impact on patient outcomes. Targeted xenobiotics, including oncolytic viruses, in combination with existing, more general, immunotherapies like exogenous cytokines show great promise for continuing to improve cancer care. However, determining optimal combination regimens can be difficult, given that testing proposed schedules would require large cohorts of patients enrolled in clinical trials. Fortunately, computational biology can help to address treatment scheduling while simultaneously helping to unravel the mechanisms driving therapeutic responses. In this work, we integrate a mathematical model of GM-CSF and talimogene laherparepvec (T-VEC) oncolytic virotherapy into a virtual clinical trial to optimize their administration in combination. Using this platform, we inferred a clinically-actionable combination schedule for patients with late-stage melanoma that significantly improved virtual patient outcome when compared to GM-CSF and T-VEC monotherapies, and a standard combination strategy. Our results outline a rational approach to therapy optimization with meaningful consequences for how we effectively design and implement clinical trials to maximize their success, and how we treat melanoma with combined immuno- and virotherapy.
Collapse
Affiliation(s)
- Tyler Cassidy
- Department of Mathematics and Statistics, McGill University, Montreal, Quebec, Canada
| | - Morgan Craig
- Département de mathématiques et de statistique, Université de Montréal, Montreal, Quebec, Canada.,Department of Physiology, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
17
|
Geraldo LHM, Garcia C, da Fonseca ACC, Dubois LGF, de Sampaio e Spohr TCL, Matias D, de Camargo Magalhães ES, do Amaral RF, da Rosa BG, Grimaldi I, Leser FS, Janeiro JM, Macharia L, Wanjiru C, Pereira CM, Moura-Neto V, Freitas C, Lima FRS. Glioblastoma Therapy in the Age of Molecular Medicine. Trends Cancer 2019; 5:46-65. [DOI: 10.1016/j.trecan.2018.11.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 11/09/2018] [Accepted: 11/12/2018] [Indexed: 12/11/2022]
|
18
|
Chiocca EA, Nassiri F, Wang J, Peruzzi P, Zadeh G. Viral and other therapies for recurrent glioblastoma: is a 24-month durable response unusual? Neuro Oncol 2019; 21:14-25. [PMID: 30346600 PMCID: PMC6303472 DOI: 10.1093/neuonc/noy170] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
A phase I trial of an engineered poliovirus for the treatment of recurrent glioblastoma (GBM) has attracted attention due to 8 survivors reaching the 24-month and 5 reaching the 36-month survival landmarks.1 Genetically engineered viruses (oncolytic viruses) have been in trials for GBM for almost two decades.2 These replication-competent (tumor-selective, oncolytic, replication-conditional) viruses or replication-defective viral vectors (gene therapy) deliver cytotoxic payloads to tumors, leading to immunogenic death and intratumoral inflammatory responses. This transforms the tumor microenvironment from immunologically naïve ("cold") to inflamed ("hot"), increasing immune cell recognition of tumor antigens and the durable responses observed in virotherapy.3,4 Several current and past virotherapy trials have reported a "tail" of apparent responders at the 24-month landmark. Other modalities have also reported a "tail" of seemingly long-term survivors. These trials seem to show that these responder "tails" characterize a defined subset of GBM patients.
Collapse
Affiliation(s)
- E Antonio Chiocca
- Department of Neurosurgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Farshad Nassiri
- Division of Neurosurgery, University of Toronto, Toronto, Ontario, Canada
| | - Justin Wang
- Division of Neurosurgery, University of Toronto, Toronto, Ontario, Canada
| | - Pierpaolo Peruzzi
- Department of Neurosurgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Gelareh Zadeh
- Division of Neurosurgery, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
19
|
de Queiroz NMGP, Xia T, Konno H, Barber GN. Ovarian Cancer Cells Commonly Exhibit Defective STING Signaling Which Affects Sensitivity to Viral Oncolysis. Mol Cancer Res 2018; 17:974-986. [PMID: 30587523 DOI: 10.1158/1541-7786.mcr-18-0504] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 10/09/2018] [Accepted: 12/07/2018] [Indexed: 12/15/2022]
Abstract
Ovarian cancer is the sixth most prevalent cancer in women and the most lethal of the gynecologic malignancies. Treatments have comprised the use of immunotherapeutic agents as well as oncolytic viruses, with varying results for reasons that remain to be clarified. To better understand the mechanisms that may help predict treatment outcome, we have evaluated innate immune signaling in select ovarian cancer cell lines, governed by the Stimulator of Interferon Genes (STING), which controls self or viral DNA-triggered cytokine production. Our results indicate that STING-dependent signaling is habitually defective in majority of ovarian cancer cells examined, frequently through the suppression of STING and/or the cyclic dinucleotide (CDN) enzyme Cyclic GMP-AMP synthase (cGAS) expression, by epigenetic processes. However, STING-independent, dsRNA-activated innate immune cytokine production, which require RIG-I/MDA5, were largely unaffected. Such defects enabled ovarian cancer cells to avoid DNA damage-mediated cytokine production, which would alert the immunosurveillance system. Loss of STING signaling also rendered ovarian cancer cells highly susceptible to viral oncolytic γ34.5 deleted-HSV1 (Herpes simplex virus) infection in vitro and in vivo. IMPLICATIONS: STING signaling evaluation in tumors may help predict disease outcome and possibly dictate the efficacy of oncoviral and other types of cancer therapies.
Collapse
Affiliation(s)
- Nina Marí Gual Pimenta de Queiroz
- Department of Cell Biology and the Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
| | - Tianli Xia
- Department of Cell Biology and the Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
| | - Hiroyasu Konno
- Department of Cell Biology and the Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
| | - Glen N Barber
- Department of Cell Biology and the Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida.
| |
Collapse
|
20
|
Moyes KW, Davis A, Hoglund V, Haberthur K, Lieberman NA, Kreuser SA, Deutsch GH, Franco S, Locke D, Carleton MO, Gilbertson DG, Simmons R, Winter C, Silber J, Gonzalez-Cuyar LF, Ellenbogen RG, Crane CA. Effects of tumor grade and dexamethasone on myeloid cells in patients with glioma. Oncoimmunology 2018; 7:e1507668. [PMID: 30377570 PMCID: PMC6204983 DOI: 10.1080/2162402x.2018.1507668] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 07/22/2018] [Accepted: 07/29/2018] [Indexed: 12/14/2022] Open
Abstract
Efforts to reduce immunosuppression in the solid tumor microenvironment by blocking the recruitment or polarization of tumor associated macrophages (TAM), or myeloid derived suppressor cells (MDSCs), have gained momentum in recent years. Expanding our knowledge of the immune cell types, cytokines, or recruitment factors that are associated with high-grade disease, both within the tumor and in circulation, is critical to identifying novel targets for immunotherapy. Furthermore, a better understanding of how therapeutic regimens, such as Dexamethasone (Dex), chemotherapy, and radiation, impact these factors will facilitate the design of therapies that can be targeted to the appropriate populations and retain efficacy when administered in combination with standard of care regimens. Here we perform quantitative analysis of tissue microarrays made of samples taken from grades I-III astrocytoma and glioblastoma (GBM, grade IV astrocytoma) to evaluate infiltration of myeloid markers CD163, CD68, CD33, and S100A9. Serum, flow cytometric, and Nanostring analysis allowed us to further elucidate the impact of Dex treatment on systemic biomarkers, circulating cells, and functional markers within tumor tissue. We found that common myeloid markers were elevated in Dex-treated grade I astrocytoma and GBM compared to non-neoplastic brain tissue and grade II-III astrocytomas. Cell frequencies in these samples differed significantly from those in Dex-naïve patients in a pattern that depended on tumor grade. In contrast, observed changes in serum chemokines or circulating monocytes were independent of disease state and were due to Dex treatment alone. Furthermore, these changes seen in blood were often not reflected within the tumor tissue. Conclusions: Our findings highlight the importance of considering perioperative treatment as well as disease grade when assessing novel therapeutic targets or biomarkers of disease.
Collapse
Affiliation(s)
- Kara W Moyes
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Amira Davis
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Virginia Hoglund
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Kristen Haberthur
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Nicole Ap Lieberman
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Shannon A Kreuser
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Gail H Deutsch
- Department of Pathology, Seattle Children's Hospital, Seattle, WA, USA
| | - Stephanie Franco
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, WA, USA
| | | | | | | | | | - Conrad Winter
- Department of Pathology, Seattle Children's Hospital, Seattle, WA, USA
| | - John Silber
- Department of Neurological Surgery, University of Washington, Seattle WA, USA
| | | | | | - Courtney A Crane
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, WA, USA.,Department of Neurological Surgery, University of Washington, Seattle WA, USA
| |
Collapse
|
21
|
Jiang S, Hua L, Guo Z, Sun L. One-pot green synthesis of doxorubicin loaded-silica nanoparticles for in vivo cancer therapy. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 90:257-263. [PMID: 29853089 DOI: 10.1016/j.msec.2018.04.047] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 03/26/2018] [Accepted: 04/16/2018] [Indexed: 01/02/2023]
Abstract
The present work reveals a new and simple one-pot green method to load doxorubicin (DOX) drugs in silica nanoparticles for efficient in vivo cancer therapy. The synthesis of DOX loaded silica nanoparticles (SiNPs/DOX) is based on the efficient encapsulation of DOX in surfactant Tween 80 micelles which act as a template for the formation of silica nanoparticles. The release profile, cellular uptake behavior, cytotoxicity and antitumor effect of SiNPs/DOX nanoparticles were investigated and compared to free DOX. The silica nanoparticles improved the cellular drug delivery efficiency and exhibited high cytotoxicity, successfully achieving the inhibition of tumor growth. Notably, the tumor size and weight of SiNPs/DOX group was 2-fold and 1.7-fold smaller than that of free DOX group, and 4-fold and 2-fold smaller than that of PBS group. The one-pot green synthesis system may have the potential to be developed as a promising drug delivery system.
Collapse
Affiliation(s)
- Shan Jiang
- College of Chemistry, Jilin University, Changchun 130012, PR China
| | - Li Hua
- Department of Immunology, Norman Bethune College of Medicine, Jilin University, Changchun 130021, PR China
| | - Zilong Guo
- College of Chemistry, Jilin University, Changchun 130012, PR China
| | - Lin Sun
- College of Chemistry, Jilin University, Changchun 130012, PR China.
| |
Collapse
|
22
|
Li K, Liang J, Lin Y, Zhang H, Xiao X, Tan Y, Cai J, Zhu W, Xing F, Hu J, Yan G. A classical PKA inhibitor increases the oncolytic effect of M1 virus via activation of exchange protein directly activated by cAMP 1. Oncotarget 2018; 7:48443-48455. [PMID: 27374176 PMCID: PMC5217030 DOI: 10.18632/oncotarget.10305] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 06/09/2016] [Indexed: 11/25/2022] Open
Abstract
Oncolytic virotherapy is an emerging and promising treatment modality that uses replicating viruses as selective antitumor agents. Here, we report that a classical protein kinase A (PKA) inhibitor, H89, synergizes with oncolytic virus M1 in various cancer cells through activation of Epac1 (exchange protein directly activated by cAMP 1). H89 substantially increases viral replication in refractory cancer cells, leading to unresolvable Endoplasmic Reticulum stress, and cell apoptosis. Microarray analysis indicates that H89 blunts antiviral response in refractory cancer cells through retarding the nuclear translocation of NF-κB. Importantly, in vivo studies show significant antitumor effects during M1/H89 combination treatment. Overall, this study reveals a previously unappreciated role for H89 and demonstrates that activation of the Epac1 activity can improve the responsiveness of biotherapeutic agents for cancer.
Collapse
Affiliation(s)
- Kai Li
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Jiankai Liang
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Yuan Lin
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Haipeng Zhang
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Xiao Xiao
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China.,Department of Pharmacy, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Yaqian Tan
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Jing Cai
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Wenbo Zhu
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Fan Xing
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Jun Hu
- Department of Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Guangmei Yan
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China.,Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510080, China
| |
Collapse
|
23
|
Angelova AL, Barf M, Geletneky K, Unterberg A, Rommelaere J. Immunotherapeutic Potential of Oncolytic H-1 Parvovirus: Hints of Glioblastoma Microenvironment Conversion towards Immunogenicity. Viruses 2017; 9:v9120382. [PMID: 29244745 PMCID: PMC5744156 DOI: 10.3390/v9120382] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 12/08/2017] [Accepted: 12/11/2017] [Indexed: 12/21/2022] Open
Abstract
Glioblastoma, one of the most aggressive primary brain tumors, is characterized by highly immunosuppressive microenvironment. This contributes to glioblastoma resistance to standard treatment modalities and allows tumor growth and recurrence. Several immune-targeted approaches have been recently developed and are currently under preclinical and clinical investigation. Oncolytic viruses, including the autonomous protoparvovirus H-1 (H-1PV), show great promise as novel immunotherapeutic tools. In a first phase I/IIa clinical trial (ParvOryx01), H-1PV was safe and well tolerated when locally or systemically administered to recurrent glioblastoma patients. The virus was able to cross the blood-brain (tumor) barrier after intravenous infusion. Importantly, H-1PV treatment of glioblastoma patients was associated with immunogenic changes in the tumor microenvironment. Tumor infiltration with activated cytotoxic T cells, induction of cathepsin B and inducible nitric oxide (NO) synthase (iNOS) expression in tumor-associated microglia/macrophages (TAM), and accumulation of activated TAM in cluster of differentiation (CD) 40 ligand (CD40L)-positive glioblastoma regions was detected. These are the first-in-human observations of H-1PV capacity to switch the immunosuppressed tumor microenvironment towards immunogenicity. Based on this pilot study, we present a tentative model of H-1PV-mediated modulation of glioblastoma microenvironment and propose a combinatorial therapeutic approach taking advantage of H-1PV-induced microglia/macrophage activation for further (pre)clinical testing.
Collapse
Affiliation(s)
- Assia L Angelova
- Department of Tumor Virology (F010), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| | - Milena Barf
- Department of Tumor Virology (F010), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| | - Karsten Geletneky
- Department of Neurosurgery, University Hospital, 69120 Heidelberg, Germany.
| | - Andreas Unterberg
- Department of Neurosurgery, University Hospital, 69120 Heidelberg, Germany.
| | - Jean Rommelaere
- Department of Tumor Virology (F010), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| |
Collapse
|
24
|
Matsuda T, Karube H, Aruga A. A Comparative Safety Profile Assessment of Oncolytic Virus Therapy Based on Clinical Trials. Ther Innov Regul Sci 2017; 52:430-437. [PMID: 29714547 DOI: 10.1177/2168479017738979] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Oncolytic virus therapy (OVT) represents a new class of therapeutic agents in cancer treatment. The molecular and cellular mechanisms of action of OVTs have been evaluated in nonclinical/clinical phase trials. Various genetically modified viruses have been developed as oncolytic agents, and the first approval of an OVT for clinical use was issued by the US Food and Drug Administration in 2015. In this context, more and more clinical development of OVTs is anticipated in the future. This article provides a risk assessment of OVT based on the safety data obtained from all clinical trials to date using a publicly available database. The most common adverse events (AEs) observed in clinical trials have been infection-related symptoms such as fatigue, chills, fever, and nausea; few serious AEs have been observed, regardless of the kind of virus or transfected genes. In vivo systemic infusion of OVTs demonstrated a high percentage of AEs, but most AEs were manageable using common drugs. This paper describes OVTs' specific safety/toxicity profiles and encourages the performance of further clinical trials of OVTs to address the most serious challenges anticipated in the development of OVTs as a new class of drugs for the treatment of cancer.
Collapse
Affiliation(s)
- Takuma Matsuda
- Cooperative Major in Advanced Biomedical Sciences, Joint Graduate School of Tokyo Women's Medical University and Waseda University, Tokyo
| | - Hiroyo Karube
- Cooperative Major in Advanced Biomedical Sciences, Joint Graduate School of Tokyo Women's Medical University and Waseda University, Tokyo
| | - Atsushi Aruga
- Cooperative Major in Advanced Biomedical Sciences, Joint Graduate School of Tokyo Women's Medical University and Waseda University, Tokyo
| |
Collapse
|
25
|
Selman M, Rousso C, Bergeron A, Son HH, Krishnan R, El-Sayes NA, Varette O, Chen A, Le Boeuf F, Tzelepis F, Bell JC, Crans DC, Diallo JS. Multi-modal Potentiation of Oncolytic Virotherapy by Vanadium Compounds. Mol Ther 2017; 26:56-69. [PMID: 29175158 DOI: 10.1016/j.ymthe.2017.10.014] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 10/16/2017] [Accepted: 10/18/2017] [Indexed: 12/20/2022] Open
Abstract
Oncolytic viruses (OV) are an emerging class of anticancer bio-therapeutics that induce antitumor immunity through selective replication in tumor cells. However, the efficacy of OVs as single agents remains limited. We introduce a strategy that boosts the therapeutic efficacy of OVs by combining their activity with immuno-modulating, small molecule protein tyrosine phosphatase inhibitors. We report that vanadium-based phosphatase inhibitors enhance OV infection in vitro and ex vivo, in resistant tumor cell lines. Furthermore, vanadium compounds increase antitumor efficacy in combination with OV in several syngeneic tumor models, leading to systemic and durable responses, even in models otherwise refractory to OV and drug alone. Mechanistically, this involves subverting the antiviral type I IFN response toward a death-inducing and pro-inflammatory type II IFN response, leading to improved OV spread, increased bystander killing of cancer cells, and enhanced antitumor immune stimulation. Overall, we showcase a new ability of vanadium compounds to simultaneously maximize viral oncolysis and systemic anticancer immunity, offering new avenues for the development of improved immunotherapy strategies.
Collapse
Affiliation(s)
- Mohammed Selman
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, ON, Canada
| | - Christopher Rousso
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON, Canada; Faculty of Science, University of Ottawa, ON, Canada
| | - Anabel Bergeron
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON, Canada; Faculty of Science, University of Ottawa, ON, Canada
| | - Hwan Hee Son
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, ON, Canada
| | - Ramya Krishnan
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, ON, Canada
| | - Nader A El-Sayes
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, ON, Canada
| | - Oliver Varette
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, ON, Canada
| | - Andrew Chen
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Fabrice Le Boeuf
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Fanny Tzelepis
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - John C Bell
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, ON, Canada
| | - Debbie C Crans
- Department of Chemistry, Colorado State University, Fort Collins, CO, USA; Cell and Molecular Biology Program, Colorado State University, Fort Collins, CO, USA
| | - Jean-Simon Diallo
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, ON, Canada.
| |
Collapse
|
26
|
Stem cell-released oncolytic herpes simplex virus has therapeutic efficacy in brain metastatic melanomas. Proc Natl Acad Sci U S A 2017; 114:E6157-E6165. [PMID: 28710334 DOI: 10.1073/pnas.1700363114] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The recent Food and Drug Administration approval of immunogenic oncolytic virus (OV) has opened a new era in the treatment of advanced melanoma; however, approximately 50% of patients with melanoma develop brain metastasis, and currently there are no beneficial treatment options for such patients. To model the progression of metastases seen in patients and to overcome the hurdles of systemic delivery of OV, we developed melanoma brain metastasis models in immunocompromised and immunocompetent mice, and tested the fate and efficacy of oncolytic herpes simplex virus (oHSV)-armed mesenchymal stem cells (MSCs). Using brain-seeking patient-derived melanoma cells and real-time in vivo imaging, we show a widespread distribution of micrometastases and macrometastases in the brain, recapitulating the progression of multifoci metastases seen in patients. We armed MSCs with different oHSV variants (MSC-oHSV) and found that intracarotid administration of MSC-oHSV, but not of purified oHSV alone, effectively tracks metastatic tumor lesions and significantly prolongs the survival of brain tumor-bearing mice. In a syngeneic model of melanoma brain metastasis, a combination of MSC-oHSV and PD-L1 blockade increases IFNγ-producing CD8+ tumor-infiltrating T lymphocytes and results in a profound extension of the median survival of treated animals. This study thus demonstrates the utility of MSCs as OV carriers to disseminated brain lesions, and provides a clinically applicable therapeutic platform to target melanoma brain metastasis.
Collapse
|
27
|
Timalsina A, Tian JP, Wang J. Mathematical and Computational Modeling for Tumor Virotherapy with Mediated Immunity. Bull Math Biol 2017; 79:1736-1758. [PMID: 28593497 DOI: 10.1007/s11538-017-0304-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 05/26/2017] [Indexed: 11/30/2022]
Abstract
We propose a new mathematical modeling framework based on partial differential equations to study tumor virotherapy with mediated immunity. The model incorporates both innate and adaptive immune responses and represents the complex interaction among tumor cells, oncolytic viruses, and immune systems on a domain with a moving boundary. Using carefully designed computational methods, we conduct extensive numerical simulation to the model. The results allow us to examine tumor development under a wide range of settings and provide insight into several important aspects of the virotherapy, including the dependence of the efficacy on a few key parameters and the delay in the adaptive immunity. Our findings also suggest possible ways to improve the virotherapy for tumor treatment.
Collapse
Affiliation(s)
- Asim Timalsina
- Department of Mathematics and Statistics, Old Dominion University, Norfolk, VA, 23529, USA
| | - Jianjun Paul Tian
- Department of Mathematical Sciences, New Mexico State University, Las Cruces, NM, 88003, USA
| | - Jin Wang
- Department of Mathematics, University of Tennessee at Chattanooga, Chattanooga, TN, 37403, USA.
| |
Collapse
|
28
|
Surface modulatable nanocapsids for targeting and tracking toward nanotheranostic delivery. Pharm Pat Anal 2017; 5:307-17. [PMID: 27610752 DOI: 10.4155/ppa-2016-0021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Nanoparticle diagnostics and therapeutics (nanotheranostics) have significantly advanced cancer detection and treatment. However, many nanotheranostics are ineffective due to defects in tumor localization and bioavailability. An engineered Hepatitis E Virus (HEV) nanocapsid is a proposed platform for targeted cancer-cell delivery. Self-assembling from HEV capsid subunits, nanocapsids retain the capacity to enter cells and resist proteolytic/acidic conditions, but lack infectious viral elements. The nanocapsid surface was modified for chemical activation to confer tumor-specific targeting and detection, immune-response manipulation and controlled theranostic delivery. Nanotheranostic molecules can be packaged in the hollow nanocapsid shell during in vitro assembly. Complementing the adapted stability and cell-entry characteristics of the HEV capsid, a modified nanocapsid serves as a tunable tumor-targeting platform for nanotheronostic delivery.
Collapse
|
29
|
Speranza MC, Kasai K, Lawler SE. Preclinical Mouse Models for Analysis of the Therapeutic Potential of Engineered Oncolytic Herpes Viruses. ILAR J 2017; 57:63-72. [PMID: 27034396 DOI: 10.1093/ilar/ilw002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
After more than two decades of research and development, oncolytic herpes viruses (oHSVs) are moving into the spotlight due to recent encouraging clinical trial data. oHSV and other oncolytic viruses function through direct oncolytic cancer cell-killing mechanisms and by stimulating antitumor immunity. As further viruses are developed and optimized for the treatment of various types of cancer, appropriate predictive preclinical models will be of great utility. This review will discuss existing data in this area, focusing on the mouse tumor models that are commonly used.
Collapse
Affiliation(s)
- Maria-Carmela Speranza
- Maria-Carmela Speranza, PhD, is a post-doctoral fellow; Kazue Kasai, PhD, is a Research Specialist; and Sean E. Lawler, PhD, is an Assistant Professor in the Harvey Cushing Neurooncology Laboratories in the Department of Neurosurgery at Brigham and Women's Hospital, Harvard Medical School in Boston, Massachusetts
| | - Kazue Kasai
- Maria-Carmela Speranza, PhD, is a post-doctoral fellow; Kazue Kasai, PhD, is a Research Specialist; and Sean E. Lawler, PhD, is an Assistant Professor in the Harvey Cushing Neurooncology Laboratories in the Department of Neurosurgery at Brigham and Women's Hospital, Harvard Medical School in Boston, Massachusetts
| | - Sean E Lawler
- Maria-Carmela Speranza, PhD, is a post-doctoral fellow; Kazue Kasai, PhD, is a Research Specialist; and Sean E. Lawler, PhD, is an Assistant Professor in the Harvey Cushing Neurooncology Laboratories in the Department of Neurosurgery at Brigham and Women's Hospital, Harvard Medical School in Boston, Massachusetts
| |
Collapse
|
30
|
Hu L, Jiang K, Ding C, Meng S. Targeting Autophagy for Oncolytic Immunotherapy. Biomedicines 2017; 5:biomedicines5010005. [PMID: 28536348 PMCID: PMC5423490 DOI: 10.3390/biomedicines5010005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 12/22/2016] [Accepted: 01/04/2017] [Indexed: 12/20/2022] Open
Abstract
Oncolytic viruses (OVs) are capable of exerting anti-cancer effects by a variety of mechanisms, including immune-mediated tumor cell death, highlighting their potential use in immunotherapy. Several adaptation mechanisms such as autophagy contribute to OV-mediated anti-tumor properties. Autophagy regulates immunogenic signaling during cancer therapy which can be utilized to design therapeutic combinations using approaches that either induce or block autophagy to potentiate the therapeutic efficacy of OVs. In this article, we review the complicated interplay between autophagy, cancer, immunity, and OV, summarize recent progress in the contribution of OV-perturbed autophagy to oncolytic immunity, and discuss the challenges in targeting autophagy to enhance oncolytic immunotherapy.
Collapse
Affiliation(s)
- Lulu Hu
- Institute of Cancer Stem Cell, Dalian Medical University Cancer Center, 9Lvshun Road South, Dalian 116044, China.
| | - Ke Jiang
- Institute of Cancer Stem Cell, Dalian Medical University Cancer Center, 9Lvshun Road South, Dalian 116044, China.
| | - Chan Ding
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200000, China.
| | - Songshu Meng
- Institute of Cancer Stem Cell, Dalian Medical University Cancer Center, 9Lvshun Road South, Dalian 116044, China.
| |
Collapse
|
31
|
Lim M, Weller M, Chiocca EA. Current State of Immune-Based Therapies for Glioblastoma. Am Soc Clin Oncol Educ Book 2017; 35:e132-9. [PMID: 27249715 DOI: 10.1200/edbk_159084] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Glioblastoma is one of the most aggressive solid tumors, and, despite treatment options such as surgery, radiation, and chemotherapy, its prognosis remains grim. Novel approaches are needed to improve survival. Immunotherapy has proven efficacy for melanoma, lung cancer, and kidney cancer and is now a focus for glioblastoma. In this article, glioblastoma-mediated immunosuppression will be discussed and two exciting immune approaches, checkpoint inhibitors and viral-based therapies, will be reviewed.
Collapse
Affiliation(s)
- Michael Lim
- From The Johns Hopkins University, Baltimore, MD; University Hospital Zurich, Zurich, Switzerland; Institute for the Neurosciences at the Brigham and Women's/Faulkner Hospital, Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA; The University of Chicago, Chicago, IL
| | - Michael Weller
- From The Johns Hopkins University, Baltimore, MD; University Hospital Zurich, Zurich, Switzerland; Institute for the Neurosciences at the Brigham and Women's/Faulkner Hospital, Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA; The University of Chicago, Chicago, IL
| | - E Antonio Chiocca
- From The Johns Hopkins University, Baltimore, MD; University Hospital Zurich, Zurich, Switzerland; Institute for the Neurosciences at the Brigham and Women's/Faulkner Hospital, Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA; The University of Chicago, Chicago, IL
| |
Collapse
|
32
|
Abstract
Immunotherapy using dendritic cell (DC)-based vaccination is an approved approach for harnessing the potential of a patient's own immune system to eliminate tumor cells in metastatic hormone-refractory cancer. Overall, although many DC vaccines have been tested in the clinic and proven to be immunogenic, and in some cases associated with clinical outcome, there remains no consensus on how to manufacture DC vaccines. In this review we will discuss what has been learned thus far about human DC biology from clinical studies, and how current approaches to apply DC vaccines in the clinic could be improved to enhance anti-tumor immunity.
Collapse
|
33
|
Xia T, Konno H, Barber GN. Recurrent Loss of STING Signaling in Melanoma Correlates with Susceptibility to Viral Oncolysis. Cancer Res 2016; 76:6747-6759. [PMID: 27680683 DOI: 10.1158/0008-5472.can-16-1404] [Citation(s) in RCA: 248] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 08/29/2016] [Accepted: 09/08/2016] [Indexed: 11/16/2022]
Abstract
The innate immunoregulator STING stimulates cytokine production in response to the presence of cytosolic DNA, which can arise following DNA damage. Extrinsic STING signaling is also needed for antigen-presenting cells to stimulate antitumor T-cell immunity. Here, we show that STING signaling is recurrently suppressed in melanoma cells, where this event may enable immune escape after DNA damage. Mechanistically, STING signaling was suppressed most frequently by epigenetic silencing of either STING or the cyclic GMP-AMP synthase, which generates STING-activating cyclic dinucleotides after binding cytosolic DNA species. Loss of STING function rendered melanoma cells unable to produce type I IFN and other immune cytokines after exposure to cytosolic DNA species. Consequently, such cells were highly susceptible to infection with DNA viruses including HSV1, a variant of which is being developed presently as a therapeutic oncolytic virus [talimogene laherparepvec (T-VEC)]. Our findings provide insight into the basis for susceptibility to viral oncolysis by agents such as HSV1. Cancer Res; 76(22); 6747-59. ©2016 AACR.
Collapse
Affiliation(s)
- Tianli Xia
- Department of Cell Biology and the Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
| | - Hiroyasu Konno
- Department of Cell Biology and the Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
| | - Glen N Barber
- Department of Cell Biology and the Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida.
| |
Collapse
|