1
|
Calzetta L, Page C, Matera MG, Cazzola M, Rogliani P. Drug-Drug Interactions and Synergy: From Pharmacological Models to Clinical Application. Pharmacol Rev 2024; 76:1159-1220. [PMID: 39009470 DOI: 10.1124/pharmrev.124.000951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 07/08/2024] [Accepted: 07/11/2024] [Indexed: 07/17/2024] Open
Abstract
This review explores the concept of synergy in pharmacology, emphasizing its importance in optimizing treatment outcomes through the combination of drugs with different mechanisms of action. Synergy, defined as an effect greater than the expected additive effect elicited by individual agents according to specific predictive models, offers a promising approach to enhance therapeutic efficacy while minimizing adverse events. The historical evolution of synergy research, from ancient civilizations to modern pharmacology, highlights the ongoing quest to understand and harness synergistic interactions. Key concepts, such as concentration-response curves, additive effects, and predictive models, are discussed in detail, emphasizing the need for accurate assessment methods throughout translational drug development. Although various mathematical models exist for synergy analysis, selecting the appropriate model and software tools remains a challenge, necessitating careful consideration of experimental design and data interpretation. Furthermore, this review addresses practical considerations in synergy assessment, including preclinical and clinical approaches, mechanism of action, and statistical analysis. Optimizing synergy requires attention to concentration/dose ratios, target site localization, and timing of drug administration, ensuring that the benefits of combination therapy detected bench-side are translatable into clinical practice. Overall, the review advocates for a systematic approach to synergy assessment, incorporating robust statistical analysis, effective and simplified predictive models, and collaborative efforts across pivotal sectors, such as academic institutions, pharmaceutical companies, and regulatory agencies. By overcoming critical challenges and maximizing therapeutic potential, effective synergy assessment in drug development holds promise for advancing patient care. SIGNIFICANCE STATEMENT: Combining drugs with different mechanisms of action for synergistic interactions optimizes treatment efficacy and safety. Accurate interpretation of synergy requires the identification of the expected additive effect. Despite innovative models to predict the additive effect, consensus in drug-drug interactions research is lacking, hindering the bench-to-bedside development of combination therapies. Collaboration among science, industry, and regulation is crucial for advancing combination therapy development, ensuring rigorous application of predictive models in clinical settings.
Collapse
Affiliation(s)
- Luigino Calzetta
- Respiratory Disease and Lung Function Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy (L.C.); Pulmonary Pharmacology Unit, Institute of Pharmaceutical Science, King's College London, United Kingdom (C.P.); Unit of Pharmacology, Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy (M.G.-M.); and Respiratory Medicine Unit, Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy (M.C., P.R.)
| | - Clive Page
- Respiratory Disease and Lung Function Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy (L.C.); Pulmonary Pharmacology Unit, Institute of Pharmaceutical Science, King's College London, United Kingdom (C.P.); Unit of Pharmacology, Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy (M.G.-M.); and Respiratory Medicine Unit, Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy (M.C., P.R.)
| | - Maria Gabriella Matera
- Respiratory Disease and Lung Function Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy (L.C.); Pulmonary Pharmacology Unit, Institute of Pharmaceutical Science, King's College London, United Kingdom (C.P.); Unit of Pharmacology, Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy (M.G.-M.); and Respiratory Medicine Unit, Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy (M.C., P.R.)
| | - Mario Cazzola
- Respiratory Disease and Lung Function Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy (L.C.); Pulmonary Pharmacology Unit, Institute of Pharmaceutical Science, King's College London, United Kingdom (C.P.); Unit of Pharmacology, Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy (M.G.-M.); and Respiratory Medicine Unit, Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy (M.C., P.R.)
| | - Paola Rogliani
- Respiratory Disease and Lung Function Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy (L.C.); Pulmonary Pharmacology Unit, Institute of Pharmaceutical Science, King's College London, United Kingdom (C.P.); Unit of Pharmacology, Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy (M.G.-M.); and Respiratory Medicine Unit, Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy (M.C., P.R.)
| |
Collapse
|
2
|
Nguyen-Vo TH, Do TTT, Nguyen BP. Multitask Learning on Graph Convolutional Residual Neural Networks for Screening of Multitarget Anticancer Compounds. J Chem Inf Model 2024. [PMID: 39197175 DOI: 10.1021/acs.jcim.4c00643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2024]
Abstract
Recently, various modern experimental screening pipelines and assays have been developed to find promising anticancer drug candidates. However, it is time-consuming and almost infeasible to screen an immense number of compounds for anticancer activity via experimental approaches. To partially address this issue, several computational advances have been proposed. In this study, we present iACP-GCR, a model based on multitask learning on graph convolutional residual neural networks with two types of shortcut connections, to identify multitarget anticancer compounds. In our architecture, the graph convolutional residual neural networks are shared by all the prediction tasks before being separately customized. The NCI-60 data set, one of the most reliable and well-known sources of experimentally verified compounds, was used to develop our model. From that data set, we collected and refined data about compounds screened across nine cancer types (panels), including breast, central nervous system, colon, leukemia, nonsmall cell lung, melanoma, ovarian, prostate, and renal, for model training and evaluation. The model performance evaluated on an independent test set shows that iACP-GCR surpasses the three advanced computational methods for multitask learning. The integration of two shortcut connection types in the shared networks also improves the prediction efficiency. We also deployed the model as a public web server to assist the research community in screening potential anticancer compounds.
Collapse
Affiliation(s)
- Thanh-Hoang Nguyen-Vo
- Ho Chi Minh City Open University, 97 Vo Van Tan, District 3, Ho Chi Minh City 70000, Vietnam
| | - Trang T T Do
- Ho Chi Minh City Open University, 97 Vo Van Tan, District 3, Ho Chi Minh City 70000, Vietnam
| | - Binh P Nguyen
- Victoria University of Wellington, Kelburn Parade, Wellington 6012, New Zealand
| |
Collapse
|
3
|
Nathani A, Aare M, Sun L, Bagde A, Li Y, Rishi A, Singh M. Unlocking the Potential of Camel Milk-Derived Exosomes as Novel Delivery Systems: Enhanced Bioavailability of ARV-825 PROTAC for Cancer Therapy. Pharmaceutics 2024; 16:1070. [PMID: 39204415 PMCID: PMC11359469 DOI: 10.3390/pharmaceutics16081070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 08/06/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024] Open
Abstract
This study investigates the use of camel milk-derived exosomes (CMEs) as carriers for ARV-825, an anticancer agent targeting bromodomain-containing protein 4 (BRD4), in oral chemotherapy. CMEs were isolated and characterized, and ARV-825-loaded CME formulations were prepared and evaluated through various in vitro and in vivo tests. The ARV-825-CME formulation exhibited an entrapment efficiency of 42.75 ± 5.05%, a particle size of 136.8 ± 1.94 nm, and a zeta potential of -32.75 ± 0.70 mV, ensuring stability and sustained drug release. In vitro studies showed a 5.4-fold enhancement in drug release kinetics compared to the free ARV-825 solution. Permeability studies indicated a 3.2-fold increase in apparent permeability, suggesting improved cellular uptake. Cytotoxicity assays demonstrated potent anticancer activity, with IC50 values decreasing by 1.5 to 2-fold in cancer cell lines SF8628 DIPG and H1975R (resistant to Osimertinib). In vivo pharmacokinetic studies in Sprague-Dawley rats revealed superior systemic absorption and bioavailability of ARV-825 from CMEs, with a 2.55-fold increase in plasma concentration and a 5.56-fold increase in AUC. Distribution studies confirmed absorption through the ileum. This research highlights the potential of CMEs as a promising delivery platform for ARV-825, enhancing its therapeutic efficacy and offering a novel approach to cancer treatment.
Collapse
Affiliation(s)
- Aakash Nathani
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA; (A.N.); (M.A.); (A.B.)
| | - Mounika Aare
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA; (A.N.); (M.A.); (A.B.)
| | - Li Sun
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32310, USA; (L.S.); (Y.L.)
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32304, USA
| | - Arvind Bagde
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA; (A.N.); (M.A.); (A.B.)
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32310, USA; (L.S.); (Y.L.)
| | - Arun Rishi
- Department of Oncology, John D. Dingell VA Medical Center, Wayne State University School of Medicine, Detroit, MI 48201, USA;
| | - Mandip Singh
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA; (A.N.); (M.A.); (A.B.)
| |
Collapse
|
4
|
Summers KM. Genetic models of fibrillinopathies. Genetics 2024; 226:iyad189. [PMID: 37972149 PMCID: PMC11021029 DOI: 10.1093/genetics/iyad189] [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: 09/01/2023] [Accepted: 10/16/2023] [Indexed: 11/19/2023] Open
Abstract
The fibrillinopathies represent a group of diseases in which the 10-12 nm extracellular microfibrils are disrupted by genetic variants in one of the genes encoding fibrillin molecules, large glycoproteins of the extracellular matrix. The best-known fibrillinopathy is Marfan syndrome, an autosomal dominant condition affecting the cardiovascular, ocular, skeletal, and other systems, with a prevalence of around 1 in 3,000 across all ethnic groups. It is caused by variants of the FBN1 gene, encoding fibrillin-1, which interacts with elastin to provide strength and elasticity to connective tissues. A number of mouse models have been created in an attempt to replicate the human phenotype, although all have limitations. There are also natural bovine models and engineered models in pig and rabbit. Variants in FBN2 encoding fibrillin-2 cause congenital contractural arachnodactyly and mouse models for this condition have also been produced. In most animals, including birds, reptiles, and amphibians, there is a third fibrillin, fibrillin-3 (FBN3 gene) for which the creation of models has been difficult as the gene is degenerate and nonfunctional in mice and rats. Other eukaryotes such as the nematode C. elegans and zebrafish D. rerio have a gene with some homology to fibrillins and models have been used to discover more about the function of this family of proteins. This review looks at the phenotype, inheritance, and relevance of the various animal models for the different fibrillinopathies.
Collapse
Affiliation(s)
- Kim M Summers
- Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba QLD 4102, Australia
| |
Collapse
|
5
|
Patel P, Pal R, Butani K, Singh S, Prajapati BG. Nanomedicine-fortified cosmeceutical serums for the mitigation of psoriasis and acne. Nanomedicine (Lond) 2023; 18:1769-1793. [PMID: 37990979 DOI: 10.2217/nnm-2023-0147] [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] [Indexed: 11/23/2023] Open
Abstract
Cosmetics have a long history of use for regenerative and therapeutic purposes that are appealing to both genders. The untapped potential of nanotechnology in cosmeceuticals promises enhanced efficacy and addresses the issues associated with conventional cosmetics. In the field of cosmetics, the incorporation of nanomedicine using various nanocarriers such as vesicle and solid lipid nanoparticles significantly enhances product effectiveness and promotes satisfaction, especially in tackling prevalent skin diseases. Moreover, vesicle-fortified serum is known for high skin absorption with the capacity to incorporate and deliver various therapeutics. Additionally, nano-embedded serum-based cosmeceuticals hold promise for treating various skin disorders, including acne and psoriasis, heralding potential therapeutic advancements. This review explores diverse nanotechnology-based approaches for delivering cosmetics with maximum benefits.
Collapse
Affiliation(s)
- Priya Patel
- Department of Pharmaceutical Sciences, Saurashtra University, Rajkot, Gujarat, 360005, India
| | - Rohit Pal
- Department of Pharmaceutical Sciences, Saurashtra University, Rajkot, Gujarat, 360005, India
| | - Krishna Butani
- Department of Pharmaceutical Sciences, Saurashtra University, Rajkot, Gujarat, 360005, India
| | - Sudarshan Singh
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai, 50200, Thailand
- Office of Research Administration, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Bhupendra G Prajapati
- Department of Pharmaceutics & Pharmaceutical Technology, Shree S.K. Patel College of Pharmaceutical Education & Research, Ganpat University, Mehsana, Gujarat, 384012, India
| |
Collapse
|
6
|
Turner JH. Cancer Care by Committee to be Superseded by Personal Physician-Patient Partnership Informed by Artificial Intelligence. Cancer Biother Radiopharm 2023; 38:497-505. [PMID: 37366774 DOI: 10.1089/cbr.2023.0058] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2023] Open
Abstract
Multidisciplinary tumor boards (MTBs) have become the reference standard of cancer management, founded upon randomized controlled trial (RCT) evidence-based guidelines. The inordinate delays inherent in awaiting formal regulatory agency approvals of novel therapeutic agents, and the rigidities and nongeneralizability of this regimented approach, often deny cancer patients timely access to effective innovative treatment. Reluctance of MTBs to accept theranostic care of patients with advanced neuroendocrine tumors (NETs) and metastatic castrate-resistant prostate cancer resulted in decades of delay in the incorporation of 177Lu-octreotate and 177Lu-prostate-specific membrane antigen (PSMA) into routine clinical oncology practice. Recent developments in immunotherapy and molecular targeted precision therapy, based on N-of-One individual multifactorial genome analyses, have greatly increased the complexity of decision-making. Burgeoning specialist workload and tight time frames now threaten to overwhelm the logistically, and emotionally, demanding MTB system. It is hypothesized that the advent of advanced artificial intelligence technology and Chatbot natural language algorithms will shift the cancer care paradigm from a MTB management model toward a personal physician-patient shared-care partnership for real-world practice of precision individualized holistic oncology.
Collapse
Affiliation(s)
- J Harvey Turner
- Department of Nuclear Medicine, Fiona Stanley Fremantle Hospitals Group, The University of Western Australia, Murdoch, Australia
| |
Collapse
|
7
|
Tan JHL, Hwang YY, Chin HX, Liu M, Tan SY, Chen Q. Towards a better preclinical cancer model - human immune aging in humanized mice. Immun Ageing 2023; 20:49. [PMID: 37752597 PMCID: PMC10523735 DOI: 10.1186/s12979-023-00374-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 09/14/2023] [Indexed: 09/28/2023]
Abstract
BACKGROUND Preclinical models are often used for cancer studies and evaluation of novel therapeutics. The relevance of these models has vastly improved with mice bearing a human immune system, especially in the context of immunotherapy. Nonetheless, cancer is an age-related disease, and studies often overlook the effects of aging. Here we have established a humanized mouse model of human immune aging to investigate the role of this phenomenon on liver tumor dynamics. METHODS Multiple organs and tissues (blood, thymus, lung, liver, spleen and bone marrow) were harvested from NOD-scid IL2rγ-/- (NIKO) mice reconstituted with human immune cells, over a period of 60 weeks post-birth, for immune profiling. Young and aging immune cells were compared for transcriptomic changes and functional differences. Effect of immune aging was investigated in a liver cancer humanized mouse model. RESULTS Focusing on the T cell population, which is central to cancer immunosurveillance and immunotherapy, we showed that the proportion of naïve T cells declined while memory subsets and senescent-like cells increased with age. RNA-sequencing revealed that downregulated genes were related to immune responses and processes, and this was corroborated by reduced cytokine production in aging T cells. Finally, we showed faster liver tumor growth in aging than younger humanized mice, which could be attributed to specific pathways of aging T cell exhaustion. CONCLUSION Our work improves on existing humanized (immune) mouse model and highlights the importance of considering immune aging in liver cancer modeling.
Collapse
Affiliation(s)
- Joel H L Tan
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore
| | - You Yi Hwang
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos Level 3 & 4, Singapore, 138648, Republic of Singapore
| | - Hui Xian Chin
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos Level 3 & 4, Singapore, 138648, Republic of Singapore
| | - Min Liu
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore
| | - Sue Yee Tan
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore
| | - Qingfeng Chen
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore.
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos Level 3 & 4, Singapore, 138648, Republic of Singapore.
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore.
| |
Collapse
|
8
|
Budiman A, Handini AL, Muslimah MN, Nurani NV, Laelasari E, Kurniawansyah IS, Aulifa DL. Amorphous Solid Dispersion as Drug Delivery Vehicles in Cancer. Polymers (Basel) 2023; 15:3380. [PMID: 37631436 PMCID: PMC10457821 DOI: 10.3390/polym15163380] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/10/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
Cancer treatment has improved over the past decades, but a major challenge lies in drug formulation, specifically for oral administration. Most anticancer drugs have poor water solubility which can affect their bioavailability. This causes suboptimal pharmacokinetic performance, resulting in limited efficacy and safety when administered orally. As a result, it is essential to develop a strategy to modify the solubility of anticancer drugs in oral formulations to improve their efficacy and safety. A promising approach that can be implemented is amorphous solid dispersion (ASD) which can enhance the aqueous solubility and bioavailability of poorly water-soluble drugs. The addition of a polymer can cause stability in the formulations and maintain a high supersaturation in bulk medium. Therefore, this study aimed to summarize and elucidate the mechanisms and impact of an amorphous solid dispersion system on cancer therapy. To gather relevant information, a comprehensive search was conducted using keywords such as "anticancer drug" and "amorphous solid dispersion" in the PubMed, Scopus, and Google Scholar databases. The review provides an overview and discussion of the issues related to the ASD system used to improve the bioavailability of anticancer drugs based on molecular pharmaceutics. A thorough understanding of anticancer drugs in this system at a molecular level is imperative for the rational design of the products.
Collapse
Affiliation(s)
- Arif Budiman
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (A.L.H.); (M.N.M.); (N.V.N.); (E.L.); (I.S.K.)
| | - Annisa Luthfiyah Handini
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (A.L.H.); (M.N.M.); (N.V.N.); (E.L.); (I.S.K.)
| | - Mutia Nur Muslimah
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (A.L.H.); (M.N.M.); (N.V.N.); (E.L.); (I.S.K.)
| | - Neng Vera Nurani
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (A.L.H.); (M.N.M.); (N.V.N.); (E.L.); (I.S.K.)
| | - Eli Laelasari
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (A.L.H.); (M.N.M.); (N.V.N.); (E.L.); (I.S.K.)
| | - Insan Sunan Kurniawansyah
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (A.L.H.); (M.N.M.); (N.V.N.); (E.L.); (I.S.K.)
| | - Diah Lia Aulifa
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia;
| |
Collapse
|
9
|
Bushra R, Ahmad M, Seidi F, Qurtulen, Song J, Jin Y, Xiao H. Polysaccharide-based nanoassemblies: From synthesis methodologies and industrial applications to future prospects. Adv Colloid Interface Sci 2023; 318:102953. [PMID: 37399637 DOI: 10.1016/j.cis.2023.102953] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 05/23/2023] [Accepted: 06/19/2023] [Indexed: 07/05/2023]
Abstract
Polysaccharides, due to their remarkable features, have gained significant prominence in the sustainable production of nanoparticles (NPs). High market demand and minimal production cost, compared to the chemically synthesised NPs, demonstrate a drive towards polysaccharide-based nanoparticles (PSNPs) benign to environment. Various approaches are used for the synthesis of PSNPs including cross-linking, polyelectrolyte complexation, and self-assembly. PSNPs have the potential to replace a wide diversity of chemical-based agents within the food, health, medical and pharmacy sectors. Nevertheless, the considerable challenges associated with optimising the characteristics of PSNPs to meet specific targeting applications are of utmost importance. This review provides a detailed compilation of recent accomplishments in the synthesis of PSNPs, the fundamental principles and critical factors that govern their rational fabrication, as well as various characterisation techniques. Noteworthy, the multiple use of PSNPs in different disciplines such as biomedical, cosmetics agrochemicals, energy storage, water detoxification, and food-related realms, is accounted in detail. Insights into the toxicological impacts of the PSNPs and their possible risks to human health are addressed, and efforts made in terms of PSNPs development and optimising strategies that allow for enhanced delivery are highlighted. Finally, limitations, potential drawbacks, market diffusion, economic viability and future possibilities for PSNPs to achieve widespread commercial use are also discussed.
Collapse
Affiliation(s)
- Rani Bushra
- International Innovation Center for Forest Chemicals and Materials and Jiangsu Co-Innovation Center for Efficient Processing and Utilization of Forest Resources, Nanjing Forestry University, Nanjing 210037, Jiangsu, China; Joint International Research Lab of Lignocellulosic Functional Materials and Provincial Key Lab of Pulp and Paper Sci & Tech, Nanjing Forestry University, Nanjing 210037, Jiangsu, China
| | - Mehraj Ahmad
- International Innovation Center for Forest Chemicals and Materials and Jiangsu Co-Innovation Center for Efficient Processing and Utilization of Forest Resources, Nanjing Forestry University, Nanjing 210037, Jiangsu, China; College of Light Industry and Food, Department of Food Science and Engineering, Nanjing Forestry University, Nanjing 210037, Jiangsu, China; Joint International Research Lab of Lignocellulosic Functional Materials and Provincial Key Lab of Pulp and Paper Sci & Tech, Nanjing Forestry University, Nanjing 210037, Jiangsu, China.
| | - Farzad Seidi
- International Innovation Center for Forest Chemicals and Materials and Jiangsu Co-Innovation Center for Efficient Processing and Utilization of Forest Resources, Nanjing Forestry University, Nanjing 210037, Jiangsu, China; Joint International Research Lab of Lignocellulosic Functional Materials and Provincial Key Lab of Pulp and Paper Sci & Tech, Nanjing Forestry University, Nanjing 210037, Jiangsu, China
| | - Qurtulen
- Department of Chemistry, Aligarh Muslim University, Aligarh 202002, India
| | - Junlong Song
- International Innovation Center for Forest Chemicals and Materials and Jiangsu Co-Innovation Center for Efficient Processing and Utilization of Forest Resources, Nanjing Forestry University, Nanjing 210037, Jiangsu, China; Joint International Research Lab of Lignocellulosic Functional Materials and Provincial Key Lab of Pulp and Paper Sci & Tech, Nanjing Forestry University, Nanjing 210037, Jiangsu, China
| | - Yongcan Jin
- International Innovation Center for Forest Chemicals and Materials and Jiangsu Co-Innovation Center for Efficient Processing and Utilization of Forest Resources, Nanjing Forestry University, Nanjing 210037, Jiangsu, China; Joint International Research Lab of Lignocellulosic Functional Materials and Provincial Key Lab of Pulp and Paper Sci & Tech, Nanjing Forestry University, Nanjing 210037, Jiangsu, China
| | - Huining Xiao
- Department of Chemical Engineering, University of New Brunswick, Fredericton, NB E3B 5A3, Canada
| |
Collapse
|
10
|
Flickinger KM, Wilson KM, Rossiter NJ, Hunger AL, Lee TD, Hall MD, Cantor JR. Conditional lethality profiling reveals anticancer mechanisms of action and drug-nutrient interactions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.04.543621. [PMID: 37333068 PMCID: PMC10274668 DOI: 10.1101/2023.06.04.543621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Chemical screening studies have identified drug sensitivities across hundreds of cancer cell lines but most putative therapeutics fail to translate. Discovery and development of drug candidates in models that more accurately reflect nutrient availability in human biofluids may help in addressing this major challenge. Here we performed high-throughput screens in conventional versus Human Plasma-Like Medium (HPLM). Sets of conditional anticancer compounds span phases of clinical development and include non-oncology drugs. Among these, we characterize a unique dual-mechanism of action for brivudine, an agent otherwise approved for antiviral treatment. Using an integrative approach, we find that brivudine affects two independent targets in folate metabolism. We also traced conditional phenotypes for several drugs to the availability of nucleotide salvage pathway substrates and verified others for compounds that seemingly elicit off-target anticancer effects. Our findings establish generalizable strategies for exploiting conditional lethality in HPLM to reveal therapeutic candidates and mechanisms of action.
Collapse
|
11
|
Mason J, Öhlund D. Key aspects for conception and construction of co-culture models of tumor-stroma interactions. Front Bioeng Biotechnol 2023; 11:1150764. [PMID: 37091337 PMCID: PMC10119418 DOI: 10.3389/fbioe.2023.1150764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 03/31/2023] [Indexed: 04/25/2023] Open
Abstract
The tumor microenvironment is crucial in the initiation and progression of cancers. The interplay between cancer cells and the surrounding stroma shapes the tumor biology and dictates the response to cancer therapies. Consequently, a better understanding of the interactions between cancer cells and different components of the tumor microenvironment will drive progress in developing novel, effective, treatment strategies. Co-cultures can be used to study various aspects of these interactions in detail. This includes studies of paracrine relationships between cancer cells and stromal cells such as fibroblasts, endothelial cells, and immune cells, as well as the influence of physical and mechanical interactions with the extracellular matrix of the tumor microenvironment. The development of novel co-culture models to study the tumor microenvironment has progressed rapidly over recent years. Many of these models have already been shown to be powerful tools for further understanding of the pathophysiological role of the stroma and provide mechanistic insights into tumor-stromal interactions. Here we give a structured overview of different co-culture models that have been established to study tumor-stromal interactions and what we have learnt from these models. We also introduce a set of guidelines for generating and reporting co-culture experiments to facilitate experimental robustness and reproducibility.
Collapse
Affiliation(s)
- James Mason
- Department of Radiation Sciences, Umeå University, Umeå, Sweden
- Wallenberg Centre for Molecular Medicine, Umeå University, Umeå, Sweden
| | - Daniel Öhlund
- Department of Radiation Sciences, Umeå University, Umeå, Sweden
- Wallenberg Centre for Molecular Medicine, Umeå University, Umeå, Sweden
- *Correspondence: Daniel Öhlund,
| |
Collapse
|
12
|
Dey M, Kim MH, Dogan M, Nagamine M, Kozhaya L, Celik N, Unutmaz D, Ozbolat IT. Chemotherapeutics and CAR-T Cell-Based Immunotherapeutics Screening on a 3D Bioprinted Vascularized Breast Tumor Model. ADVANCED FUNCTIONAL MATERIALS 2022; 32:2203966. [PMID: 38938621 PMCID: PMC11209929 DOI: 10.1002/adfm.202203966] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Indexed: 06/29/2024]
Abstract
Despite substantial advancements in development of cancer treatments, lack of standardized and physiologically-relevant in vitro testing platforms limit the early screening of anticancer agents. A major barrier is the complex interplay between the tumor microenvironment and immune response. To tackle this, a dynamic-flow based 3D bioprinted multi-scale vascularized breast tumor model, responding to chemo and immunotherapeutics is developed. Heterotypic tumors are precisely bioprinted at pre-defined distances from a perfused vasculature, exhibit tumor angiogenesis and cancer cell invasion into the perfused vasculature. Bioprinted tumors treated with varying dosages of doxorubicin for 72 h portray a dose-dependent drug response behavior. More importantly, a cell based immune therapy approach is explored by perfusing HER2-targeting chimeric antigen receptor (CAR) modified CD8+ T cells for 24 or 72 h. Extensive CAR-T cell recruitment to the endothelium, substantial T cell activation and infiltration to the tumor site, resulted in up to ≈70% reduction in tumor volumes. The presented platform paves the way for a robust, precisely fabricated, and physiologically-relevant tumor model for future translation of anti-cancer therapies to personalized medicine.
Collapse
Affiliation(s)
- Madhuri Dey
- Department of Chemistry, Penn State University, University Park, PA 16802, USA; The Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, USA
| | - Myoung Hwan Kim
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, USA; Biomedical Engineering Department, Penn State University, University Park, PA 16802, USA
| | - Mikail Dogan
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Momoka Nagamine
- Department of Chemistry, Penn State University, University Park, PA 16802, USA; The Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, USA
| | - Lina Kozhaya
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Nazmiye Celik
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, USA; Engineering Science and Mechanics Department, Penn State University, University Park, PA 16802, USA
| | - Derya Unutmaz
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; University of Connecticut Health Center, Farmington, CT 06032, USA
| | - Ibrahim T Ozbolat
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, USA; Biomedical Engineering Department, Penn State University, University Park, PA 16802, USA; Engineering Science and Mechanics Department, Penn State University, University Park, PA 16802, USA; Materials Research Institute, Penn State University, University Park, PA 16802, USA; Cancer Institute, Penn State University, Hershey, PA 17033, USA; Neurosurgery Department, Penn State University, Hershey, PA 17033, USA; Department of Medical Oncology, Cukurova University, Adana 01330, Turkey
| |
Collapse
|
13
|
Belete TM. Recent Updates on the Development of Deuterium-Containing Drugs for the Treatment of Cancer. Drug Des Devel Ther 2022; 16:3465-3472. [PMID: 36217450 PMCID: PMC9547620 DOI: 10.2147/dddt.s379496] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 09/25/2022] [Indexed: 11/13/2022] Open
Abstract
Cancer is one of the deadliest diseases in the world. In 2020, 19.3 million cancer cases and 10 million deaths were reported in the world. It is supposed that the prevalence of cancer cases will rise to 28.4 million by 2040. Chemotherapy-based regimens have a narrow therapeutic index, severe adverse drug reactions, and lack metabolic stability. Besides, the metabolism of anticancer produces several non-active and toxic metabolites that reduce exposure of the target site to the parent drug. Therefore, developing better-tolerated and effective new anticancer drugs and modification of the existing anticancer drugs to minimize toxicity and increase efficacy has become a very urgent need. Deuterium incorporation reduces the metabolism of certain drugs that are breakdown by pathways involving hydrogen-carbon bond scission. For example, CYP450 mediated oxidative metabolism of drugs that involves the breakdown of a hydrogen-carbon bond affected by deuteration. Deuterium incorporation into the drug increases the half-life and reduces the dose, which provides better safety and efficacy. Deutetrabenazine is the first deuterated form of tetrabenazine approved to treat chorea associated with Huntington’s disease and tardive dyskinesia. The study revealed that Deutetrabenazine has fewer neuropsychiatric side effects with favorable safety than tetrabenazine. The current review highlights the deuterium kinetic isotope effect on drug metabolism, deuterated compound pharmacokinetic property, and safety profile. Besides, this review explains the deuterated anticancer drug development update status.
Collapse
Affiliation(s)
- Tafere Mulaw Belete
- Department of Pharmacology, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia,Correspondence: Tafere Mulaw Belete, Tel +251 918045943, Email
| |
Collapse
|
14
|
Bodei L, Herrmann K, Schöder H, Scott AM, Lewis JS. Radiotheranostics in oncology: current challenges and emerging opportunities. Nat Rev Clin Oncol 2022; 19:534-550. [PMID: 35725926 PMCID: PMC10585450 DOI: 10.1038/s41571-022-00652-y] [Citation(s) in RCA: 129] [Impact Index Per Article: 64.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2022] [Indexed: 12/20/2022]
Abstract
Structural imaging remains an essential component of diagnosis, staging and response assessment in patients with cancer; however, as clinicians increasingly seek to noninvasively investigate tumour phenotypes and evaluate functional and molecular responses to therapy, theranostics - the combination of diagnostic imaging with targeted therapy - is becoming more widely implemented. The field of radiotheranostics, which is the focus of this Review, combines molecular imaging (primarily PET and SPECT) with targeted radionuclide therapy, which involves the use of small molecules, peptides and/or antibodies as carriers for therapeutic radionuclides, typically those emitting α-, β- or auger-radiation. The exponential, global expansion of radiotheranostics in oncology stems from its potential to target and eliminate tumour cells with minimal adverse effects, owing to a mechanism of action that differs distinctly from that of most other systemic therapies. Currently, an enormous opportunity exists to expand the number of patients who can benefit from this technology, to address the urgent needs of many thousands of patients across the world. In this Review, we describe the clinical experience with established radiotheranostics as well as novel areas of research and various barriers to progress.
Collapse
Affiliation(s)
- Lisa Bodei
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Radiology, Weill Cornell Medical School, New York, NY, USA
| | - Ken Herrmann
- German Cancer Consortium, University Hospital Essen, Essen, Germany
- Department of Nuclear Medicine, University of Duisburg-Essen, University Hospital Essen, Essen, Germany
| | - Heiko Schöder
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Radiology, Weill Cornell Medical School, New York, NY, USA
| | - Andrew M Scott
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Melbourne, Victoria, Australia
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, Victoria, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, Victoria, Australia
- Department of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| | - Jason S Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Radiology, Weill Cornell Medical School, New York, NY, USA.
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Pharmacology, Weill Cornell Medical School, New York, NY, USA.
| |
Collapse
|
15
|
Parekh A, Das S, Das CK, Mandal M. Progressing Towards a Human-Centric Approach in Cancer Research. Front Oncol 2022; 12:896633. [PMID: 35928861 PMCID: PMC9343698 DOI: 10.3389/fonc.2022.896633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 06/21/2022] [Indexed: 11/17/2022] Open
Abstract
Despite the advancement in research methodologies and technologies for cancer research, there is a high rate of anti-cancer drug attrition. In this review, we discuss different conventional and modern approaches in cancer research and how human-centric models can improve on the voids conferred by more traditional animal-centric models, thereby offering a more reliable platform for drug discovery. Advanced three-dimensional cell culture methodologies, along with in silico computational analysis form the core of human-centric cancer research. This can provide a holistic understanding of the research problems and help design specific and accurate experiments that could lead to the development of better cancer therapeutics. Here, we propose a new human-centric research roadmap that promises to provide a better platform for cancer research and drug discovery.
Collapse
Affiliation(s)
- Aditya Parekh
- School of Design, Anant National University, Ahmedabad, India
- Genetics and Development, National Centre For Biological Sciences, Bengaluru, India
- *Correspondence: Aditya Parekh,
| | - Subhayan Das
- School of Medical Science and Technology (SMST), Indian Institute of Technology, Kharagpur, India
| | - Chandan K. Das
- Cancer Biology, University of Pennsylvania, Philadelphia, PA, United States
| | - Mahitosh Mandal
- School of Medical Science and Technology (SMST), Indian Institute of Technology, Kharagpur, India
| |
Collapse
|
16
|
Hamid SJ, Salih T. Design, Synthesis, and Anti-Inflammatory Activity of Some Coumarin Schiff Base Derivatives: In silico and in vitro Study. Drug Des Devel Ther 2022; 16:2275-2288. [PMID: 35860526 PMCID: PMC9293384 DOI: 10.2147/dddt.s364746] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 06/29/2022] [Indexed: 11/23/2022] Open
Abstract
Introduction Inflammation is a fundamental response of the immune system during tissue damage or pathogen infection to protect and maintain tissue homeostasis. However, inflammation may lead to life-threatening conditions. The most common treatment of inflammation is non-steroidal anti-inflammatory drugs (NSAIDs). Nowadays, the development of safer new NSAIDs is critical as most of the existing NSAIDs have serious adverse effects, such as gastrointestinal (GI) toxicity and cardiotoxicity. In the present study, four compounds as Schiff base derivatives of 7-hydroxy-4-formyl coumarin and 7-methoxy-4-formyl coumarin were designed and synthesized aiming to develop a lead compound that exhibits anti-inflammatory activity and circumvents the side effects of NSAIDs, especially GI toxicity. Materials and Methods Lipinski’s rule of five was applied for each designed molecule to evaluate the drug-likeness properties. Molecular docking studies were performed using the ligands and the cyclooxygenase-2 (COX-2) protein to select the best-scored molecule using AutoDock 4.2.6. The molecules were then synthesized and characterized. An in vitro anti-inflammatory assay of the compounds against the COX-2 receptor was realized through a protein denaturation assay. Results and Discussion All four synthesized ligands passed Lipinski’s rule of five and exhibited higher binding free energy compared to the positive standard control (ibuprofen), and the Ki values of compounds 5, 7, and 8 were in the nanomolar range. However, only compounds 6 and 7 obtained a higher percentage of inhibition of protein denaturation relative to ibuprofen. Conclusion The present study suggested that compound 7 may be a lead molecule because this ligand not only exhibited the best computational and experimental results but also exhibited the strongest correlation between the concentration and percentage of protein denaturation (R = 0.986 and R2 = 0.972) with the lowest P-value (0.014).
Collapse
Affiliation(s)
- Shokhan J Hamid
- Department of Pharmacognosy and Pharmaceutical Chemistry, College of Pharmacy, University of Sulaimani, Sulaymaniyah, Iraq
| | - Twana Salih
- Department of Pharmacognosy and Pharmaceutical Chemistry, College of Pharmacy, University of Sulaimani, Sulaymaniyah, Iraq
- Correspondence: Twana Salih, Department of Pharmacognosy and Pharmaceutical Chemistry, College of Pharmacy, University of Sulaimani, Town Campus, Sulaymaniyah, 46001, Iraq, Tel +964 0 770 146 2171, Email
| |
Collapse
|
17
|
Utilizing Three-Dimensional Culture Methods to Improve High-Throughput Drug Screening in Anaplastic Thyroid Carcinoma. Cancers (Basel) 2022; 14:cancers14081855. [PMID: 35454763 PMCID: PMC9031362 DOI: 10.3390/cancers14081855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/21/2022] [Accepted: 04/02/2022] [Indexed: 11/16/2022] Open
Abstract
Anaplastic thyroid carcinoma (ATC) is the most aggressive endocrine neoplasm, with a median survival of just four to six months post-diagnosis. Even with surgical and chemotherapeutic interventions, the five-year survival rate is less than 5%. Although combination dabrafenib/trametinib therapy was recently approved for treatment of the ~25% of ATCs harboring BRAFV600E mutations, there are no approved, effective treatments for BRAF-wildtype disease. Herein, we perform a screen of 1525 drugs and evaluate therapeutic candidates using monolayer cell lines and four corresponding spheroid models of anaplastic thyroid carcinoma. We utilize three-dimensional culture methods, as they have been shown to more accurately recapitulate tumor responses in vivo. These three-dimensional cultures include four distinct ATC spheroid lines representing unique morphology and mutational drivers to provide drug prioritization that will be more readily translatable to the clinic. Using this screen, we identify three exceptionally potent compounds (bortezomib, cabazitaxel, and YM155) that have established safety profiles and could potentially be moved into clinical trial for the treatment of anaplastic thyroid carcinoma, a disease with few treatment options.
Collapse
|
18
|
Geleta B, Tout FS, Lim SC, Sahni S, Jansson PJ, Apte MV, Richardson DR, Kovačević Ž. Targeting Wnt/tenascin C-mediated cross talk between pancreatic cancer cells and stellate cells via activation of the metastasis suppressor NDRG1. J Biol Chem 2022; 298:101608. [PMID: 35065073 PMCID: PMC8881656 DOI: 10.1016/j.jbc.2022.101608] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 01/02/2022] [Accepted: 01/05/2022] [Indexed: 02/06/2023] Open
Abstract
A major barrier to successful pancreatic cancer (PC) treatment is the surrounding stroma, which secretes growth factors/cytokines that promote PC progression. Wnt and tenascin C (TnC) are key ligands secreted by stromal pancreatic stellate cells (PSCs) that then act on PC cells in a paracrine manner to activate the oncogenic β-catenin and YAP/TAZ signaling pathways. Therefore, therapies targeting oncogenic Wnt/TnC cross talk between PC cells and PSCs constitute a promising new therapeutic approach for PC treatment. The metastasis suppressor N-myc downstream-regulated gene-1 (NDRG1) inhibits tumor progression and metastasis in numerous cancers, including PC. We demonstrate herein that targeting NDRG1 using the clinically trialed anticancer agent di-2-pyridylketone-4-cyclohexyl-4-methyl-3-thiosemicarbazone (DpC) inhibited Wnt/TnC-mediated interactions between PC cells and the surrounding PSCs. Mechanistically, NDRG1 and DpC markedly inhibit secretion of Wnt3a and TnC by PSCs, while also attenuating Wnt/β-catenin and YAP/TAZ activation and downstream signaling in PC cells. This antioncogenic activity was mediated by direct inhibition of β-catenin and YAP/TAZ nuclear localization and by increasing the Wnt inhibitor, DKK1. Expression of NDRG1 also inhibited transforming growth factor (TGF)-β secretion by PC cells, a key mechanism by which PC cells activate PSCs. Using an in vivo orthotopic PC mouse model, we show DpC downregulated β-catenin, TnC, and YAP/TAZ, while potently increasing NDRG1 expression in PC tumors. We conclude that NDRG1 and DpC inhibit Wnt/TnC-mediated interactions between PC cells and PSCs. These results further illuminate the antioncogenic mechanism of NDRG1 and the potential of targeting this metastasis suppressor to overcome the oncogenic effects of the PC-PSC interaction.
Collapse
Affiliation(s)
- Bekesho Geleta
- Cancer Metastasis and Tumor Microenvironment Program, Department of Pathology, University of Sydney, Sydney, New South Wales, Australia; Molecular Pharmacology and Pathology Program, Department of Pathology, University of Sydney, Sydney, New South Wales, Australia
| | - Faten S Tout
- Cancer Metastasis and Tumor Microenvironment Program, Department of Pathology, University of Sydney, Sydney, New South Wales, Australia; Molecular Pharmacology and Pathology Program, Department of Pathology, University of Sydney, Sydney, New South Wales, Australia; Department of Medical Laboratory Science, Faculty of Allied Health Sciences, The Hashemite University, Zarqa, Jordan
| | - Syer Choon Lim
- Cancer Metastasis and Tumor Microenvironment Program, Department of Pathology, University of Sydney, Sydney, New South Wales, Australia; Molecular Pharmacology and Pathology Program, Department of Pathology, University of Sydney, Sydney, New South Wales, Australia
| | - Sumit Sahni
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - Patric J Jansson
- Molecular Pharmacology and Pathology Program, Department of Pathology, University of Sydney, Sydney, New South Wales, Australia; Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia; Cancer Drug Resistance & Stem Cell Program, Faculty of Medicine and Health, School of Medical Science, University of Sydney, Sydney, New South Wales, Australia
| | - Minoti V Apte
- Pancreatic Research Group, South Western Sydney Clinical School, UNSW Sydney, Sydney, New South Wales, Australia; Pancreatic Research Group, Ingham Institute for Applied Medical Research, Sydney, New South Wales, Australia
| | - Des R Richardson
- Molecular Pharmacology and Pathology Program, Department of Pathology, University of Sydney, Sydney, New South Wales, Australia; Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia; Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Žaklina Kovačević
- Cancer Metastasis and Tumor Microenvironment Program, Department of Pathology, University of Sydney, Sydney, New South Wales, Australia; Molecular Pharmacology and Pathology Program, Department of Pathology, University of Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
19
|
Nguyen L, Nguyen Vo TH, Trinh QH, Nguyen BH, Nguyen-Hoang PU, Le L, Nguyen BP. iANP-EC: Identifying Anticancer Natural Products Using Ensemble Learning Incorporated with Evolutionary Computation. J Chem Inf Model 2022; 62:5080-5089. [PMID: 35157472 DOI: 10.1021/acs.jcim.1c00920] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cancer is one of the most deadly diseases that annually kills millions of people worldwide. The investigation on anticancer medicines has never ceased to seek better and more adaptive agents with fewer side effects. Besides chemically synthetic anticancer compounds, natural products are scientifically proved as a highly potential alternative source for anticancer drug discovery. Along with experimental approaches being used to find anticancer drug candidates, computational approaches have been developed to virtually screen for potential anticancer compounds. In this study, we construct an ensemble computational framework, called iANP-EC, using machine learning approaches incorporated with evolutionary computation. Four learning algorithms (k-NN, SVM, RF, and XGB) and four molecular representation schemes are used to build a set of classifiers, among which the top-four best-performing classifiers are selected to form an ensemble classifier. Particle swarm optimization (PSO) is used to optimise the weights used to combined the four top classifiers. The models are developed by a set of curated 997 compounds which are collected from the NPACT and CancerHSP databases. The results show that iANP-EC is a stable, robust, and effective framework that achieves an AUC-ROC value of 0.9193 and an AUC-PR value of 0.8366. The comparative analysis of molecular substructures between natural anticarcinogens and nonanticarcinogens partially unveils several key substructures that drive anticancerous activities. We also deploy the proposed ensemble model as an online web server with a user-friendly interface to support the research community in identifying natural products with anticancer activities.
Collapse
Affiliation(s)
- Loc Nguyen
- Computational Biology Center, International University - VNU HCMC, Ho Chi Minh City 700000, Vietnam
| | - Thanh-Hoang Nguyen Vo
- School of Mathematics and Statistics, Victoria University of Wellington, Wellington 6140, New Zealand
| | - Quang H Trinh
- Computational Biology Center, International University - VNU HCMC, Ho Chi Minh City 700000, Vietnam.,School of Information and Communication Technology, Hanoi University of Science and Technology, Hanoi 100000, Vietnam
| | - Bach Hoai Nguyen
- School of Engineering and Computer Science, Victoria University of Wellington, Wellington 6140, New Zealand
| | - Phuong-Uyen Nguyen-Hoang
- Computational Biology Center, International University - VNU HCMC, Ho Chi Minh City 700000, Vietnam
| | - Ly Le
- Computational Biology Center, International University - VNU HCMC, Ho Chi Minh City 700000, Vietnam.,Vingroup Big Data Institute, Ha Noi 100000, Vietnam
| | - Binh P Nguyen
- School of Mathematics and Statistics, Victoria University of Wellington, Wellington 6140, New Zealand
| |
Collapse
|
20
|
Sivanesan D, Verma RS, Prasad E. 5FU encapsulated polyglycerol sebacate nanoparticles as anti-cancer drug carriers. RSC Adv 2021; 11:18984-18993. [PMID: 35478658 PMCID: PMC9033480 DOI: 10.1039/d1ra01722e] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 05/10/2021] [Indexed: 12/19/2022] Open
Abstract
The majority of anti-cancer drugs fail to reach clinical trials due to their low water solubility. A biocompatible drug delivery system that encapsulates and efficiently delivers hydrophobic drugs to the target site is the need of the hour. This study addresses the issue by focusing on a polymeric polyglycerol sebacate (PGS) nanoparticles loaded with 5-fluorouracil (5FU), a primary line chemotherapy drug for many types of cancers. The generated nanoparticle (PGS-NP) was biocompatible and had minimal cytotoxicity against the MDA-MB-231 and A549 cell lines, even at a high concentration of 100 μg mL-1. The cell viability post treatment with PGS nanoparticles encapsulated with 5FU (PGS-5FU) decreased to as low as around 40% whereas, in the case of treatment with 5FU, the viability percentage increased. The nanoparticles also showed controlled drug release when encapsulated with 5FU. This striking observation suggested that these nanoparticles can improve the efficacy of drug delivery to tumor sites. Apoptosis assay and caspase-3 activity quantification supported these data wherein PGS-5FU treatment showed almost three times caspase-3 activity as compared to control cells. Additionally, throughout all the experiments, MDA-MB-231 cells were more sensitive to PGS-5FU than A549 cells, indicating that these nanoparticles are ideal for breast cancer treatment. In summary, 5FU encapsulated PGS nanoparticles are a potential drug carrier to deliver 5FU efficiently to cancer cells.
Collapse
Affiliation(s)
- Divya Sivanesan
- Department of Biotechnology, Indian Institute of Technology Madras Chennai-600036 India
| | - Rama S Verma
- Department of Biotechnology, Indian Institute of Technology Madras Chennai-600036 India
| | - Edamana Prasad
- Department of Chemistry, Indian Institute of Technology Madras Chennai-600036 India
| |
Collapse
|
21
|
Lu W, Yao J, Zhu X, Qi Y. Nanomedicines: Redefining traditional medicine. Biomed Pharmacother 2020; 134:111103. [PMID: 33338747 DOI: 10.1016/j.biopha.2020.111103] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/24/2020] [Accepted: 12/01/2020] [Indexed: 12/11/2022] Open
Abstract
Nanomedicines offer nanoscale drug delivery system. They offer ways of promising drug transportation, and address the issues of lack of targeting and permeability of traditional drugs. The physical and chemical properties in the domain of nanomedicine applications in vivo have not been sufficiently delivered. What's more, the metabolic of nanomedicines is not clear enough. Those factors which mentioned above determine that many nanomedicines have not yet realized clinical application due to their safety problems and in vivo efficacy. For example, they may cause immune response and cytotoxicity, as well as the ability to clear organs in vivo, the penetration ability of them and the lack of targeting ability may also cause poor efficacy of drugs in vivo. In this review, the new progresses of different kinds of nanomedicines (including gold nanoparticles, nanorobots, black phosphorus nanoparticles, brain diseases, gene editing and immunotherapy etc.) in anti-tumor, antibacterial, ocular diseases and arteriosclerosis in recent years were summarized. Their shortcomings were pointed out, and the new methods to improve the biosafety and efficacy were summarized.
Collapse
Affiliation(s)
- Weijia Lu
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang 524023, China
| | - Jing Yao
- First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xiao Zhu
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang 524023, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang 524023, China; The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang 524023, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang 524023, China.
| | - Yi Qi
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang 524023, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang 524023, China; The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang 524023, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang 524023, China.
| |
Collapse
|
22
|
Senescence and the Impact on Biodistribution of Different Nanosystems: the Discrepancy on Tissue Deposition of Graphene Quantum Dots, Polycaprolactone Nanoparticle and Magnetic Mesoporous Silica Nanoparticles in Young and Elder Animals. Pharm Res 2020; 37:40. [PMID: 31970499 DOI: 10.1007/s11095-019-2754-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 12/30/2019] [Indexed: 12/16/2022]
Abstract
PURPOSES Senescence is an inevitable and irreversible process, which may lead to loss in muscle and bone density, decline in brain volume and loss in renal clearance. Although aging is a well-known process, few studies on the consumption of nanodrugs by elderly people were performed. METHODS We evaluated three different nanosystems: i) carbon based nanosystem (Graphene Quantum Dots, GQD), ii) polymeric nanoparticles and mesoporous silica (magnetic core mesoporous silica, MMSN). In previous studies, our group has already characterized GQD and MMSN nanoparticles by dynamic light scattering analysis, atomic force microscopy, transmission electron microscopy, X-ray diffraction, Raman analysis, fluorescence and absorbance. The polymeric nanoparticle has been characterized by AFM and DLS. All the nanosystems were radiolabeled with 99 m-Tc by. The in vivo biodistribution/tissue deposition analysis evaluation was done using elder (PN270) and young (PN90) mice injected with radioactive nanosystems. RESULTS The nanosystems used in this study were well-formed as the radiolabeling processes were stable. Biodistribution analysis showed that there is a decrease in the uptake of the nanoparticles in elder mice when compared to young mice, showing that is necessary to increase the initial dose in elder people to achieve the same concentration when compared to young animals. CONCLUSION The discrepancy on tissue distribution of nanosystems between young and elder individuals must be monitored, as the therapeutic effect will be different in the groups. Noteworthy, this data is an alarm that some specific conditions must be evaluated before commercialization of nano-drugs. Graphical Abstract Changes between younger and elderly individuals are undoubtedly, especially in drug tissue deposition, biodistribution and pharmacokinetics. The same thought should be applied to nanoparticles. A comprehensive analysis on how age discrepancy change the biological behavior of nanoparticles has been performed.
Collapse
|
23
|
Harnessing the therapeutic potential of anticancer drugs through amorphous solid dispersions. Biochim Biophys Acta Rev Cancer 2019; 1873:188319. [PMID: 31678141 DOI: 10.1016/j.bbcan.2019.188319] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/28/2019] [Accepted: 10/28/2019] [Indexed: 12/19/2022]
Abstract
The treatment of cancer is still a major challenge. But tremendous progress in anticancer drug discovery and development has occurred in the last few decades. However, this progress has resulted in few effective oncology products due to challenges associated with anticancer drug delivery. Oral administration is the most preferred route for anticancer drug delivery, but the majority of anticancer drugs currently in product pipelines and the majority of those that have been commercially approved have inherently poor water solubility, and this cannot be mitigated without compromising their potency and stability. The poor water solubility of anticancer drugs, in conjunction with other factors, leads to suboptimal pharmacokinetic performance. Thus, these drugs have limited efficacy and safety when administered orally. The amorphous solid dispersion (ASD) is a promising formulation technology that primarily enhances the aqueous solubility of poorly water-soluble drugs. In this review, we discuss the challenges associated with the oral administration of anticancer drugs and the use of ASD technology in alleviating these challenges. We emphasize the ability of ASDs to improve not only the pharmacokinetics of poorly water-soluble anticancer drugs, but also their efficacy and safety. The goal of this paper is to rationalize the application of ASD technology in the formulation of anticancer drugs, thereby creating superior oncology products that lead to improved therapeutic outcomes.
Collapse
|
24
|
Kunnumakkara AB, Bordoloi D, Sailo BL, Roy NK, Thakur KK, Banik K, Shakibaei M, Gupta SC, Aggarwal BB. Cancer drug development: The missing links. Exp Biol Med (Maywood) 2019; 244:663-689. [PMID: 30961357 PMCID: PMC6552400 DOI: 10.1177/1535370219839163] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
IMPACT STATEMENT The success rate for cancer drugs which enter into phase 1 clinical trials is utterly less. Why the vast majority of drugs fail is not understood but suggests that pre-clinical studies are not adequate for human diseases. In 1975, as per the Tufts Center for the Study of Drug Development, pharmaceutical industries expended 100 million dollars for research and development of the average FDA approved drug. By 2005, this figure had more than quadrupled, to $1.3 billion. In order to recover their high and risky investment cost, pharmaceutical companies charge more for their products. However, there exists no correlation between drug development cost and actual sale of the drug. This high drug development cost could be due to the reason that all patients might not respond to the drug. Hence, a given drug has to be tested in large number of patients to show drug benefits and obtain significant results.
Collapse
Affiliation(s)
- Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory, DBT-AIST International Laboratory for Advanced Biomedicine (DAILAB), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, India
| | - Devivasha Bordoloi
- Cancer Biology Laboratory, DBT-AIST International Laboratory for Advanced Biomedicine (DAILAB), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, India
| | - Bethsebie Lalduhsaki Sailo
- Cancer Biology Laboratory, DBT-AIST International Laboratory for Advanced Biomedicine (DAILAB), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, India
| | - Nand Kishor Roy
- Cancer Biology Laboratory, DBT-AIST International Laboratory for Advanced Biomedicine (DAILAB), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, India
| | - Krishan Kumar Thakur
- Cancer Biology Laboratory, DBT-AIST International Laboratory for Advanced Biomedicine (DAILAB), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, India
| | - Kishore Banik
- Cancer Biology Laboratory, DBT-AIST International Laboratory for Advanced Biomedicine (DAILAB), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, India
| | - Mehdi Shakibaei
- Faculty of Medicine, Institute of Anatomy, Ludwig Maximilian University of Munich, Munich D-80336, Germany
| | - Subash C Gupta
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | | |
Collapse
|
25
|
Shaheer Malik M, Seddigi ZS, Bajee S, Azeeza S, Riyaz S, Ahmed SA, Althagafi II, Sajid Jamal QM, Kamal A. Multicomponent access to novel proline/cyclized cysteine tethered monastrol conjugates as potential anticancer agents. JOURNAL OF SAUDI CHEMICAL SOCIETY 2019. [DOI: 10.1016/j.jscs.2019.01.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
26
|
Nanomedicines for cancer therapy: current status, challenges and future prospects. Ther Deliv 2019; 10:113-132. [DOI: 10.4155/tde-2018-0062] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The emergence of nanomedicine as an innovative and promising alternative technology shows many advantages over conventional cancer therapies and provides new opportunities for early detection, improved treatment, and diagnosis of cancer. Despite the cancer nanomedicines’ capability of delivering chemotherapeutic agents while providing lower systemic toxicity, it is paramount to consider the cancer complexity and dynamics for bridging the translational bench-to-bedside gap. It is important to conduct appropriate investigations for exploiting the tumor microenvironment, and achieving a more comprehensive understanding of the fundamental biological processes in cancer and their roles in modulating nanoparticle–protein interactions, blood circulation, and tumor penetration. This review provides an overview of the current cancer nanomedicines, the major challenges, and the future opportunities in this research area.
Collapse
|
27
|
Farjadian F, Ghasemi A, Gohari O, Roointan A, Karimi M, Hamblin MR. Nanopharmaceuticals and nanomedicines currently on the market: challenges and opportunities. Nanomedicine (Lond) 2019; 14:93-126. [PMID: 30451076 PMCID: PMC6391637 DOI: 10.2217/nnm-2018-0120] [Citation(s) in RCA: 292] [Impact Index Per Article: 58.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 10/15/2018] [Indexed: 12/23/2022] Open
Abstract
There has been a revolution in nanotechnology and nanomedicine. Since 1980, there has been a remarkable increase in approved nano-based pharmaceutical products. These novel nano-based systems can either be therapeutic agents themselves, or else act as vehicles to carry different active pharmaceutical agents into specific parts of the body. Currently marketed nanostructures include nanocrystals, liposomes and lipid nanoparticles, PEGylated polymeric nanodrugs, other polymers, protein-based nanoparticles and metal-based nanoparticles. A range of issues must be addressed in the development of these nanostructures. Ethics, market size, possibility of market failure, costs and commercial development, are some topics which are on the table to be discussed. After passing all the ethical and biological assessments, and satisfying the investors as to future profitability, only a handful of these nanoformulations, successfully obtained marketing approval. We survey the range of nanomedicines that have received regulatory approval and are marketed. We discuss ethics, costs, commercial development and possible market failure. We estimate the global nanomedicine market size and future growth. Our goal is to summarize the different approved nanoformulations on the market, and briefly cover the challenges and future outlook.
Collapse
Affiliation(s)
- Fatemeh Farjadian
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz 71468-64685, Iran
| | - Amir Ghasemi
- Department of Materials Science & Engineering, Sharif University of Technology, Tehran 11365-9466, Iran
- Advances Nanobiotechnology & Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran 14496-4535, Iran
| | - Omid Gohari
- Department of Materials Science & Engineering, Sharif University of Technology, Tehran 11365-9466, Iran
| | - Amir Roointan
- Department of Medical Biotechnology, School of Advanced Medical Sciences & Technologies, Shiraz University of Medical Science, Shiraz 71348-14336, Iran
| | - Mahdi Karimi
- Cellular & Molecular Research Center, Iran University of Medical Sciences, Tehran 14496-14535, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 14496-14535, Iran
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Department of Dermatology, Harvard Medical School, Boston, MA 02115, USA
- Harvard – MIT Division of Health Sciences & Technology, Cambridge, MA 02139, USA
| |
Collapse
|
28
|
Strauss BE, Silva GRO, de Luna Vieira I, Cerqueira OLD, Del Valle PR, Medrano RFV, Mendonça SA. Perspectives for cancer immunotherapy mediated by p19Arf plus interferon-beta gene transfer. Clinics (Sao Paulo) 2018; 73:e479s. [PMID: 30208166 PMCID: PMC6113850 DOI: 10.6061/clinics/2018/e479s] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 05/22/2018] [Indexed: 12/13/2022] Open
Abstract
While cancer immunotherapy has gained much deserved attention in recent years, many areas regarding the optimization of such modalities remain unexplored, including the development of novel approaches and the strategic combination of therapies that target multiple aspects of the cancer-immunity cycle. Our own work involves the use of gene transfer technology to promote cell death and immune stimulation. Such immunogenic cell death, mediated by the combined transfer of the alternate reading frame (p14ARF in humans and p19Arf in mice) and the interferon-β cDNA in our case, was shown to promote an antitumor immune response in mouse models of melanoma and lung carcinoma. With these encouraging results, we are now setting out on the road toward translational and preclinical development of our novel immunotherapeutic approach. Here, we outline the perspectives and challenges that we face, including the use of human tumor and immune cells to verify the response seen in mouse models and the incorporation of clinically relevant models, such as patient-derived xenografts and spontaneous tumors in animals. In addition, we seek to combine our immunotherapeutic approach with other treatments, such as chemotherapy or checkpoint blockade, with the goal of reducing dosage and increasing efficacy. The success of any translational research requires the cooperation of a multidisciplinary team of professionals involved in laboratory and clinical research, a relationship that is fostered at the Cancer Institute of Sao Paulo.
Collapse
Affiliation(s)
- Bryan E Strauss
- Laboratório de Vetores Virais, Centro de Investigação Translacional em Oncologia, Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
- *Corresponding author. E-mail: /
| | - Gissele Rolemberg Oliveira Silva
- Laboratório de Vetores Virais, Centro de Investigação Translacional em Oncologia, Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Igor de Luna Vieira
- Laboratório de Vetores Virais, Centro de Investigação Translacional em Oncologia, Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Otto Luiz Dutra Cerqueira
- Laboratório de Vetores Virais, Centro de Investigação Translacional em Oncologia, Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Paulo Roberto Del Valle
- Laboratório de Vetores Virais, Centro de Investigação Translacional em Oncologia, Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Ruan Felipe Vieira Medrano
- Laboratório de Vetores Virais, Centro de Investigação Translacional em Oncologia, Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Samir Andrade Mendonça
- Laboratório de Vetores Virais, Centro de Investigação Translacional em Oncologia, Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| |
Collapse
|
29
|
Nuccio ML, Paul M, Bate NJ, Cohn J, Cutler SR. Where are the drought tolerant crops? An assessment of more than two decades of plant biotechnology effort in crop improvement. PLANT SCIENCE : AN INTERNATIONAL JOURNAL OF EXPERIMENTAL PLANT BIOLOGY 2018; 273:110-119. [PMID: 29907303 DOI: 10.1016/j.plantsci.2018.01.020] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 01/29/2018] [Accepted: 01/31/2018] [Indexed: 05/22/2023]
Abstract
Since the dawn of modern biotechnology public and private enterprise have pursued the development of a new breed of drought tolerant crop products. After more than 20 years of research and investment only a few such products have reached the market. This is due to several technical and market constraints. The technical challenges include the difficulty in defining tractable single-gene trait development strategies, the logistics of moving traits from initial to commercial genetic backgrounds, and the disconnect between conditions in farmer's fields and controlled environments. Market constraints include the significant difficulty, and associated costs, in obtaining access to markets around the world. Advances in the biology of plant water management, including response to water deficit reveal new opportunities to improve crop response to water deficit and new genome-based tools promise to usher in the next era of crop improvement. As biotechnology looks to improve crop productivity under drought conditions, the environmental and food security advantages will influence public perception and shift the debate toward benefits rather than risks.
Collapse
Affiliation(s)
- Michael L Nuccio
- Syngenta Crop Protection, LLC., 9 Davis Drive, Research Triangle Park, NC, 27709, USA.
| | - Matthew Paul
- Rothamsted Research, Harpenden, Hertfordshire, AL5 2JQ, UK.
| | - Nicholas J Bate
- Syngenta Crop Protection, LLC., 9 Davis Drive, Research Triangle Park, NC, 27709, USA.
| | - Jonathan Cohn
- Syngenta Crop Protection, LLC., 9 Davis Drive, Research Triangle Park, NC, 27709, USA.
| | - Sean R Cutler
- Plant Cell Biology and Chemistry, Botany and Plant Sciences Chemistry Genomics Building, University of California Riverside, CA, 92521, USA.
| |
Collapse
|
30
|
Nguyen EV, Centenera MM, Moldovan M, Das R, Irani S, Vincent AD, Chan H, Horvath LG, Lynn DJ, Daly RJ, Butler LM. Identification of Novel Response and Predictive Biomarkers to Hsp90 Inhibitors Through Proteomic Profiling of Patient-derived Prostate Tumor Explants. Mol Cell Proteomics 2018; 17:1470-1486. [PMID: 29632047 DOI: 10.1074/mcp.ra118.000633] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 03/26/2018] [Indexed: 12/16/2022] Open
Abstract
Inhibition of the heat shock protein 90 (Hsp90) chaperone is a promising therapeutic strategy to target expression of the androgen receptor (AR) and other oncogenic drivers in prostate cancer cells. However, identification of clinically-relevant responses and predictive biomarkers is essential to maximize efficacy and treatment personalization. Here, we combined mass spectrometry (MS)-based proteomic analyses with a unique patient-derived explant (PDE) model that retains the complex microenvironment of primary prostate tumors. Independent discovery and validation cohorts of PDEs (n = 16 and 30, respectively) were cultured in the absence or presence of Hsp90 inhibitors AUY922 or 17-AAG. PDEs were analyzed by LC-MS/MS with a hyper-reaction monitoring data independent acquisition (HRM-DIA) workflow, and differentially expressed proteins identified using repeated measure analysis of variance (ANOVA; raw p value <0.01). Using gene set enrichment, we found striking conservation of the most significantly AUY922-altered gene pathways between the discovery and validation cohorts, indicating that our experimental and analysis workflows were robust. Eight proteins were selectively altered across both cohorts by the most potent inhibitor, AUY922, including TIMP1, SERPINA3 and CYP51A (adjusted p < 0.01). The AUY922-mediated decrease in secretory TIMP1 was validated by ELISA of the PDE culture medium. We next exploited the heterogeneous response of PDEs to 17-AAG in order to detect predictive biomarkers of response and identified PCBP3 as a marker with increased expression in PDEs that had no response or increased in proliferation. Also, 17-AAG treatment led to increased expression of DNAJA1 in PDEs that exhibited a cytostatic response, revealing potential drug resistance mechanisms. This selective regulation of DNAJA1 was validated by Western blot analysis. Our study establishes "proof-of-principle" that proteomic profiling of drug-treated PDEs represents an effective and clinically-relevant strategy for identification of biomarkers that associate with certain tumor-specific responses.
Collapse
Affiliation(s)
- Elizabeth V Nguyen
- From the ‡Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia.,§Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
| | - Margaret M Centenera
- ¶Adelaide Medical School and Freemasons Foundation Centre for Men's Health, University of Adelaide, Adelaide, South Australia 5005, Australia.,‖South Australian Health and Medical Research Institute, Adelaide, South Australia 5000, Australia
| | - Max Moldovan
- ‖South Australian Health and Medical Research Institute, Adelaide, South Australia 5000, Australia
| | - Rajdeep Das
- ¶Adelaide Medical School and Freemasons Foundation Centre for Men's Health, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Swati Irani
- ¶Adelaide Medical School and Freemasons Foundation Centre for Men's Health, University of Adelaide, Adelaide, South Australia 5005, Australia.,‖South Australian Health and Medical Research Institute, Adelaide, South Australia 5000, Australia
| | - Andrew D Vincent
- ¶Adelaide Medical School and Freemasons Foundation Centre for Men's Health, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Howard Chan
- From the ‡Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia.,§Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
| | - Lisa G Horvath
- **Cancer Division, The Kinghorn Cancer Centre/Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia.,‡‡Royal Prince Alfred Hospital, Camperdown, New South Wales 2050, Australia.,§§Department of Medical Oncology, Chris O'Brien Lifehouse, Camperdown, New South Wales 2050, Australia
| | - David J Lynn
- ‖South Australian Health and Medical Research Institute, Adelaide, South Australia 5000, Australia.,¶¶School of Medicine, Flinders University, Bedford Park, SA 5042, Australia
| | - Roger J Daly
- From the ‡Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia; .,§Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
| | - Lisa M Butler
- ¶Adelaide Medical School and Freemasons Foundation Centre for Men's Health, University of Adelaide, Adelaide, South Australia 5005, Australia.,‖South Australian Health and Medical Research Institute, Adelaide, South Australia 5000, Australia
| |
Collapse
|
31
|
Masters A, Nutt D. A Plutocratic Proposal: an ethical way for rich patients to pay for a place on a clinical trial. JOURNAL OF MEDICAL ETHICS 2017; 43:730-736. [PMID: 28588147 PMCID: PMC5749301 DOI: 10.1136/medethics-2016-104050] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 03/02/2017] [Accepted: 03/29/2017] [Indexed: 06/07/2023]
Abstract
Many potential therapeutic agents are discarded before they are tested in humans. These are not quack medications. They are drugs and other interventions that have been developed by responsible scientists in respectable companies or universities and are often backed up by publications in peer-reviewed journals. These possible treatments might ease suffering and prolong the lives of innumerable patients, yet they have been put aside. In this paper, we outline a novel mechanism-the Plutocratic Proposal-to revive such neglected research and fund early phase clinical trials. The central idea of the Proposal is that any patient who rescues a potential therapeutic agent from neglect by funding early phase clinical trials (either entirely or in large part) should be offered a place on the trial.
Collapse
|
32
|
Kashif M, Andersson C, Mansoori S, Larsson R, Nygren P, Gustafsson MG. Bliss and Loewe interaction analyses of clinically relevant drug combinations in human colon cancer cell lines reveal complex patterns of synergy and antagonism. Oncotarget 2017; 8:103952-103967. [PMID: 29262612 PMCID: PMC5732778 DOI: 10.18632/oncotarget.21895] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 09/03/2017] [Indexed: 12/15/2022] Open
Abstract
We analyzed survival effects for 15 different pairs of clinically relevant anti-cancer drugs in three iso-genic pairs of human colorectal cancer carcinoma cell lines, by applying for the first time our novel software (R package) called COMBIA. In our experiments iso-genic pairs of cell lines were used, differing only with respect to a single clinically important KRAS or BRAF mutation. Frequently, concentration dependent but mutation independent joint Bliss and Loewe synergy/antagonism was found statistically significant. Four combinations were found synergistic/antagonistic specifically to the parental (harboring KRAS or BRAF mutation) cell line of the corresponding iso-genic cell lines pair. COMBIA offers considerable improvements over established software for synergy analysis such as MacSynergy™ II as it includes both Bliss (independence) and Loewe (additivity) analyses, together with a tailored non-parametric statistical analysis employing heteroscedasticity, controlled resampling, and global (omnibus) testing. In many cases Loewe analyses found significant synergistic as well as antagonistic effects in a cell line at different concentrations of a tested drug combination. By contrast, Bliss analysis found only one type of significant effect per cell line. In conclusion, the integrated Bliss and Loewe interaction analysis based on non-parametric statistics may provide more robust interaction analyses and reveal complex patterns of synergy and antagonism.
Collapse
Affiliation(s)
- Muhammad Kashif
- Department of Medical Sciences, Cancer Pharmacology, and Computational Medicine, Uppsala University Academic Hospital, Uppsala, Sweden.,Current/Present address: Department of Biosciences and Nutrition, Karolinska Institute, Stockholm, Sweden
| | - Claes Andersson
- Department of Medical Sciences, Cancer Pharmacology, and Computational Medicine, Uppsala University Academic Hospital, Uppsala, Sweden
| | - Sharmineh Mansoori
- Department of Medical Sciences, Cancer Pharmacology, and Computational Medicine, Uppsala University Academic Hospital, Uppsala, Sweden
| | - Rolf Larsson
- Department of Medical Sciences, Cancer Pharmacology, and Computational Medicine, Uppsala University Academic Hospital, Uppsala, Sweden
| | - Peter Nygren
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, Sweden
| | - Mats G Gustafsson
- Department of Medical Sciences, Cancer Pharmacology, and Computational Medicine, Uppsala University Academic Hospital, Uppsala, Sweden
| |
Collapse
|
33
|
Liu Z, Delavan B, Roberts R, Tong W. Lessons Learned from Two Decades of Anticancer Drugs. Trends Pharmacol Sci 2017; 38:852-872. [DOI: 10.1016/j.tips.2017.06.005] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 06/09/2017] [Accepted: 06/19/2017] [Indexed: 02/08/2023]
|
34
|
Ciavatta ML, Lefranc F, Carbone M, Mollo E, Gavagnin M, Betancourt T, Dasari R, Kornienko A, Kiss R. Marine Mollusk-Derived Agents with Antiproliferative Activity as Promising Anticancer Agents to Overcome Chemotherapy Resistance. Med Res Rev 2017; 37:702-801. [PMID: 27925266 PMCID: PMC5484305 DOI: 10.1002/med.21423] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 09/20/2016] [Accepted: 09/23/2016] [Indexed: 12/18/2022]
Abstract
The chemical investigation of marine mollusks has led to the isolation of a wide variety of bioactive metabolites, which evolved in marine organisms as favorable adaptations to survive in different environments. Most of them are derived from food sources, but they can be also biosynthesized de novo by the mollusks themselves, or produced by symbionts. Consequently, the isolated compounds cannot be strictly considered as "chemotaxonomic markers" for the different molluscan species. However, the chemical investigation of this phylum has provided many compounds of interest as potential anticancer drugs that assume particular importance in the light of the growing literature on cancer biology and chemotherapy. The current review highlights the diversity of chemical structures, mechanisms of action, and, most importantly, the potential of mollusk-derived metabolites as anticancer agents, including those biosynthesized by mollusks and those of dietary origin. After the discussion of dolastatins and kahalalides, compounds previously studied in clinical trials, the review covers potentially promising anticancer agents, which are grouped based on their structural type and include terpenes, steroids, peptides, polyketides and nitrogen-containing compounds. The "promise" of a mollusk-derived natural product as an anticancer agent is evaluated on the basis of its ability to target biological characteristics of cancer cells responsible for poor treatment outcomes. These characteristics include high antiproliferative potency against cancer cells in vitro, preferential inhibition of the proliferation of cancer cells over normal ones, mechanism of action via nonapoptotic signaling pathways, circumvention of multidrug resistance phenotype, and high activity in vivo, among others. The review also includes sections on the targeted delivery of mollusk-derived anticancer agents and solutions to their procurement in quantity.
Collapse
Affiliation(s)
- Maria Letizia Ciavatta
- Consiglio Nazionale delle Ricerche (CNR)Istituto di Chimica Biomolecolare (ICB)Via Campi Flegrei 3480078PozzuoliItaly
| | - Florence Lefranc
- Service de Neurochirurgie, Hôpital ErasmeUniversité Libre de Bruxelles (ULB)1070BrusselsBelgium
| | - Marianna Carbone
- Consiglio Nazionale delle Ricerche (CNR)Istituto di Chimica Biomolecolare (ICB)Via Campi Flegrei 3480078PozzuoliItaly
| | - Ernesto Mollo
- Consiglio Nazionale delle Ricerche (CNR)Istituto di Chimica Biomolecolare (ICB)Via Campi Flegrei 3480078PozzuoliItaly
| | - Margherita Gavagnin
- Consiglio Nazionale delle Ricerche (CNR)Istituto di Chimica Biomolecolare (ICB)Via Campi Flegrei 3480078PozzuoliItaly
| | - Tania Betancourt
- Department of Chemistry and BiochemistryTexas State UniversitySan MarcosTX78666
| | - Ramesh Dasari
- Department of Chemistry and BiochemistryTexas State UniversitySan MarcosTX78666
| | - Alexander Kornienko
- Department of Chemistry and BiochemistryTexas State UniversitySan MarcosTX78666
| | - Robert Kiss
- Laboratoire de Cancérologie et de Toxicologie ExpérimentaleFaculté de Pharmacie, Université Libre de Bruxelles (ULB)1050BrusselsBelgium
| |
Collapse
|
35
|
Jackson SJ, Andrews N, Ball D, Bellantuono I, Gray J, Hachoumi L, Holmes A, Latcham J, Petrie A, Potter P, Rice A, Ritchie A, Stewart M, Strepka C, Yeoman M, Chapman K. Does age matter? The impact of rodent age on study outcomes. Lab Anim 2017; 51:160-169. [PMID: 27307423 PMCID: PMC5367550 DOI: 10.1177/0023677216653984] [Citation(s) in RCA: 200] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Rodent models produce data which underpin biomedical research and non-clinical drug trials, but translation from rodents into successful clinical outcomes is often lacking. There is a growing body of evidence showing that improving experimental design is key to improving the predictive nature of rodent studies and reducing the number of animals used in research. Age, one important factor in experimental design, is often poorly reported and can be overlooked. The authors conducted a survey to assess the age used for a range of models, and the reasoning for age choice. From 297 respondents providing 611 responses, researchers reported using rodents most often in the 6-20 week age range regardless of the biology being studied. The age referred to as 'adult' by respondents varied between six and 20 weeks. Practical reasons for the choice of rodent age were frequently given, with increased cost associated with using older animals and maintenance of historical data comparability being two important limiting factors. These results highlight that choice of age is inconsistent across the research community and often not based on the development or cellular ageing of the system being studied. This could potentially result in decreased scientific validity and increased experimental variability. In some cases the use of older animals may be beneficial. Increased scientific rigour in the choice of the age of rodent may increase the translation of rodent models to humans.
Collapse
Affiliation(s)
- Samuel J Jackson
- National Centre for the Replacement, Refinement and Reduction of Animals in Research, London, UK
| | - Nick Andrews
- Division of Neurology, Kirby Center for Neurobiology, Boston Children’s Hospital, Boston, US
| | - Doug Ball
- Immunoinflammation TAU, GlaxoSmithKline, Stevenage, UK
| | - Ilaria Bellantuono
- Centre for Integrated Research into Musculoskeletal Ageing, University of Sheffield, Sheffield, UK
| | - James Gray
- Immunoinflammation TAU, GlaxoSmithKline, Stevenage, UK
| | - Lamia Hachoumi
- Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, UK
| | - Alan Holmes
- Centre for Rheumatology, UCL Division of Medicine, Royal Free Campus, London, UK
| | - Judy Latcham
- Laboratory Animal Science, GlaxoSmithKline, Stevenage, UK
| | - Anja Petrie
- Rowett Institute of Nutrition & Health, University of Aberdeen, Aberdeen, UK
| | - Paul Potter
- Disease Models and Translation, Mammalian Genetics Unit, MRC Harwell, Harwell, UK
| | - Andrew Rice
- Pain Research, Department of Surgery & Cancer, Imperial College London, London, UK
| | - Alison Ritchie
- Division of Cancer & Stem Cells, University of Nottingham, Nottingham, UK
| | | | | | - Mark Yeoman
- Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, UK
| | - Kathryn Chapman
- National Centre for the Replacement, Refinement and Reduction of Animals in Research, London, UK
| |
Collapse
|
36
|
Domalaon R, Findlay B, Ogunsina M, Arthur G, Schweizer F. Ultrashort cationic lipopeptides and lipopeptoids: Evaluation and mechanistic insights against epithelial cancer cells. Peptides 2016; 84:58-67. [PMID: 27486068 DOI: 10.1016/j.peptides.2016.07.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 07/26/2016] [Accepted: 07/27/2016] [Indexed: 12/12/2022]
Abstract
Peptides present an attractive scaffold for the development of new anticancer lead agents due to their accessibility and ease of modification. Synthetic ultrashort cationic lipopeptides, with four amino acids or less conjugated to a fatty acid, were developed to retain the biological activity of longer peptides in a smaller molecular size. Herein, we report the activity of amphiphilic lipotripeptides, lipotripeptoids and lipotetrapeptides against breast (MDA-MB-231, JIMT-1), prostate (DU145) and pancreas (MiaPaCa2) epithelial cancer cell lines. The lipotripeptide C16-KKK-NH2 and lipotetrapeptide C16-PCatPHexPHexPCat-NH2 were identified to possess anticancer activity. The latter lipotetrapeptide possess a short polyproline scaffold consisting of only two L-4R-aminoproline (PCat) and two L-4R-hexyloxyproline (PHex). However, all the prepared lipotripeptoids lack anticancer activity. The amphiphilic C16-PCatPHexPHexPCat-NH2 exhibited similar anticancer potency to the surfactant benzethonium chloride while superior activity was observed in comparison to myristylamine. Mechanistic studies revealed that the peptides do not lyse ovine erythrocytes nor epithelial cancer cells, thus ruling out necrosis as the mechanism of cell death. Surprisingly, the two lipopeptides exhibit different mechanisms of action that result in cancer cell death. The lipotripeptide C16-KKK-NH2 was found to induce caspase-mediated apoptosis while C16-PCatPHexPHexPCat-NH2 kills tumor cells independent of caspases.
Collapse
Affiliation(s)
- Ronald Domalaon
- Department of Chemistry, University of Manitoba, Winnipeg, Canada
| | - Brandon Findlay
- Department of Chemistry, University of Manitoba, Winnipeg, Canada
| | | | - Gilbert Arthur
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Canada
| | - Frank Schweizer
- Department of Chemistry, University of Manitoba, Winnipeg, Canada; Department of Medical Microbiology, University of Manitoba, Winnipeg, Canada.
| |
Collapse
|
37
|
Fecher D, Hofmann E, Buck A, Bundschuh R, Nietzer S, Dandekar G, Walles T, Walles H, Lückerath K, Steinke M. Human Organotypic Lung Tumor Models: Suitable For Preclinical 18F-FDG PET-Imaging. PLoS One 2016; 11:e0160282. [PMID: 27501455 PMCID: PMC4976941 DOI: 10.1371/journal.pone.0160282] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 07/15/2016] [Indexed: 12/20/2022] Open
Abstract
Development of predictable in vitro tumor models is a challenging task due to the enormous complexity of tumors in vivo. The closer the resemblance of these models to human tumor characteristics, the more suitable they are for drug-development and –testing. In the present study, we generated a complex 3D lung tumor test system based on acellular rat lungs. A decellularization protocol was established preserving the architecture, important ECM components and the basement membrane of the lung. Human lung tumor cells cultured on the scaffold formed cluster and exhibited an up-regulation of the carcinoma-associated marker mucin1 as well as a reduced proliferation rate compared to respective 2D culture. Additionally, employing functional imaging with 2-deoxy-2-[18F]fluoro-D-glucose positron emission tomography (FDG-PET) these tumor cell cluster could be detected and tracked over time. This approach allowed monitoring of a targeted tyrosine kinase inhibitor treatment in the in vitro lung tumor model non-destructively. Surprisingly, FDG-PET assessment of single tumor cell cluster on the same scaffold exhibited differences in their response to therapy, indicating heterogeneity in the lung tumor model. In conclusion, our complex lung tumor test system features important characteristics of tumors and its microenvironment and allows monitoring of tumor growth and -metabolism in combination with functional imaging. In longitudinal studies, new therapeutic approaches and their long-term effects can be evaluated to adapt treatment regimes in future.
Collapse
Affiliation(s)
- David Fecher
- Department of Tissue Engineering and Regenerative Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
- * E-mail:
| | - Elisabeth Hofmann
- Department of Tissue Engineering and Regenerative Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Andreas Buck
- Department of Nuclear Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Ralph Bundschuh
- Department of Nuclear Medicine, University Hospital Bonn, Bonn, Germany
| | - Sarah Nietzer
- Department of Tissue Engineering and Regenerative Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Gudrun Dandekar
- Department of Tissue Engineering and Regenerative Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
- Translational Center ´Regenerative Therapies for Oncology and Musculoskeletal Diseases`Wuerzburg, branch of the Fraunhofer Institute Interfacial Engineering and Biotechnology (IGB), Wuerzburg, Germany
| | - Thorsten Walles
- Department of Cardiothoracic Surgery, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Heike Walles
- Department of Tissue Engineering and Regenerative Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
- Translational Center ´Regenerative Therapies for Oncology and Musculoskeletal Diseases`Wuerzburg, branch of the Fraunhofer Institute Interfacial Engineering and Biotechnology (IGB), Wuerzburg, Germany
| | - Katharina Lückerath
- Department of Nuclear Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Maria Steinke
- Department of Tissue Engineering and Regenerative Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
- Translational Center ´Regenerative Therapies for Oncology and Musculoskeletal Diseases`Wuerzburg, branch of the Fraunhofer Institute Interfacial Engineering and Biotechnology (IGB), Wuerzburg, Germany
| |
Collapse
|
38
|
Perlman RL. Mouse models of human disease: An evolutionary perspective. EVOLUTION MEDICINE AND PUBLIC HEALTH 2016; 2016:170-6. [PMID: 27121451 PMCID: PMC4875775 DOI: 10.1093/emph/eow014] [Citation(s) in RCA: 249] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Accepted: 04/12/2016] [Indexed: 12/21/2022]
Abstract
The use of mice as model organisms to study human biology is predicated on the genetic and physiological similarities between the species. Nonetheless, mice and humans have evolved in and become adapted to different environments and so, despite their phylogenetic relatedness, they have become very different organisms. Mice often respond to experimental interventions in ways that differ strikingly from humans. Mice are invaluable for studying biological processes that have been conserved during the evolution of the rodent and primate lineages and for investigating the developmental mechanisms by which the conserved mammalian genome gives rise to a variety of different species. Mice are less reliable as models of human disease, however, because the networks linking genes to disease are likely to differ between the two species. The use of mice in biomedical research needs to take account of the evolved differences as well as the similarities between mice and humans.
Collapse
Affiliation(s)
- Robert L Perlman
- Department of Pediatrics, The University of Chicago, 5841 S. Maryland Ave, MC 5058, Chicago, IL 60637, USA
| |
Collapse
|
39
|
Satalkar P, Elger BS, Hunziker P, Shaw D. Challenges of clinical translation in nanomedicine: A qualitative study. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 12:893-900. [DOI: 10.1016/j.nano.2015.12.376] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 12/10/2015] [Accepted: 12/22/2015] [Indexed: 11/25/2022]
|
40
|
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: 1.3] [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
|
41
|
|
42
|
Lopez J, Harris S, Roda D, Yap TA. Precision Medicine for Molecularly Targeted Agents and Immunotherapies in Early-Phase Clinical Trials. TRANSLATIONAL ONCOGENOMICS 2015; 7:1-11. [PMID: 26609214 PMCID: PMC4648610 DOI: 10.4137/tog.s30533] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Revised: 10/06/2015] [Accepted: 10/09/2015] [Indexed: 12/12/2022]
Abstract
Precision medicine in oncology promises the matching of genomic, molecular, and clinical data with underlying mechanisms of a range of novel anticancer therapeutics to develop more rational and effective antitumor strategies in a timely manner. However, despite the remarkable progress made in the understanding of novel drivers of different oncogenic processes, success rates for the approval of oncology drugs remain low with substantial fiscal consequences. In this article, we focus on how recent rapid innovations in technology have brought greater clarity to the biological and clinical complexities of different cancers and advanced the development of molecularly targeted agents and immunotherapies in clinical trials. We discuss the key challenges of identifying and validating predictive biomarkers of response and resistance using both tumor and surrogate tissues, as well as the hurdles associated with intratumor heterogeneity. Finally, we outline evolving strategies employed in early-phase trial designs that incorporate omics-based technologies.
Collapse
Affiliation(s)
- Juanita Lopez
- Royal Marsden NHS Foundation Trust, The Institute of Cancer Research, London, UK
| | - Sam Harris
- Royal Marsden NHS Foundation Trust, The Institute of Cancer Research, London, UK
| | - Desam Roda
- Royal Marsden NHS Foundation Trust, The Institute of Cancer Research, London, UK
| | - Timothy A Yap
- Royal Marsden NHS Foundation Trust, The Institute of Cancer Research, London, UK
| |
Collapse
|
43
|
A method to investigate the anti-metabolic activity of anti-cancer agents on ovarian cancer cells cultured in a 96-well high throughput format. J Ovarian Res 2015; 8:43. [PMID: 26141064 PMCID: PMC4491427 DOI: 10.1186/s13048-015-0172-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2014] [Accepted: 06/25/2015] [Indexed: 11/26/2022] Open
Abstract
Background An early step of advanced ovarian cancer begins when floating cancerous cells as single cells or small clusters grow on the peritoneal surface. This surface is rich in extracellular matrix (ECM) proteins, which have profound effects on cellular behaviour and can facilitate cancer progression. Subsequently, this ECM may alter cellular metabolism making cancer cells susceptible to chemotherapeutic agents differently. Therefore, generating a cell culture tool in vitro that includes the interaction between ECM and cancer cells will facilitate our understanding of how cancer cells behave during cancer treatment. There is some evidence to suggest that in an in vitro model that includes ECM components such as collagens will provide a better predictive tool for drug evaluation than a traditional cell monolayer (2D) culture model. Findings As a proof -of- concept, we made a collagen gel in a 96-well plate format and utilised this to evaluate the efficacy of clinical cytotoxic drugs, a targeted drug, and food compounds in single and combination treatments. The primary endpoints were to measure the reduction of cellular metabolism and secretion of vascular endothelial growth factor (VEGF). The invasive capacity of cancer cells was observed in collagen gels and it was cell line-dependent. The responses to drugs were prominently observed in collagen gels, but they had little effect on 2D cell monolayers. These responses were cell line- and type of drug-dependent. Conclusions The collagen gel in a 96 well plate format was easy to set up and could have potential to identify drug sensitivity in the clinical management of women with platinum resistant ovarian cancer.
Collapse
|
44
|
Jin X, Wang Y, Tan L, He Y, Peng J, Hai L, Wu Y, Qian Z. An efficient injectable formulation with block copolymer micelles for hydrophobic antitumor candidate-pyridazinone derivatives. Nanomedicine (Lond) 2015. [PMID: 26214355 DOI: 10.2217/nnm.15.66] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Aim: To make delivery improvements via delivery systems for 6-(4-morpholino-3-(trifluoromethyl)phenyl)pyridazin-3(2H)-one (DZO) – a model compound of hydrophobic antitumor candidate pyridazinone derivatives. Materials & methods: Methoxy poly(ethylene glycol)-poly(d,l-lactide) (MPEG-PDLLA) micelle was employed as a vector, and DZO was encapsulated in. The DZO-loaded micelles were characterized in detail and its cytotoxicity, maximum tolerated dose (MTD) and pharmacokinetic experiments were done. In vivo anticancer activity was studied through a subcutaneous 4T1 tumor model. Results: Compared with free DZO, the DZO-loaded micelles possessed a sustained release property, an improved MTD, better pharmacokinetic parameters and an enhanced antitumor activity for subcutaneous 4T1 model in vivo. Conclusion: An effective injectable delivery system for DZO was developed successfully.
Collapse
Affiliation(s)
- XiuXiu Jin
- State Key Laboratory of Biotherapy & Cancer Center, West China Hospital, Sichuan University, & Collaborative Innovation Center for Biotherapy. Chengdu, China
- Key Laboratory of Drug Targeting of Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - YaLi Wang
- State Key Laboratory of Biotherapy & Cancer Center, West China Hospital, Sichuan University, & Collaborative Innovation Center for Biotherapy. Chengdu, China
| | - LiWei Tan
- State Key Laboratory of Biotherapy & Cancer Center, West China Hospital, Sichuan University, & Collaborative Innovation Center for Biotherapy. Chengdu, China
| | - Yun He
- Key Laboratory of Drug Targeting of Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - JinRong Peng
- State Key Laboratory of Biotherapy & Cancer Center, West China Hospital, Sichuan University, & Collaborative Innovation Center for Biotherapy. Chengdu, China
| | - Li Hai
- Key Laboratory of Drug Targeting of Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Yong Wu
- Key Laboratory of Drug Targeting of Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - ZhiYong Qian
- State Key Laboratory of Biotherapy & Cancer Center, West China Hospital, Sichuan University, & Collaborative Innovation Center for Biotherapy. Chengdu, China
| |
Collapse
|
45
|
Fowler CJ. Delta(9) -tetrahydrocannabinol and cannabidiol as potential curative agents for cancer: A critical examination of the preclinical literature. Clin Pharmacol Ther 2015; 97:587-96. [PMID: 25669486 DOI: 10.1002/cpt.84] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 01/26/2015] [Indexed: 12/13/2022]
Abstract
An Internet search with search words "cannabis cures cancer" produce a wealth of sites claiming that cannabis has this effect. These sites are freely accessible to the general public and thus contribute to public opinion. But do delta(9) -tetrahydrocannabinol (Δ(9) -THC) and cannabidiol (CBD) cure cancer? In the absence of clinical data other than a safety study and case reports, preclinical data should be evaluated in terms of its predictive value. Using a strict approach where only concentrations and/or models relevant to the clinical situation are considered, the current preclinical data do not yet provide robust evidence that systemically administered Δ(9) -THC will be useful for the curative treatment of cancer. There is more support for an intratumoral route of administration of higher doses of Δ(9) -THC. CBD produces effects in relevant concentrations and models, although more data are needed concerning its use in conjunction with other treatment strategies.
Collapse
Affiliation(s)
- C J Fowler
- Department of Pharmacology and Clinical Neuroscience, Umeå University, Umeå, Sweden
| |
Collapse
|
46
|
Miura S, Suzuki H, Bae YH. A Multilayered Cell Culture Model for Transport Study in Solid Tumors: Evaluation of Tissue Penetration of Polyethyleneimine Based Cationic Micelles. NANO TODAY 2014; 9:695-704. [PMID: 25866552 PMCID: PMC4387546 DOI: 10.1016/j.nantod.2014.10.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Limited drug distribution is partially responsible for the efficacy gap between preclinical and clinical studies of nano-sized drug carriers for cancer therapy. In this study, we examined the transport behavior of cationic micelles formed from a triblock copolymer of poly(D,L-lactide-co-glycolide)-block-branched polyethyleneimine-block-poly(D,L-lactide-co-glycolide) using a unique in vitro tumor model composed of a multilayered cell culture (MCC) and an Ussing chamber system. The Cy3-labeled cationic micelles showed remarkable Cy3 distribution in the MCC whereas charge-shielded micelles with a poly(ethylene glycol) surface accumulated on the surface of the MCC. Penetration occurred against convectional flow caused by a hydraulic pressure gradient. The study using fluorescence resonance energy transfer (FRET) showed that the cationic micelles dissociate at the interface between the culture media and the MCC or possibly inside of the first-layer cells and penetrates into the MCC as unimers. The penetration and distribution were energy-dependent and suppressed by various endocytic inhibitors. These suggest that cationic unimers mainly utilized clathrin-mediated endocytosis and macropinocytosis for cellular entry and a significant fraction were exocytosed by an unknown mechanism.
Collapse
Affiliation(s)
- Seiji Miura
- Department of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, University of Utah, 30 S 2000 E, Salt Lake City, UT 84112, United States
- Fuji Research Laboratories, Pharmaceutical Division, Kowa Co. Ltd., 332–1 Ohnoshinden, Fuji, Shizuoka, Japan
| | - Hidenori Suzuki
- Department of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, University of Utah, 30 S 2000 E, Salt Lake City, UT 84112, United States
- Fuji Research Laboratories, Pharmaceutical Division, Kowa Co. Ltd., 332–1 Ohnoshinden, Fuji, Shizuoka, Japan
| | - You Han Bae
- Department of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, University of Utah, 30 S 2000 E, Salt Lake City, UT 84112, United States
| |
Collapse
|
47
|
Fakhraei S, Huang B, Raschid L, Getoor L. Network-Based Drug-Target Interaction Prediction with Probabilistic Soft Logic. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2014; 11:775-787. [PMID: 26356852 DOI: 10.1109/tcbb.2014.2325031] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Drug-target interaction studies are important because they can predict drugs' unexpected therapeutic or adverse side effects. In silico predictions of potential interactions are valuable and can focus effort on in vitro experiments. We propose a prediction framework that represents the problem using a bipartite graph of drug-target interactions augmented with drug-drug and target-target similarity measures and makes predictions using probabilistic soft logic (PSL). Using probabilistic rules in PSL, we predict interactions with models based on triad and tetrad structures. We apply (blocking) techniques that make link prediction in PSL more efficient for drug-target interaction prediction. We then perform extensive experimental studies to highlight different aspects of the model and the domain, first comparing the models with different structures and then measuring the effect of the proposed blocking on the prediction performance and efficiency. We demonstrate the importance of rule weight learning in the proposed PSL model and then show that PSL can effectively make use of a variety of similarity measures. We perform an experiment to validate the importance of collective inference and using multiple similarity measures for accurate predictions in contrast to non-collective and single similarity assumptions. Finally, we illustrate that our PSL model achieves state-of-the-art performance with simple, interpretable rules and evaluate our novel predictions using online data sets.
Collapse
|
48
|
Romero L, Vela JM. Alternative Models in Drug Discovery and Development Part I:In SilicoandIn VitroModels. ACTA ACUST UNITED AC 2014. [DOI: 10.1002/9783527679348.ch02] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
49
|
Abstract
UNLABELLED Creation of lethal and synthetic lethal mutations in an experimental organism is a cornerstone of genetic dissection of gene function, and is related to the concept of an essential gene. Common inbred mouse strains carry background mutations, which can act as genetic modifiers, interfering with the assignment of gene essentiality. The inbred strain C57BL/6J, commonly known as "Black Six", stands out, as it carries a spontaneous homozygous deletion in the nicotinamide nucleotide transhydrogenase (Nnt) gene [GenBank: AH009385.2], resulting in impairment of steroidogenic mitochondria of the adrenal gland, and a multitude of indirect modifier effects, coming from alteration of glucocorticoid-regulated processes. Over time, the popular strain has been used, by means of gene targeting technology, to assign "essential" and "redundant" qualifiers to numerous genes, thus creating an internally consistent "parallel universe" of knowledge. It is unrealistic to suggest phasing-out of this strain, given the scope of shared resources built around it, however, continuing on the road of "strain-unawareness" will result in profound waste of effort, particularly where translational research is concerned. The review analyzes the historical roots of this phenomenon and proposes that building of "parallel universes" should be urgently made visible to a critical reader by obligatory use of unambiguous and persistent tags in publications and databases, such as hypertext links, pointing to a vendor's strain description web page, or to a digital object identifier (d.o.i.) of the original publication, so that any research done exclusively in C57BL/6J, could be easily identified. REVIEWERS This article was reviewed by Dr. Neil Smalheiser and Dr. Miguel Andrade-Navarro.
Collapse
Affiliation(s)
- Alexander Kraev
- Charles H, Best Institute, Banting and Best Department of Medical Research, University of Toronto, Toronto, Ontario M5G 1L6, Canada.
| |
Collapse
|
50
|
The influence of monoacylglycerol lipase inhibition upon the expression of epidermal growth factor receptor in human PC-3 prostate cancer cells. BMC Res Notes 2014; 7:441. [PMID: 25012825 PMCID: PMC4109781 DOI: 10.1186/1756-0500-7-441] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 06/28/2014] [Indexed: 12/20/2022] Open
Abstract
Background It has been reported that direct activation of the cannabinoid CB1 receptor in epidermal growth factor (EGR)-stimulated PC-3 prostate cancer cells results in an anti-proliferative effect accompanied by a down-regulation of EGF receptors (EGFR). In the present study, we investigated whether similar effects are seen following inhibition of the endocannabinoid hydrolytic enzyme monoacylglycerol lipase (MGL). Results CB1 receptor expression levels were found to differ greatly between two experimental series conducted using PC-3 cells. The monoacylglycerol lipase inhibitor JZL184 increased levels of 2-arachidonoylglycerol in the PC-3 cells without producing changes in the levels of anandamide and related N-acylethanolamines. In the first series of experiments, JZL184 produced a small mitogenic effect for cells that had not been treated with EGF, whereas an anti-proliferative effect was seen for EGF-treated cells. An anti-proliferative effect for the EGF-treated cells was also seen with the CB receptor agonist CP55,940. In the second batch of cells, there was an interaction between JZL184 and CB1 receptor expression densities in linear regression analyses with EGFR expression as the dependent variable. Conclusions Inhibition of MGL by JZL184 can affect EGFR expression. However, the use in our hands of PC-3 cells as a model to investigate the therapeutic potential of MGL inhibitors and related compounds is compromised by their variability of CB1 receptor expression.
Collapse
|