1
|
Saeidi T, Wang S, Contreras HA, Daly MJ, Betz V, Lilge L. Photosensitizer spatial heterogeneity and its impact on personalized interstitial photodynamic therapy treatment planning. JOURNAL OF BIOMEDICAL OPTICS 2025; 30:018001. [PMID: 39802351 PMCID: PMC11724368 DOI: 10.1117/1.jbo.30.1.018001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 11/21/2024] [Accepted: 11/25/2024] [Indexed: 01/16/2025]
Abstract
Significance Personalized photodynamic therapy (PDT) treatment planning requires knowledge of the spatial and temporal co-localization of photons, photosensitizers (PSs), and oxygen. The inter- and intra-subject variability in the photosensitizer concentration can lead to suboptimal outcomes using standard treatment plans. Aim We aim to quantify the PS spatial variation in tumors and its effect on PDT treatment planning solutions. Approach The spatial variability of two PSs is imaged at various spatial resolutions for an orthotopic rat glioma model and applied in silico to human glioblastoma models to determine the spatial PDT dose, including in organs at risk. An open-source interstitial photodynamic therapy (iPDT) planning tool is applied to these models, deriving the spatial photosensitizer quantification resolution that consistently impacts iPDT source placement and power allocation. Results The ex vivo studies revealed a bimodal photosensitizer distribution in the tumor. The concentration of the PS can vary by a factor of 2 between the tumor core and rim, with slight variation within the core but a factor of 5 in the rim. An average sampling volume of 1 mm 3 for photosensitizer quantification will result in significantly different iPDT planning solutions for each case. Conclusions Assuming homogeneous photosensitizer distribution results in suboptimal therapeutic outcomes, we highlight the need to predict the photosensitizer distribution before source placement for effective treatment plans.
Collapse
Affiliation(s)
- Tina Saeidi
- University of Toronto, University Health Network, Princess Margaret Cancer Centre, Department of Medical Biophysics, Toronto, Ontario, Canada
| | - Shuran Wang
- University of Toronto, Edward S. Rogers Sr. Department of Electrical and Computer Engineering, Toronto, Ontario, Canada
| | - Hector A. Contreras
- University of Toronto, University Health Network, Princess Margaret Cancer Centre, Department of Medical Biophysics, Toronto, Ontario, Canada
| | - Michael J. Daly
- University Health Network, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Vaughn Betz
- University of Toronto, Edward S. Rogers Sr. Department of Electrical and Computer Engineering, Toronto, Ontario, Canada
| | - Lothar Lilge
- University of Toronto, University Health Network, Princess Margaret Cancer Centre, Department of Medical Biophysics, Toronto, Ontario, Canada
- University Health Network, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| |
Collapse
|
2
|
Radwan AM, Abosharaf HA, Sharaky M, Abdelmonem R, Effat H. Functional combination of resveratrol and tamoxifen to overcome tamoxifen-resistance in breast cancer cells. Arch Pharm (Weinheim) 2024; 357:e2400261. [PMID: 38943449 DOI: 10.1002/ardp.202400261] [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: 04/08/2024] [Revised: 06/10/2024] [Accepted: 06/12/2024] [Indexed: 07/01/2024]
Abstract
Researchers are encountering challenges in addressing the issue of cancer cells becoming unresponsive to various chemotherapy treatments due to drug resistance. This study was designed to study the influence of antioxidant resveratrol (RSV) to sensitize resistant breast cancer (BC) cells toward tamoxifen (TAM). The cytotoxic effects of RSV and TAM against TAM-resistant LCC2 cells and their parental michigan cancer foundation-7 BC cells were determined by sulphorhodamine B assay. Further, the expression levels of multidrug resistance (MDR) genes including ABCB1, ABCC2, ABCG2, and MRP1 using quantitative polymerase chain reaction, apoptosis induction, and reactive oxygen species (ROS) content using flow cytometry were evaluated in either LCC2 cells treated with RSV, TAM, or their combination. The obtained results showed that resistant cells have a magnificent level of MDR genes. This elevated expression dramatically lowered upon receiving the combined therapy of RSV and TAM. Additionally, our work assessed the possible role of RSV in modulating the expression of MDR genes by controlling the expression of certain microRNAs (miRNAs) that target ATP-binding cassette (ABC) transporters. According to the obtained data, the TAM and RSV combination increased the expression of tumor inhibitor miRNAs such miR-10b-3p, miR-195-3p, and miR-223-3p, which made LCC2 cells more sensitive to TAM. Furthermore, this combination showed an elevation in apoptotic levels and total ROS content. The combination between RSV and TAM could be a functional therapy in the fight against TAM-resistant BC cells via modulating miRNA and ABC transporters.
Collapse
Affiliation(s)
- Aliaa M Radwan
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, Egypt
| | - Hamed A Abosharaf
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, Egypt
| | - Marwa Sharaky
- Pharmacology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Rehab Abdelmonem
- Department of Industrial Pharmacy, Faculty of Pharmacy, Misr University for Science & Technology, 6th October City, Egypt
| | - Heba Effat
- Medical Biochemistry and Molecular Biology Unit, Department of Cancer Biology, National Cancer Institute, Cairo University, Cairo, Egypt
| |
Collapse
|
3
|
Perelló-Trias MT, Serrano-Muñoz AJ, Rodríguez-Fernández A, Segura-Sampedro JJ, Ramis JM, Monjo M. Intraperitoneal drug delivery systems for peritoneal carcinomatosis: Bridging the gap between research and clinical implementation. J Control Release 2024; 373:70-92. [PMID: 38986910 DOI: 10.1016/j.jconrel.2024.07.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 07/05/2024] [Accepted: 07/06/2024] [Indexed: 07/12/2024]
Abstract
Several abdominal-located cancers develop metastasis within the peritoneum, what is called peritoneal carcinomatosis (PC), constituting a clinical challenge in their therapeutical management, often leading to poor prognoses. Current multidisciplinary strategies, including cytoreductive surgery (CRS), hyperthermic intraperitoneal chemotherapy (HIPEC), and pressurized intraperitoneal aerosol chemotherapy (PIPAC), demonstrate efficacy but have limitations. In response, alternative strategies are explored in the drug delivery field for intraperitoneal chemotherapy. Controlled drug delivery offers a promising avenue, maintaining localized drug concentrations for optimal PC management. Drug delivery systems (DDS), including hydrogels, implants, nanoparticles, and hybrid systems, show potential for sustained and region-specific drug release. The present review aims to offer an overview of the advances and current designs of DDS for PC chemotherapy administration, focusing on their composition, main characteristics, and principal experimental outcomes, highlighting the importance of biomaterial rationale design and in vitro/vivo models for their testing. Moreover, since clinical data for human subjects are scarce, we offer a critical discussion of the gap between bench and bedside in DDS translation, emphasizing the need for further research.
Collapse
Affiliation(s)
- M Teresa Perelló-Trias
- Cell Therapy and Tissue Engineering Group (TERCIT), Research Institute of Health Sciences (IUNICS), University of the Balearic Islands (UIB), Carretera de Valldemossa, Km 7,5, 07122 Palma, Balearic Islands, Spain; Health Research Institute of the Balearic Islands (IdISBa) - Carretera de Valldemossa, 79. Hospital Universitari Son Espases. Edifici S. 07120 Palma, Balearic Islands, Spain; Department of Fundamental Biology and Health Sciences, University of the Balearic Islands (UIB), Palma, Balearic Islands, Spain
| | - Antonio Jose Serrano-Muñoz
- Cell Therapy and Tissue Engineering Group (TERCIT), Research Institute of Health Sciences (IUNICS), University of the Balearic Islands (UIB), Carretera de Valldemossa, Km 7,5, 07122 Palma, Balearic Islands, Spain; Health Research Institute of the Balearic Islands (IdISBa) - Carretera de Valldemossa, 79. Hospital Universitari Son Espases. Edifici S. 07120 Palma, Balearic Islands, Spain; Department of Fundamental Biology and Health Sciences, University of the Balearic Islands (UIB), Palma, Balearic Islands, Spain
| | - Ana Rodríguez-Fernández
- Cell Therapy and Tissue Engineering Group (TERCIT), Research Institute of Health Sciences (IUNICS), University of the Balearic Islands (UIB), Carretera de Valldemossa, Km 7,5, 07122 Palma, Balearic Islands, Spain; Health Research Institute of the Balearic Islands (IdISBa) - Carretera de Valldemossa, 79. Hospital Universitari Son Espases. Edifici S. 07120 Palma, Balearic Islands, Spain; Department of Fundamental Biology and Health Sciences, University of the Balearic Islands (UIB), Palma, Balearic Islands, Spain
| | - Juan José Segura-Sampedro
- Health Research Institute of the Balearic Islands (IdISBa) - Carretera de Valldemossa, 79. Hospital Universitari Son Espases. Edifici S. 07120 Palma, Balearic Islands, Spain; General & Digestive Surgery Service, Hospital Universitario La Paz, Paseo de la Castellana, 261, Fuencarral-El Pardo, 28046 Madrid, Spain; School of Medicine, University of the Balearic Islands (UIB), Carretera de Valldemossa, km 7,5, 07122 Palma, Balearic Islands, Spain
| | - Joana Maria Ramis
- Cell Therapy and Tissue Engineering Group (TERCIT), Research Institute of Health Sciences (IUNICS), University of the Balearic Islands (UIB), Carretera de Valldemossa, Km 7,5, 07122 Palma, Balearic Islands, Spain; Health Research Institute of the Balearic Islands (IdISBa) - Carretera de Valldemossa, 79. Hospital Universitari Son Espases. Edifici S. 07120 Palma, Balearic Islands, Spain; Department of Fundamental Biology and Health Sciences, University of the Balearic Islands (UIB), Palma, Balearic Islands, Spain.
| | - Marta Monjo
- Cell Therapy and Tissue Engineering Group (TERCIT), Research Institute of Health Sciences (IUNICS), University of the Balearic Islands (UIB), Carretera de Valldemossa, Km 7,5, 07122 Palma, Balearic Islands, Spain; Health Research Institute of the Balearic Islands (IdISBa) - Carretera de Valldemossa, 79. Hospital Universitari Son Espases. Edifici S. 07120 Palma, Balearic Islands, Spain; Department of Fundamental Biology and Health Sciences, University of the Balearic Islands (UIB), Palma, Balearic Islands, Spain.
| |
Collapse
|
4
|
Kiaheyrati N, Babaei A, Ranji R, Bahadoran E, Taheri S, Farokhpour Z. Cancer therapy with the viral and bacterial pathogens: The past enemies can be considered the present allies. Life Sci 2024; 349:122734. [PMID: 38788973 DOI: 10.1016/j.lfs.2024.122734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024]
Abstract
Cancer continues to be one of the leading causes of mortality worldwide despite significant advancements in cancer treatment. Many difficulties have arisen as a result of the detrimental consequences of chemotherapy and radiotherapy as a common cancer therapy, such as drug inability to penetrate deep tumor tissue, and also the drug resistance in tumor cells continues to be a major concern. These obstacles have increased the need for the development of new techniques that are more selective and effective against cancer cells. Bacterial-based therapies and the use of oncolytic viruses can suppress cancer in comparison to other cancer medications. The tumor microenvironment is susceptible to bacterial accumulation and proliferation, which can trigger immune responses against the tumor. Oncolytic viruses (OVs) have also gained considerable attention in recent years because of their potential capability to selectively target and induce apoptosis in cancer cells. This review aims to provide a comprehensive summary of the latest literature on the role of bacteria and viruses in cancer treatment, discusses the limitations and challenges, outlines various strategies, summarizes recent preclinical and clinical trials, and emphasizes the importance of optimizing current strategies for better clinical outcomes.
Collapse
Affiliation(s)
- Niloofar Kiaheyrati
- Medical Microbiology Research Center, Qazvin University of Medical Sciences, Qazvin, Iran; Department of Microbiology and Immunology, School of Medicine, Qazvin University of Medical Science, Qazvin, Iran
| | - Abouzar Babaei
- Medical Microbiology Research Center, Qazvin University of Medical Sciences, Qazvin, Iran; Department of Microbiology and Immunology, School of Medicine, Qazvin University of Medical Science, Qazvin, Iran.
| | - Reza Ranji
- Department of Genetics, Faculty of Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ensiyeh Bahadoran
- School of Medicine, Qazvin University of Medical Science, Qazvin, Iran
| | - Shiva Taheri
- Medical Microbiology Research Center, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Zahra Farokhpour
- Medical Microbiology Research Center, Qazvin University of Medical Sciences, Qazvin, Iran
| |
Collapse
|
5
|
Mohr P, van Sluis J, Lub-de Hooge MN, Lammertsma AA, Brouwers AH, Tsoumpas C. Advances and challenges in immunoPET methodology. FRONTIERS IN NUCLEAR MEDICINE (LAUSANNE, SWITZERLAND) 2024; 4:1360710. [PMID: 39355220 PMCID: PMC11440922 DOI: 10.3389/fnume.2024.1360710] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 02/05/2024] [Indexed: 10/03/2024]
Abstract
Immuno-positron emission tomography (immunoPET) enables imaging of specific targets that play a role in targeted therapy and immunotherapy, such as antigens on cell membranes, targets in the disease microenvironment, or immune cells. The most common immunoPET applications use a monoclonal antibody labeled with a relatively long-lived positron emitter such as 89Zr (T 1/2 = 78.4 h), but smaller antibody-based constructs labeled with various other positron emitting radionuclides are also being investigated. This molecular imaging technique can thus guide the development of new drugs and may have a pivotal role in selecting patients for a particular therapy. In early phase immunoPET trials, multiple imaging time points are used to examine the time-dependent biodistribution and to determine the optimal imaging time point, which may be several days after tracer injection due to the slow kinetics of larger molecules. Once this has been established, usually only one static scan is performed and semi-quantitative values are reported. However, total PET uptake of a tracer is the sum of specific and nonspecific uptake. In addition, uptake may be affected by other factors such as perfusion, pre-/co-administration of the unlabeled molecule, and the treatment schedule. This article reviews imaging methodologies used in immunoPET studies and is divided into two parts. The first part summarizes the vast majority of clinical immunoPET studies applying semi-quantitative methodologies. The second part focuses on a handful of studies applying pharmacokinetic models and includes preclinical and simulation studies. Finally, the potential and challenges of immunoPET quantification methodologies are discussed within the context of the recent technological advancements provided by long axial field of view PET/CT scanners.
Collapse
Affiliation(s)
- Philipp Mohr
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Joyce van Sluis
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Marjolijn N Lub-de Hooge
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Adriaan A Lammertsma
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Adrienne H Brouwers
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Charalampos Tsoumpas
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
6
|
Riccardi F, Dal Bo M, Macor P, Toffoli G. A comprehensive overview on antibody-drug conjugates: from the conceptualization to cancer therapy. Front Pharmacol 2023; 14:1274088. [PMID: 37790810 PMCID: PMC10544916 DOI: 10.3389/fphar.2023.1274088] [Citation(s) in RCA: 67] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 09/07/2023] [Indexed: 10/05/2023] Open
Abstract
Antibody-Drug Conjugates (ADCs) represent an innovative class of potent anti-cancer compounds that are widely used in the treatment of hematologic malignancies and solid tumors. Unlike conventional chemotherapeutic drug-based therapies, that are mainly associated with modest specificity and therapeutic benefit, the three key components that form an ADC (a monoclonal antibody bound to a cytotoxic drug via a chemical linker moiety) achieve remarkable improvement in terms of targeted killing of cancer cells and, while sparing healthy tissues, a reduction in systemic side effects caused by off-tumor toxicity. Based on their beneficial mechanism of action, 15 ADCs have been approved to date by the market approval by the Food and Drug Administration (FDA), the European Medicines Agency (EMA) and/or other international governmental agencies for use in clinical oncology, and hundreds are undergoing evaluation in the preclinical and clinical phases. Here, our aim is to provide a comprehensive overview of the key features revolving around ADC therapeutic strategy including their structural and targeting properties, mechanism of action, the role of the tumor microenvironment and review the approved ADCs in clinical oncology, providing discussion regarding their toxicity profile, clinical manifestations and use in novel combination therapies. Finally, we briefly review ADCs in other pathological contexts and provide key information regarding ADC manufacturing and analytical characterization.
Collapse
Affiliation(s)
- Federico Riccardi
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico (CRO), IRCCS, Aviano, Italy
| | - Michele Dal Bo
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico (CRO), IRCCS, Aviano, Italy
| | - Paolo Macor
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico (CRO), IRCCS, Aviano, Italy
| |
Collapse
|
7
|
Xu W, Du L, Yu L, Cen H, Lin F, Wang S, Ruan Z, Lin Z, Zhang X, Zhou N, Chang J, Yu X, Zhang L, Liang L. The mirrored cationic peptide as miRNA vehicle for efficient lung cancer therapy. MedComm (Beijing) 2023; 4:e273. [PMID: 37521428 PMCID: PMC10382604 DOI: 10.1002/mco2.273] [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: 11/27/2022] [Revised: 04/14/2023] [Accepted: 04/20/2023] [Indexed: 08/01/2023] Open
Abstract
Gene therapy has emerged as a potential approach for lung cancer therapy. However, the application of gene therapy is still limited by their properties, such as low specificity to the cancer cells, negatively charged groups, short systemic circulation time, and rapid degradation by nucleases. The progression of lung adenocarcinoma (LUAD) can be promoted through the methylation process of miR-148a-3p promoter, as confirmed by our previous research. In the current study, we are the first to design a mirrored Arg-Gly-Asp (RGD)-modified cationic peptide (RD24) as a microRNA (miRNA) vehicle, which enabled to pack the miRNA (miR-148a-3p) efficiently and generate RD24/miR-148a-3p nanoparticles (RPRIN) by self-assembling. RPRIN exhibited a high transfection efficiency in lung cancer cells via the conjugation between RGD and integrins on the surface of lung cancer cells. Furthermore, RD24 showed matrix metallopeptidase 2 (MMP2) responsiveness, which improved lung cancer cell inhibition induced by the miRNA intracellularly. In addition, RPRIN exhibits several advantages, such as prolonged circulation duration, reduced toxicity, and immune escape. Experiments conducted both in vitro and in vivo revealed that RPRIN effectively suppressed the growth and progression of lung cancer. Thus, the mirrored RGD-modified cationic peptide showed great potential in transducing miRNA for lung cancer therapy.
Collapse
Affiliation(s)
- Wenyan Xu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated HospitalGuangzhou Medical UniversityGuangzhouPR China
| | - Lingran Du
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated HospitalGuangzhou Medical UniversityGuangzhouPR China
| | - Lina Yu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated HospitalGuangzhou Medical UniversityGuangzhouPR China
- Department of Preventive Dentistry, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and ReconstructionGuangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative MedicineGuangzhouPR China
| | - Huiyu Cen
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated HospitalGuangzhou Medical UniversityGuangzhouPR China
| | - Fangyu Lin
- Department of OphthalmologyEmory UniversityAtlantaGeorgiaUSA
| | - Siran Wang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated HospitalGuangzhou Medical UniversityGuangzhouPR China
- Department of Preventive Dentistry, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and ReconstructionGuangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative MedicineGuangzhouPR China
| | - Zhixiong Ruan
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated HospitalGuangzhou Medical UniversityGuangzhouPR China
| | - Zhongxiao Lin
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated HospitalGuangzhou Medical UniversityGuangzhouPR China
- State Key Laboratory of Quality Research in Chinese MedicineMacau University of Science and TechnologyAvenida WailongTaipaMacauPR China
| | - Xin Zhang
- State Key Laboratory of Quality Research in Chinese MedicineMacau University of Science and TechnologyAvenida WailongTaipaMacauPR China
| | - Na Zhou
- State Key Laboratory of Quality Research in Chinese MedicineMacau University of Science and TechnologyAvenida WailongTaipaMacauPR China
| | - Jishuo Chang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated HospitalGuangzhou Medical UniversityGuangzhouPR China
| | - Xiyong Yu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated HospitalGuangzhou Medical UniversityGuangzhouPR China
| | - Lingmin Zhang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated HospitalGuangzhou Medical UniversityGuangzhouPR China
| | - Lu Liang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated HospitalGuangzhou Medical UniversityGuangzhouPR China
| |
Collapse
|
8
|
Breusa S, Zilio S, Catania G, Bakrin N, Kryza D, Lollo G. Localized chemotherapy approaches and advanced drug delivery strategies: a step forward in the treatment of peritoneal carcinomatosis from ovarian cancer. Front Oncol 2023; 13:1125868. [PMID: 37287910 PMCID: PMC10242058 DOI: 10.3389/fonc.2023.1125868] [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: 12/16/2022] [Accepted: 05/04/2023] [Indexed: 06/09/2023] Open
Abstract
Peritoneal carcinomatosis (PC) is a common outcome of epithelial ovarian carcinoma and is the leading cause of death for these patients. Tumor location, extent, peculiarities of the microenvironment, and the development of drug resistance are the main challenges that need to be addressed to improve therapeutic outcome. The development of new procedures such as HIPEC (Hyperthermic Intraperitoneal Chemotherapy) and PIPAC (Pressurized Intraperitoneal Aerosol Chemotherapy) have enabled locoregional delivery of chemotherapeutics, while the increasingly efficient design and development of advanced drug delivery micro and nanosystems are helping to promote tumor targeting and penetration and to reduce the side effects associated with systemic chemotherapy administration. The possibility of combining drug-loaded carriers with delivery via HIPEC and PIPAC represents a powerful tool to improve treatment efficacy, and this possibility has recently begun to be explored. This review will discuss the latest advances in the treatment of PC derived from ovarian cancer, with a focus on the potential of PIPAC and nanoparticles in terms of their application to develop new therapeutic strategies and future prospects.
Collapse
Affiliation(s)
- Silvia Breusa
- Univ Lyon, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique (CNRS), LAGEPP Unité Mixte de Recherche (UMR) 5007, Villeurbanne, France
- Apoptosis, Cancer and Development Laboratory- Equipe labellisée ‘La Ligue’, LabEx DEVweCAN, Institut PLAsCAN, Centre de Recherche en Cancérologie de Lyon, Institut national de santé et de la recherche médicale (INSERM) U1052-Centre National de la Recherche Scientifique - Unité Mixte de Recherche (CNRS UMR)5286, Université de Lyon, Centre Léon Bérard, Lyon, France
| | - Serena Zilio
- Univ Lyon, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique (CNRS), LAGEPP Unité Mixte de Recherche (UMR) 5007, Villeurbanne, France
- Sociétés d'Accélération du Transfert de Technologies (SATT) Ouest Valorisation, Rennes, France
| | - Giuseppina Catania
- Univ Lyon, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique (CNRS), LAGEPP Unité Mixte de Recherche (UMR) 5007, Villeurbanne, France
| | - Naoual Bakrin
- Department of Surgical Oncology, Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, Lyon, France
- Centre pour l'Innovation en Cancérologie de Lyon (CICLY), Claude Bernard University Lyon 1, Lyon, France
| | - David Kryza
- Univ Lyon, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique (CNRS), LAGEPP Unité Mixte de Recherche (UMR) 5007, Villeurbanne, France
- Imthernat Plateform, Hospices Civils de Lyon, Lyon, France
| | - Giovanna Lollo
- Univ Lyon, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique (CNRS), LAGEPP Unité Mixte de Recherche (UMR) 5007, Villeurbanne, France
| |
Collapse
|
9
|
Kekani LN, Witika BA. Current advances in nanodrug delivery systems for malaria prevention and treatment. DISCOVER NANO 2023; 18:66. [PMID: 37382765 PMCID: PMC10409709 DOI: 10.1186/s11671-023-03849-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 04/13/2023] [Indexed: 06/30/2023]
Abstract
Malaria is a life-threatening, blood-borne disease with over two hundred million cases throughout the world and is more prevalent in Sub-Saharan Africa than anywhere else in the world. Over the years, several treatment agents have been developed for malaria; however, most of these active pharmaceutical ingredients exhibit poor aqueous solubility and low bioavailability and may result in drug-resistant parasites, thus increasing malaria cases and eventually, deaths. Factors such as these in therapeutics have led to a better appreciation of nanomaterials. The ability of nanomaterials to function as drug carriers with a high loading capacity and targeted drug delivery, good biocompatibility, and low toxicity renders them an appealing alternative to conventional therapy. Nanomaterials such as dendrimers and liposomes have been demonstrated to be capable of enhancing the efficacy of antimalarial drugs. This review discusses the recent development of nanomaterials and their benefits in drug delivery for the potential treatment of malaria.
Collapse
Affiliation(s)
- Linda N Kekani
- Department of Pharmaceutical Sciences, School of Pharmacy, Sefako Makgatho Health Sciences University, Pretoria, 0208, South Africa
| | - Bwalya A Witika
- Department of Pharmaceutical Sciences, School of Pharmacy, Sefako Makgatho Health Sciences University, Pretoria, 0208, South Africa.
| |
Collapse
|
10
|
de Maar JS, Zandvliet MMJM, Veraa S, Tobón Restrepo M, Moonen CTW, Deckers R. Ultrasound and Microbubbles Mediated Bleomycin Delivery in Feline Oral Squamous Cell Carcinoma—An In Vivo Veterinary Study. Pharmaceutics 2023; 15:pharmaceutics15041166. [PMID: 37111651 PMCID: PMC10142092 DOI: 10.3390/pharmaceutics15041166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/06/2023] [Accepted: 03/31/2023] [Indexed: 04/09/2023] Open
Abstract
To investigate the feasibility and tolerability of ultrasound and microbubbles (USMB)-enhanced chemotherapy delivery for head and neck cancer, we performed a veterinary trial in feline companion animals with oral squamous cell carcinomas. Six cats were treated with a combination of bleomycin and USMB therapy three times, using the Pulse Wave Doppler mode on a clinical ultrasound system and EMA/FDA approved microbubbles. They were evaluated for adverse events, quality of life, tumour response and survival. Furthermore, tumour perfusion was monitored before and after USMB therapy using contrast-enhanced ultrasound (CEUS). USMB treatments were feasible and well tolerated. Among 5 cats treated with optimized US settings, 3 had stable disease at first, but showed disease progression 5 or 11 weeks after first treatment. One cat had progressive disease one week after the first treatment session, maintaining a stable disease thereafter. Eventually, all cats except one showed progressive disease, but each survived longer than the median overall survival time of 44 days reported in literature. CEUS performed immediately before and after USMB therapy suggested an increase in tumour perfusion based on an increase in median area under the curve (AUC) in 6 out of 12 evaluated treatment sessions. In this small hypothesis-generating study, USMB plus chemotherapy was feasible and well-tolerated in a feline companion animal model and showed potential for enhancing tumour perfusion in order to increase drug delivery. This could be a forward step toward clinical translation of USMB therapy to human patients with a clinical need for locally enhanced treatment.
Collapse
Affiliation(s)
- Josanne S. de Maar
- Imaging and Oncology Division, University Medical Center Utrecht, Utrecht University, 3508 GA Utrecht, The Netherlands
| | - Maurice M. J. M. Zandvliet
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, The Netherlands
| | - Stefanie Veraa
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, The Netherlands
| | - Mauricio Tobón Restrepo
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, The Netherlands
| | - Chrit T. W. Moonen
- Imaging and Oncology Division, University Medical Center Utrecht, Utrecht University, 3508 GA Utrecht, The Netherlands
| | - Roel Deckers
- Imaging and Oncology Division, University Medical Center Utrecht, Utrecht University, 3508 GA Utrecht, The Netherlands
| |
Collapse
|
11
|
Soleimani N, Javadi MM. Future prospects of bacteria-mediated cancer therapies: Affliction or opportunity? Microb Pathog 2022; 172:105795. [PMID: 36155065 DOI: 10.1016/j.micpath.2022.105795] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/05/2022] [Accepted: 09/19/2022] [Indexed: 01/10/2023]
Abstract
Cancer, as a disease characterized by uncontrolled growth of cells, is recognized as one of the significant challenges in the field of health and medicine. There are various treatments for cancer like surgery, hormone therapy, chemotherapy, etc., but they have negative effects on the patient's lifestyle. Numerous side effects, and recently the emergence of drug resistance to these methods are weaknesses of these treatments. The utilization of bacteria as a treatment for cancer has attracted scientists' attention in the last decade. There are various methods of using bacteria to treat cancer, including the use of live, attenuated, or genetically engineered microbes, the use of bacterial toxins as an immunotoxin or conjugated to tumor antigens, bacteria-based cancer immunotherapy, bacterial vectors for gene-directed enzyme prodrug, and also the undeniable role of probiotics in treatment, are the cases that today are used for treatment. Bacterial therapy has shown a greater promise in cancer treatment due to its ability to lyse the tumor cells and deliver therapeutic products. However, the potential cytotoxicity of bacteria for healthy tissues, their inability to entirely lyse cancerous cells, and the possibility of mutations in their genomes are among the challenges of bacteriotherapy for cancer. Herein, we summarize the mechanism of bacteria, their potential benefits and harms, and the future of research in this field.
Collapse
Affiliation(s)
- Neda Soleimani
- Department of Microbiology and Microbial Biotechnology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran.
| | - Mahtab Moshref Javadi
- Department of Microbiology and Microbial Biotechnology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran.
| |
Collapse
|
12
|
Rommasi F. Bacterial-Based Methods for Cancer Treatment: What We Know and Where We Are. Oncol Ther 2022; 10:23-54. [PMID: 34780046 PMCID: PMC9098760 DOI: 10.1007/s40487-021-00177-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 10/25/2021] [Indexed: 01/10/2023] Open
Abstract
A severe disease, cancer is caused by the exponential and uncontrolled growth of cells, leading to organ dysfunction as well as disorders. This disease has been recognized as one of the significant challenges to health and medicine. Various treatment procedures for cancer are associated with diverse side effects; the most conventional cancer treatments include chemotherapy, surgery, and radiotherapy, among others. Numerous adverse and side effects, low specificity and sensitivity, narrow therapeutic windows, and, recently, the emergence of tumor cells resistant to such treatments have been documented as the shortcomings of conventional treatment strategies. As a group of prokaryotic microorganisms, bacteria have great potential for use in cancer therapy. Currently, utilizing bacteria for cancer treatment has attracted the attention of scientists. The high potential of bacteria to become non-pathogenic by genetic manipulation, their distinguished virulence factors (which can be used as weapons against tumors), their ability to proliferate in tissues, and the contingency to control their population by administrating antibiotics, etc., have made bacteria viable candidates and live micro-medication for cancer therapies. However, the possible cytotoxicity impacts of bacteria, their inability to entirely lyse cancerous cells, as well as the probability of mutations in their genomes are among the significant challenges of bacteria-based methods for cancer treatment. In this article, various available data on bacterial therapeutics, along with their pros and cons, are discussed.
Collapse
Affiliation(s)
- Foad Rommasi
- Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran.
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
13
|
Wu H, Li W, Hao M, Wang Y, Xue L, Ju C, Zhang C. An EPR-Independent extravasation Strategy: Deformable leukocytes as vehicles for improved solid tumor therapy. Adv Drug Deliv Rev 2022; 187:114380. [PMID: 35662610 DOI: 10.1016/j.addr.2022.114380] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 05/20/2022] [Accepted: 05/27/2022] [Indexed: 02/07/2023]
Abstract
Effective delivery of therapeutic modality throughout the tumorous nidus plays a crucial role in successful solid tumor treatment. However, conventional nanomedicines based on enhanced permeability and retention (EPR) effect have yielded limited delivery/therapeutic efficiency, due mainly to the heterogeneity of the solid tumor. Leukocytes, which could intrinsically migrate across the vessel wall and crawl through tissue interstitium in a self-deformable manner, have currently emerged as an alternative drug delivery vehicle. In this review, we start with the intrinsic properties of leukocytes (e.g., extravasation and crawling inside tumor), focusing on unveiling the conceptual rationality of leveraging leukocytes as EPR-independent delivery vehicles. Then we discussed various cargoes-loading/unloading strategies for leukocyte-based vehicles as well as their promising applications. This review aims to serve as an up-to-date compilation, which might provide inspiration for scientists in the field of drug delivery.
Collapse
|
14
|
de Brito RV, Mancini MW, Palumbo MDN, de Moraes LHO, Rodrigues GJ, Cervantes O, Sercarz JA, Paiva MB. The Rationale for "Laser-Induced Thermal Therapy (LITT) and Intratumoral Cisplatin" Approach for Cancer Treatment. Int J Mol Sci 2022; 23:5934. [PMID: 35682611 PMCID: PMC9180481 DOI: 10.3390/ijms23115934] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/19/2022] [Accepted: 05/20/2022] [Indexed: 01/27/2023] Open
Abstract
Cisplatin is one of the most widely used anticancer drugs in the treatment of various types of solid human cancers, as well as germ cell tumors, sarcomas, and lymphomas. Strong evidence from research has demonstrated higher efficacy of a combination of cisplatin and derivatives, together with hyperthermia and light, in overcoming drug resistance and improving tumoricidal efficacy. It is well known that the antioncogenic potential of CDDP is markedly enhanced by hyperthermia compared to drug treatment alone. However, more recently, accelerators of high energy particles, such as synchrotrons, have been used to produce powerful and monochromatizable radiation to induce an Auger electron cascade in cis-platinum molecules. This is the concept that makes photoactivation of cis-platinum theoretically possible. Both heat and light increase cisplatin anticancer activity via multiple mechanisms, generating DNA lesions by interacting with purine bases in DNA followed by activation of several signal transduction pathways which finally lead to apoptosis. For the past twenty-seven years, our group has developed infrared photo-thermal activation of cisplatin for cancer treatment from bench to bedside. The future development of photoactivatable prodrugs of platinum-based agents injected intratumorally will increase selectivity, lower toxicity and increase efficacy of this important class of antitumor drugs, particularly when treating tumors accessible to laser-based fiber-optic devices, as in head and neck cancer. In this article, the mechanistic rationale of combined intratumor injections of cisplatin and laser-induced thermal therapy (CDDP-LITT) and the clinical application of such minimally invasive treatment for cancer are reviewed.
Collapse
Affiliation(s)
- Renan Vieira de Brito
- Department of Otolaryngology and Head and Neck Surgery, Federal University of São Paulo (UNIFESP), Sao Paulo 04023-062, SP, Brazil; (R.V.d.B.); (M.d.N.P.); (O.C.)
| | - Marília Wellichan Mancini
- Biophotonics Department, Institute of Research and Education in the Health Area (NUPEN), Sao Carlos 13562-030, SP, Brazil;
| | - Marcel das Neves Palumbo
- Department of Otolaryngology and Head and Neck Surgery, Federal University of São Paulo (UNIFESP), Sao Paulo 04023-062, SP, Brazil; (R.V.d.B.); (M.d.N.P.); (O.C.)
| | - Luis Henrique Oliveira de Moraes
- Department of Physiological Sciences, Federal University of Sao Carlos (UFSCar), Sao Carlos 13565-905, SP, Brazil; (L.H.O.d.M.); (G.J.R.)
| | - Gerson Jhonatan Rodrigues
- Department of Physiological Sciences, Federal University of Sao Carlos (UFSCar), Sao Carlos 13565-905, SP, Brazil; (L.H.O.d.M.); (G.J.R.)
| | - Onivaldo Cervantes
- Department of Otolaryngology and Head and Neck Surgery, Federal University of São Paulo (UNIFESP), Sao Paulo 04023-062, SP, Brazil; (R.V.d.B.); (M.d.N.P.); (O.C.)
| | - Joel Avram Sercarz
- Department of Head and Neck Surgery, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA;
| | - Marcos Bandiera Paiva
- Department of Otolaryngology and Head and Neck Surgery, Federal University of São Paulo (UNIFESP), Sao Paulo 04023-062, SP, Brazil; (R.V.d.B.); (M.d.N.P.); (O.C.)
- Department of Head and Neck Surgery, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA;
| |
Collapse
|
15
|
Chen S, Bian H, Duan J. High-Intensity Focused Ultrasound Enhanced Anti-Tumor Activities of Paclitaxel in Breast Cancer in vitro and in vivo. Cancer Manag Res 2022; 14:1303-1312. [PMID: 35386184 PMCID: PMC8978695 DOI: 10.2147/cmar.s349409] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 01/31/2022] [Indexed: 12/21/2022] Open
Abstract
Background Paclitaxel (PTX) is an important oncologic chemotherapeutic agent against breast cancer, but breast cancer patients develop significant resistance to PTX during chemotherapy. Alterations in tubulin and associated proteins have been implicated in resistance to PTX. High-intensity focused ultrasound (HIFU) induces deep tumor penetration of anti-tumor agents in solid tumors. Methods We investigated the influence of HIFU on the anti-tumor activities of PTX in breast cancer. Both in vivo and in vitro experiments were performed in this research: mice were treated with 2 mg/Kg PTX through tail vein injection, while breast cancer cells were treated with 400 nM PTX. Cell viability was analyzed through Cell Counting Kit-8. Cell apoptosis was evaluated through Annexin-V/PI Apoptosis Analysis Kit. The activities of catalase (CAT) and superoxide dismutase (SOD) and the concentration of malondialdehyde (MDA) were evaluated by relative commercial kits. Results HIFU enhanced PTX-inhibited breast cancer cell viability and PTX-induced cell apoptosis. Simultaneous treatment of HIFU and PTX decreased the activities of CAT and SOD and increased the concentration of MDA. In mice bearing MDA-MB-231 tumors, the treatment of HIFU and PTX significantly decreased tumor size, increased body weight and elevated animal survival. HIFU enhanced the distribution of PTX in tumor tissues. Conclusion The performance of HIFU promoted the distribution of PTX and enhanced its anti-tumor activities in breast cancer.
Collapse
Affiliation(s)
- Sha Chen
- Department 2 of Ultrasound, Cangzhou Central Hospital, Cangzhou, 061000, Hebei, People’s Republic of China
- Correspondence: Sha Chen, Department 2 of Ultrasound, Cangzhou Central Hospital, No. 16, Xinhua West Road, Yunhe District, Cangzhou, 061000, Hebei, People’s Republic of China, Tel +86-18232858958, Email
| | - Hao Bian
- Magnetic Resonance Imaging Department, Cangzhou Central Hospital, Cangzhou, 061000, Hebei, People’s Republic of China
| | - Jingyu Duan
- Department 2 of Ultrasound, Cangzhou Central Hospital, Cangzhou, 061000, Hebei, People’s Republic of China
| |
Collapse
|
16
|
Rad AT, Hargrove D, Daneshmandi L, Ramsdell A, Lu X, Nieh MP. Codelivery of Paclitaxel and Parthenolide in Discoidal Bicelles for a Synergistic Anticancer Effect: Structure Matters. ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202100080] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
- Armin Tahmasbi Rad
- Department of Biomedical Engineering University of Connecticut Storrs CT 06269 USA
- Polymer Program Institute of Materials Sciences University of Connecticut 191 Auditorium Road Storrs CT 06269 USA
- Encapsulate, University of Connecticut Technology Incubation Program Farmington CT 06032
| | - Derek Hargrove
- School of Pharmacy University of Connecticut Storrs CT 06269 USA
| | - Leila Daneshmandi
- Department of Biomedical Engineering University of Connecticut Storrs CT 06269 USA
- Encapsulate, University of Connecticut Technology Incubation Program Farmington CT 06032
| | - Amanda Ramsdell
- Department of Chemical and Bimolecular Engineering University of Connecticut Storrs CT 06269 USA
| | - Xiuling Lu
- Polymer Program Institute of Materials Sciences University of Connecticut 191 Auditorium Road Storrs CT 06269 USA
- School of Pharmacy University of Connecticut Storrs CT 06269 USA
| | - Mu-Ping Nieh
- Department of Biomedical Engineering University of Connecticut Storrs CT 06269 USA
- Polymer Program Institute of Materials Sciences University of Connecticut 191 Auditorium Road Storrs CT 06269 USA
- Department of Chemical and Bimolecular Engineering University of Connecticut Storrs CT 06269 USA
| |
Collapse
|
17
|
Patel H, Wu ZX, Chen Y, Bo L, Chen ZS. Drug resistance: from bacteria to cancer. MOLECULAR BIOMEDICINE 2021; 2:27. [PMID: 35006446 PMCID: PMC8607383 DOI: 10.1186/s43556-021-00041-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Accepted: 04/22/2021] [Indexed: 12/14/2022] Open
Abstract
The phenomenon of drug resistance has been a hindrance to therapeutic medicine since the late 1940s. There is a plethora of factors and mechanisms contributing to progression of drug resistance. From prokaryotes to complex cancers, drug resistance is a prevailing issue in clinical medicine. Although there are numerous factors causing and influencing the phenomenon of drug resistance, cellular transporters contribute to a noticeable majority. Efflux transporters form a huge family of proteins and are found in a vast number of species spanning from prokaryotes to complex organisms such as humans. During the last couple of decades, various approaches in analyses of biochemistry and pharmacology of transporters have led us to understand much more about drug resistance. In this review, we have discussed the structure, function, potential causes, and mechanisms of multidrug resistance in bacteria as well as cancers.
Collapse
Affiliation(s)
- Harsh Patel
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York, NY, 11439, USA
| | - Zhuo-Xun Wu
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York, NY, 11439, USA
| | - Yanglu Chen
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA
| | - Letao Bo
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York, NY, 11439, USA
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York, NY, 11439, USA.
| |
Collapse
|
18
|
Mo J, Mai Le NP, Priefer R. Evaluating the mechanisms of action and subcellular localization of ruthenium(II)-based photosensitizers. Eur J Med Chem 2021; 225:113770. [PMID: 34403979 DOI: 10.1016/j.ejmech.2021.113770] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/23/2021] [Accepted: 08/09/2021] [Indexed: 01/16/2023]
Abstract
The identification of ruthenium(II) polypyridyl complexes as photosensitizers in photodynamic therapy (PDT) for the treatment of cancer is progressing rapidly. Due to their favorable photophysical and photochemical properties, Ru(II)-based photosensitizers have absorption in the visible spectrum, can be irradiated via one- and two-photon excitation within the PDT window, and yield potent oxygen-dependent and/or oxygen-independent photobiological activities. Herein, we present a current overview of the mechanisms of action and subcellular localization of Ru(II)-based photosensitizers in the treatment of cancer. These photosensitizers are highlighted from a medicinal chemistry and chemical biology perspective. However, although this field is burgeoning, challenges and limitations remain in the photosensitization strategies and clinical translation.
Collapse
Affiliation(s)
- Jiancheng Mo
- Massachusetts College of Pharmacy and Health Sciences University, Boston, MA, USA
| | - Ngoc Phuong Mai Le
- Massachusetts College of Pharmacy and Health Sciences University, Boston, MA, USA
| | - Ronny Priefer
- Massachusetts College of Pharmacy and Health Sciences University, Boston, MA, USA.
| |
Collapse
|
19
|
Wolf P. Targeted Toxins for the Treatment of Prostate Cancer. Biomedicines 2021; 9:biomedicines9080986. [PMID: 34440190 PMCID: PMC8391386 DOI: 10.3390/biomedicines9080986] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/02/2021] [Accepted: 08/07/2021] [Indexed: 12/12/2022] Open
Abstract
Prostate cancer is the second most common cancer and the fifth leading cause of cancer deaths worldwide. Despite improvements in diagnosis and treatment, new treatment options are urgently needed for advanced stages of the disease. Targeted toxins are chemical conjugates or fully recombinant proteins consisting of a binding domain directed against a target antigen on the surface of cancer cells and a toxin domain, which is transported into the cell for the induction of apoptosis. In the last decades, targeted toxins against prostate cancer have been developed. Several challenges, however, became apparent that prevented their direct clinical use. They comprise immunogenicity, low target antigen binding, endosomal entrapment, and lysosomal/proteasomal degradation of the targeted toxins. Moreover, their efficacy is impaired by prostate tumors, which are marked by a dense microenvironment, low target antigen expression, and apoptosis resistance. In this review, current findings in the development of targeted toxins against prostate cancer in view of effective targeting, reduction of immunogenicity, improvement of intracellular trafficking, and overcoming apoptosis resistance are discussed. There are promising approaches that should lead to the clinical use of targeted toxins as therapeutic alternatives for advanced prostate cancer in the future.
Collapse
Affiliation(s)
- Philipp Wolf
- Department of Urology, Medical Center, University of Freiburg, 79106 Freiburg, Germany; ; Tel.: +49-761-270-28921
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| |
Collapse
|
20
|
Sarangthem V, Yi A, Kim Y, Rehemtulla A, Lee BH, Jeon YH, Singh TD, Park RW. Therapeutic Effect of IL-4 Receptor-Targeting Pro-Apoptotic Peptide (AP1-ELP-KLAK) in Glioblastoma Tumor Model. Int J Nanomedicine 2021; 16:5039-5052. [PMID: 34335025 PMCID: PMC8318221 DOI: 10.2147/ijn.s316388] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 06/28/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Thermal-responsive self-assembled elastin-like polypeptide (ELP)-based nanoparticles are an emerging platform for controlled delivery of therapeutic peptides, proteins and small molecular drugs. The antitumor effect of bioengineered chimeric polypeptide AP1-ELP-KLAK containing an interleukin-4 receptor (IL-4R) targeting peptide and pro-apoptotic peptide (KLAKLAK) was evaluated in glioblastoma (GBM) in vitro and in vivo. METHODS AND RESULTS Herein, the therapeutic effect of AP1-ELP-KLAK was tested in advanced, and less curable glioblastoma cells with higher expression of IL-4R. Glioblastoma cell lines stably expressing different reporter systems i.e., caspase-3 sensor (surrogate marker for cellular apoptosis) or effluc/enhanced firefly luciferase (cellular viability) were established to measure cell death non-invasively. Bioluminescence imaging (BLI) of D54/effluc and U97MG/effluc treated with AP1-ELP-KLAK exhibited higher cell death up to 2~3-fold than the control. Treatment with AP1-ELP-KLAK resulted in time-dependent increase of caspase-3 sensor BLI activity in D54/C cells and D54/C tumor-bearing mice. Intravenous injection of AP1-ELP-KLAK dramatically reduced tumor growth by inducing cellular apoptosis in D54/effluc tumor-bearing mice. Further, the immuno-histological examination of the excised tumor tissue confirmed the presence of apoptotic cells as well as caspase-3 activation. CONCLUSION Collectively, AP1-ELP-KLAK effectively induced cellular apoptosis of glioblastoma cells and non-invasive imaging provides a window for real-time monitoring of anti-tumor effect with the provision of improving therapeutic efficacy in a glioblastoma mice model.
Collapse
Affiliation(s)
- Vijaya Sarangthem
- Department of Biochemistry and Cell Biology, School of Medicine, and Cell & Matrix Research Institute, Kyungpook National University, Daegu, 41944, Republic of Korea
- Department of Pathology, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
| | - Aena Yi
- Department of Biochemistry and Cell Biology, School of Medicine, and Cell & Matrix Research Institute, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Yunjae Kim
- Department of Biochemistry and Cell Biology, School of Medicine, and Cell & Matrix Research Institute, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Alnawaz Rehemtulla
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Byung-Heon Lee
- Department of Biochemistry and Cell Biology, School of Medicine, and Cell & Matrix Research Institute, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Young Hyun Jeon
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu, Republic of Korea
| | - Thoudam Debraj Singh
- Department of Medical Oncology Laboratory, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
| | - Rang-Woon Park
- Department of Biochemistry and Cell Biology, School of Medicine, and Cell & Matrix Research Institute, Kyungpook National University, Daegu, 41944, Republic of Korea
| |
Collapse
|
21
|
Liu Z, Zhang L, Cui T, Ma M, Ren J, Qu X. A Nature-Inspired Metal-Organic Framework Discriminator for Differential Diagnosis of Cancer Cell Subtypes. Angew Chem Int Ed Engl 2021; 60:15436-15444. [PMID: 33960090 DOI: 10.1002/anie.202102286] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Indexed: 12/13/2022]
Abstract
Metabolic glycan labeling (MGL) followed by bioorthogonal chemistry provides a powerful tool for tumor imaging and therapy. However, selectively metabolic labeling of cells or tissues of interest remains a challenge. Particularly, owing to tumor heterogeneity including tumor subtypes and interpatient heterogeneity, it is far more difficult to realize tumor-cell-selective metabolic labeling for precise diagnosis. Inspired by nature, we designed azidosugar-functionalized metal-organic frameworks camouflaged with cancer cell membranes to accomplish cancer-cell-selective MGL in vivo. With abundant receptors, this biomimetic platform not only selectively targets homotypic cells but also realizes different breast cancer subtype-selective MGL. Moreover, the endo/lysosomal-escaped ZIF-8 can make azidosugar escape from lysosomes and accelerate its metabolic incorporation. This strategy also takes advantage of cancer-tissue-derived cell membranes, which may have huge potential for personalized diagnosis and therapy.
Collapse
Affiliation(s)
- Zhengwei Liu
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China.,University of Chinese Academy of Sciences, Beijing, 100039, P. R. China
| | - Lu Zhang
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China.,University of Chinese Academy of Sciences, Beijing, 100039, P. R. China
| | - Tingting Cui
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China.,University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Mengmeng Ma
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China.,University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Jinsong Ren
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China.,University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Xiaogang Qu
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China.,University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| |
Collapse
|
22
|
Liu Z, Zhang L, Cui T, Ma M, Ren J, Qu X. A Nature‐Inspired Metal–Organic Framework Discriminator for Differential Diagnosis of Cancer Cell Subtypes. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202102286] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Zhengwei Liu
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun Jilin 130022 P. R. China
- University of Chinese Academy of Sciences Beijing 100039 P. R. China
| | - Lu Zhang
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun Jilin 130022 P. R. China
- University of Chinese Academy of Sciences Beijing 100039 P. R. China
| | - Tingting Cui
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun Jilin 130022 P. R. China
- University of Science and Technology of China Hefei Anhui 230026 P. R. China
| | - Mengmeng Ma
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun Jilin 130022 P. R. China
- University of Science and Technology of China Hefei Anhui 230026 P. R. China
| | - Jinsong Ren
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun Jilin 130022 P. R. China
- University of Science and Technology of China Hefei Anhui 230026 P. R. China
| | - Xiaogang Qu
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun Jilin 130022 P. R. China
- University of Science and Technology of China Hefei Anhui 230026 P. R. China
| |
Collapse
|
23
|
Patel JP, Spiller SE, Barker ED. Drug penetration in pediatric brain tumors: Challenges and opportunities. Pediatr Blood Cancer 2021; 68:e28983. [PMID: 33719183 DOI: 10.1002/pbc.28983] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 02/11/2021] [Accepted: 02/12/2021] [Indexed: 12/23/2022]
Abstract
Larger clinical trial enrollments and a greater understanding of biological heterogeneity have led to improved survival rates for children diagnosed with brain tumors in the last 50 years. However, reducing long-term morbidities and improving survival rates of high-risk tumors remain major challenges. Chemotherapy can reduce tumor burden, but effective drug penetration at the tumor site is limited by barriers in the route of drug administration and within the tumor microenvironment. Bioavailability of drugs is impeded by the blood-brain barrier, plasma protein binding, and structural components by the tumor including the matrix and vasculature contributing to increased interstitial fluid pressure, hypoxia, and acidity. Designing drug delivery systems to circumvent these barriers could lead to improved drug penetration at the tumor site and reduce adverse systemic side effects. In this review, we expand on how systemic and local barriers limit drug penetration and present potential methods to enhance drug penetration in pediatric brain tumors.
Collapse
Affiliation(s)
- Jenny P Patel
- Department of Mechanical, Aerospace, and Biomedical Engineering, The University of Tennessee at Knoxville, Knoxville, Tennessee
| | - Susan E Spiller
- Pediatric Hematology/Oncology, East Tennessee Children's Hospital, Knoxville, Tennessee
| | - Elizabeth D Barker
- Department of Mechanical, Aerospace, and Biomedical Engineering, The University of Tennessee at Knoxville, Knoxville, Tennessee
| |
Collapse
|
24
|
Bacteria in Carcinogenesis and Cancer Prevention: A Review Study. INTERNATIONAL JOURNAL OF CANCER MANAGEMENT 2021. [DOI: 10.5812/ijcm.107956] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Context: Although conventional therapies improve the conditions of patients with cancer, adverse side effects, and resistance to different therapies have convinced scientists to use alternative methods to overcome these problems. One of the most promising research directions is the application of specific types of bacteria and their components to prevent and treat different cancers. Apart from the ability of bacteria to modulate immune responses, various particular properties such as toxin production and anaerobic lifestyle, have made them one of the potential candidates to help cancer therapy. Evidence Acquisition: In this review, the latest information on the role of bacteria in carcinogenesis and cancer prevention in PubMed, Google scholar, and Science Direct databases in 2020 were considered using a combination of keywords “bacteria”, “carcinogenesis”, “cancer” and “prevention”. Results: Bacteria-cancer interactions can be studied in 2 areas of bacteria and carcinogenesis and the other bacteria and cancer treatment or prevention. In this review, bacterial carcinogenicity has been mentioned with 3 main mechanisms: bacterial toxin, bacterial metabolites, and chronic inflammation caused by bacteria. Bacterial-mediated tumor therapy (BMTT) is briefly discussed in 8 mechanisms including tumor-targeting bacterial therapy, gene therapy and vectors, bacterial products, arginine metabolism, magnetotactic bacteria, combination bacteriolytic therapy (COBALT), immunomodulation of bacteria in cancer, and immune survival. Conclusions: The importance of bacteria in terms of diversity in their interaction with humans, as well as their components that can affect homeostasis and the immune system, has made them a powerful factor in describing the human condition in health and disease. These important elements can be used in the prevention and treatment of many complex diseases with different origins like cancer. The present study can provide an overview of the role of bacteria in cancer development or prevention and potential approaches for bacteria in cancer therapy.
Collapse
|
25
|
Petrillo M, Sozzi G, Dessole M, Capobianco G, Dessole S, Madonia M, Cherchi PL, Paoletti AM, Scambia G, Chiantera V. The role of surgery in platinum-resistant ovarian cancer: A call to the scientific community. Semin Cancer Biol 2021; 77:194-202. [PMID: 33607247 DOI: 10.1016/j.semcancer.2021.02.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 01/26/2021] [Accepted: 02/10/2021] [Indexed: 12/30/2022]
Abstract
In the last decade, a growing attention has been focused on identifying effective therapeutic strategies also in the orphan clinical setting of women with platinum-resistant disease. In this context, secondary cytoreductive surgery (SCS) remains a potential approach only in women with platinum sensitive relapse, but experimental data have been published supporting the role of SCS also in patients with platinum-resistant recurrence. In particular, surgery is emerging as a potential option in specific subgroups of women, such as those patients with low-grade serous histology, or low-volume relapse with disease located in the so-called pharmacological sanctuaries. Furthermore, contrasting evidences have suggested a potential role in this clinical setting of SCS combined with intraperitoneal hyperthermic chemotherapy. In this complex scenario we review here the available evidences regarding the role surgery in ovarian cancer patients with platinum resistant disease, trying also to understand which patients may benefit from this challenging, experimental approach.
Collapse
Affiliation(s)
- Marco Petrillo
- Gynecologic and Obstetric Clinic, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy; PhD School in Biomedical Sciences, University of Sassari, Sassari, Italy.
| | - Giulio Sozzi
- Department of Gynecologic Oncology, University of Palermo, Palermo, Italy
| | - Margherita Dessole
- Community Gynecologic Service, Azienda Socio Sanitari Locale di Sassari, Sassari, Italy
| | - Giampiero Capobianco
- Gynecologic and Obstetric Clinic, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy; PhD School in Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Salvatore Dessole
- Gynecologic and Obstetric Clinic, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy; PhD School in Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Massimo Madonia
- Urology Clinic, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy; PhD School in Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Pier Luigi Cherchi
- Gynecologic and Obstetric Clinic, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy; PhD School in Biomedical Sciences, University of Sassari, Sassari, Italy
| | | | - Giovanni Scambia
- Department of Woman and Child Health, IRCCS Fondazione Policlinico Agostino Gemelli, 00168, Rome, Italy
| | - Vito Chiantera
- Department of Gynecologic Oncology, University of Palermo, Palermo, Italy
| |
Collapse
|
26
|
Wang Y, Bruggeman KF, Franks S, Gautam V, Hodgetts SI, Harvey AR, Williams RJ, Nisbet DR. Is Viral Vector Gene Delivery More Effective Using Biomaterials? Adv Healthc Mater 2021; 10:e2001238. [PMID: 33191667 DOI: 10.1002/adhm.202001238] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 10/03/2020] [Indexed: 12/16/2022]
Abstract
Gene delivery has been extensively investigated for introducing foreign genetic material into cells to promote expression of therapeutic proteins or to silence relevant genes. This approach can regulate genetic or epigenetic disorders, offering an attractive alternative to pharmacological therapy or invasive protein delivery options. However, the exciting potential of viral gene therapy has yet to be fully realized, with a number of clinical trials failing to deliver optimal therapeutic outcomes. Reasons for this include difficulty in achieving localized delivery, and subsequently lower efficacy at the target site, as well as poor or inconsistent transduction efficiency. Thus, ongoing efforts are focused on improving local viral delivery and enhancing its efficiency. Recently, biomaterials have been exploited as an option for more controlled, targeted and programmable gene delivery. There is a growing body of literature demonstrating the efficacy of biomaterials and their potential advantages over other delivery strategies. This review explores current limitations of gene delivery and the progress of biomaterial-mediated gene delivery. The combination of biomaterials and gene vectors holds the potential to surmount major challenges, including the uncontrolled release of viral vectors with random delivery duration, poorly localized viral delivery with associated off-target effects, limited viral tropism, and immune safety concerns.
Collapse
Affiliation(s)
- Yi Wang
- Laboratory of Advanced Biomaterials Research School of Engineering The Australian National University Canberra ACT 2601 Australia
| | - Kiara F. Bruggeman
- Laboratory of Advanced Biomaterials Research School of Engineering The Australian National University Canberra ACT 2601 Australia
| | - Stephanie Franks
- Laboratory of Advanced Biomaterials Research School of Engineering The Australian National University Canberra ACT 2601 Australia
| | - Vini Gautam
- Department of Biomedical Engineering The University of Melbourne Melbourne Victoria 3010 Australia
| | - Stuart I. Hodgetts
- School of Human Sciences The University of Western Australia Perth WA 6009 Australia
- Perron Institute for Neurological and Translational Science Perth WA 6009 Australia
| | - Alan R. Harvey
- School of Human Sciences The University of Western Australia Perth WA 6009 Australia
- Perron Institute for Neurological and Translational Science Perth WA 6009 Australia
| | - Richard J. Williams
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT) School of Medicine Deakin University Waurn Ponds VIC 3216 Australia
- Biofab3D St. Vincent's Hospital Fitzroy 3065 Australia
| | - David R. Nisbet
- Laboratory of Advanced Biomaterials Research School of Engineering The Australian National University Canberra ACT 2601 Australia
- Biofab3D St. Vincent's Hospital Fitzroy 3065 Australia
| |
Collapse
|
27
|
Park JE, Kim JY, Kim HS, Shim WH. Comparison of Dynamic Contrast-Enhancement Parameters between Gadobutrol and Gadoterate Meglumine in Posttreatment Glioma: A Prospective Intraindividual Study. AJNR Am J Neuroradiol 2020; 41:2041-2048. [PMID: 33060100 DOI: 10.3174/ajnr.a6792] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 07/22/2020] [Indexed: 01/10/2023]
Abstract
BACKGROUND AND PURPOSE Differences in molecular properties between one-molar and half-molar gadolinium-based contrast agents are thought to affect parameters obtained from dynamic contrast-enhanced imaging. The aim of our study was to investigate differences in dynamic contrast-enhanced parameters between one-molar nonionic gadobutrol and half-molar ionic gadoterate meglumine in patients with posttreatment glioma. MATERIALS AND METHODS This prospective study enrolled 32 patients who underwent 2 20-minute dynamic contrast-enhanced examinations, one with gadobutrol and one with gadoterate meglumine. The model-free parameter of area under the signal intensity curve from 30 to 1100 seconds and the Tofts model-based pharmacokinetic parameters were calculated and compared intraindividually using paired t tests. Patients were further divided into progression (n = 12) and stable (n = 20) groups, which were compared using Student t tests. RESULTS Gadobutrol and gadoterate meglumine did not show any significant differences in the area under the signal intensity curve or pharmacokinetic parameters of K trans, Ve, Vp, or Kep (all P > .05). Gadobutrol showed a significantly higher mean wash-in rate (0.83 ± 0.64 versus 0.29 ± 0.63, P = .013) and a significantly lower mean washout rate (0.001 ± 0.0001 versus 0.002 ± 0.002, P = .02) than gadoterate meglumine. Trends toward higher area under the curve, K trans, Ve, Vp, wash-in, and washout rates and lower Kep were observed in the progression group in comparison with the treatment-related-change group, regardless of the contrast agent used. CONCLUSIONS Model-free and pharmacokinetic parameters did not show any significant differences between the 2 gadolinium-based contrast agents, except for a higher wash-in rate with gadobutrol and a higher washout rate with gadoterate meglumine, supporting the interchangeable use of gadolinium-based contrast agents for dynamic contrast-enhanced imaging in patients with posttreatment glioma.
Collapse
Affiliation(s)
- J E Park
- From the Department of Radiology and Research Institute of Radiology (J.E.P., H.S.K., W.H.S.), University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - J Y Kim
- Department of Radiology (J.Y.K.), Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - H S Kim
- From the Department of Radiology and Research Institute of Radiology (J.E.P., H.S.K., W.H.S.), University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - W H Shim
- From the Department of Radiology and Research Institute of Radiology (J.E.P., H.S.K., W.H.S.), University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| |
Collapse
|
28
|
Deep Tumor Penetration of Doxorubicin-Loaded Glycol Chitosan Nanoparticles Using High-Intensity Focused Ultrasound. Pharmaceutics 2020; 12:pharmaceutics12100974. [PMID: 33076520 PMCID: PMC7650702 DOI: 10.3390/pharmaceutics12100974] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 10/11/2020] [Accepted: 10/14/2020] [Indexed: 12/13/2022] Open
Abstract
The dense extracellular matrix (ECM) in heterogeneous tumor tissues can prevent the deep tumor penetration of drug-loaded nanoparticles, resulting in a limited therapeutic efficacy in cancer treatment. Herein, we suggest that the deep tumor penetration of doxorubicin (DOX)-loaded glycol chitosan nanoparticles (CNPs) can be improved using high-intensity focused ultrasound (HIFU) technology. Firstly, we prepared amphiphilic glycol chitosan-5β-cholanic acid conjugates that can self-assemble to form stable nanoparticles with an average of 283.7 ± 5.3 nm. Next, the anticancer drug DOX was simply loaded into the CNPs via a dialysis method. DOX-loaded CNPs (DOX-CNPs) had stable nanoparticle structures with an average size of 265.9 ± 35.5 nm in aqueous condition. In cultured cells, HIFU-treated DOX-CNPs showed rapid drug release and enhanced cellular uptake in A549 cells, resulting in increased cytotoxicity, compared to untreated DOX-CNPs. In ECM-rich A549 tumor-bearing mice, the tumor-targeting efficacy of intravenously injected DOX-CNPs with HIFU treatment was 1.84 times higher than that of untreated DOX-CNPs. Furthermore, the deep tumor penetration of HIFU-treated DOX-CNPs was clearly observed at targeted tumor tissues, due to the destruction of the ECM structure via HIFU treatment. Finally, HIFU-treated DOX-CNPs greatly increased the therapeutic efficacy at ECM-rich A549 tumor-bearing mice, compared to free DOX and untreated DOX-CNPs. This deep penetration of drug-loaded nanoparticles via HIFU treatment is a promising strategy to treat heterogeneous tumors with dense ECM structures.
Collapse
|
29
|
Simões JCS, Sarpaki S, Papadimitroulas P, Therrien B, Loudos G. Conjugated Photosensitizers for Imaging and PDT in Cancer Research. J Med Chem 2020; 63:14119-14150. [PMID: 32990442 DOI: 10.1021/acs.jmedchem.0c00047] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Early cancer detection and perfect understanding of the disease are imperative toward efficient treatments. It is straightforward that, for choosing a specific cancer treatment methodology, diagnostic agents undertake a critical role. Imaging is an extremely intriguing tool since it assumes a follow up to treatments to survey the accomplishment of the treatment and to recognize any conceivable repeating injuries. It also permits analysis of the disease, as well as to pursue treatment and monitor the possible changes that happen on the tumor. Likewise, it allows screening the adequacy of treatment and visualizing the state of the tumor. Additionally, when the treatment is finished, observing the patient is imperative to evaluate the treatment methodology and adjust the treatment if necessary. The goal of this review is to present an overview of conjugated photosensitizers for imaging and therapy.
Collapse
Affiliation(s)
- João C S Simões
- Institute of Chemistry, University of Neuchatel, Avenue de Bellevaux 51, CH-2000 Neuchatel, Switzerland.,BioEmission Technology Solutions, Alexandras Avenue 116, 11472 Athens, Greece
| | - Sophia Sarpaki
- BioEmission Technology Solutions, Alexandras Avenue 116, 11472 Athens, Greece
| | | | - Bruno Therrien
- Institute of Chemistry, University of Neuchatel, Avenue de Bellevaux 51, CH-2000 Neuchatel, Switzerland
| | - George Loudos
- BioEmission Technology Solutions, Alexandras Avenue 116, 11472 Athens, Greece
| |
Collapse
|
30
|
Ahmed A, Sarwar S, Hu Y, Munir MU, Nisar MF, Ikram F, Asif A, Rahman SU, Chaudhry AA, Rehman IU. Surface-modified polymeric nanoparticles for drug delivery to cancer cells. Expert Opin Drug Deliv 2020; 18:1-24. [PMID: 32905714 DOI: 10.1080/17425247.2020.1822321] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
INTRODUCTION The utilization of polymeric nanoparticles, as drug payloads, has been extensively prevailed in cancer therapy. However, the precise distribution of these nanocarriers is restrained by various physiological and cellular obstacles. Nanoparticles must avoid nonspecific interactions with healthy cells and in vivo compartments to circumvent these barriers. Since in vivo interactions of nanoparticles are mainly dependent on surface properties of nanoparticles, efficient control on surface constituents is necessary for the determination of nanoparticles' fate in the body. AREAS COVERED In this review, the surface-modified polymeric nanoparticles and their utilization in cancer treatment were elaborated. First, the interaction of nanoparticles with numerous in vivo barriers was highlighted. Second, different strategies to overcome these obstacles were described. Third, some inspiring examples of surface-modified nanoparticles were presented. Later, fabrication and characterization methods of surface-modified nanoparticles were discussed. Finally, the applications of these nanoparticles in different routes of treatments were explored. EXPERT OPINION Surface modification of anticancer drug-loaded polymeric nanoparticles can enhance the efficacy, selective targeting, and biodistribution of the anticancer drug at the tumor site.
Collapse
Affiliation(s)
- Arsalan Ahmed
- Interdisciplinary Research Centre in Biomedical Materials, COMSATS University Islamabad , Lahore, Pakistan
| | - Shumaila Sarwar
- Interdisciplinary Research Centre in Biomedical Materials, COMSATS University Islamabad , Lahore, Pakistan.,Faculty of Pharmacy, University of Sargodha , Sargodha, Pakistan
| | - Yong Hu
- Institute of Materials Engineering, College of Engineering and Applied Sciences, Nanjing University , Nanjing, Jiangsu, China
| | - Muhammad Usman Munir
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University , Sakaka, Aljouf, Saudi Arabia
| | - Muhammad Farrukh Nisar
- Department of Physiology and Biochemistry, Cholistan University of Veterinary and Animal Sciences , Bahawalpur, Pakistan
| | - Fakhera Ikram
- Interdisciplinary Research Centre in Biomedical Materials, COMSATS University Islamabad , Lahore, Pakistan
| | - Anila Asif
- Interdisciplinary Research Centre in Biomedical Materials, COMSATS University Islamabad , Lahore, Pakistan
| | - Saeed Ur Rahman
- Interdisciplinary Research Centre in Biomedical Materials, COMSATS University Islamabad , Lahore, Pakistan
| | - Aqif Anwar Chaudhry
- Interdisciplinary Research Centre in Biomedical Materials, COMSATS University Islamabad , Lahore, Pakistan
| | - Ihtasham Ur Rehman
- Interdisciplinary Research Centre in Biomedical Materials, COMSATS University Islamabad , Lahore, Pakistan.,Bioengineering, Engineering Department, Lancaster University , Lancaster, UK
| |
Collapse
|
31
|
Yu J, Mu Q, Perazzolo S, Griffin JI, Zhu L, McConnachie LA, Shen DD, Ho RJ. Novel Long-Acting Drug Combination Nanoparticles Composed of Gemcitabine and Paclitaxel Enhance Localization of Both Drugs in Metastatic Breast Cancer Nodules. Pharm Res 2020; 37:197. [PMID: 32968837 PMCID: PMC8686529 DOI: 10.1007/s11095-020-02888-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 07/21/2020] [Indexed: 12/25/2022]
Abstract
PURPOSE To develop drug-combination nanoparticles (DcNPs) composed of hydrophilic gemcitabine (G) and hydrophobic paclitaxel (T) and deliver both drugs to metastatic cancer cells. METHODS GT DcNPs were evaluated based on particle size and drug association efficiency (AE%). The effect of DcNP on GT plasma time-course and tissue distribution was characterized in mice and a pharmacokinetic model was developed. A GT distribution study into cancer nodules (derived from 4 T1 cells) was performed. RESULTS An optimized GT DcNP composition (d = 59.2 nm ±9.2 nm) was found to be suitable for IV formulation. Plasma exposure of G and T were enhanced 61-fold and 3.8-fold when given in DcNP form compared to the conventional formulation, respectively. Mechanism based pharmacokinetic modeling and simulation show that both G and T remain highly associated to DcNPs in vivo (G: 98%, T:75%). GT DcNPs have minimal distribution to healthy organs with selective distribution and retention in tumor burdened tissue. Tumor bearing lungs had a 5-fold higher tissue-to-plasma ratio of gemcitabine in GT DcNPs compared to healthy lungs. CONCLUSIONS DcNPs can deliver hydrophilic G and hydrophobic T together to cancer nodules and produce long acting exposure, likely due to stable GT association to DcNPs in vivo.
Collapse
Affiliation(s)
- Jesse Yu
- Departments of Pharmaceutics, University of Washington, Seattle, Washington, 98195, USA
| | - Qingxin Mu
- Departments of Pharmaceutics, University of Washington, Seattle, Washington, 98195, USA
| | - Simone Perazzolo
- Departments of Pharmaceutics, University of Washington, Seattle, Washington, 98195, USA
| | - James I Griffin
- Departments of Pharmaceutics, University of Washington, Seattle, Washington, 98195, USA
| | - Linxi Zhu
- Departments of Pharmaceutics, University of Washington, Seattle, Washington, 98195, USA
| | - Lisa A McConnachie
- Departments of Pharmaceutics, University of Washington, Seattle, Washington, 98195, USA
| | - Danny D Shen
- Departments of Pharmaceutics, University of Washington, Seattle, Washington, 98195, USA
| | - Rodney Jy Ho
- Departments of Pharmaceutics, University of Washington, Seattle, Washington, 98195, USA.
- Departments of Bioengineering, University of Washington, Seattle, Washington, 98195, USA.
| |
Collapse
|
32
|
Meng B, Folaron MR, Strawbridge RR, Sadeghipour N, Samkoe KS, Tichauer K, Davis SC. Noninvasive quantification of target availability during therapy using paired-agent fluorescence tomography. Am J Cancer Res 2020; 10:11230-11243. [PMID: 33042280 PMCID: PMC7532673 DOI: 10.7150/thno.45273] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 08/06/2020] [Indexed: 12/13/2022] Open
Abstract
Immuno-oncological treatment strategies that target abnormal receptor profiles of tumors are an increasingly important feature of cancer therapy. Yet, assessing receptor availability (RA) and drug-target engagement, important determinants of therapeutic efficacy, is challenging with current imaging strategies, largely due to the complex nonspecific uptake behavior of imaging agents in tumors. Herein, we evaluate whether a quantitative noninvasive imaging approach designed to compensate for nonspecific uptake, MRI-coupled paired-agent fluorescence tomography (MRI-PAFT), is capable of rapidly assessing the availability of epidermal growth factor receptor (EGFR) in response to one dose of anti-EGFR antibody therapy in orthotopic brain tumor models. Methods: Mice bearing orthotopic brain tumor xenografts with relatively high EGFR expression (U251) (N=10) or undetectable human EGFR (9L) (N=9) were considered in this study. For each tumor type, mice were either treated with one dose of cetuximab, or remained untreated. All animals were scanned using MRI-PAFT, which commenced immediately after paired-agent administration, and values of RA were recovered using a model-based approach, which uses the entire dynamic sequence of agent uptake, as well as a simplified “snapshot” approach which requires uptake measurements at only two time points. Recovered values of RA were evaluated between groups and techniques. Hematoxylin & eosin (H&E) and immunohistochemical (IHC) staining was performed on tumor specimens from every animal to confirm tumor presence and EGFR status. Results: In animals bearing EGFR(+) tumors, a significant difference in RA values between treated and untreated animals was observed (RA = 0.24 ± 0.15 and 0.61 ± 0.18, respectively, p=0.027), with an area under the curve - receiver operating characteristic (AUC-ROC) value of 0.92. We did not observe a statistically significant difference in RA values between treated and untreated animals bearing EGFR(-) tumors (RA = 0.18 ± 0.19 and 0.27 ± 0.21, respectively; p = 0.89; AUC-ROC = 0.55), nor did we observe a difference between treated EGFR(+) tumors compared to treated and untreated EGFR(-) tumors. Notably, the snapshot paired-agent strategy quantified drug-receptor engagement within just 30 minutes of agent administration. Examination of the targeted agent alone showed no capacity to distinguish tumors either by treatment or receptor status, even 24h after agent administration. Conclusions: This study demonstrated that a noninvasive imaging strategy enables rapid quantification of receptor availability in response to therapy, a capability that could be leveraged in preclinical drug development, patient stratification, and treatment monitoring.
Collapse
|
33
|
do Nascimento T, Todeschini AR, Santos-Oliveira R, de Souza de Bustamante Monteiro MS, de Souza VT, Ricci-Júnior E. Trends in Nanomedicines for Cancer Treatment. Curr Pharm Des 2020; 26:3579-3600. [DOI: 10.2174/1381612826666200318145349] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 01/21/2020] [Indexed: 12/12/2022]
Abstract
Background:
Cancer is characterized by abnormal cell growth and considered one of the leading
causes of death around the world. Pharmaceutical Nanotechnology has been extensively studied for the optimization
of cancer treatment.
Objective:
Comprehend the panorama of Pharmaceutical Nanotechnology in cancer treatment, through a survey
about nanomedicines applied in clinical studies, approved for use and patented.
Methods:
Acknowledged products under clinical study and nanomedicines commercialized found in scientific
articles through research on the following databases: Pubmed, Science Direct, Scielo and Lilacs. Derwent tool
was used for patent research.
Results:
Nanomedicines based on nanoparticles, polymer micelles, liposomes, dendrimers and nanoemulsions
were studied, along with cancer therapies such as Photodynamic Therapy, Infrared Phototherapy Hyperthermia,
Magnetic Hyperthermia, Radiotherapy, Gene Therapy and Nanoimmunotherapy. Great advancement has been
observed over nanotechnology applied to cancer treatment, mainly for nanoparticles and liposomes.
Conclusion:
The combination of drugs in nanosystems helps to increase efficacy and decrease toxicity. Based on
the results encountered, nanoparticles and liposomes were the most commonly used nanocarriers for drug encapsulation.
In addition, although few nanomedicines are commercially available, this specific research field is continuously
growing.
Collapse
Affiliation(s)
- Tatielle do Nascimento
- Laboratorio de Desenvolvimento Galenico, Farmacia Universitária, Centro de Ciencias da Saude, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Adriane R. Todeschini
- Laboratorio de Glicobiologia Estrutural e Funcional, Instituto de Biofisica Carlos Chagas Filho, Centro de Ciencias da Saude, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ralph Santos-Oliveira
- Instituto de Engenharia Nuclear, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Vilênia T. de Souza
- Laboratorio de Tecnologia Industrial Farmaceutica, Centro de Ciencias da Saude, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Eduardo Ricci-Júnior
- Laboratorio de Desenvolvimento Galenico, Farmacia Universitária, Centro de Ciencias da Saude, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
34
|
Chaturvedi S, Verma A, Saharan VA. Lipid Drug Carriers for Cancer Therapeutics: An Insight into Lymphatic Targeting, P-gp, CYP3A4 Modulation and Bioavailability Enhancement. Adv Pharm Bull 2020; 10:524-541. [PMID: 33072532 PMCID: PMC7539309 DOI: 10.34172/apb.2020.064] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 02/08/2020] [Accepted: 02/10/2020] [Indexed: 12/12/2022] Open
Abstract
In the treatment of cancer, chemotherapy plays an important role though the efficacy of anti-cancer drug administered orally is limited, due to their poor solubility in physiological medium, inability to cross biological membrane, high Para-glycoprotein (P-gp) mediated drug efflux, and pre-systemic metabolism. These all factors cumulatively reduce drug exposure at the target site leading to multidrug resistance (MDR). Lipid based carriers systems has been explored to overcome solubility and permeability related issues of anti-cancer drugs. The lipid based formulations have also been reported to circumvent the effect of P-gp and CYP3A4. Further long chain triglycerides (LCT) has shown their ability to access Lymphatic route over Medium Chain Triglycerides, as the former has been extensively used for targeting anti-cancer drugs at proliferating cells through lymphatic route. Therefore this review tries to reflect the usefulness of lipid based drug carriers systems (viz. liposome, solid lipid nanoparticle, nano-lipid carriers, self-emulsifying, lipidic pro-drugs) in targeting lymphatic system and overcoming issues related to solubility and permeability of anti-cancer drugs. Moreover, we have also tried to reflect how critically lipid based carriers are important in maximizing therapeutic safety and efficacy of anti-cancer drugs.
Collapse
Affiliation(s)
- Shashank Chaturvedi
- Department of Pharmaceutics, Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Anurag Verma
- Department of Pharmaceutics, School of Pharmaceutical Sciences, IFTM University, Moradabad, Uttar Pradesh, India
| | - Vikas Anand Saharan
- Department of Pharmaceutics, School of Pharmaceutical Sciences and Technology, Sardar Bhagwan Singh University, Dehradun, Uttarakhand, India
| |
Collapse
|
35
|
Drug delivery: Experiments, mathematical modelling and machine learning. Comput Biol Med 2020; 123:103820. [PMID: 32658778 DOI: 10.1016/j.compbiomed.2020.103820] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 04/22/2020] [Accepted: 05/10/2020] [Indexed: 01/28/2023]
Abstract
We address the problem of determining from laboratory experiments the data necessary for a proper modeling of drug delivery and efficacy in anticancer therapy. There is an inherent difficulty in extracting the necessary parameters, because the experiments often yield an insufficient quantity of information. To overcome this difficulty, we propose to combine real experiments, numerical simulation, and Machine Learning (ML) based on Artificial Neural Networks (ANN), aiming at a reliable identification of the physical model factors, e.g. the killing action of the drug. To this purpose, we exploit the employed mathematical-numerical model for tumor growth and drug delivery, together with the ANN - ML procedure, to integrate the results of the experimental tests and feed back the model itself, thus obtaining a reliable predictive tool. The procedure represents a hybrid data-driven, physics-informed approach to machine learning. The physical and mathematical model employed for the numerical simulations is without extracellular matrix (ECM) and healthy cells because of the experimental conditions we reproduce.
Collapse
|
36
|
Rapid Evaluation of Antibody Fragment Endocytosis for Antibody Fragment-Drug Conjugates. Biomolecules 2020; 10:biom10060955. [PMID: 32630402 PMCID: PMC7355425 DOI: 10.3390/biom10060955] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 06/12/2020] [Accepted: 06/23/2020] [Indexed: 12/31/2022] Open
Abstract
Antibody-drug conjugates (ADCs) have emerged as the most promising strategy in targeted cancer treatment. Recent strategies for the optimization ADCs include the development of antibody fragment-drug conjugates (FDCs). The critical factor in the successful development of ADCs and FDCs is the identification of tumor antigen-specific and internalizing antibodies (Abs). However, systematic comparison or correlation studies of internalization rates with different antibody formats have not been reported previously. In this study, we generated a panel of scFv-phage Abs using phage display technology and their corresponding scFv and scFv-Fc fragments and evaluated their relative internalization kinetics in relation to their antibody forms. We found that the relative rates and levels of internalization of scFv-phage antibodies positively correlate with their scFv and scFv-Fc forms. Our systematic study demonstrates that endocytosis of scFv-phage can serve as a predictive indicator for the assessment of Ab fragment internalization. Additionally, the present study demonstrates that endocytic antibodies can be rapidly screened and selected from phage antibody libraries prior to the conversion of phage antibodies for the generation of the conventional antibody format. Our strategic approach for the identification and evaluation of endocytic antibodies would expedite the selection for optimal antibodies and antibody fragments and be broadly applicable to ADC and FDC development.
Collapse
|
37
|
Antitumor Effects of Curcumin and Glycyrrhetinic Acid-Modified Curcumin-Loaded Cationic Liposome by Intratumoral Administration. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:4504936. [PMID: 32565859 PMCID: PMC7277028 DOI: 10.1155/2020/4504936] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 05/04/2020] [Indexed: 01/21/2023]
Abstract
Curcumin is a hydrophobic polyphenolic compound extracted from the rhizome of Curcuma longa and shows a line of active biological functions, but its application has been limited and questioned because of its low solubility, low bioavailability, and rapid metabolism. In terms of antitumor effect, these disadvantages can be overcome by intratumoral injection. In this study, we present the intratumoral injection of curcumin and glycyrrhetinic acid-modified curcumin-loaded cationic liposome (GAMCLCL) in H22 tumor-bearing mice. The experimental results demonstrated that curcumin exhibited positive antitumor activities in vitro and in vivo by intratumoral injection, but its activities were much weaker than GAMCLCL and adriamycin. Compared with free curcumin, GAMCLCL showed much better effects in improving the blood parameters (WBC, RBC, PLT, ALT, CRE, and LDH), inhibiting tumor growth, reducing tumor microvascular density, downregulating the expression of VEGF-protein and mRNA, and upregulating the expression of caspase-3 protein and mRNA in H22 tumor tissues. Under the experimental conditions of this study, the antitumor effect of high-dose GAMCLCL was similar to adriamycin. In conclusion, the experimental results demonstrated that free curcumin possessed definite antitumor efficacy, but its antitumor activities were weaker, and some strategies should be adopted to overcome its disadvantages, improve, and ensure its clinical efficacy.
Collapse
|
38
|
Sorrin AJ, Ruhi MK, Ferlic NA, Karimnia V, Polacheck WJ, Celli JP, Huang HC, Rizvi I. Photodynamic Therapy and the Biophysics of the Tumor Microenvironment. Photochem Photobiol 2020; 96:232-259. [PMID: 31895481 PMCID: PMC7138751 DOI: 10.1111/php.13209] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 11/27/2019] [Indexed: 02/07/2023]
Abstract
Targeting the tumor microenvironment (TME) provides opportunities to modulate tumor physiology, enhance the delivery of therapeutic agents, impact immune response and overcome resistance. Photodynamic therapy (PDT) is a photochemistry-based, nonthermal modality that produces reactive molecular species at the site of light activation and is in the clinic for nononcologic and oncologic applications. The unique mechanisms and exquisite spatiotemporal control inherent to PDT enable selective modulation or destruction of the TME and cancer cells. Mechanical stress plays an important role in tumor growth and survival, with increasing implications for therapy design and drug delivery, but remains understudied in the context of PDT and PDT-based combinations. This review describes pharmacoengineering and bioengineering approaches in PDT to target cellular and noncellular components of the TME, as well as molecular targets on tumor and tumor-associated cells. Particular emphasis is placed on the role of mechanical stress in the context of targeted PDT regimens, and combinations, for primary and metastatic tumors.
Collapse
Affiliation(s)
- Aaron J. Sorrin
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, USA
| | - Mustafa Kemal Ruhi
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC and North Carolina State University, Raleigh, NC, 27599, USA
| | - Nathaniel A. Ferlic
- Department of Electrical and Computer Engineering, University of Maryland, College Park, MD, 20742, USA
| | - Vida Karimnia
- Department of Physics, College of Science and Mathematics, University of Massachusetts at Boston, Boston, MA, 02125, USA
| | - William J. Polacheck
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC and North Carolina State University, Raleigh, NC, 27599, USA
- Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| | - Jonathan P. Celli
- Department of Physics, College of Science and Mathematics, University of Massachusetts at Boston, Boston, MA, 02125, USA
| | - Huang-Chiao Huang
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, USA
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Imran Rizvi
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC and North Carolina State University, Raleigh, NC, 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| |
Collapse
|
39
|
Tumor extravasation and infiltration as barriers of nanomedicine for high efficacy: The current status and transcytosis strategy. Biomaterials 2020; 240:119902. [PMID: 32105817 DOI: 10.1016/j.biomaterials.2020.119902] [Citation(s) in RCA: 145] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 02/15/2020] [Accepted: 02/15/2020] [Indexed: 12/12/2022]
Abstract
Nanotechnology-based drug delivery platforms have been explored for cancer treatments and resulted in several nanomedicines in clinical uses and many in clinical trials. However, current nanomedicines have not met the expected clinical therapeutic efficacy. Thus, improving therapeutic efficacy is the foremost pressing task of nanomedicine research. An effective nanomedicine must overcome biological barriers to go through at least five steps to deliver an effective drug into the cytosol of all the cancer cells in a tumor. Of these barriers, nanomedicine extravasation into and infiltration throughout the tumor are the two main unsolved blockages. Up to now, almost all the nanomedicines are designed to rely on the high permeability of tumor blood vessels to extravasate into tumor interstitium, i.e., the enhanced permeability and retention (EPR) effect or so-called "passive tumor accumulation"; however, the EPR features are not so characteristic in human tumors as in the animal tumor models. Following extravasation, the large size nanomedicines are almost motionless in the densely packed tumor microenvironment, making them restricted in the periphery of tumor blood vessels rather than infiltrating in the tumors and thus inaccessible to the distal but highly malignant cells. Recently, we demonstrated using nanocarriers to induce transcytosis of endothelial and cancer cells to enable nanomedicines to actively extravasate into and infiltrate in solid tumors, which led to radically increased anticancer activity. In this perspective, we make a brief discussion about how active transcytosis can be employed to overcome the difficulties, as mentioned above, and solve the inherent extravasation and infiltration dilemmas of nanomedicines.
Collapse
|
40
|
Abstract
Acidic metabolic waste products accumulate in the tumor microenvironment because of high metabolic activity and insufficient perfusion. In tumors, the acidity of the interstitial space and the relatively well-maintained intracellular pH influence cancer and stromal cell function, their mutual interplay, and their interactions with the extracellular matrix. Tumor pH is spatially and temporally heterogeneous, and the fitness advantage of cancer cells adapted to extracellular acidity is likely particularly evident when they encounter less acidic tumor regions, for instance, during invasion. Through complex effects on genetic stability, epigenetics, cellular metabolism, proliferation, and survival, the compartmentalized pH microenvironment favors cancer development. Cellular selection exacerbates the malignant phenotype, which is further enhanced by acid-induced cell motility, extracellular matrix degradation, attenuated immune responses, and modified cellular and intercellular signaling. In this review, we discuss how the acidity of the tumor microenvironment influences each stage in cancer development, from dysplasia to full-blown metastatic disease.
Collapse
Affiliation(s)
- Ebbe Boedtkjer
- Department of Biomedicine, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Stine F. Pedersen
- Department of Biology, University of Copenhagen, DK-2100 Copenhagen, Denmark
| |
Collapse
|
41
|
Wirthl B, Kremheller J, Schrefler BA, Wall WA. Extension of a multiphase tumour growth model to study nanoparticle delivery to solid tumours. PLoS One 2020; 15:e0228443. [PMID: 32023318 PMCID: PMC7001947 DOI: 10.1371/journal.pone.0228443] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 01/15/2020] [Indexed: 01/04/2023] Open
Abstract
One of the main challenges in increasing the efficacy of conventional chemotherapeutics is the fact that they do not reach cancerous cells at a sufficiently high dosage. In order to remedy this deficiency, nanoparticle-based drugs have evolved as a promising novel approach to more specific tumour targeting. Nevertheless, several biophysical phenomena prevent the sufficient penetration of nanoparticles in order to target the entire tumour. We therefore extend our vascular multiphase tumour growth model, enabling it to investigate the influence of different biophysical factors on the distribution of nanoparticles in the tumour microenvironment. The novel model permits the examination of the interplay between the size of vessel-wall pores, the permeability of the blood-vessel endothelium and the lymphatic drainage on the delivery of particles of different sizes. Solid tumours develop a non-perfused core and increased interstitial pressure. Our model confirms that those two typical features of solid tumours limit nanoparticle delivery. Only in case of small nanoparticles is the transport dominated by diffusion, and particles can reach the entire tumour. The size of the vessel-wall pores and the permeability of the blood-vessel endothelium have a major impact on the amount of delivered nanoparticles. This extended in-silico tumour growth model permits the examination of the characteristics and of the limitations of nanoparticle delivery to solid tumours, which currently complicate the translation of nanoparticle therapy to a clinical stage.
Collapse
Affiliation(s)
- Barbara Wirthl
- Institute for Computational Mechanics, Technical University of Munich, Garching b. München, Germany
| | - Johannes Kremheller
- Institute for Computational Mechanics, Technical University of Munich, Garching b. München, Germany
| | - Bernhard A. Schrefler
- Institute for Advanced Study, Technical University of Munich, Garching b. München, Germany
- Department of Civil, Environmental and Architectural Engineering, University of Padova, Padova, Italy
| | - Wolfgang A. Wall
- Institute for Computational Mechanics, Technical University of Munich, Garching b. München, Germany
| |
Collapse
|
42
|
Kayaalp Nalbant E, Rounds C, Sadeghipour N, Meng B, Folaron MR, Haldar C, Strawbridge RR, Samkoe KS, Davis SC, Tichauer KM. A paired-agent fluorescent molecular imaging strategy for quantifying antibody drug target engagement in in vivo window chamber xenograft models. PROCEEDINGS OF SPIE--THE INTERNATIONAL SOCIETY FOR OPTICAL ENGINEERING 2020; 11219:112190G. [PMID: 34183872 PMCID: PMC8236091 DOI: 10.1117/12.2545182] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
A paired-agent fluorescent molecular imaging strategy is presented as a method to measure drug target engagement in whole tumor imaging. The protocol involves dynamic imaging of a pair of targeted and control imaging agents prior to and following antibody therapy. Simulations demonstrated that antibody "drug target engagement" can be estimated within a 15%-error over a wide range of tumor physiology (blood flow, vascular permeability, target density) and antibody characteristics (affinity, binding rates). Experimental results demonstrated the first in vivo detection of binding site barrier, highlighting the potential for this methodology to provide novel insights in drug distribution/binding imaging.
Collapse
Affiliation(s)
- Elif Kayaalp Nalbant
- Biomedical Engineering, Illinois Institute of Technology, Chicago, IL
- Authors contributed equally
| | - Cody Rounds
- Biomedical Engineering, Illinois Institute of Technology, Chicago, IL
- Authors contributed equally
| | - Negar Sadeghipour
- Molecular Imaging Program at Stanford, Stanford University, Stanford, CA
| | - Boyu Meng
- Thayer School of Engineering, Dartmouth College, Hanover, NH
| | | | | | | | | | - Scott C Davis
- Thayer School of Engineering, Dartmouth College, Hanover, NH
| | | |
Collapse
|
43
|
Arazi L. Diffusing alpha-emitters radiation therapy: approximate modeling of the macroscopic alpha particle dose of a point source. Phys Med Biol 2020; 65:015015. [PMID: 31766047 DOI: 10.1088/1361-6560/ab5b73] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Diffusing alpha-emitters radiation therapy ('DaRT') is a new cancer-treatment modality, which enables treating solid tumors by alpha particles. The treatment utilizes implantable seeds embedded with a low activity of radium-224. Each seed continuously emits the short-lived alpha-emitting daughters of radium-224, which spread over several mm around it, creating a 'kill region' of high alpha-particle dose. DaRT is presently tested in clinical trials, starting with locally advanced and recurrent squamous cell carcinoma (SCC) of the skin and head and neck, with promising results with respect to both efficacy and safety. This work aims to provide a simple model which can serve as a zero-order approximation for DaRT dosimetry, allowing for calculating the macroscopic alpha particle dose of a point source, as a basis for more realistic source geometries. The model consists of diffusion equations for radon-220, lead-212 and bismuth-212, with the other short-lived daughters in local secular equilibrium. For simplicity, the medium is assumed to be homogeneous, isotropic and time-independent. Vascular effects are accounted for by effective diffusion and clearance terms. To leading order, the alpha particle dose can be described by simple analytic expressions, which shed light on the underlying physics. The calculations demonstrate that, for a reasonable choice of model parameters, therapeutic alpha-particle dose levels are obtained over a region measuring 4-7 mm in diameter for sources carrying a few [Formula: see text]Ci of radium-224. The model predictions served as the basis for treatment planning in the SCC clinical trial, where treatments employing DaRT seeds carrying 2 [Formula: see text]Ci of radium-224 and spaced 5 mm apart resulted in ∼[Formula: see text] complete response of the treated tumors with no observed radiation-induced toxicity. The promising results of the SCC clinical trial indicate that in spite of its approximate nature, the simple diffusion-based dosimetry model provides a quantitative starting point for DaRT treatment planning.
Collapse
Affiliation(s)
- Lior Arazi
- Unit of Nuclear Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negav, POB 653 Beer-Sheva 8410501, Israel
| |
Collapse
|
44
|
Saw PE, Song EW. Phage display screening of therapeutic peptide for cancer targeting and therapy. Protein Cell 2019; 10:787-807. [PMID: 31140150 PMCID: PMC6834755 DOI: 10.1007/s13238-019-0639-7] [Citation(s) in RCA: 172] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 04/21/2019] [Indexed: 12/14/2022] Open
Abstract
Recently, phage display technology has been announced as the recipient of Nobel Prize in Chemistry 2018. Phage display technique allows high affinity target-binding peptides to be selected from a complex mixture pool of billions of displayed peptides on phage in a combinatorial library and could be further enriched through the biopanning process; proving to be a powerful technique in the screening of peptide with high affinity and selectivity. In this review, we will first discuss the modifications in phage display techniques used to isolate various cancer-specific ligands by in situ, in vitro, in vivo, and ex vivo screening methods. We will then discuss prominent examples of solid tumor targeting-peptides; namely peptide targeting tumor vasculature, tumor microenvironment (TME) and over-expressed receptors on cancer cells identified through phage display screening. We will also discuss the current challenges and future outlook for targeting peptide-based therapeutics in the clinics.
Collapse
Affiliation(s)
- Phei Er Saw
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Er-Wei Song
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
| |
Collapse
|
45
|
Kremheller J, Vuong AT, Schrefler BA, Wall WA. An approach for vascular tumor growth based on a hybrid embedded/homogenized treatment of the vasculature within a multiphase porous medium model. INTERNATIONAL JOURNAL FOR NUMERICAL METHODS IN BIOMEDICAL ENGINEERING 2019; 35:e3253. [PMID: 31441222 DOI: 10.1002/cnm.3253] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 07/04/2019] [Accepted: 08/16/2019] [Indexed: 05/13/2023]
Abstract
The aim of this work is to develop a novel computational approach to facilitate the modeling of angiogenesis during tumor growth. The preexisting vasculature is modeled as a 1D inclusion and embedded into the 3D tissue through a suitable coupling method, which allows for nonmatching meshes in 1D and 3D domain. The neovasculature, which is formed during angiogenesis, is represented in a homogenized way as a phase in our multiphase porous medium system. This splitting of models is motivated by the highly complex morphology, physiology, and flow patterns in the neovasculature, which are challenging and computationally expensive to resolve with a discrete, 1D angiogenesis and blood flow model. Moreover, it is questionable if a discrete representation generates any useful additional insight. By contrast, our model may be classified as a hybrid vascular multiphase tumor growth model in the sense that a discrete, 1D representation of the preexisting vasculature is coupled with a continuum model describing angiogenesis. It is based on an originally avascular model which has been derived via the thermodynamically constrained averaging theory. The new model enables us to study mass transport from the preexisting vasculature into the neovasculature and tumor tissue. We show by means of several illustrative examples that it is indeed capable of reproducing important aspects of vascular tumor growth phenomenologically.
Collapse
Affiliation(s)
- Johannes Kremheller
- Institute for Computational Mechanics, Technical University of Munich, Garching, Germany
| | - Anh-Tu Vuong
- Institute for Computational Mechanics, Technical University of Munich, Garching, Germany
| | - Bernhard A Schrefler
- Institute for Advanced Study, Technical University of Munich, Garching, Germany
- Department of Civil, Environmental and Architectural Engineering, University of Padova, Padua, Italy
| | - Wolfgang A Wall
- Institute for Computational Mechanics, Technical University of Munich, Garching, Germany
| |
Collapse
|
46
|
Critical Issues in the Development of Immunotoxins for Anticancer Therapy. J Pharm Sci 2019; 109:104-115. [PMID: 31669121 DOI: 10.1016/j.xphs.2019.10.037] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 09/23/2019] [Accepted: 10/21/2019] [Indexed: 12/16/2022]
Abstract
Immunotoxins (ITs) are attractive anticancer modalities aimed at cancer-specific delivery of highly potent cytotoxic protein toxins. An IT consists of a targeting domain (an antibody, cytokine, or another cell-binding protein) chemically conjugated or recombinantly fused to a highly cytotoxic payload (a bacterial and plant toxin or human cytotoxic protein). The mode of action of ITs is killing designated cancer cells through the effector function of toxins in the cytosol after cellular internalization via the targeted cell-specific receptor-mediated endocytosis. Although numerous ITs of diverse structures have been tested in the past decades, only 3 ITs-denileukin diftitox, tagraxofusp, and moxetumomab pasudotox-have been clinically approved for treating hematological cancers. No ITs against solid tumors have been approved for clinical use. In this review, we discuss critical research and development issues associated with ITs that limit their clinical success as well as strategies to overcome these obstacles. The issues include off-target and on-target toxicities, immunogenicity, human cytotoxic proteins, antigen target selection, cytosolic delivery efficacy, solid-tumor targeting, and developability. To realize the therapeutic promise of ITs, novel strategies for safe and effective cytosolic delivery into designated tumors, including solid tumors, are urgently needed.
Collapse
|
47
|
Sousa AR, Oliveira MJ, Sarmento B. Impact of CEA-targeting Nanoparticles for Drug Delivery in Colorectal Cancer. J Pharmacol Exp Ther 2019; 370:657-670. [PMID: 30670480 DOI: 10.1124/jpet.118.254441] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 01/18/2019] [Indexed: 12/18/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common causes of cancer-related death in the world, mainly owing to distant metastasis events. Developing targeted strategies to treat and follow individuals in more developed stages is needed. The carcinoembryonic antigen (CEA) is a cell surface-overexpressed glycoprotein in most CRC patients, and the evaluation of its serum levels is recommended in the clinic. These reasons motivated the production of CEA-targeted nanotechnologies for monitorization of CRC progression, but only a few centers have reported their use for drug delivery. The cellular internalization of CEA-linked nanosystems occurs by the natural recycling of the CEA itself, enabling longer retention and sustained release of the cargo. The functionalization of nanoparticles with lower affinity ligands for CEA is possibly the best choice to avoid their binding to the soluble CEA. Here, we also highlight the use of nanoparticles made of poly(lactic-co-glycolic acid) (PLGA) polymer, a well known material, owing to its biocompatibility and low toxicity. This work offers support to the contribution of antibody fragment-functionalized nanoparticles as promising high affinity molecules to decorate nanosystems. The linkers and conjugation chemistries chosen for ligand-nanoparticle coupling will be addressed herein as an elements essential to the modulation of nanosystem features. This review, to our knowledge, is the first that focuses on CEA-targeted nanotechnologies to serve colorectal cancer therapy and monitorization.
Collapse
Affiliation(s)
- Ana Rita Sousa
- Instituto de Investigação e Inovação em Saúde (A.R.S., M.J.O., B.S.), Instituto de Engenharia Biomédica (A.R.S., M.J.O., B.S.), Instituto de Ciências Biomédicas Abel Salazar (A.R.S., M.J.O.), and Faculdade de Medicina da (M.J.O.), Universidade do Porto, Porto, Portugal; Instituto Português de Oncologia do Porto, Porto, Portugal (A.R.S.); and Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde & Instituto Universitário de Ciências da Saúde, Gandra, Portugal (B.S.)
| | - Maria José Oliveira
- Instituto de Investigação e Inovação em Saúde (A.R.S., M.J.O., B.S.), Instituto de Engenharia Biomédica (A.R.S., M.J.O., B.S.), Instituto de Ciências Biomédicas Abel Salazar (A.R.S., M.J.O.), and Faculdade de Medicina da (M.J.O.), Universidade do Porto, Porto, Portugal; Instituto Português de Oncologia do Porto, Porto, Portugal (A.R.S.); and Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde & Instituto Universitário de Ciências da Saúde, Gandra, Portugal (B.S.)
| | - Bruno Sarmento
- Instituto de Investigação e Inovação em Saúde (A.R.S., M.J.O., B.S.), Instituto de Engenharia Biomédica (A.R.S., M.J.O., B.S.), Instituto de Ciências Biomédicas Abel Salazar (A.R.S., M.J.O.), and Faculdade de Medicina da (M.J.O.), Universidade do Porto, Porto, Portugal; Instituto Português de Oncologia do Porto, Porto, Portugal (A.R.S.); and Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde & Instituto Universitário de Ciências da Saúde, Gandra, Portugal (B.S.)
| |
Collapse
|
48
|
Kim DH, Khan H, Ullah H, Hassan STS, Šmejkal K, Efferth T, Mahomoodally MF, Xu S, Habtemariam S, Filosa R, Lagoa R, Rengasamy KR. MicroRNA targeting by quercetin in cancer treatment and chemoprotection. Pharmacol Res 2019; 147:104346. [PMID: 31295570 DOI: 10.1016/j.phrs.2019.104346] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 07/05/2019] [Accepted: 07/05/2019] [Indexed: 02/08/2023]
Abstract
A growing number of evidences from clinical and preclinical studies have shown that dysregulation of microRNA (miRNA) function contributes to the progression of cancer and thus miRNA can be an effective target in therapy. Dietary phytochemicals, such as quercetin, are natural products that have potential anti-cancer properties due to their proven antioxidant, anti-inflammatory, and anti-proliferative effects. Available experimental studies indicate that quercetin could modulate multiple cancer-relevant miRNAs including let-7, miR-21, miR-146a and miR-155, thereby inhibiting cancer initiation and development. This paper reviews the data supporting the use of quercetin for miRNA-mediated chemopreventive and therapeutic strategies in various cancers, with the aim to comprehensively understand its health-promoting benefits and pharmacological potential. Integration of technology platforms for miRNAs biomarker and drug discovery is also presented.
Collapse
Affiliation(s)
- Doo Hwan Kim
- Department of Bioresources and Food Science, Konkuk University, Seoul, 05029, South Korea
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan, 23200, Pakistan.
| | - Hammad Ullah
- Department of Pharmacy, Abdul Wali Khan University, Mardan, 23200, Pakistan
| | - Sherif T S Hassan
- Department of Natural Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences Brno, Palackého tř. 1946/1, 612 42, Brno, Czech Republic
| | - Karel Šmejkal
- Department of Natural Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences Brno, Palackého tř. 1946/1, 612 42, Brno, Czech Republic
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Johannes Gutenberg University, Staudinger Weg 5, 55128, Mainz, Germany
| | | | - Suowen Xu
- University of Rochester, Aab Cardiovascular Research Institute, Rochester, NY, 14623, USA
| | - Solomon Habtemariam
- Pharmacognosy Research Laboratories and Herbal Analysis Services UK, University of Greenwich, UK
| | - Rosanna Filosa
- Institute of Food Sciences, National Research Council, Roma str. 64, Avellino, 83100, Italy; Consorzio Sannio Tech, AMP Biotec, Appia Str, Apollosa, Benevento, 82030, Italy
| | - Ricardo Lagoa
- School of Technology and Management, Polytechnic Institute of Leiria, Portugal; UCIBIO-Faculty of Science and Technology, University NOVA of Lisbon, Portugal.
| | - Kannan Rr Rengasamy
- Department of Bioresources and Food Science, Konkuk University, Seoul, 05029, South Korea.
| |
Collapse
|
49
|
Akbari-Karadeh S, Aghamiri SMR, Tajer-Mohammad-Ghazvini P, Ghorbanzadeh-Mashkani S. Radiolabeling of Biogenic Magnetic Nanoparticles with Rhenium-188 as a Novel Agent for Targeted Radiotherapy. Appl Biochem Biotechnol 2019; 190:540-550. [DOI: 10.1007/s12010-019-03079-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Accepted: 07/05/2019] [Indexed: 12/11/2022]
|
50
|
Cheng HB, Cui Y, Wang R, Kwon N, Yoon J. The development of light-responsive, organic dye based, supramolecular nanosystems for enhanced anticancer therapy. Coord Chem Rev 2019. [DOI: 10.1016/j.ccr.2019.04.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|