51
|
Balma M, Liberini V, Racca M, Laudicella R, Bauckneht M, Buschiazzo A, Nicolotti DG, Peano S, Bianchi A, Albano G, Quartuccio N, Abgral R, Morbelli SD, D'Alessandria C, Terreno E, Huellner MW, Papaleo A, Deandreis D. Non-conventional and Investigational PET Radiotracers for Breast Cancer: A Systematic Review. Front Med (Lausanne) 2022; 9:881551. [PMID: 35492341 PMCID: PMC9039137 DOI: 10.3389/fmed.2022.881551] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 03/22/2022] [Indexed: 02/05/2023] Open
Abstract
Breast cancer is one of the most common malignancies in women, with high morbidity and mortality rates. In breast cancer, the use of novel radiopharmaceuticals in nuclear medicine can improve the accuracy of diagnosis and staging, refine surveillance strategies and accuracy in choosing personalized treatment approaches, including radioligand therapy. Nuclear medicine thus shows great promise for improving the quality of life of breast cancer patients by allowing non-invasive assessment of the diverse and complex biological processes underlying the development of breast cancer and its evolution under therapy. This review aims to describe molecular probes currently in clinical use as well as those under investigation holding great promise for personalized medicine and precision oncology in breast cancer.
Collapse
Affiliation(s)
- Michele Balma
- Nuclear Medicine Department, S. Croce e Carle Hospital, Cuneo, Italy
| | - Virginia Liberini
- Nuclear Medicine Department, S. Croce e Carle Hospital, Cuneo, Italy
- Division of Nuclear Medicine, Department of Medical Science, University of Turin, Turin, Italy
| | - Manuela Racca
- Nuclear Medicine Unit, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Riccardo Laudicella
- Department of Biomedical and Dental Sciences and of Morpho-Functional Imaging, Nuclear Medicine Unit, University of Messina, Messina, Italy
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Nuclear Medicine Unit, Fondazione Istituto G. Giglio, Cefalù, Italy
| | - Matteo Bauckneht
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Health Science (DISSAL), University of Genoa, Genoa, Italy
| | - Ambra Buschiazzo
- Nuclear Medicine Department, S. Croce e Carle Hospital, Cuneo, Italy
| | | | - Simona Peano
- Nuclear Medicine Department, S. Croce e Carle Hospital, Cuneo, Italy
| | - Andrea Bianchi
- Nuclear Medicine Department, S. Croce e Carle Hospital, Cuneo, Italy
| | - Giovanni Albano
- Nuclear Medicine Unit, Fondazione Istituto G. Giglio, Cefalù, Italy
| | - Natale Quartuccio
- Nuclear Medicine Unit, A.R.N.A.S. Civico di Cristina and Benfratelli Hospitals, Palermo, Italy
| | - Ronan Abgral
- Department of Nuclear Medicine, University Hospital of Brest, Brest, France
| | - Silvia Daniela Morbelli
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Health Science (DISSAL), University of Genoa, Genoa, Italy
| | | | - Enzo Terreno
- Department of Molecular Biotechnology and Health Sciences, Molecular & Preclinical Imaging Centers, University of Turin, Turin, Italy
| | - Martin William Huellner
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Alberto Papaleo
- Nuclear Medicine Department, S. Croce e Carle Hospital, Cuneo, Italy
| | - Désirée Deandreis
- Division of Nuclear Medicine, Department of Medical Science, University of Turin, Turin, Italy
| |
Collapse
|
52
|
Gamal-Eldeen AM, Alrehaili AA, Alharthi A, Raafat BM. Perftoran® Inhibits Hypoxia-Associated Resistance in Lung Cancer Cells to Carboplatin. Front Pharmacol 2022; 13:860898. [PMID: 35401227 PMCID: PMC8987772 DOI: 10.3389/fphar.2022.860898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 02/24/2022] [Indexed: 11/29/2022] Open
Abstract
Perftoran® (perfluorodecalin) is an oxygen carrier, and carboplatin is a common chemotherapy drug used worldwide for lung cancer treatment. Hypoxia is one of the factors that induce resistance of lung cancer cells to carboplatin. This study explored the role of Perftoran®, as an oxygen carrier, in lowering the resistance of lung cancer cells to carboplatin through suppression of hypoxia pathway mediators. The effect of Perftoran® on the resistance of human lung cancer A549 cells to carboplatin was investigated through the evaluation of cytotoxicity by MTT, cell death mode by dual DNA staining, DNA damage by comet assay, DNA platination (DNA/carboplatin adducts) by atomic absorption spectroscopy, hypoxia degree by pimonidazole, HIF-1α/HIF-2α concentrations by ELISA, expression of miRNAs (hypoxamiRs miR-210, miR-21, and miR-181a) by qRT-PCR, and the content of drug resistance transporter MRP-2 by immunocytochemical staining. Results indicated that compared to carboplatin, Perftoran®/carboplatin decreased cell resistance to carboplatin by potentiating its cytotoxicity using only 45% of carboplatin IC50 and inducing apoptosis. Perftoran® induced DNA platination and DNA damage index in cells compared to carboplatin alone. Moreover, compared to treatment with carboplatin alone, co-treatment of cells with Perftoran® and carboplatin inhibited cellular pimonidazole hypoxia adducts, diminished HIF-1α/HIF-2α concentrations, suppressed hypoxamiR expression, and decreased MRP-2. In conclusion, Perftoran® inhibited resistance of lung cancer cells to carboplatin through the inhibition of both hypoxia pathway mediators and the drug resistance transporter MRP-2 and through the induction of DNA/carboplatin adduct formation.
Collapse
Affiliation(s)
- Amira M. Gamal-Eldeen
- Clinical Laboratory Sciences Department, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
- *Correspondence: Amira M. Gamal-Eldeen,
| | - Amani A. Alrehaili
- Clinical Laboratory Sciences Department, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Afaf Alharthi
- Clinical Laboratory Sciences Department, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Bassem M. Raafat
- Radiological Sciences Department, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| |
Collapse
|
53
|
Wijayasinghe YS, Bhansali MP, Borkar MR, Chaturbhuj GU, Muntean BS, Viola RE, Bhansali PR. A Comprehensive Biological and Synthetic Perspective on 2-Deoxy-d-Glucose (2-DG), A Sweet Molecule with Therapeutic and Diagnostic Potentials. J Med Chem 2022; 65:3706-3728. [PMID: 35192360 DOI: 10.1021/acs.jmedchem.1c01737] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Glucose, the primary substrate for ATP synthesis, is catabolized during glycolysis to generate ATP and precursors for the synthesis of other vital biomolecules. Opportunistic viruses and cancer cells often hijack this metabolic machinery to obtain energy and components needed for their replication and proliferation. One way to halt such energy-dependent processes is by interfering with the glycolytic pathway. 2-Deoxy-d-glucose (2-DG) is a synthetic glucose analogue that can inhibit key enzymes in the glycolytic pathway. The efficacy of 2-DG has been reported across an array of diseases and disorders, thereby demonstrating its broad therapeutic potential. Recent approval of 2-DG in India as a therapeutic approach for the management of the COVID-19 pandemic has brought renewed attention to this molecule. The purpose of this perspective is to present updated therapeutic avenues as well as a variety of chemical synthetic strategies for this medically useful sugar derivative, 2-DG.
Collapse
Affiliation(s)
- Yasanandana S Wijayasinghe
- Department of Biochemistry and Clinical Chemistry, Faculty of Medicine, University of Kelaniya, Ragama 11010, Sri Lanka
| | | | - Maheshkumar R Borkar
- Department of Pharmaceutical Chemistry, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai 400056, Maharashtra, India
| | - Ganesh U Chaturbhuj
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Matunga (E), Mumbai 400019, Maharashtra, India
| | - Brian S Muntean
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia 30912, United States
| | - Ronald E Viola
- Department of Chemistry and Biochemistry, University of Toledo, Toledo, Ohio 43606, United States
| | - Pravin R Bhansali
- Department of Science, Faculty of Science and Technology, Alliance University, Chikkahagade Cross, Chandapura-Anekal Main Road, Anekal, Bengaluru 562106, Karnataka, India
| |
Collapse
|
54
|
Treatment of Pheochromocytoma Cells with Recurrent Cycles of Hypoxia: A New Pseudohypoxic In Vitro Model. Cells 2022; 11:cells11030560. [PMID: 35159368 PMCID: PMC8834104 DOI: 10.3390/cells11030560] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/26/2022] [Accepted: 02/03/2022] [Indexed: 02/04/2023] Open
Abstract
Continuous activation of hypoxia pathways in pheochromocytomas and paragangliomas (PPGLs) is associated with higher disease aggressiveness, for which effective treatment strategies are still missing. Most of the commonly used in vitro models lack characteristics of these pseudohypoxic tumors, including elevated expression of hypoxia-inducible factor (HIF) 2α. To address this shortcoming, we investigated whether recurrent hypoxia cycles lead to continuous activation of hypoxia pathways under normoxic conditions and whether this pseudohypoxia is associated with increased cellular aggressiveness. Rat pheochromocytoma cells (PC12) were incubated under hypoxia for 24 h every 3–4 days, up to 20 hypoxia–reoxygenation cycles, resulting in PC12 Z20 cells. PC12 Z20 control cells were obtained by synchronous cultivation under normoxia. RNA sequencing revealed upregulation of HIF2α in PC12 Z20 cells and a pseudohypoxic gene signature that overlapped with the gene signature of pseudohypoxic PPGLs. PC12 Z20 cells showed a higher growth rate, and the migration and adhesion capacity were significantly increased compared with control cells. Changes in global methylation, together with the pseudohypoxic conditions, may be responsible for the increased aggressiveness of this new model. The established sub-cell line with characteristics of pseudohypoxic PPGLs represent a complementary model for further investigations, for example, with regard to new therapeutic approaches.
Collapse
|
55
|
Lu S, Feng W, Dong C, Song X, Gao X, Guo J, Chen Y, Hu Z. Photosynthetic Oxygenation-Augmented Sonodynamic Nanotherapy of Hypoxic Tumors. Adv Healthc Mater 2022; 11:e2102135. [PMID: 34787379 DOI: 10.1002/adhm.202102135] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/02/2021] [Indexed: 12/31/2022]
Abstract
Reactive oxygen species (ROS) has been employed as a powerful therapeutic agent for eradicating tumor via oxidative stress. As an emerging ROS-involving noninvasive anticancer therapeutic modality, sonodynamic therapy (SDT) with high tissue penetration depth and benign remote spatiotemporal selectivity has been progressively utilized as the distinct alternative for ROS-based tumor treatment. However, the hypoxic tumor microenvironment substantially restricts the sonodynamic effect. In this work, an oxygen self-sufficient hybrid sonosensitizer on the basis of photosynthetic microorganisms cyanobacteria (Cyan) integrated with ultrasmall oxygen-deficient bimetallic oxide Mn1.4 WOx nanosonosensitizers, termed as M@C, is designed and engineered to overcome the critical issue of hypoxia-induced tumor resistance and strengthen the SDT effect. The sustained photosynthetic oxygen production by Cyan under light illumination can promote Mn1.4 WOx nanosonosensitizers to produce more ROS against cancer cells both in vitro and in vivo under ultrasound (US) irradiation. Especially, the sustained oxygen evolution for suppressing the gene expression of hypoxia-inducible factor 1alpha (HIF-1α) further boosts and augments the SDT efficiency. Thus, this work provides the paradigm that the rationally engineered biohybrid microorganism-based multifunctional sonosensitizers can serve as an effective bioplatform for augmenting the therapeutic efficiency of SDT, particularly for the treatment of hypoxic tumors.
Collapse
Affiliation(s)
- Shuting Lu
- Center of Interventional Radiology and Vascular Surgery Department of Radiology and Ultrasound Zhongda Hospital Medical School Southeast University Nanjing 210009 P. R. China
| | - Wei Feng
- Materdicine Lab School of Life Sciences Shanghai University Shanghai 200444 P. R. China
| | - Caihong Dong
- Department of Ultrasound Zhongshan Hospital Fudan University Shanghai 200032 P. R. China
| | - Xinran Song
- Department of Medical Ultrasound Shanghai Tenth People's Hospital Ultrasound Research and Education Institute Tongji University Cancer Center Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment Tongji University School of Medicine Shanghai 200072 P. R. China
| | - Xiang Gao
- Key Laboratory of Quantitative Engineering Biology Institute of Synthetic Biology Shenzhen Institute of Advanced Technology Chinese Academic of Science Shenzhen 518000 P. R. China
| | - Jinhe Guo
- Center of Interventional Radiology and Vascular Surgery Department of Radiology and Ultrasound Zhongda Hospital Medical School Southeast University Nanjing 210009 P. R. China
| | - Yu Chen
- Materdicine Lab School of Life Sciences Shanghai University Shanghai 200444 P. R. China
| | - Zhongqian Hu
- Center of Interventional Radiology and Vascular Surgery Department of Radiology and Ultrasound Zhongda Hospital Medical School Southeast University Nanjing 210009 P. R. China
| |
Collapse
|
56
|
Ni J, Xu H, Zhong Y, Zhou Y, Hu S. Activatable UCL/CT/MR-enhanced in vivo imaging-guided radiotherapy and photothermal therapy. J Mater Chem B 2022; 10:549-561. [PMID: 34985095 DOI: 10.1039/d1tb02006d] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Although sophisticated radiotherapy (RT) technology has been widely applied in clinical oncotherapy, unsatisfactory therapeutic effects due to hypoxic tumor microenvironments and complications are still prevalent. Herein, copper sulphide nanoparticles (CuS NPs) wrapped on the surface of upconversion nanoparticles (UCNPs) via manganese dioxide (MnO2) coatings were synthesized for O2 self-supplementing and enhanced combinational RT/photothermal therapy (PTT). In our design, the nanoplatforms can be rapidly enriched at tumor sites by the enhanced permeability and retention (EPR) effect and respond to the tumor microenvironment. The surface MnO2 coatings can interact with over-expressed H2O2 in tumors and cause an abundant generation of oxygen for hypoxic improvement, leading to an enhanced RT. More importantly, by irradiation with near-infrared light, the scattered CuS NPs can convert light energy into heat to destroy tumor cells and reinforce the therapeutic effects of RT. Furthermore, these NPs also displayed excellent performances in upconversion fluorescence imaging (UCL), computerized tomographic (CT) scanning and magnetic resonance imaging (MRI), demonstrating a potential imaging-guided cancer therapy system.
Collapse
Affiliation(s)
- Jianming Ni
- Radiology Department, Affiliated Wuxi No. 2 People's Hospital, Nanjing Medical University, Wuxi, 214002, China
| | - Huiting Xu
- Radiology Department, Affiliated Wuxi No. 2 People's Hospital, Nanjing Medical University, Wuxi, 214002, China
| | - Yanqi Zhong
- Radiology Department, Affiliated Hospital, Jiangnan University, Wuxi, 214122, China.
| | - Yongping Zhou
- Hepatobiliary surgery Department, Affiliated Wuxi No. 2 People's Hospital, Nanjing Medical University, Wuxi, 214002, China.
| | - Shudong Hu
- Radiology Department, Affiliated Hospital, Jiangnan University, Wuxi, 214122, China.
| |
Collapse
|
57
|
Hao Y, Gao Y, Fan Y, Zhang C, Zhan M, Cao X, Shi X, Guo R. A tumor microenvironment-responsive poly(amidoamine) dendrimer nanoplatform for hypoxia-responsive chemo/chemodynamic therapy. J Nanobiotechnology 2022; 20:43. [PMID: 35062953 PMCID: PMC8781438 DOI: 10.1186/s12951-022-01247-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 01/04/2022] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Chemodynamic therapy is a promising cancer treatment with specific therapeutic effect at tumor sites, as toxic hydroxyl radical (·OH) could only be generated by Fenton or Fenton-like reaction in the tumor microenvironment (TME) with low pH and high level of endogenous hydrogen peroxide. However, the low concentration of catalytic metal ions, excessive glutathione (GSH) and aggressive hypoxia at tumor site seriously restrict the curative outcomes of conventional chemodynamic therapy. RESULTS In this study, polyethylene glycol-phenylboronic acid (PEG-PBA)-modified generation 5 (G5) poly(amidoamine) (PAMAM) dendrimers were synthesized as a targeted nanocarrier to chelate Cu(II) and then encapsulate hypoxia-sensitive drug tirapazamine (TPZ) by the formation of hydrophobic Cu(II)/TPZ complex for hypoxia-enhanced chemo/chemodynamic therapy. The formed G5.NHAc-PEG-PBA@Cu(II)/TPZ (GPPCT) nanoplatform has good stability and hemocompatibility, and could release Cu(II) ions and TPZ quickly in weakly acidic tumor sites via pH-sensitive dissociation of Cu(II)/TPZ. In vitro experiments showed that the GPPCT nanoplatforms can efficiently target murine breast cancer cells (4T1) cells overexpressing sialic acid residues, and show a significantly enhanced inhibitory effect on hypoxic cells by the activation of TPZ. The excessive GSH in tumors could be depleted by the reduction of Cu(II) to Cu(I), and abundant of toxic ·OH would be generated in tumor cells by Fenton reaction for chemodynamic therapy. In vivo experiments demonstrated that the GPPCT nanoplatform could specifically accumulate at tumors, effectively inhibit the growth and metastasis of tumors by the combination of CDT and chemotherapy, and be metabolized with no systemic toxicity. CONCLUSIONS The targeted GPPCT nanoplatform may represent an effective model for the synergistic inhibition of different tumor types by hypoxia-enhanced chemo/chemodynamic therapy.
Collapse
Affiliation(s)
- Yingchao Hao
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, China
| | - Yue Gao
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, China
| | - Yu Fan
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, China
| | - Changchang Zhang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, China
| | - Mengsi Zhan
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, China
| | - Xueyan Cao
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, China
| | - Xiangyang Shi
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, China.
| | - Rui Guo
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, China.
| |
Collapse
|
58
|
Fathi M, Bahmanpour S, Barshidi A, Rasouli H, Karoon Kiani F, Mahmoud Salehi Khesht A, Izadi S, Rashidi B, Kermanpour S, Mokhtarian R, Karpisheh V, Hassannia H, Mohammadi H, Jalili A, Jadidi-Niaragh F. Simultaneous blockade of TIGIT and HIF-1α induces synergistic anti-tumor effect and decreases the growth and development of cancer cells. Int Immunopharmacol 2021; 101:108288. [PMID: 34710844 DOI: 10.1016/j.intimp.2021.108288] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/11/2021] [Accepted: 10/18/2021] [Indexed: 01/19/2023]
Abstract
PURPOSE T-cell immunoglobulin and ITIM domain (TIGIT) is an immune checkpoint that is overexpressed on both immune cells and some cancer cells. TIGIT can alter the anti-tumor responses inside the tumor microenvironment. Hypoxia-inducible factor 1-alpha (HIF-1α) plays a significant role in the TME and involves suppressing the anti-tumor responses. Under hypoxic conditions, HIF-1α can enhance the expression of different immune checkpoints. Accordingly, hypoxic TME and TIGIT overexpression cause cancer development. Thus, we decided to inhibit tumor cell expansion by inhibiting TIGIT and HIF-1α molecules and discovering the relationship between TIGIT and HIF-1α. METHODS In this research, we utilized superparamagnetic iron oxide-based NPs (SPIONs) combined with chitosan lactate (CL) and folic acid (FA) nanoparticles (NPs) loaded with TIGIT-siRNA and HIF-1α- siRNA for suppressing TIGIT and HIF-1α in tumor cells and evaluated the consequences of this treatment strategy on tumor growth, apoptosis, and metastasis. RESULTS The results showed that cancer cells treated with TIGIT and HIF-1α siRNA-loaded SPIONs-CL-FA NPs, strongly suppressed the TIGIT and HIF-1α expression, colony formation ability, angiogenesis, and the growth rate of cancer cells. CONCLUSIONS Present data suggest the combination treatment of TIGIT and HIF-1α as a novel treatment strategy against colorectal and breast cancer, but more researches are required to realize the complete role of TIGIT and HIF-1α inside the TME.
Collapse
Affiliation(s)
- Mehrdad Fathi
- Student Research Committee, Kurdistan University of Medical Sciences, Sanandaj, Iran; Cancer and Immunology Research Center, Kurdistan University of Medical Sciences, Sanandaj, Iran; Department of Immunology, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Simin Bahmanpour
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Asal Barshidi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Rasouli
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fariba Karoon Kiani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Sepideh Izadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bentolhoda Rashidi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shiva Kermanpour
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Roya Mokhtarian
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahid Karpisheh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Hassannia
- Immunogenetic Research Center, Faculty of Medicine and Amol Faculty of Paramedical Sciences, Mazandaran University of Medical Sciences, Sari, Iran
| | - Hamed Mohammadi
- Non-communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Ali Jalili
- Cancer and Immunology Research Center, Kurdistan University of Medical Sciences, Sanandaj, Iran; Department of Immunology, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
59
|
Feng S, Qiao W, Tang J, Yu Y, Gao S, Liu Z, Zhu X. Chemotherapy Augmentation Using Low-Intensity Ultrasound Combined with Microbubbles with Different Mechanical Indexes in a Pancreatic Cancer Model. ULTRASOUND IN MEDICINE & BIOLOGY 2021; 47:3221-3230. [PMID: 34362582 DOI: 10.1016/j.ultrasmedbio.2021.07.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 07/06/2021] [Accepted: 07/12/2021] [Indexed: 06/13/2023]
Abstract
The aim of the study was to explore the optimal mechanical indexes (MIs) for low-intensity ultrasound (LIUS) combined with microbubbles to enhance tumor blood perfusion and improve drug concentration in pancreatic cancer-bearing nude mice. Fifty-four nude mice bearing bilateral pancreatic tumors on the hind legs were randomly divided into three groups (the MI was set at 0.3, 0.7 and 1.1 in groups A, B and C, respectively). Five nude mice in each group were intravenously injected with the fluorescent dye DiR iodide (DiIC18(7),1,1'-dioctadecyl-3,3,3',3'-tetramethylindotricarbocyanine iodide); for each mouse, one tumor was treated with LIUS combined with microbubbles, and the contralateral tumor was exposed to sham ultrasound. In vivo fluorescence imaging was performed to detect the enrichment of intratumoral DiR iodide. Twelve mice in each group were intravenously injected with doxorubicin (DOX) and underwent ultrasound therapy as described above. Tumor blood perfusion changes were quantitatively evaluated with pre- and post-treatment contrast-enhanced ultrasound (CEUS, MI = 0.08). One hour after the post-treatment CEUS, nude mice were sacrificed to determine the DOX concentration in tumor tissue; one mouse in each group was sacrificed after ultrasound treatment for tumor hematoxylin-eosin staining examination. CEUS quantitative analysis and in vivo fluorescence images confirmed that LIUS at MI = 0.3 combined with microbubbles was able to enhance tumor blood flow and increase regional fluorescence dye DiR iodide concentration. The DOX concentration on the therapeutic side was significantly higher than that on the control side after ultrasound-stimulated (MI = 0.3) microbubble cavitation (USMC) treatment (1.45 ± 0.53 μg/g vs. 1.07 ± 0.46 μg/g, t = -5.163, p = 0.001). However, in groups B and C, there were no significant differences in DOX concentration between the therapeutic and control sides (Z = -0.297, -0.357, p = 0.766, 0.721). No hemorrhage or other tissue damage was observed in hematoxylin-eosin-stained tumor specimens of both sides in all groups. LIUS at MI = 0.3 combined with microbubbles was able to enhance tumor blood perfusion and improve local drug concentration in nude mice bearing pancreatic cancer.
Collapse
Affiliation(s)
- Shuang Feng
- Department of Ultrasound, General Hospital of Southern Theatre Command, Guangzhou, China
| | - Wei Qiao
- Department of Ultrasound, Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Jiawei Tang
- Department of Ultrasound, Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Yanlan Yu
- Department of Ultrasound, Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Shunji Gao
- Department of Ultrasound, General Hospital of Central Theatre Command, Wuhan, China
| | - Zheng Liu
- Department of Ultrasound, Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Xiansheng Zhu
- Department of Ultrasound, General Hospital of Southern Theatre Command, Guangzhou, China.
| |
Collapse
|
60
|
Cao W, Jin M, Yang K, Chen B, Xiong M, Li X, Cao G. Fenton/Fenton-like metal-based nanomaterials combine with oxidase for synergistic tumor therapy. J Nanobiotechnology 2021; 19:325. [PMID: 34656118 PMCID: PMC8520258 DOI: 10.1186/s12951-021-01074-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 10/04/2021] [Indexed: 02/07/2023] Open
Abstract
Chemodynamic therapy (CDT) catalyzed by transition metal and starvation therapy catalyzed by intracellular metabolite oxidases are both classic tumor treatments based on nanocatalysts. CDT monotherapy has limitations including low catalytic efficiency of metal ions and insufficient endogenous hydrogen peroxide (H2O2). Also, single starvation therapy shows limited ability on resisting tumors. The “metal-oxidase” cascade catalytic system is to introduce intracellular metabolite oxidases into the metal-based nanoplatform, which perfectly solves the shortcomings of the above-mentioned monotherapiesIn this system, oxidases can not only consume tumor nutrients to produce a “starvation effect”, but also provide CDT with sufficient H2O2 and a suitable acidic environment, which further promote synergy between CDT and starvation therapy, leading to enhanced antitumor effects. More importantly, the “metal-oxidase” system can be combined with other antitumor therapies (such as photothermal therapy, hypoxia-activated drug therapy, chemotherapy, and immunotherapy) to maximize their antitumor effects. In addition, both metal-based nanoparticles and oxidases can activate tumor immunity through multiple pathways, so the combination of the “metal-oxidase” system with immunotherapy has a powerful synergistic effect. This article firstly introduced the metals which induce CDT and the oxidases which induce starvation therapy and then described the “metal-oxidase” cascade catalytic system in detail. Moreover, we highlight the application of the “metal-oxidase” system in combination with numerous antitumor therapies, especially in combination with immunotherapy, expecting to provide new ideas for tumor treatment.
Collapse
Affiliation(s)
- Wei Cao
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China
| | - Mengyao Jin
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China
| | - Kang Yang
- Department of General Surgery, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China
| | - Bo Chen
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China.
| | - Maoming Xiong
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China.
| | - Xiang Li
- State Key Laboratory of Silicon Materials, School of Materials Science and Engineering, Zhejiang University, Hangzhou, 310027, People's Republic of China.
| | - Guodong Cao
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China.
| |
Collapse
|
61
|
Bastani S, Akbarzadeh M, Rastgar Rezaei Y, Farzane A, Nouri M, Mollapour Sisakht M, Fattahi A, Akbarzadeh M, Reiter RJ. Melatonin as a Therapeutic Agent for the Inhibition of Hypoxia-Induced Tumor Progression: A Description of Possible Mechanisms Involved. Int J Mol Sci 2021; 22:10874. [PMID: 34639215 PMCID: PMC8509383 DOI: 10.3390/ijms221910874] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/24/2021] [Accepted: 09/29/2021] [Indexed: 12/27/2022] Open
Abstract
Hypoxia has an important role in tumor progression via the up-regulation of growth factors and cellular adaptation genes. These changes promote cell survival, proliferation, invasion, metastasis, angiogenesis, and energy metabolism in favor of cancer development. Hypoxia also plays a central role in determining the resistance of tumors to chemotherapy. Hypoxia of the tumor microenvironment provides an opportunity to develop new therapeutic strategies that may selectively induce apoptosis of the hypoxic cancer cells. Melatonin is well known for its role in the regulation of circadian rhythms and seasonal reproduction. Numerous studies have also documented the anti-cancer properties of melatonin, including anti-proliferation, anti-angiogenesis, and apoptosis promotion. In this paper, we hypothesized that melatonin exerts anti-cancer effects by inhibiting hypoxia-induced pathways. Considering this action, co-administration of melatonin in combination with other therapeutic medications might increase the effectiveness of anti-cancer drugs. In this review, we discussed the possible signaling pathways by which melatonin inhibits hypoxia-induced cancer cell survival, invasion, migration, and metabolism, as well as tumor angiogenesis.
Collapse
Affiliation(s)
- Sepideh Bastani
- Research Center for Pharmaceutical Nanotechnology (RCPN), Tabriz University of Medical Sciences, Tabriz 51368, Iran;
- Stem Cell And Regenerative Medicine Institute (SCARM), Tabriz University of Medical Sciences, Tabriz 51368, Iran;
| | - Moloud Akbarzadeh
- Stem Cell And Regenerative Medicine Institute (SCARM), Tabriz University of Medical Sciences, Tabriz 51368, Iran;
- Department of Cellular and Molecular Biology, Faculty of Biological Science, Azarbaijan Shahid Madani University, Tabriz 51368, Iran
| | - Yeganeh Rastgar Rezaei
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz 51368, Iran;
| | - Ali Farzane
- Department of Health Information Management, School of Allied Medical Science, Tehran University of Medical Sciences, Tehran 11369, Iran;
| | - Mohammad Nouri
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz 51368, Iran;
| | - Mahsa Mollapour Sisakht
- Stem Cell and Regenerative Medicine Center of Excellence, Tehran University of Medical Sciences, Tehran 11369, Iran;
- Department of Biochemistry, Erasmus University Medical Center, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Amir Fattahi
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz 51368, Iran;
- Department of Obstetrics and Gynecology, Erlangen University Hospital, Friedrich-Alexander University of Erlangen–Nürnberg, Comprehensive Cancer Center ER-EMN, 91054 Erlangen, Germany
| | - Maryam Akbarzadeh
- Department of Biochemistry, Erasmus University Medical Center, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Russel J. Reiter
- Department of Cell Systems and Anatomy, UT Health, Long School of Medicine, San Antonio, TX 78229, USA;
| |
Collapse
|
62
|
Silva VL, Ruiz A, Ali A, Pereira S, Seitsonen J, Ruokolainen J, Furlong F, Coulter J, Al-Jamal WT. Hypoxia-targeted cupric-tirapazamine liposomes potentiate radiotherapy in prostate cancer spheroids. Int J Pharm 2021; 607:121018. [PMID: 34416329 DOI: 10.1016/j.ijpharm.2021.121018] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 08/13/2021] [Accepted: 08/16/2021] [Indexed: 12/16/2022]
Abstract
In this study, novel cupric-tirapazamine [Cu(TPZ)2]-liposomes were developed as an effective hypoxia-targeted therapeutic, which potentiated radiotherapy in a three dimensional (3D) prostate cancer (PCa) model. To overcome the low water solubility of the Cu(TPZ)2, a remote loading method was developed to efficiently load the lipophilic complex into different liposomal formulations. The effect of pH, temperature, PEGylation, lipid composition, liposome size, lipid: complex ratio on the liposome properties, and drug loading was evaluated. The highest loading efficiency was obtained at neutral pH, which was independent of lipid composition and incubation time. In addition, enhanced drug loading was achieved upon decreasing the lipid:complex molar ratio with minimal effects on liposomes' morphology. Interestingly, the in vitro potency of the developed liposomes was easily manipulated by changing the lipid composition. The hydrophilic nature of our liposomal formulations improved the complex's solubility, leading to enhanced cellular uptake and toxicity, both in PCa monolayers and tumour spheroids. Moreover, Cu(TPZ)2-loaded liposomes combined with radiation, showed a significant reduction in PCa spheroids growth rate, compared to the free complex or radiation alone, which could potentiate radiotherapy in patients with localised advanced PCa.
Collapse
Affiliation(s)
- Vera L Silva
- School of Pharmacy - University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, United Kingdom
| | - Amalia Ruiz
- School of Pharmacy - Queen's University Belfast, Belfast BT9 7BL, United Kingdom
| | - Ahlam Ali
- School of Pharmacy - Queen's University Belfast, Belfast BT9 7BL, United Kingdom
| | - Sara Pereira
- School of Pharmacy - Queen's University Belfast, Belfast BT9 7BL, United Kingdom
| | - Jani Seitsonen
- Department of Applied Physics, Aalto University School of Science, P.O. Box 15100, FI-00076 Aalto, Finland
| | - Janne Ruokolainen
- Department of Applied Physics, Aalto University School of Science, P.O. Box 15100, FI-00076 Aalto, Finland
| | - Fiona Furlong
- School of Pharmacy - Queen's University Belfast, Belfast BT9 7BL, United Kingdom
| | - Jonathan Coulter
- School of Pharmacy - Queen's University Belfast, Belfast BT9 7BL, United Kingdom
| | - Wafa' T Al-Jamal
- School of Pharmacy - University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, United Kingdom; School of Pharmacy - Queen's University Belfast, Belfast BT9 7BL, United Kingdom.
| |
Collapse
|
63
|
The Role of the Hedgehog Pathway in Cholangiocarcinoma. Cancers (Basel) 2021; 13:cancers13194774. [PMID: 34638259 PMCID: PMC8507550 DOI: 10.3390/cancers13194774] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/17/2021] [Accepted: 09/21/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Cholangiocarcinoma (CCA) is one of the most refractory malignancies with a high mortality rate. Among all the pathways involved in CCA development, emerging evidence highlights Hedgehog (HH) signaling as a substantial player in CCA-genesis and development. The pro-tumoral function of HH provides potential therapeutic implications, and recently the use of HH inhibitors has paved the way for clinical application in various solid tumors. Targeting HH members, namely Hedgehog ligands, SMO transmembrane protein and GLI transcription factors may thus confer therapeutic options for the improvement of CCA treatment outcome. Abstract Cholangiocarcinoma (CCA) is a poorly treatable type of cancer and, along with hepatocellular carcinoma (HCC), is the predominant type of primitive liver cancer in adults. The lack of understanding of CCA biology has slowed down the identification of novel targets and the development of effective treatments. While tumors share some general characteristics, detailed knowledge of specific features is essential for the development of effectively tailored therapeutic approaches. The Hedgehog (HH) signaling cascade regulates stemness biology, embryonal development, tissue homeostasis, and cell proliferation and differentiation. Its aberrant activation has been associated with a variety of solid and hematological human malignancies. Several HH-inhibiting compounds have been indeed developed as potential anticancer agents in different types of tumors, with Smoothened and GLI inhibitors showing the most promising results. Beside its well-established function in other tumors, findings regarding the HH signaling in CCA are still controversial. Here we will give an overview of the most important clinical and molecular features of cholangiocarcinoma, and we will discuss the available evidence of the crosstalk between the HH signaling pathway and the cholangiocarcinoma cell biology.
Collapse
|
64
|
He J, Fu LH, Qi C, Lin J, Huang P. Metal peroxides for cancer treatment. Bioact Mater 2021; 6:2698-2710. [PMID: 33665502 PMCID: PMC7895646 DOI: 10.1016/j.bioactmat.2021.01.026] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/02/2021] [Accepted: 01/21/2021] [Indexed: 12/13/2022] Open
Abstract
In recent years, metal peroxide (MO2) such as CaO2 has received more and more attention in cancer treatment. MO2 is readily decompose to release metal ions and hydrogen peroxide in the acidic tumor microenvironment (TME), resulting metal ions overloading, decreased acidity and elevated oxidative stress in TME. All of these changes making MO2 an excellent tumor therapeutic agent. Moreover, by combining MO2 with photosensitizers, enzymes or Fenton reagents, MO2 can assist and promote various tumor therapies such as photodynamic therapy and chemodynamic therapy. In this review, the synthesis and modification methods of MO2 are introduced, and the representative studies of MO2-based tumor monotherapy and combination therapy are discussed in detail. Finally, the current challenges and prospects of MO2 in the field of tumor therapy are emphasized to promote the development of MO2-based cancer treatment.
Collapse
Affiliation(s)
- Jin He
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, China
| | - Lian-Hua Fu
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, China
| | - Chao Qi
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Jing Lin
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, China
| | - Peng Huang
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, China
| |
Collapse
|
65
|
Hristova SH, Zhivkov AM. Montmorillonite colloid plates with adsorbed cytochrome c: in vitro cytotoxic effect on colon cancer cell culture. Cancer Nanotechnol 2021. [DOI: 10.1186/s12645-021-00095-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
The apoptosis (a cascade of biochemical reactions leading to suicide of damaged biological cells) is blocked in the cancer cells because of impossibility of cytochrome c (cytC) go out from the mitochondria. However, the apoptosis can be started by introducing of exogenous cytC into cytoplasm using colloid particles as a protein carrier due to ability of the cancer cells to phagocytize extracellular particles with submicron size.
Results
The clay mineral montmorillonite (MM) were used to prepare aqueous suspension of protein/mineral composite particles by electrostatic adsorption of the positively charged cytC globules on the negatively charged MM colloid plates, and then added to colon cancel culture. The results shows out that separately cytC and MM have no effect but the composite cytC-MM particles kill 95% of the cancer cells after 96 h treatment using equine cytC which is 97% structurally identical with the human cytC. To reach this high cytotoxicity we have formulated requirements to: (a) bare colloid particles (electric charge, form and size), (b) conditions for protein adsorption (concentrations, pH, ionic strength), and (c) suspension with the composite particles (positive total charge and optimal concentration). Due to satisfying these requirements we have reached cytotoxicity which is 1/3 higher than the reached by other authors using different artificial particles. The cytotoxicity rapidly increases with concentration of the cytC-MM particles but further it shows tendency to saturation.
Methods
The optimal pH 6.5 and the 10:3 mg/mg cytC/MM concentration ratio at adsorption were found out by employing computer (protein electrostatics) and physicochemical methods (microelectrophoresis and colloid electrooptics) to prepare cytC-MM suspension. The anticancer capability of cytC-MM nanoplates were investigated using cell culture of metastasizing colon cancer.
Conclusion
The in vitro experiments with colon cancer cell culture disclose that cytC-MM composite particles have potential for application in anticancer therapy of superficial neoplasms of the skin and the alimentary system (mouth cavity, esophagus, stomach, jejunum and colon).
Graphic abstract
Collapse
|
66
|
Dorrigiv D, Simeone K, Communal L, Kendall-Dupont J, St-Georges-Robillard A, Péant B, Carmona E, Mes-Masson AM, Gervais T. Microdissected Tissue vs Tissue Slices-A Comparative Study of Tumor Explant Models Cultured On-Chip and Off-Chip. Cancers (Basel) 2021; 13:4208. [PMID: 34439362 PMCID: PMC8394960 DOI: 10.3390/cancers13164208] [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: 07/05/2021] [Revised: 08/17/2021] [Accepted: 08/20/2021] [Indexed: 12/29/2022] Open
Abstract
Predicting patient responses to anticancer drugs is a major challenge both at the drug development stage and during cancer treatment. Tumor explant culture platforms (TECPs) preserve the native tissue architecture and are well-suited for drug response assays. However, tissue longevity in these models is relatively low. Several methodologies have been developed to address this issue, although no study has compared their efficacy in a controlled fashion. We investigated the effect of two variables in TECPs, specifically, the tissue size and culture vessel on tissue survival using micro-dissected tumor tissue (MDT) and tissue slices which were cultured in microfluidic chips and plastic well plates. Tumor models were produced from ovarian and prostate cancer cell line xenografts and were matched in terms of the specimen, total volume of tissue, and respective volume of medium in each culture system. We examined morphology, viability, and hypoxia in the various tumor models. Our observations suggest that the viability and proliferative capacity of MDTs were not affected during the time course of the experiments. In contrast, tissue slices had reduced proliferation and showed increased cell death and hypoxia under both culture conditions. Tissue slices cultured in microfluidic devices had a lower degree of hypoxia compared to those in 96-well plates. Globally, our results show that tissue slices have lower survival rates compared to MDTs due to inherent diffusion limitations, and that microfluidic devices may decrease hypoxia in tumor models.
Collapse
Affiliation(s)
- Dina Dorrigiv
- Centre de Recherche du Centre hospitalier de l’Université de Montréal, (CRCHUM) and Institut du Cancer de Montréal, Montreal, QC H2X 0A9, Canada; (D.D.); (K.S.); (L.C.); (J.K.-D.); (A.S.-G.-R.); (B.P.); (E.C.)
- Institute of Biomedical Engineering Polytechnique Montréal, Montreal, QC H3T 1J4, Canada
| | - Kayla Simeone
- Centre de Recherche du Centre hospitalier de l’Université de Montréal, (CRCHUM) and Institut du Cancer de Montréal, Montreal, QC H2X 0A9, Canada; (D.D.); (K.S.); (L.C.); (J.K.-D.); (A.S.-G.-R.); (B.P.); (E.C.)
- Department of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Laudine Communal
- Centre de Recherche du Centre hospitalier de l’Université de Montréal, (CRCHUM) and Institut du Cancer de Montréal, Montreal, QC H2X 0A9, Canada; (D.D.); (K.S.); (L.C.); (J.K.-D.); (A.S.-G.-R.); (B.P.); (E.C.)
| | - Jennifer Kendall-Dupont
- Centre de Recherche du Centre hospitalier de l’Université de Montréal, (CRCHUM) and Institut du Cancer de Montréal, Montreal, QC H2X 0A9, Canada; (D.D.); (K.S.); (L.C.); (J.K.-D.); (A.S.-G.-R.); (B.P.); (E.C.)
| | - Amélie St-Georges-Robillard
- Centre de Recherche du Centre hospitalier de l’Université de Montréal, (CRCHUM) and Institut du Cancer de Montréal, Montreal, QC H2X 0A9, Canada; (D.D.); (K.S.); (L.C.); (J.K.-D.); (A.S.-G.-R.); (B.P.); (E.C.)
- Department of Engineering Physics, Polytechnique Montréal, Montreal, QC H3T 1J4, Canada
| | - Benjamin Péant
- Centre de Recherche du Centre hospitalier de l’Université de Montréal, (CRCHUM) and Institut du Cancer de Montréal, Montreal, QC H2X 0A9, Canada; (D.D.); (K.S.); (L.C.); (J.K.-D.); (A.S.-G.-R.); (B.P.); (E.C.)
| | - Euridice Carmona
- Centre de Recherche du Centre hospitalier de l’Université de Montréal, (CRCHUM) and Institut du Cancer de Montréal, Montreal, QC H2X 0A9, Canada; (D.D.); (K.S.); (L.C.); (J.K.-D.); (A.S.-G.-R.); (B.P.); (E.C.)
| | - Anne-Marie Mes-Masson
- Centre de Recherche du Centre hospitalier de l’Université de Montréal, (CRCHUM) and Institut du Cancer de Montréal, Montreal, QC H2X 0A9, Canada; (D.D.); (K.S.); (L.C.); (J.K.-D.); (A.S.-G.-R.); (B.P.); (E.C.)
- Department of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Thomas Gervais
- Centre de Recherche du Centre hospitalier de l’Université de Montréal, (CRCHUM) and Institut du Cancer de Montréal, Montreal, QC H2X 0A9, Canada; (D.D.); (K.S.); (L.C.); (J.K.-D.); (A.S.-G.-R.); (B.P.); (E.C.)
- Institute of Biomedical Engineering Polytechnique Montréal, Montreal, QC H3T 1J4, Canada
- Department of Engineering Physics, Polytechnique Montréal, Montreal, QC H3T 1J4, Canada
| |
Collapse
|
67
|
Hu J, Guan Z, Chen J. Multifunctional biomaterials that modulate oxygen levels in the tumor microenvironment. Cancer Lett 2021; 521:39-49. [PMID: 34419500 DOI: 10.1016/j.canlet.2021.08.019] [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: 07/03/2021] [Revised: 08/11/2021] [Accepted: 08/17/2021] [Indexed: 12/27/2022]
Abstract
A characteristic feature of solid tumors is their low oxygen tension, which confers resistance to radiotherapy, photodynamic therapy, and chemotherapy. Therefore, to improve treatment outcomes, it is critical to develop biomaterials capable of targeted modulation of oxygen levels in tumors. In this review, we summarize four types of oxygen-modulating biomaterials, namely, oxygen-carrying biomaterials to deliver oxygen into tumors (e.g., perfluorocarbon and hemoglobin), oxygen-generating biomaterials to promote in situ oxygen generation (e.g., MnO2, catalase, and CuO), oxygen-consuming biomaterials to starve tumors (e.g., photosensitizer, glucose oxidase, and magnesium silicide), and oxygen-circulating biomaterials capable of both providing and consuming oxygen (e.g., ENBS-B). The current literature suggests that these biomaterials are useful for anticancer therapeutics. We present the key molecular mechanisms involved in modulating oxygen levels and the potential applications of these biomaterials in the context of hypoxic tumor treatment.
Collapse
Affiliation(s)
- Jinghui Hu
- School of Rehabilitation, Institute of Rehabilitation Engineering, Binzhou Medical University, Yantai, 264003, PR China
| | - Zhenxin Guan
- School of Pharmacy, The Key Laboratory of Prescription Effect and Clinical Evaluation of State Administration of Traditional Chinese Medicine of China, Binzhou Medical University, Yantai, 264003, PR China
| | - Jing Chen
- School of Pharmacy, The Key Laboratory of Prescription Effect and Clinical Evaluation of State Administration of Traditional Chinese Medicine of China, Binzhou Medical University, Yantai, 264003, PR China.
| |
Collapse
|
68
|
Alghamdi M, Gumbleton M, Newland B. Local delivery to malignant brain tumors: potential biomaterial-based therapeutic/adjuvant strategies. Biomater Sci 2021; 9:6037-6051. [PMID: 34357362 DOI: 10.1039/d1bm00896j] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Glioblastoma (GBM) is the most aggressive malignant brain tumor and is associated with a very poor prognosis. The standard treatment for newly diagnosed patients involves total tumor surgical resection (if possible), plus irradiation and adjuvant chemotherapy. Despite treatment, the prognosis is still poor, and the tumor often recurs within two centimeters of the original tumor. A promising approach to improving the efficacy of GBM therapeutics is to utilize biomaterials to deliver them locally at the tumor site. Local delivery to GBM offers several advantages over systemic administration, such as bypassing the blood-brain barrier and increasing the bioavailability of the therapeutic at the tumor site without causing systemic toxicity. Local delivery may also combat tumor recurrence by maintaining sufficient drug concentrations at and surrounding the original tumor area. Herein, we critically appraised the literature on local delivery systems based within the following categories: polymer-based implantable devices, polymeric injectable systems, and hydrogel drug delivery systems. We also discussed the negative effect of hypoxia on treatment strategies and how one might utilize local implantation of oxygen-generating biomaterials as an adjuvant to enhance current therapeutic strategies.
Collapse
Affiliation(s)
- Majed Alghamdi
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff, CF10 3NB, UK. and Faculty of Pharmacy, King Abdulaziz University, Jeddah, 22522, Kingdom of Saudi Arabia
| | - Mark Gumbleton
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff, CF10 3NB, UK.
| | - Ben Newland
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff, CF10 3NB, UK. and Leibniz-Institut für Polymerforschung Dresden, Max Bergmann Center of Biomaterials Dresden, Hohe Straße 6, D-01069 Dresden, Germany
| |
Collapse
|
69
|
Differential Angiogenic Potential of 3-Dimension Spheroid of HNSCC Cells in Mouse Xenograft. Int J Mol Sci 2021; 22:ijms22158245. [PMID: 34361027 PMCID: PMC8348975 DOI: 10.3390/ijms22158245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 07/24/2021] [Accepted: 07/28/2021] [Indexed: 11/16/2022] Open
Abstract
The experimental animal model is still essential in the development of new anticancer drugs. We characterized mouse tumors derived from two-dimensional (2D) monolayer cells or three-dimensional (3D) spheroids to establish an in vivo model with highly standardized conditions. Primary cancer-associated fibroblasts (CAFs) were cultured from head and neck squamous cell carcinoma (HNSCC) tumor tissues and co-injected with monolayer cancer cells or spheroids into the oral mucosa of mice. Mice tumor blood vessels were stained, followed by tissue clearing and 3D Lightsheet fluorescent imaging. We compared the effect of exosomes secreted from 2D or 3D culture conditions on the angiogenesis-related genes in HNSCC cells. Our results showed that both the cells and spheroids co-injected with primary CAFs formed tumors. Interestingly, vasculature was abundantly distributed inside the spheroid-derived but not the monolayer-derived mice tumors. In addition, cisplatin injection more significantly decreased spheroid-derived but not monolayer-derived tumor size in mice. Additionally, exosomes isolated from co-culture media of FaDu spheroid and CAF upregulated angiogenesis-related genes in HNSCC cells as compared to exosomes from FaDu cell and CAF co-culture media under in vitro conditions. The mouse tumor xenograft model derived from 3D spheroids of HNSCC cells with primary CAFs is expected to produce reliable chemotherapy drug screening results given the robust angiogenesis and lack of necrosis inside tumor tissues.
Collapse
|
70
|
Debacker JM, Gondry O, Lahoutte T, Keyaerts M, Huvenne W. The Prognostic Value of CD206 in Solid Malignancies: A Systematic Review and Meta-Analysis. Cancers (Basel) 2021; 13:cancers13143422. [PMID: 34298638 PMCID: PMC8305473 DOI: 10.3390/cancers13143422] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/02/2021] [Accepted: 07/05/2021] [Indexed: 01/03/2023] Open
Abstract
Simple Summary The role of innate immune cells in the tumor microenvironment (TME), more specifically the presence of the tumor associated macrophages (TAMs), is becoming more important in the prognosis and treatment of patients diagnosed with malignancies. The aim of this systematic review and meta-analysis was to assess the potential prognostic value of CD206-expressing TAMs, a subclass of macrophages, which were previously proposed to negatively impact the patient’s prognosis. We identified 27 manuscripts describing the role of CD206 in patient prognosis for 14 different tumor types. Despite a large heterogeneity in the results, we identified a significantly worse overall and disease-free survival for patients with increased CD206-expressing TAMs in the TME. The use of CD206-expressing TAMs could therefore be used as a prognostic marker in patients diagnosed with solid malignancies. Abstract An increased presence of CD206-expressing tumor associated macrophages in solid cancers was proposed to be associated with worse outcomes in multiple types of malignancies, but contradictory results are published. We performed a reproducible systematic review and meta-analysis to provide increased evidence to confirm or reject this hypothesis following the Preferred Reporting Items for Systematic Reviews and Meta-analyses statement. The Embase, Web of Science, and MEDLINE-databases were systematically searched for eligible manuscripts. A total of 27 papers studying the prognostic impact of CD206 in 14 different tumor types were identified. Meta-analyses showed a significant impact on the overall survival (OS) and disease-free survival (DFS). While no significant differences were revealed in progression-free survival (PFS) and disease-specific survival (DSS), a shift towards negative survival was correlated with increased CD206-expresion. As a result of the different tumor types, large heterogeneity was present between the different tumor types. Subgroup analysis of hepatocellular carcinoma and gastric cancers revealed no heterogeneity, associated with a significant negative impact on OS in both groups. The current systematic review displays the increased presence CD206-expressing macrophages as a significant negative prognostic biomarker for both OS and DFS in patients diagnosed with solid cancers. Because a heterogenous group of tumor types was included in the meta-analysis, the results cannot be generalized. These results can, however, be used to further lead follow-up research to validate the specific prognostic value of CD206 in individual tumor types and therapeutic approaches.
Collapse
Affiliation(s)
- Jens M. Debacker
- Department of Head and Skin, Ghent University, 9000 Ghent, Belgium;
- Department of Head and Neck Surgery, Ghent University Hospital, 9000 Ghent, Belgium
- Department of Nuclear Medicine, University Hospital Brussels, 1090 Brussels, Belgium; (O.G.); (T.L.); (M.K.)
- Correspondence: ; Tel.: +32-9-332-39-90
| | - Odrade Gondry
- Department of Nuclear Medicine, University Hospital Brussels, 1090 Brussels, Belgium; (O.G.); (T.L.); (M.K.)
- In Vivo Cellular and Molecular Imaging, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Tony Lahoutte
- Department of Nuclear Medicine, University Hospital Brussels, 1090 Brussels, Belgium; (O.G.); (T.L.); (M.K.)
- In Vivo Cellular and Molecular Imaging, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Marleen Keyaerts
- Department of Nuclear Medicine, University Hospital Brussels, 1090 Brussels, Belgium; (O.G.); (T.L.); (M.K.)
- In Vivo Cellular and Molecular Imaging, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Wouter Huvenne
- Department of Head and Skin, Ghent University, 9000 Ghent, Belgium;
- Department of Head and Neck Surgery, Ghent University Hospital, 9000 Ghent, Belgium
| |
Collapse
|
71
|
Presley KF, Fan F, DiRando NM, Shahhosseini M, Rao JZ, Tedeschi A, Castro CE, Lannutti JJ. Injectable, dispersible polysulfone-polysulfone core-shell particles for optical oxygen sensing. J Appl Polym Sci 2021; 138:50603. [PMID: 36091476 PMCID: PMC9455784 DOI: 10.1002/app.50603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 01/31/2021] [Indexed: 11/12/2022]
Abstract
Injectable sensors can significantly improve the volume of critical biomedical information emerging from the human body in response to injury or disease. Optical oxygen sensors with rapid response times can be achieved by incorporating oxygen-sensitive luminescent molecules within polymeric matrices with suitably high surface area to volume ratios. In this work, electrospraying utilizes these advances to produce conveniently injectable, oxygen sensing particles made up of a core-shell polysulfone-polysulfone structure containing a phosphorescent oxygen-sensitive palladium porphyrin species within the core. Particle morphology is highly dependent on solvent identity and electrospraying parameters; DMF offers the best potential for the creation of uniform, sub-micron particles. Total internal reflection fluorescence (TIRF) microscopy confirms the existence of both core-shell structure and oxygen sensitivity. The dissolved oxygen response time is rapid (<0.30 s), ideal for continuous real-time monitoring of oxygen concentration. The incorporation of Pluronic F-127 surfactant enables efficient dispersion; selection of an appropriate electrospraying solvent (DMF) yields particles readily injected even through a <100 μm diameter needle.
Collapse
Affiliation(s)
- Kayla F Presley
- Department of Materials Science and Engineering, The Ohio State University, 116 W 19th Avenue, Columbus, OH 43210, USA
| | - Fan Fan
- Department of Materials Science and Engineering, The Ohio State University, 116 W 19th Avenue, Columbus, OH 43210, USA; Center for Chronic Brain Injury Program, The Ohio State University, Columbus, OH 43210, USA
| | - Nicole M DiRando
- Department of Materials Science and Engineering, The Ohio State University, 116 W 19th Avenue, Columbus, OH 43210, USA
| | - Melika Shahhosseini
- Department of Mechanical and Aerospace Engineering, The Ohio State University, 201 W 19th Avenue, Columbus, OH 43210, USA
| | - Jim Z Rao
- Department of Materials Science and Engineering, The Ohio State University, 116 W 19th Avenue, Columbus, OH 43210, USA
| | - Andrea Tedeschi
- Center for Chronic Brain Injury Program, The Ohio State University, Columbus, OH 43210, USA; Department of Neuroscience, Wexner Medical Center, The Ohio State University, 460W 12th Avenue, Columbus, OH 43210, USA
| | - Carlos E Castro
- Department of Mechanical and Aerospace Engineering, The Ohio State University, 201 W 19th Avenue, Columbus, OH 43210, USA
| | - John J Lannutti
- Department of Materials Science and Engineering, The Ohio State University, 116 W 19th Avenue, Columbus, OH 43210, USA; Center for Chronic Brain Injury Program, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
72
|
Lequeux A, Noman MZ, Xiao M, Van Moer K, Hasmim M, Benoit A, Bosseler M, Viry E, Arakelian T, Berchem G, Chouaib S, Janji B. Targeting HIF-1 alpha transcriptional activity drives cytotoxic immune effector cells into melanoma and improves combination immunotherapy. Oncogene 2021; 40:4725-4735. [PMID: 34155342 PMCID: PMC8282500 DOI: 10.1038/s41388-021-01846-x] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 04/27/2021] [Accepted: 05/17/2021] [Indexed: 02/08/2023]
Abstract
Hypoxia is a key factor responsible for the failure of therapeutic response in most solid tumors and promotes the acquisition of tumor resistance to various antitumor immune effectors. Reshaping the hypoxic immune suppressive tumor microenvironment to improve cancer immunotherapy is still a relevant challenge. We investigated the impact of inhibiting HIF-1α transcriptional activity on cytotoxic immune cell infiltration into B16-F10 melanoma. We showed that tumors expressing a deleted form of HIF-1α displayed increased levels of NK and CD8+ effector T cells in the tumor microenvironment, which was associated with high levels of CCL2 and CCL5 chemokines. We showed that combining acriflavine, reported as a pharmacological agent preventing HIF-1α/HIF-1β dimerization, dramatically improved the benefit of cancer immunotherapy based on TRP-2 peptide vaccination and anti-PD-1 blocking antibody. In melanoma patients, we revealed that tumors exhibiting high CCL5 are less hypoxic, and displayed high NK, CD3+, CD4+ and CD8+ T cell markers than those having low CCL5. In addition, melanoma patients with high CCL5 in their tumors survive better than those having low CCL5. This study provides the pre-clinical proof of concept for a novel triple combination strategy including blocking HIF-1α transcription activity along vaccination and PD-1 blocking immunotherapy.
Collapse
Affiliation(s)
- Audrey Lequeux
- Tumor Immunotherapy and Microenvironment (TIME) group, Department of Oncology, Luxembourg Institute of Health (LIH), Luxembourg City, Luxembourg
| | - Muhammad Zaeem Noman
- Tumor Immunotherapy and Microenvironment (TIME) group, Department of Oncology, Luxembourg Institute of Health (LIH), Luxembourg City, Luxembourg
| | - Malina Xiao
- Tumor Immunotherapy and Microenvironment (TIME) group, Department of Oncology, Luxembourg Institute of Health (LIH), Luxembourg City, Luxembourg
| | - Kris Van Moer
- Tumor Immunotherapy and Microenvironment (TIME) group, Department of Oncology, Luxembourg Institute of Health (LIH), Luxembourg City, Luxembourg
| | - Meriem Hasmim
- Tumor Immunotherapy and Microenvironment (TIME) group, Department of Oncology, Luxembourg Institute of Health (LIH), Luxembourg City, Luxembourg
| | - Alice Benoit
- Tumor Immunotherapy and Microenvironment (TIME) group, Department of Oncology, Luxembourg Institute of Health (LIH), Luxembourg City, Luxembourg
| | - Manon Bosseler
- Tumor Immunotherapy and Microenvironment (TIME) group, Department of Oncology, Luxembourg Institute of Health (LIH), Luxembourg City, Luxembourg
| | - Elodie Viry
- Tumor Immunotherapy and Microenvironment (TIME) group, Department of Oncology, Luxembourg Institute of Health (LIH), Luxembourg City, Luxembourg
| | - Tsolere Arakelian
- Tumor Immunotherapy and Microenvironment (TIME) group, Department of Oncology, Luxembourg Institute of Health (LIH), Luxembourg City, Luxembourg
| | - Guy Berchem
- Tumor Immunotherapy and Microenvironment (TIME) group, Department of Oncology, Luxembourg Institute of Health (LIH), Luxembourg City, Luxembourg
- Department of Hemato-oncology, Centre Hospitalier du Luxembourg, Luxembourg City, Luxembourg
| | - Salem Chouaib
- INSERM UMR 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy, Villejuif, France
- Thumbay Research Institute of Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | - Bassam Janji
- Tumor Immunotherapy and Microenvironment (TIME) group, Department of Oncology, Luxembourg Institute of Health (LIH), Luxembourg City, Luxembourg.
| |
Collapse
|
73
|
Liapis E, Karlas A, Klemm U, Ntziachristos V. Chemotherapeutic effects on breast tumor hemodynamics revealed by eigenspectra multispectral optoacoustic tomography (eMSOT). Theranostics 2021; 11:7813-7828. [PMID: 34335966 PMCID: PMC8315054 DOI: 10.7150/thno.56173] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 03/24/2021] [Indexed: 12/11/2022] Open
Abstract
Non-invasive monitoring of hemodynamic tumor responses to chemotherapy could provide unique insights into the development of therapeutic resistance and inform therapeutic decision-making in the clinic. Methods: Here, we examined the longitudinal and dynamic effects of the common chemotherapeutic drug Taxotere on breast tumor (KPL-4) blood volume and oxygen saturation using eigenspectra multispectral optoacoustic tomography (eMSOT) imaging over a period of 41 days. Tumor vascular function was assessed by dynamic oxygen-enhanced eMSOT (OE-eMSOT). The obtained in vivo optoacoustic data were thoroughly validated by ex vivo cryoimaging and immunohistochemical staining against markers of vascularity and hypoxia. Results: We provide the first preclinical evidence that prolonged treatment with Taxotere causes a significant drop in mean whole tumor oxygenation. Furthermore, application of OE-eMSOT showed a diminished vascular response in Taxotere-treated tumors and revealed the presence of static blood pools, indicating increased vascular permeability. Conclusion: Our work has important translational implications and supports the feasibility of eMSOT imaging for non-invasive assessment of tumor microenvironmental responses to chemotherapy.
Collapse
|
74
|
Owen J, Logan K, Nesbitt H, Able S, Vasilyeva A, Bluemke E, Kersemans V, Smart S, Vallis KA, McHale AP, Callan JF, Stride E. Orally administered oxygen nanobubbles enhance tumor response to sonodynamic therapy. NANO SELECT 2021. [DOI: 10.1002/nano.202100038] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Affiliation(s)
- Joshua Owen
- Institute of Biomedical Engineering University of Oxford Oxford UK
| | - Keiran Logan
- Biomedical Sciences Research Institute Ulster University Coleraine Northern Ireland UK
| | - Heather Nesbitt
- Biomedical Sciences Research Institute Ulster University Coleraine Northern Ireland UK
| | - Sarah Able
- Oxford Institute for Radiation Oncology University of Oxford Oxford UK
| | | | - Emma Bluemke
- Institute of Biomedical Engineering University of Oxford Oxford UK
| | - Veerle Kersemans
- Oxford Institute for Radiation Oncology University of Oxford Oxford UK
| | - Sean Smart
- Oxford Institute for Radiation Oncology University of Oxford Oxford UK
| | | | - Anthony P. McHale
- Biomedical Sciences Research Institute Ulster University Coleraine Northern Ireland UK
| | - John F. Callan
- Biomedical Sciences Research Institute Ulster University Coleraine Northern Ireland UK
| | - Eleanor Stride
- Institute of Biomedical Engineering University of Oxford Oxford UK
| |
Collapse
|
75
|
Li Q, Zhang J, Li J, Ye H, Li M, Hou W, Li H, Wang Z. Glutathione-Activated NO-/ROS-Generation Nanoparticles to Modulate the Tumor Hypoxic Microenvironment for Enhancing the Effect of HIFU-Combined Chemotherapy. ACS APPLIED MATERIALS & INTERFACES 2021; 13:26808-26823. [PMID: 34085524 DOI: 10.1021/acsami.1c07494] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The combination of high-intensity focused ultrasound (HIFU) and chemotherapy has promising potential in the synergistic treatment of various types of solid tumors. However, the clinical efficacy of HIFU in combination chemotherapy is often impeded by the pre-existing hypoxia tumor microenvironment-induced multidrug resistance (MDR). Therefore, it is imperative for HIFU combined with chemotherapy to overcome MDR by improving the tumor hypoxic microenvironment. Hence, we developed highly stable nanoparticles (P@BDOX/β-lapachone-NO-NPs) with intracellular nitric oxide (NO)- and reactive oxygen species (ROS)-generating capabilities at the tumor site to relieve the hypoxic tumor microenvironment in solid tumors. Doxorubicin prodrug (boronate-DOX, BDOX) and β-lapachone were concurrently loaded onto actively targeted pH (low) insertion peptides (pHLIPs)-poly(ethylene glycol) and nitrated gluconic acid copolymers. Our results showed that the ability of P@BDOX/β-lapachone-NO-NPs to generate NO and ROS simultaneously is vital for the sensitization of hypoxic solid tumors for chemotherapy, as evidenced by the suppression of tumor cells and tissues (in vitro and in the nude mice model). Thus, this combined therapy holds considerable potential in the management of hypoxic solid tumors.
Collapse
Affiliation(s)
- Qianyan Li
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Jingni Zhang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Jingnan Li
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Hemin Ye
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Meixuan Li
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Wei Hou
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Huanan Li
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Zhibiao Wang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
76
|
Uwada J, Mukai S, Terada N, Nakazawa H, Islam MS, Nagai T, Fujii M, Yamasaki K, Taniguchi T, Kamoto T, Yazawa T. Pleiotropic effects of probenecid on three-dimensional cultures of prostate cancer cells. Life Sci 2021; 278:119554. [PMID: 33932444 DOI: 10.1016/j.lfs.2021.119554] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 04/19/2021] [Accepted: 04/24/2021] [Indexed: 12/30/2022]
Abstract
AIMS Chemoresistance remains a persistent challenge in advanced prostate cancer therapy. Probenecid reportedly inhibits multiple drug-efflux transporters; hence, it can be employed as a potential sensitizer for chemotherapy. In the present study, we evaluated the effects of probenecid on three-dimensional (3D)-cultures of prostate cancer cells. MAIN METHODS Prostate cancer cell lines, 22Rv1 and PC-3 were cultured as multicellular tumor spheroids. The effects of probenecid were evaluated using the MTT assay for viability, microscopy for spheroid size, and soft agar colony formation assay for anchorage-independent growth. KEY FINDINGS The 3D-cultured 22Rv1 cells were less sensitive to cisplatin and doxorubicin than two-dimensional (2D) cell culture. Co-administration of probenecid at a low (100 or 300 μM), but not high (500 μM), concentration increased the sensitivity to cisplatin or doxorubicin in 22Rv1 spheroids. Probenecid increased the expression of ABCG2, a multidrug resistance transporter, in a dose-dependent manner. Furthermore, treatment with probenecid alone reduced the growth of 22Rv1 spheroids. Conversely, probenecid inhibited spheroid compaction rather than growth inhibition in 3D-cultured PC-3 cells. Moreover, probenecid inhibited colony formation of 22Rv1 and PC-3 cells in soft agar, as well as downregulated focal adhesion kinase (FAK), a crucial factor in anchorage-independent growth. SIGNIFICANCE In 3D-cultured prostate cancer cells, probenecid demonstrated pleiotropic effects such as chemosensitization, growth suppression, inhibition of spheroid compaction, and suppression of anchorage-independent growth. Elucidating the detailed mechanism underlying these probenecid actions could result in the identification of novel therapeutic targets toward the advanced prostate cancer.
Collapse
Affiliation(s)
- Junsuke Uwada
- Department of Biochemistry, Asahikawa Medical University, Asahikawa 078-8510, Japan.
| | - Shoichiro Mukai
- Department of Urology, Faculty of Medicine, University of Miyazaki, Kiyotake, Miyazaki 889-1692, Japan
| | - Naoki Terada
- Department of Urology, Faculty of Medicine, University of Miyazaki, Kiyotake, Miyazaki 889-1692, Japan
| | - Hitomi Nakazawa
- Department of Functional Anatomy and Neuroscience, Asahikawa Medical University, Asahikawa 078-8510, Japan
| | | | - Takahiro Nagai
- Department of Urology, Faculty of Medicine, University of Miyazaki, Kiyotake, Miyazaki 889-1692, Japan
| | - Masato Fujii
- Department of Urology, Faculty of Medicine, University of Miyazaki, Kiyotake, Miyazaki 889-1692, Japan
| | - Koji Yamasaki
- Department of Urology, Faculty of Medicine, University of Miyazaki, Kiyotake, Miyazaki 889-1692, Japan
| | | | - Toshiyuki Kamoto
- Department of Urology, Faculty of Medicine, University of Miyazaki, Kiyotake, Miyazaki 889-1692, Japan
| | - Takashi Yazawa
- Department of Biochemistry, Asahikawa Medical University, Asahikawa 078-8510, Japan
| |
Collapse
|
77
|
Ding J, He X, Cheng X, Cao G, Chen B, Chen S, Xiong M. A 4-gene-based hypoxia signature is associated with tumor immune microenvironment and predicts the prognosis of pancreatic cancer patients. World J Surg Oncol 2021; 19:123. [PMID: 33865399 PMCID: PMC8053300 DOI: 10.1186/s12957-021-02204-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 03/19/2021] [Indexed: 12/17/2022] Open
Abstract
Background Pancreatic cancer (PAC) is one of the most devastating cancer types with an extremely poor prognosis, characterized by a hypoxic microenvironment and resistance to most therapeutic drugs. Hypoxia has been found to be one of the factors contributing to chemoresistance in PAC, but also a major driver of the formation of the tumor immunosuppressive microenvironment. However, the method to identify the degree of hypoxia in the tumor microenvironment (TME) is incompletely understood. Methods The mRNA expression profiles and corresponding clinicopathological information of PAC patients were downloaded from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) database, respectively. To further explore the effect of hypoxia on the prognosis of patients with PAC as well as the tumor immune microenvironment, we established a hypoxia risk model and divided it into high- and low-risk groups in line with the hypoxia risk score. Results We established a hypoxia risk model according to four hypoxia-related genes, which could be used to demonstrate the immune microenvironment in PAC and predict prognosis. Moreover, the hypoxia risk score can act as an independent prognostic factor in PAC, and a higher hypoxia risk score was correlated with poorer prognosis in patients as well as the immunosuppressive microenvironment of the tumor. Conclusions In summary, we established and validated a hypoxia risk model that can be considered as an independent prognostic indicator and reflected the immune microenvironment of PAC, suggesting the feasibility of hypoxia-targeted therapy for PAC patients.
Collapse
Affiliation(s)
- Jianfeng Ding
- Department of General Surgery, Chaohu Hospital of Anhui Medical University, Chaohu, 238000, Anhui, China
| | - Xiaobo He
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Xiao Cheng
- Department of Pathology, School of Basic Medicine, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Guodong Cao
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Bo Chen
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Sihan Chen
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China.
| | - Maoming Xiong
- Department of General Surgery, Chaohu Hospital of Anhui Medical University, Chaohu, 238000, Anhui, China. .,Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China.
| |
Collapse
|
78
|
Hettie KS, Klockow JL, Moon EJ, Giaccia AJ, Chin FT. A NIR fluorescent smart probe for imaging tumor hypoxia. Cancer Rep (Hoboken) 2021; 4:e1384. [PMID: 33811473 PMCID: PMC8551997 DOI: 10.1002/cnr2.1384] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/09/2021] [Accepted: 03/11/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Tumor hypoxia is a characteristic of paramount importance due to low oxygenation levels in tissue negatively correlating with resistance to traditional therapies. The ability to noninvasively identify such could provide for personalized treatment(s) and enhance survival rates. Accordingly, we recently developed an NIR fluorescent hypoxia-sensitive smart probe (NO2 -Rosol) for identifying hypoxia via selectively imaging nitroreductase (NTR) activity, which could correlate to oxygen deprivation levels in cells, thereby serving as a proxy. We demonstrated proof of concept by subjecting a glioblastoma (GBM) cell line to extreme stress by evaluating such under radiobiological hypoxic (pO2 ≤ ~0.5%) conditions, which is a far cry from representative levels for hypoxia for brain glioma (pO2 = ~1.7%) which fluctuate little from physiological hypoxic (pO2 = 1.0-3.0%) conditions. AIM We aimed to evaluate the robustness, suitability, and feasibility of NO2 -Rosol for imaging hypoxia in vitro and in vivo via assessing NTR activity in diverse GBM models under relevant oxygenation levels (pO2 = 2.0%) within physiological hypoxic conditions that mimic oxygenation levels in GBM tumor tissue in the brain. METHODS We evaluated multiple GBM cell lines to determine their relative sensitivity to oxygenation levels via measuring carbonic anhydrase IX (CAIX) levels, which is a surrogate marker for indirectly identifying hypoxia by reporting on oxygen deprivation levels and upregulated NTR activity. We evaluated for hypoxia via measuring NTR activity when employing NO2 -Rosol in in vitro and tumor hypoxia imaging studies in vivo. RESULTS The GBM39 cell line demonstrated the highest CAIX expression under hypoxic conditions representing that of GBM in the brain. NO2 -Rosol displayed an 8-fold fluorescence enhancement when evaluated in GBM39 cells (pO2 = 2.0%), thereby establishing its robustness and suitability for imaging hypoxia under relevant physiological conditions. We demonstrated the feasibility of NO2 -Rosol to afford tumor hypoxia imaging in vivo via it demonstrating a tumor-to-background of 5 upon (i) diffusion throughout, (ii) bioreductive activation by NTR activity in, and (iii) retention within, GBM39 tumor tissue. CONCLUSION We established the robustness, suitability, and feasibility of NO2 -Rosol for imaging hypoxia under relevant oxygenation levels in vitro and in vivo via assessing NTR activity in GBM39 models.
Collapse
Affiliation(s)
- Kenneth S Hettie
- Department of Radiology, Stanford University School of Medicine, Stanford, California, USA.,Department of Otolaryngology - Head & Neck Surgery, Stanford University, Stanford, California, USA
| | - Jessica L Klockow
- Department of Radiology, Stanford University School of Medicine, Stanford, California, USA
| | - Eui Jung Moon
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California, USA
| | - Amato J Giaccia
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California, USA
| | - Frederick T Chin
- Department of Radiology, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
79
|
Li X, Guo X, Ling J, Tang Z, Huang G, He L, Chen T. Nanomedicine-based cancer immunotherapies developed by reprogramming tumor-associated macrophages. NANOSCALE 2021; 13:4705-4727. [PMID: 33625411 DOI: 10.1039/d0nr08050k] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Tumor microenvironment is a complex ecosystem composed of tumor extracellular matrix, fibroblasts, blood vessels, and immune cells, promoting tumor development by secreting various growth factors, hydrolase, and inflammatory factors. Tumor-associated macrophages (TAMs) constitute the largest number of immune cells in the TME, and they have a "double-edged sword" effect on tumor growth, invasion, metastasis, angiogenesis, and immunosuppression. Under the regulation of different cytokines in the TME, the bidirectional TAMs can switch their phenotypes between tumoricidal M1-like and pro-tumorigenic M2-like macrophages. TAM polarization suggests that scientists can use this property to design drugs targeting this regulation as a promising immunotherapy strategy to enhance tumor therapy efficiency. In this review, we summarize a brief introduction of TAMs and their implications for tumorigenesis. Next, we review recent advances in designing various functionalized nanomedicines and their applications in nanomedicine-based cancer therapies that target TAMs by killing them, inhibiting macrophage recruitment, and repolarizing them from pro-tumorigenic M2-like to tumoricidal M1-like macrophages. Simultaneously, the regulation of nanomedicines on the signaling pathways accounting for these effects is also summarized. This review will not only provide background scientific information for the understanding of TAMs and their roles in cancer treatment but also help scientists design nanomedicines based on tumor TAMs, which can help achieve better clinical treatment outcomes for tumors.
Collapse
Affiliation(s)
- Xiaoying Li
- Department of Neurology and Stroke Center, The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou 510632, China.
| | - Xiaoming Guo
- Department of Neurology and Stroke Center, The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou 510632, China.
| | - Jiabao Ling
- Department of Neurology and Stroke Center, The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou 510632, China.
| | - Zheng Tang
- Department of Neurology and Stroke Center, The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou 510632, China.
| | - Guanning Huang
- Department of Neurology and Stroke Center, The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou 510632, China.
| | - Lizhen He
- Department of Neurology and Stroke Center, The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou 510632, China.
| | - Tianfeng Chen
- Department of Neurology and Stroke Center, The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
80
|
El Sayed R, Haibe Y, Amhaz G, Bouferraa Y, Shamseddine A. Metabolic Factors Affecting Tumor Immunogenicity: What Is Happening at the Cellular Level? Int J Mol Sci 2021; 22:2142. [PMID: 33670011 PMCID: PMC7927105 DOI: 10.3390/ijms22042142] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 02/07/2021] [Accepted: 02/11/2021] [Indexed: 12/11/2022] Open
Abstract
Immunotherapy has changed the treatment paradigm in multiple solid and hematologic malignancies. However, response remains limited in a significant number of cases, with tumors developing innate or acquired resistance to checkpoint inhibition. Certain "hot" or "immune-sensitive" tumors become "cold" or "immune-resistant", with resultant tumor growth and disease progression. Multiple factors are at play both at the cellular and host levels. The tumor microenvironment (TME) contributes the most to immune-resistance, with nutrient deficiency, hypoxia, acidity and different secreted inflammatory markers, all contributing to modulation of immune-metabolism and reprogramming of immune cells towards pro- or anti-inflammatory phenotypes. Both the tumor and surrounding immune cells require high amounts of glucose, amino acids and fatty acids to fulfill their energy demands. Thus, both compete over one pool of nutrients that falls short on needs, obliging cells to resort to alternative adaptive metabolic mechanisms that take part in shaping their inflammatory phenotypes. Aerobic or anaerobic glycolysis, oxidative phosphorylation, tryptophan catabolism, glutaminolysis, fatty acid synthesis or fatty acid oxidation, etc. are all mechanisms that contribute to immune modulation. Different pathways are triggered leading to genetic and epigenetic modulation with consequent reprogramming of immune cells such as T-cells (effector, memory or regulatory), tumor-associated macrophages (TAMs) (M1 or M2), natural killers (NK) cells (active or senescent), and dendritic cells (DC) (effector or tolerogenic), etc. Even host factors such as inflammatory conditions, obesity, caloric deficit, gender, infections, microbiota and smoking status, may be as well contributory to immune modulation, anti-tumor immunity and response to immune checkpoint inhibition. Given the complex and delicate metabolic networks within the tumor microenvironment controlling immune response, targeting key metabolic modulators may represent a valid therapeutic option to be combined with checkpoint inhibitors in an attempt to regain immune function.
Collapse
Affiliation(s)
- Rola El Sayed
- Global Health Institute, American University of Beirut, Beirut 11-0236, Lebanon;
| | - Yolla Haibe
- Division of Hematology/Oncology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut 11-0236, Lebanon; (Y.H.); (G.A.); (Y.B.)
| | - Ghid Amhaz
- Division of Hematology/Oncology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut 11-0236, Lebanon; (Y.H.); (G.A.); (Y.B.)
| | - Youssef Bouferraa
- Division of Hematology/Oncology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut 11-0236, Lebanon; (Y.H.); (G.A.); (Y.B.)
| | - Ali Shamseddine
- Division of Hematology/Oncology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut 11-0236, Lebanon; (Y.H.); (G.A.); (Y.B.)
| |
Collapse
|
81
|
Leal LG, Lopes MA, Peres SB, Batista ML. Exercise Training as Therapeutic Approach in Cancer Cachexia: A Review of Potential Anti-inflammatory Effect on Muscle Wasting. Front Physiol 2021; 11:570170. [PMID: 33613297 PMCID: PMC7890241 DOI: 10.3389/fphys.2020.570170] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 12/14/2020] [Indexed: 12/12/2022] Open
Abstract
Cachexia is a multifactorial inflammatory syndrome with high prevalence in cancer patients. It is characterized by a metabolic chaos culminating in drastic reduction in body weight, mainly due to skeletal muscle and fat depletion. Currently, there is not a standard intervention for cachexia, but it is believed that a dynamic approach should be applied early in the course of the disease to maintain or slow the loss of physical function. The present review sought to explain the different clinical and experimental applications of different models of exercise and their contribution to a better prognosis of the disease. Here the advances in knowledge about the application of physical training in experimental models are elucidated, tests that contribute substantially to elucidate the cellular and biochemical mechanisms of exercise in different ways, as well as clinical trials that present not only the impacts of exercise in front cachexia but also the challenges of its application in clinical practice.
Collapse
Affiliation(s)
- Luana G Leal
- Integrated Group of Biotechnology, Laboratory of Adipose Tissue Biology, University of Mogi das Cruzes, Mogi das Cruzes, Brazil.,Technological Research Group, University of Mogi das Cruzes, Mogi das Cruzes, Brazil
| | - Magno A Lopes
- Laboratory of Metabolism of Bioactive Lipids, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - Sidney B Peres
- Department of Physiological Sciences, State University of Maringá, Maringá, Brazil
| | - Miguel L Batista
- Integrated Group of Biotechnology, Laboratory of Adipose Tissue Biology, University of Mogi das Cruzes, Mogi das Cruzes, Brazil.,Technological Research Group, University of Mogi das Cruzes, Mogi das Cruzes, Brazil
| |
Collapse
|
82
|
The Oxygen-Generating Calcium Peroxide-Modified Magnetic Nanoparticles Attenuate Hypoxia-Induced Chemoresistance in Triple-Negative Breast Cancer. Cancers (Basel) 2021; 13:cancers13040606. [PMID: 33546453 PMCID: PMC7913619 DOI: 10.3390/cancers13040606] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/26/2021] [Accepted: 01/27/2021] [Indexed: 12/22/2022] Open
Abstract
Cancer response to chemotherapy is regulated not only by intrinsic sensitivity of cancer cells but also by tumor microenvironment. Tumor hypoxia, a condition of low oxygen level in solid tumors, is known to increase the resistance of cancer cells to chemotherapy. Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer. Due to lack of target in TNBC, chemotherapy is the only approved systemic treatment. We evaluated the effect of hypoxia on chemotherapy resistance in TNBC in a series of in vitro and in vivo experiments. Furthermore, we synthesized the calcium peroxide-modified magnetic nanoparticles (CaO2-MNPs) with the function of oxygen generation to improve and enhance the therapeutic efficiency of doxorubicin treatment in the hypoxia microenvironment of TNBC. The results of gene set enrichment analysis (GSEA) software showed that the hypoxia and autophagy gene sets are significantly enriched in TNBC patients. We found that the chemical hypoxia stabilized the expression of hypoxia-inducible factor 1α (HIF-1α) protein and increased doxorubicin resistance in TNBC cells. Moreover, hypoxia inhibited the induction of apoptosis and autophagy by doxorubicin. In addition, CaO2-MNPs promoted ubiquitination and protein degradation of HIF-1α. Furthermore, CaO2-MNPs inhibited autophagy and induced apoptosis in TNBC cells. Our animal studies with an orthotopic mouse model showed that CaO2-MNPs in combination with doxorubicin exhibited a stronger tumor-suppressive effect on TNBC, compared to the doxorubicin treatment alone. Our findings suggest that combined with CaO2-MNPs and doxorubicin attenuates HIF-1α expression to improve the efficiency of chemotherapy in TNBC.
Collapse
|
83
|
The Shuganhuazheng Formula in Triple-Negative Breast Cancer: A Study Based on Network Pharmacology and In Vivo Experiments. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2020:8173147. [PMID: 33414839 PMCID: PMC7752265 DOI: 10.1155/2020/8173147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 11/18/2020] [Accepted: 11/26/2020] [Indexed: 02/06/2023]
Abstract
Breast cancer is the most common cancer in women. Among breast cancer subtypes, triple-negative breast cancer (TNBC) has the highest degree of malignancy and the worst prognosis. The Shuganhuazheng formula (SGHZF) is a traditional Chinese herbal formula for the treatment of TNBC, but the mechanism of SGHZF in the treatment of TNBC remains unclear. In this study, the therapeutic effect and mechanism of SGHZF against TNBC were preliminarily determined based on in vivo experimental verification and network pharmacology. In terms of therapeutic effects, the antitumour effect was verified by measuring and calculating tumour volume, and the expression of proto-oncogene c-Myc was verified by PCR. In terms of the mechanism, potential therapeutic targets were identified by overlapping the SGHZF-related and TNBC-related targets. After comprehensively analysing the results of the protein-protein interaction (PPI), gene ontology (GO) function, and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses, Akt and HIF-1α were selected for verification by using immunohistochemical and Western blot analyses. The results of the study indicated that SGHZF can inhibit breast tumour growth in mice and that the mechanism may be related to the inhibition of Akt and HIF-1α expression.
Collapse
|
84
|
Satija S, Kaur H, Tambuwala MM, Sharma P, Vyas M, Khurana N, Sharma N, Bakshi HA, Charbe NB, Zacconi FC, Aljabali AA, Nammi S, Dureja H, Singh TG, Gupta G, Dhanjal DS, Dua K, Chellappan DK, Mehta M. Hypoxia-Inducible Factor (HIF): Fuel for Cancer Progression. Curr Mol Pharmacol 2021; 14:321-332. [PMID: 33494692 DOI: 10.2174/1874467214666210120154929] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 09/24/2020] [Accepted: 10/09/2020] [Indexed: 11/22/2022]
Abstract
Hypoxia is an integral part of the tumor microenvironment, caused primarily due to rapidly multiplying tumor cells and a lack of proper blood supply. Among the major hypoxic pathways, HIF-1 transcription factor activation is one of the widely investigated pathways in the hypoxic tumor microenvironment (TME). HIF-1 is known to activate several adaptive reactions in response to oxygen deficiency in tumor cells. HIF-1 has two subunits, HIF-1β (constitutive) and HIF-1α (inducible). The HIF-1α expression is largely regulated via various cytokines (through PI3K-ACT-mTOR signals), which involves the cascading of several growth factors and oncogenic cascades. These events lead to the loss of cellular tumor suppressant activity through changes in the level of oxygen via oxygen-dependent and oxygen-independent pathways. The significant and crucial role of HIF in cancer progression and its underlying mechanisms have gained much attention lately among the translational researchers in the fields of cancer and biological sciences, which have enabled them to correlate these mechanisms with various other disease modalities. In the present review, we have summarized the key findings related to the role of HIF in the progression of tumors.
Collapse
Affiliation(s)
- Saurabh Satija
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara-144411, Punjab, India
| | - Harpreet Kaur
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara-144411, Punjab, India
| | - Murtaza M Tambuwala
- School of Pharmacy and Pharmaceutical Sciences, Ulster University, Coleraine, County Londonderry, BT52 1SA, Northern Ireland, United Kingdom
| | - Prabal Sharma
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara-144411, Punjab, India
| | - Manish Vyas
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara-144411, Punjab, India
| | - Navneet Khurana
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara-144411, Punjab, India
| | - Neha Sharma
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara-144411, Punjab, India
| | - Hamid A Bakshi
- School of Pharmacy and Pharmaceutical Sciences, Ulster University, Coleraine, County Londonderry, BT52 1SA, Northern Ireland, United Kingdom
| | - Nitin B Charbe
- Departamento de Quimica Organica, Facultad de Quimica y de Farmacia, Pontificia Universidad Católica de Chile, Av. Vicuna McKenna 4860, 7820436 Macul, Santiago, Chile
| | - Flavia C Zacconi
- Departamento de Quimica Organica, Facultad de Quimica y de Farmacia, Pontificia Universidad Católica de Chile, Av. Vicuna McKenna 4860, 7820436 Macul, Santiago, Chile
| | - Alaa A Aljabali
- Yarmouk University - Faculty of Pharmacy, Department of Pharmaceutical Sciences, Irbid, Jordan
| | - Srinivas Nammi
- School of Science and Health, Western Sydney University, Penrith NSW 2751, Australia
| | - Harish Dureja
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana, India
| | - Thakur G Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Gaurav Gupta
- School of Pharmaceutical Sciences, Suresh Gyan Vihar University, Jaipur, India
| | - Daljeet S Dhanjal
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara-144411, Punjab, India
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo NSW 2007, Australia
| | - Dinesh K Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| | - Meenu Mehta
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara-144411, Punjab, India
| |
Collapse
|
85
|
Wang X, Li S, Liu X, Wu X, Ye N, Yang X, Li Z. Boosting Nanomedicine Efficacy with Hyperbaric Oxygen Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1295:77-95. [PMID: 33543456 DOI: 10.1007/978-3-030-58174-9_4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nanomedicine has been a hot topic in the field of tumor therapy in the past few decades. Because of the enhanced permeability and retention effect (EPR effect), nanomedicine can passively yet selectively accumulate at tumor tissues. As a result, it can improve drug concentration in tumor tissues and reduce drug distribution in normal tissues, thereby contributing to enhanced antitumor effect and reduced adverse effects. However, the therapeutic efficacy of anticancer nanomedicine is not satisfactory in clinical settings. Therefore, how to improve the clinical therapeutic effect of nanomedicine has become an urgent problem. The grand challenges of nanomedicine lie in how to overcome various pathophysiological barriers and simultaneously kill cancer cells effectively in hypoxic tumor microenvironment (TME). To this end, the development of novel stimuli-responsive nanomedicine has become a new research hotspot. While a great deal of progress has been made in this direction and preclinical results report many different kinds of promising multifunctional smart nanomedicine, the design of these intelligent nanomedicines is often too complicated, the requirements for the preparation processes are strict, the cost is high, and the clinical translation is difficult. Thus, it is more practical to find solutions to promote the therapeutic efficacy of commercialized nanomedicines, for example, Doxil®, Oncaspar®, DaunoXome®, Abraxane®, to name a few. Increasing attention has been paid to the combination of modern advanced medical technology and nanomedicine for the treatment of various malignancies. Recently, we found that hyperbaric oxygen (HBO) therapy could enhance Doxil® antitumor efficacy. Inspired by this study, we further carried out researches on the combination of HBO therapy with other nanomedicines for various cancer therapies, and revealed that HBO therapy could significantly boost antitumor efficacy of nanomedicine-mediated photodynamic therapy and photothermal therapy in different kinds of tumors, including hepatocellular carcinoma, breast cancer, and gliomas. Our results implicate that HBO therapy might be a universal strategy to boost therapeutic efficacy of nanomedicine against hypoxic solid malignancies.
Collapse
Affiliation(s)
- Xiaoxian Wang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Si Li
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Liu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Xian Wu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Ningbing Ye
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangliang Yang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medical, Huazhong University of Science and Technology, Wuhan, China
| | - Zifu Li
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China.
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medical, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
86
|
Fudulu A, Olar R, Maxim C, Scăeţeanu GV, Bleotu C, Matei L, Chifiriuc MC, Badea M. New Cobalt (II) Complexes with Imidazole Derivatives: Antimicrobial Efficiency against Planktonic and Adherent Microbes and In Vitro Cytotoxicity Features. Molecules 2020; 26:molecules26010055. [PMID: 33374438 PMCID: PMC7796409 DOI: 10.3390/molecules26010055] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 12/18/2020] [Accepted: 12/19/2020] [Indexed: 01/28/2023] Open
Abstract
Three novel Co(II) complexes of the type [Co(C4H5O2)2L2] (where C4H5O2 is methacrylate anion; L = C3H4N2 (imidazole; HIm) (1), C4H6N2 (2-methylimidazole; 2-MeIm) (2), C5H8N2 (2-ethylimidazole; 2-EtIm) (3)) have been synthesized and characterized by elemental analysis, IR and UV-Vis spectroscopic techniques, thermal analysis and single crystal X-ray diffraction. X-ray crystallography revealed for complexes (1) and (2) distorted trigonal bipyramid stereochemistry for Co(II), meanwhile for complex (3) evidenced that the unit cell comprises three molecular units with interesting structural features. In each unit, both stereochemistry adopted by metallic ion and coordination modes of carboxylate anions are different. The screening of antimicrobial activity revealed that Candida albicans planktonic cells were the most susceptible, with minimal inhibitory concentration (MIC) values of 7.8 μg/mL for complexes (1) and (2) and 15.6 μg/mL for complex (3). Complexes (1) and (2) proved to be more active than complex (3) against the tested bacterial strains, both in planktonic and biofilm growth state, with MIC and minimal biofilm eradication concentration (MBEC) values ranging from 15.6 to 62.5 μg/mL, the best antibacterial effects being noticed against Staphylococcus aureus and Pseudomonas aeruginosa. Remarkably, the MBEC values obtained for the four tested bacterial strains were either identical or even lower than the MIC ones. The cytotoxicity assay indicated that the tested complexes affected the cellular cycle of HeLa, HCT-8, and MG63 cells, probably by inhibiting the expression of vimentin and transient receptor potential canonical 1 (TRPC1). The obtained biological results recommend these complexes as potential candidates for the development of novel anti-biofilm agents.
Collapse
Affiliation(s)
- Alina Fudulu
- Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania; (A.F.); (C.B.); (L.M.)
| | - Rodica Olar
- Department of Inorganic Chemistry, Faculty of Chemistry, University of Bucharest, 90–92 Panduri Str., 050663 Bucharest, Romania; (R.O.); (C.M.)
| | - Cătălin Maxim
- Department of Inorganic Chemistry, Faculty of Chemistry, University of Bucharest, 90–92 Panduri Str., 050663 Bucharest, Romania; (R.O.); (C.M.)
| | - Gina Vasile Scăeţeanu
- Department of Soil Sciences, University of Agronomical Sciences and Veterinary Medicine, 59 Mărăști Str., 011464 Bucharest, Romania;
| | - Coralia Bleotu
- Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania; (A.F.); (C.B.); (L.M.)
| | - Lilia Matei
- Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania; (A.F.); (C.B.); (L.M.)
| | - Mariana Carmen Chifiriuc
- Department of Microbiology, Faculty of Biology, University of Bucharest, 1–3 Aleea Portocalelor Str., 60101 Bucharest, Romania;
- Academy of Romanian Scientists, 010071 Bucharest, Romania
| | - Mihaela Badea
- Department of Inorganic Chemistry, Faculty of Chemistry, University of Bucharest, 90–92 Panduri Str., 050663 Bucharest, Romania; (R.O.); (C.M.)
- Correspondence: ; Tel.: +40-072-314-0111
| |
Collapse
|
87
|
Cowell GW. Editorial for: "3D Oxygen-Enhanced MR Imaging at 3T MR System: Comparison With Thin-Section CT of Quantitative Capability for Pulmonary Functional Loss Assessment and Clinical Stage Classification of COPD in Smokers". J Magn Reson Imaging 2020; 53:1052-1053. [PMID: 33354851 DOI: 10.1002/jmri.27471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 11/30/2020] [Indexed: 11/12/2022] Open
Affiliation(s)
- Gordon W Cowell
- Department of Imaging, Queen Elizabeth University Hospital, Glasgow, UK.,College of MVLS, University of Glasgow, Glasgow, UK
| |
Collapse
|
88
|
Shahzadi A, Ikram F, Subhani RUH, Ahmed A, Asif A, Fatima N, Chaudhry AA, Hu Y. Acid susceptible polymeric stealthy nanoparticles for improved anticancer drug delivery. INT J POLYM MATER PO 2020. [DOI: 10.1080/00914037.2019.1683556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- Anam Shahzadi
- Interdisciplinary Research Centre in Biomedical Materials, COMSATS University Islamabad, Lahore, Pakistan
- Department of Pharmacy, COMSATS University Islamabad, Abbottabad, Pakistan
| | - Fakhera Ikram
- Interdisciplinary Research Centre in Biomedical Materials, COMSATS University Islamabad, Lahore, Pakistan
| | | | - Arsalan Ahmed
- Interdisciplinary Research Centre in Biomedical Materials, COMSATS University Islamabad, Lahore, Pakistan
| | - Anila Asif
- Interdisciplinary Research Centre in Biomedical Materials, COMSATS University Islamabad, Lahore, Pakistan
| | - Nighat Fatima
- Department of Pharmacy, COMSATS University Islamabad, Abbottabad, Pakistan
| | - Aqif Anwar Chaudhry
- Interdisciplinary Research Centre in Biomedical Materials, COMSATS University Islamabad, Lahore, Pakistan
| | - Yong Hu
- Collaborative Innovation Center of Chemistry for Life Sciences, College of Engineering and Applied Sciences, Nanjing University, Nanjing, China
| |
Collapse
|
89
|
Solimando AG, Summa SD, Vacca A, Ribatti D. Cancer-Associated Angiogenesis: The Endothelial Cell as a Checkpoint for Immunological Patrolling. Cancers (Basel) 2020; 12:cancers12113380. [PMID: 33203154 PMCID: PMC7696032 DOI: 10.3390/cancers12113380] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 11/08/2020] [Accepted: 11/12/2020] [Indexed: 12/11/2022] Open
Abstract
Simple Summary A clinical decision and study design investigating the level and extent of angiogenesis modulation aimed at vascular normalization without rendering tissues hypoxic is key and represents an unmet medical need. Specifically, determining the active concentration and optimal times of the administration of antiangiogenetic drugs is crucial to inhibit the growth of any microscopic residual tumor after surgical resection and in the pre-malignant and smolder neoplastic state. This review uncovers the pre-clinical translational insights crucial to overcome the caveats faced so far while employing anti-angiogenesis. This literature revision also explores how abnormalities in the tumor endothelium harm the crosstalk with an effective immune cell response, envisioning a novel combination with other anti-cancer drugs and immunomodulatory agents. These insights hold vast potential to both repress tumorigenesis and unleash an effective immune response. Abstract Cancer-associated neo vessels’ formation acts as a gatekeeper that orchestrates the entrance and egress of patrolling immune cells within the tumor milieu. This is achieved, in part, via the directed chemokines’ expression and cell adhesion molecules on the endothelial cell surface that attract and retain circulating leukocytes. The crosstalk between adaptive immune cells and the cancer endothelium is thus essential for tumor immune surveillance and the success of immune-based therapies that harness immune cells to kill tumor cells. This review will focus on the biology of the endothelium and will explore the vascular-specific molecular mediators that control the recruitment, retention, and trafficking of immune cells that are essential for effective antitumor immunity. The literature revision will also explore how abnormalities in the tumor endothelium impair crosstalk with adaptive immune cells and how targeting these abnormalities can improve the success of immune-based therapies for different malignancies, with a particular focus on the paradigmatic example represented by multiple myeloma. We also generated and provide two original bio-informatic analyses, in order to sketch the physiopathology underlying the endothelial–neoplastic interactions in an easier manner, feeding into a vicious cycle propagating disease progression and highlighting novel pathways that might be exploited therapeutically.
Collapse
Affiliation(s)
- Antonio Giovanni Solimando
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine ‘G. Baccelli’, University of Bari Medical School, 70124 Bari, Italy;
- Istituto di Ricovero e Cura a Carattere Scientifico-IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy
- Correspondence: (A.G.S.); (D.R.); Tel.: +39-3395626475 (A.G.S.); +39-080-5478326 (D.R.)
| | - Simona De Summa
- Molecular Diagnostics and Pharmacogenetics Unit, IRCCS Istituto Tumori Giovanni Paolo II, 70124 Bari, Italy;
| | - Angelo Vacca
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine ‘G. Baccelli’, University of Bari Medical School, 70124 Bari, Italy;
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, University of Bari Medical School, 70124 Bari, Italy
- Correspondence: (A.G.S.); (D.R.); Tel.: +39-3395626475 (A.G.S.); +39-080-5478326 (D.R.)
| |
Collapse
|
90
|
Metronidazole-conjugates: A comprehensive review of recent developments towards synthesis and medicinal perspective. Eur J Med Chem 2020; 210:112994. [PMID: 33234343 DOI: 10.1016/j.ejmech.2020.112994] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/22/2020] [Accepted: 11/02/2020] [Indexed: 12/13/2022]
Abstract
Nitroimidazoles based compounds remain a hot topic of research in medicinal chemistry due to their numerous biological activities. Moreover, many clinical candidates based on this chemical core have been reported to be valuable in the treatment of human diseases. Metronidazole (MTZ) derived conjugates demonstrated a potential application in medicinal chemistry research over the last decade. In this review, we summarize the synthesis, key structure-activity-relationship (SAR) and associated biological activities such as antimicrobial, anticancer, antidiabetic, anti-inflammatory, anti-HIV and anti-parasitic (Anti-trichomonas, antileishmanial, antiamoebic and anti-giardial) of explored MTZ-conjugates. The molecular docking analysis is also presented simultaneously, which will assist in developing an understanding towards designing of new MTZ-conjugates for target-based drug discovery against multiple disease areas.
Collapse
|
91
|
Roma-Rodrigues C, Pombo I, Fernandes AR, Baptista PV. Hyperthermia Induced by Gold Nanoparticles and Visible Light Photothermy Combined with Chemotherapy to Tackle Doxorubicin Sensitive and Resistant Colorectal Tumor 3D Spheroids. Int J Mol Sci 2020; 21:E8017. [PMID: 33126535 PMCID: PMC7672550 DOI: 10.3390/ijms21218017] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 10/22/2020] [Accepted: 10/26/2020] [Indexed: 12/17/2022] Open
Abstract
Current cancer therapies are frequently ineffective and associated with severe side effects and with acquired cancer drug resistance. The development of effective therapies has been hampered by poor correlations between pre-clinical and clinical outcomes. Cancer cell-derived spheroids are three-dimensional (3D) structures that mimic layers of tumors in terms of oxygen and nutrient and drug resistance gradients. Gold nanoparticles (AuNP) are promising therapeutic agents which permit diminishing the emergence of secondary effects and increase therapeutic efficacy. In this work, 3D spheroids of Doxorubicin (Dox)-sensitive and -resistant colorectal carcinoma cell lines (HCT116 and HCT116-DoxR, respectively) were used to infer the potential of the combination of chemotherapy and Au-nanoparticle photothermy in the visible (green laser of 532 nm) to tackle drug resistance in cancer cells. Cell viability analysis of 3D tumor spheroids suggested that AuNPs induce cell death in the deeper layers of spheroids, further potentiated by laser irradiation. The penetration of Dox and earlier spheroid disaggregation is potentiated in combinatorial therapy with Dox, AuNP functionalized with polyethylene glycol (AuNP@PEG) and irradiation. The time point of Dox administration and irradiation showed to be important for spheroids destabilization. In HCT116-sensitive spheroids, pre-irradiation induced earlier disintegration of the 3D structure, while in HCT116 Dox-resistant spheroids, the loss of spheroid stability occurred almost instantly in post-irradiated spheroids, even with lower Dox concentrations. These results point towards the application of new strategies for cancer therapeutics, reducing side effects and resistance acquisition.
Collapse
|
92
|
Gao X, Jiang S, Li C, Chen Y, Zhang Y, Huang P, Lin J. Highly photostable croconium dye-anchored cell membrane vesicle for tumor pH-responsive duplex imaging-guided photothermal therapy. Biomaterials 2020; 267:120454. [PMID: 33160122 DOI: 10.1016/j.biomaterials.2020.120454] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 09/17/2020] [Accepted: 10/15/2020] [Indexed: 12/12/2022]
Abstract
The development of tumor acidic microenvironment-responsive theranostic agents is a research hotspot. Herein, we developed highly photostable amphiphilic croconium dye-anchored red blood cell membrane vesicle (denoted as LET-5) for tumor pH-responsive near-infrared fluorescence (NIRF) and photoacoustic (PA) duplex imaging-guided photothermal therapy. In tumor acidic microenvironment, both NIRF and PA signals of LET-5 were significantly enhanced and the photothermal effect of LET-5 was activated. Notably, cell membrane-based vesicle with enhanced stability and long blood circulation significantly improved the tumor accumulation of croconium dye, thus achieving better therapeutic effect than free croconium dye. These findings provide a promising approach to construct amphiphilic dye-anchored cell membrane vesicle for cancer theranostics.
Collapse
Affiliation(s)
- Xiaoting Gao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University Wenzhou, Zhejiang, 325000, China; Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Shanshan Jiang
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Chunying Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University Wenzhou, Zhejiang, 325000, China; Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Yunzhi Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University Wenzhou, Zhejiang, 325000, China
| | - Yifan Zhang
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Peng Huang
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Jing Lin
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University Wenzhou, Zhejiang, 325000, China; Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, 518060, China.
| |
Collapse
|
93
|
Sun X, Ni N, Ma Y, Wang Y, Leong DT. Retooling Cancer Nanotherapeutics' Entry into Tumors to Alleviate Tumoral Hypoxia. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2003000. [PMID: 32803846 DOI: 10.1002/smll.202003000] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/20/2020] [Indexed: 06/11/2023]
Abstract
Anti-hypoxia cancer nanomedicine (AHCN) holds exciting potential in improving oxygen-dependent therapeutic efficiencies of malignant tumors. However, most studies regarding AHCN focus on optimizing structure and function of nanomaterials with presupposed successful entry into tumor cells. From such a traditional perspective, the main barrier that AHCN needs to overcome is mainly the tumor cell membrane. However, such an oversimplified perspective would neglect that real tumors have many biological, physiological, physical, and chemical defenses preventing the current state-of-the-art AHCNs from even reaching the targeted tumor cells. Fortunately, in recent years, some studies are beginning to intentionally focus on overcoming physiological barriers to alleviate hypoxia. In this Review, the limitations behind the traditional AHCN delivery mindset are addressed and the key barriers that need to be surmounted before delivery to cancer cells and some good ways to improve cell membrane attachment, internalization, and intracellular retention are summarized. It is aimed to contribute to Review literature on this emerging topic through refreshing perspectives based on this work and what is also learnt from others. This Review would therefore assist AHCNs researchers to have a quick overview of the essential information and glean thought-provoking ideas to advance this sub-field in cancer nanomedicine.
Collapse
Affiliation(s)
- Xiao Sun
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
| | - Nengyi Ni
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
| | - Yanling Ma
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
| | - Yan Wang
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
| | - David Tai Leong
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
| |
Collapse
|
94
|
Liu Y, Wang X, Li W, Xu Y, Zhuo Y, Li M, He Y, Wang X, Guo Q, Zhao L, Qiang L. Oroxylin A reverses hypoxia-induced cisplatin resistance through inhibiting HIF-1α mediated XPC transcription. Oncogene 2020; 39:6893-6905. [PMID: 32978517 DOI: 10.1038/s41388-020-01474-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 09/04/2020] [Accepted: 09/15/2020] [Indexed: 12/20/2022]
Abstract
Hypoxia is a key concern during the treatment of non-small cell lung cancer (NSCLC), and hypoxia-inducible factor 1 alpha (HIF-1α) has been associated with increased tumor resistance to therapeutic modalities such as cisplatin. Compensatory activation of nucleotide excision repair (NER) pathway is the major mechanism that accounts for cisplatin resistance. In the present study, we suggest a novel strategy to improve the treatment of NSCLC and overcome the hypoxia-induced cisplatin resistance by cotreatment with Oroxylin A, one of the main bioactive flavonoids of Scutellariae radix. Based on the preliminary screening, we found that xeroderma pigmentosum group C (XPC), an important DNA damage recognition protein involved in NER, dramatically increased in hypoxic condition and contributed to hypoxia-induced cisplatin resistance. Further data suggested that Oroxylin A significantly reversed the hypoxia-induced cisplatin resistance through directly binding to HIF-1α bHLH-PAS domain and blocking its binding to HRE3 transcription factor binding sites on XPC promoter which is important to hypoxia-induced XPC transcription. Taken together, our findings not only demonstrate a crucial role of XPC dependent NER in hypoxia-induced cisplatin resistance, but also suggest a previously unrecognized tumor suppressive mechanism of Oroxylin A in NSCLC which through sensitization of cisplatin-mediated growth inhibition and apoptosis under hypoxia.
Collapse
Affiliation(s)
- Yunyao Liu
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xiaoping Wang
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Wenshu Li
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yujiao Xu
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yating Zhuo
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Mengyuan Li
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yuan He
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xiaosheng Wang
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Qinglong Guo
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Li Zhao
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China.
| | - Lei Qiang
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
95
|
Boteon Y, Flores Carvalho MA, Panconesi R, Muiesan P, Schlegel A. Preventing Tumour Recurrence after Liver Transplantation: The Role of Machine Perfusion. Int J Mol Sci 2020; 21:5791. [PMID: 32806712 PMCID: PMC7460879 DOI: 10.3390/ijms21165791] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 07/31/2020] [Accepted: 08/05/2020] [Indexed: 02/07/2023] Open
Abstract
Tumour recurrence is currently a hot topic in liver transplantation. The basic mechanisms are increasingly discussed, and, for example, recurrence of hepatocellular carcinoma is often described in pre-injured donor livers, which frequently suffer from significant ischemia/reperfusion injury. This review article highlights the underlying mechanisms and describes the specific tissue milieu required to promote tumour recurrence after liver transplantation. We summarise the current literature in this field and show risk factors that contribute to a pro-tumour-recurrent environment. Finally, the potential role of new machine perfusion technology is discussed, including the most recent data, which demonstrate a protective effect of hypothermic oxygenated perfusion before liver transplantation.
Collapse
Affiliation(s)
- Yuri Boteon
- Liver Unit, Albert Einstein Hospital, 05652–900 São Paulo, Brazil;
- Albert Einstein Jewish Institute for Education and Research, 05652–900 São Paulo, Brazil
| | - Mauricio Alfredo Flores Carvalho
- Hepatobiliary Unit, Department of Clinical and Experimental Medicine, University of Florence, AOU Careggi, 50134 Florence, Italy; (M.A.F.C.); (R.P.); (P.M.)
| | - Rebecca Panconesi
- Hepatobiliary Unit, Department of Clinical and Experimental Medicine, University of Florence, AOU Careggi, 50134 Florence, Italy; (M.A.F.C.); (R.P.); (P.M.)
| | - Paolo Muiesan
- Hepatobiliary Unit, Department of Clinical and Experimental Medicine, University of Florence, AOU Careggi, 50134 Florence, Italy; (M.A.F.C.); (R.P.); (P.M.)
- The Liver Unit, Queen Elizabeth Hospital Birmingham, Edgbaston, Birmingham B15 2TH, UK
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham B15 2TT, UK
| | - Andrea Schlegel
- Hepatobiliary Unit, Department of Clinical and Experimental Medicine, University of Florence, AOU Careggi, 50134 Florence, Italy; (M.A.F.C.); (R.P.); (P.M.)
| |
Collapse
|
96
|
Yao L, Feng L, Tao D, Tao H, Zhong X, Liang C, Zhu Y, Hu B, Liu Z, Zheng Y. Perfluorocarbon nanodroplets stabilized with cisplatin-prodrug-constructed lipids enable efficient tumor oxygenation and chemo-radiotherapy of cancer. NANOSCALE 2020; 12:14764-14774. [PMID: 32627775 DOI: 10.1039/d0nr01476a] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Concurrent chemo-radiotherapy has been widely applied for the treatment of a wide range of cancers, but its therapeutic efficacy against most solid tumors is severely impaired by their intrinsic hypoxic microenvironments. Utilizing the high oxygen loading capacity of perfluoro-15-crown-5-ether (PFCE), herein, we prepare PFCE nanodroplets with cisplatin prodrug (cisPt(iv)) conjugated phospholipids and other commercial lipids as the stabilizer to enable tumor targeted oxygen shuttling. The obtained PFCE@cisPt(iv)-Lip shows high physiological stability and efficient oxygen loading capacity. As vividly visualized under an in vivo photoacoustic imaging system, tumors on the mice with intravenous injection of such PFCE@cisPt(iv)-Lip show effective tumor oxygenation. Together with X-ray exposure, such PFCE@cisPt(iv)-Lip upon intravenous injection could induce severe DNA damage of cells, thereby remarkably suppressing the tumor growth and significantly prolonging their survival time without causing obvious toxic side effects. This work highlights PFCE@cisPt(iv)-Lip as an adjuvant nanomedicine for enhanced chemo-radiotherapy of tumors by attenuating hostile tumor hypoxia, indicating its promising potential for future clinical translation ascribed to its straightforward synthesis and notable tumor growth inhibition at a safe dose.
Collapse
Affiliation(s)
- Li Yao
- Department of Ultrasound in Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Institute of Ultrasound in Medicine, Shanghai 200233, P.R. China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
97
|
Zou MZ, Liu WL, Chen HS, Bai XF, Gao F, Ye JJ, Cheng H, Zhang XZ. Advances in nanomaterials for treatment of hypoxic tumor. Natl Sci Rev 2020; 8:nwaa160. [PMID: 34691571 PMCID: PMC8288333 DOI: 10.1093/nsr/nwaa160] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 06/01/2020] [Accepted: 07/01/2020] [Indexed: 02/06/2023] Open
Abstract
Abstract
The hypoxic tumor microenvironment is characterized by disordered vasculature and rapid proliferation of tumors, resulting from tumor invasion, progression and metastasis. The hypoxic conditions restrict efficiency of tumor therapies, such as chemotherapy, radiotherapy, phototherapy and immunotherapy, leading to serious results of tumor recurrence and high mortality. Recently, research has concentrated on developing functional nanomaterials to treat hypoxic tumors. In this review, we categorize such nanomaterials into (i) nanomaterials that elevate oxygen levels in tumors for enhanced oxygen-dependent tumor therapy and (ii) nanomaterials with diminished oxygen dependence for hypoxic tumor therapy. To elevate oxygen levels in tumors, oxygen-carrying nanomaterials, oxygen-generating nanomaterials and oxygen-economizing nanomaterials can be used. To diminish oxygen dependence of nanomaterials for hypoxic tumor therapy, therapeutic gas-generating nanomaterials and radical-generating nanomaterials can be used. The biocompatibility and therapeutic efficacy of these nanomaterials are discussed.
Collapse
Affiliation(s)
- Mei-Zhen Zou
- The Institute for Advanced Studies, Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, China
| | - Wen-Long Liu
- School of Chemistry and Materials Science, South-Central University for Nationalities, Wuhan 430074, China
| | - Han-Shi Chen
- The Institute for Advanced Studies, Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, China
| | - Xue-Feng Bai
- The Institute for Advanced Studies, Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, China
| | - Fan Gao
- The Institute for Advanced Studies, Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, China
| | - Jing-Jie Ye
- The Institute for Advanced Studies, Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, China
| | - Han Cheng
- The Institute for Advanced Studies, Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, China
| | - Xian-Zheng Zhang
- The Institute for Advanced Studies, Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, China
| |
Collapse
|
98
|
Meng Q, Zhang Y, Hu LG. Targeting Autophagy Facilitates T Lymphocyte Migration by Inducing the Expression of CXCL10 in Gastric Cancer Cell Lines. Front Oncol 2020; 10:886. [PMID: 32582551 PMCID: PMC7280490 DOI: 10.3389/fonc.2020.00886] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 05/05/2020] [Indexed: 12/20/2022] Open
Abstract
Autophagy is a type of cellular catabolic degradation process that occurs in response to nutrient starvation or metabolic stress, and is a valuable resource for highly proliferating cancer cells. Autophagy also facilitates the resistance of cancer cells to antitumor therapies. However, the involvement of autophagy in regulating CXCL10 expression in gastric cancer (GC) cells and T lymphocyte migration remains unclear. In this study, we aimed to investigate the effect of autophagy inhibition on CXCL10 expression and T lymphocyte infiltration in GC and elucidate the underlying mechanism. Analysis of public databases revealed a positive correlation between CXCL10 expression and both prognosis of patients with GC and the expression profile of T lymphocyte markers in the GCs. Chemotaxis and spheroid infiltration assays revealed that CXCL10 induced T lymphocyte migration and infiltration into GC spheroids, an in vitro three-dimensional cell culture model. In addition, in vitro autophagy inhibition in GC cells increased CXCL10 expression under both normal and hypoxic culture conditions. Further investigation on the underlying mechanism showed that in vitro autophagy inhibition suppressed the JNK signaling pathway and further enhanced CXCL10 expression in GC cells. Collectively, our results provide novel insights for understanding the role of autophagy in regulation of intra-tumor immunity.
Collapse
Affiliation(s)
- Qingyuan Meng
- Department of Comparative Biology and Safety Science, Amgen Biopharmaceutical R&D (Shanghai) Co., Ltd, Shanghai, China
| | - Yihong Zhang
- Department of Comparative Biology and Safety Science, Amgen Biopharmaceutical R&D (Shanghai) Co., Ltd, Shanghai, China
| | - Liangbiao George Hu
- Department of Comparative Biology and Safety Science, Amgen Biopharmaceutical R&D (Shanghai) Co., Ltd, Shanghai, China
| |
Collapse
|
99
|
Wang J, Sun J, Hu W, Wang Y, Chou T, Zhang B, Zhang Q, Ren L, Wang H. A Porous Au@Rh Bimetallic Core-Shell Nanostructure as an H 2 O 2 -Driven Oxygenerator to Alleviate Tumor Hypoxia for Simultaneous Bimodal Imaging and Enhanced Photodynamic Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e2001862. [PMID: 32329171 PMCID: PMC7386557 DOI: 10.1002/adma.202001862] [Citation(s) in RCA: 131] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 03/27/2020] [Accepted: 03/30/2020] [Indexed: 05/19/2023]
Abstract
In treatment of hypoxic tumors, oxygen-dependent photodynamic therapy (PDT) is considerably limited. Herein, a new bimetallic and biphasic Rh-based core-shell nanosystem (Au@Rh-ICG-CM) is developed to address tumor hypoxia while achieving high PDT efficacy. Such porous Au@Rh core-shell nanostructures are expected to exhibit catalase-like activity to efficiently catalyze oxygen generation from endogenous hydrogen peroxide in tumors. Coating Au@Rh nanostructures with tumor cell membrane (CM) enables tumor targeting via homologous binding. As a result of the large pores of Rh shells and the trapping ability of CM, the photosensitizer indocyanine green (ICG) is successfully loaded and retained in the cavity of Au@Rh-CM. Au@Rh-ICG-CM shows good biocompatibility, high tumor accumulation, and superior fluorescence and photoacoustic imaging properties. Both in vitro and in vivo results demonstrate that Au@Rh-ICG-CM is able to effectively convert endogenous hydrogen peroxide into oxygen and then elevate the production of tumor-toxic singlet oxygen to significantly enhance PDT. As noted, the mild photothermal effect of Au@Rh-ICG-CM also improves PDT efficacy. By integrating the superiorities of hypoxia regulation function, tumor accumulation capacity, bimodal imaging, and moderate photothermal effect into a single nanosystem, Au@Rh-ICG-CM can readily serve as a promising nanoplatform for enhanced cancer PDT.
Collapse
Affiliation(s)
- Jinping Wang
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ, 07030, USA
| | - Jingyu Sun
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, NJ, 07030, USA
| | - Wei Hu
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, NJ, 07030, USA
| | - Yuhao Wang
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ, 07030, USA
| | - Tsengming Chou
- Laboratory for Multiscale Imaging, Stevens Institute of Technology, Hoboken, NJ, 07030, USA
| | - Beilu Zhang
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, NJ, 07030, USA
| | - Qiang Zhang
- Department of Biomaterials, Key Laboratory of Biomedical Engineering of Fujian Province, State Key Lab of Physical Chemistry of Solid Surface, College of Materials, Xiamen University, Xiamen, Fujian, 361005, P. R. China
| | - Lei Ren
- Department of Biomaterials, Key Laboratory of Biomedical Engineering of Fujian Province, State Key Lab of Physical Chemistry of Solid Surface, College of Materials, Xiamen University, Xiamen, Fujian, 361005, P. R. China
| | - Hongjun Wang
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ, 07030, USA
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, NJ, 07030, USA
| |
Collapse
|
100
|
Phung CD, Tran TH, Pham LM, Nguyen HT, Jeong JH, Yong CS, Kim JO. Current developments in nanotechnology for improved cancer treatment, focusing on tumor hypoxia. J Control Release 2020; 324:413-429. [PMID: 32461115 DOI: 10.1016/j.jconrel.2020.05.029] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 05/15/2020] [Accepted: 05/19/2020] [Indexed: 12/12/2022]
Abstract
Hypoxia is a common feature of the tumor microenvironment, which is characterized by tissue oxygen deficiency due to an aggressive proliferation of cancer cells. Hypoxia activates hypoxia-inducible factor-dependent signaling, which in turn regulates metabolic reprogramming, immune suppression, resistance to apoptosis, angiogenesis, metastasis, and invasion to secondary sites. In this review, we provide an overview of the use of nanotechnology to harmonize intra-tumoral oxygen or suppress hypoxia-related signaling for an improved efficacy of cancer treatment. The biological background was followed by conducting a literature review on the (1) nanoparticles responsible for enhancing oxygen levels within the tumor, (2) nanoparticles sensitizing hypoxia, (3) nanoparticles suppressing hypoxia-inducing factor, (4) nanoparticles that relieve tumor hypoxia for enhancement of chemotherapy, photodynamic therapy, and immunotherapy, either individually or in combination. Lastly, the heterogeneity of cancer and limitations of nanotechnology are discussed to facilitate translational therapeutic treatment.
Collapse
Affiliation(s)
- Cao Dai Phung
- College of Pharmacy, Yeungnam University, 280 Deahak-ro, Gyeongsan 38541, Republic of Korea
| | - Tuan Hiep Tran
- Faculty of Pharmacy, PHENIKAA University, Yen Nghia, Ha Dong, Hanoi 12116, Viet Nam; PHENIKAA Research and Technology Institute (PRATI), A&A Green Phoenix Group JSC, No.167 Hoang Ngan, Trung Hoa, Cau Giay, Hanoi 11313, Viet Nam
| | - Le Minh Pham
- College of Pharmacy, Yeungnam University, 280 Deahak-ro, Gyeongsan 38541, Republic of Korea
| | - Hanh Thuy Nguyen
- Department of Industrial & Physical Pharmacy, College of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, United States
| | - Jee-Heon Jeong
- College of Pharmacy, Yeungnam University, 280 Deahak-ro, Gyeongsan 38541, Republic of Korea
| | - Chul Soon Yong
- College of Pharmacy, Yeungnam University, 280 Deahak-ro, Gyeongsan 38541, Republic of Korea
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, 280 Deahak-ro, Gyeongsan 38541, Republic of Korea.
| |
Collapse
|