1
|
Kakoti BB, Zothantluanga JH, Deka K, Halder RK, Roy D. In silico design and computational screening of berberine derivatives for potential antidiabetic activity through allosteric activation of the AMPK pathway. In Silico Pharmacol 2025; 13:12. [PMID: 39780772 PMCID: PMC11704122 DOI: 10.1007/s40203-024-00295-0] [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: 12/19/2023] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Globally, there is an increase in the prevalence of metabolic illnesses, including diabetes mellitus. However, current therapies for diabetes and other metabolic illnesses are not well understood. Pharmacological treatment of type 2 diabetes is challenging, moreover, the majority of antidiabetic medications are incompatible with individuals who have cardiac disease, renal illness, or liver damage. Despite the ongoing development of innovative medicines, the quest for an optimal treatment that serves both as a hypoglycaemic agent and mitigates diabetes-related problems remains unattained. Recent research demonstrates that berberine has significant promise in the treatment of diabetes. Berberine influences glucose metabolism by enhancing insulin secretion, promoting glycolysis, decreasing adipogenesis, disrupting the function of the mitochondria, stimulating the 5' adenosine monophosphate-activated protein kinase (AMPK) pathway, thereby augmenting glucokinase activity. In this study, we virtually designed and synthesized 5 berberine derivatives (data not yet published) to study their impact on the AMP-activated protein kinase (AMPK) pathway through molecular docking and dynamic simulation study. Activation of AMPK plays an important role by enhancing glucose uptake in cells. Berberine and its derivatives showed potential for allosteric activation of the AMPK pathway. The allosteric activation of AMPK α- & β-subunit involves complex interactions with standard activators like A-769662. Berberine and its derivatives showed potential binding affinity at the allosteric site of AMPK α- & β-subunit, forming similar interactions to A-769662. Molecular dynamic simulations indicated stability of these complexes. However, interactions of these derivatives with the AMPK γ-subunit were less stable, suggesting limited potential for allosteric activation at this site. Further studies are required to assess the long-term stability and efficacy of berberine and its derivatives as allosteric AMPK activators. Additionally, ADMET predictions suggest these derivatives to be safe, warranting further experimental and preclinical investigations as potential antidiabetic agents. Supplementary Information The online version contains supplementary material available at 10.1007/s40203-024-00295-0.
Collapse
Affiliation(s)
- Bibhuti Bhusan Kakoti
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam 786004 India
| | - James H. Zothantluanga
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam 786004 India
| | - Kangkan Deka
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam 786004 India
- NETES Institute of Pharmaceutical Science, NEMCARE Group of Institutions, Mirza, Kamrup, Guwahati, Assam 781125 India
| | - Raj Kumar Halder
- Ruhvenile Biomedical, Plot-8 OCF Pocket Institution, Sarita Vihar, Delhi, 110076 India
| | - Dhritiman Roy
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam 786004 India
| |
Collapse
|
2
|
Liew KB, Koh EV, Kong XE, Ismail NA, Abu Bakar RA, Kee PE, Khalid SH, Phang HC. Recent Advancements in Nanopharmaceuticals for Novel Drug Delivery Systems. Pharm Nanotechnol 2025; 13:271-286. [PMID: 39323341 DOI: 10.2174/0122117385324246240826042254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 06/26/2024] [Accepted: 07/04/2024] [Indexed: 09/27/2024]
Abstract
Nanoparticles have found applications across diverse sectors, including agriculture, food, cosmetics, chemicals, mechanical engineering, automotive, and oil and gas industries. In the medical field, nanoparticles have garnered considerable attention due to their great surface area, high solubility, rapid dissolution, and enhanced bioavailability. Nanopharmaceuticals are specifically designed to precisely deliver drug substances to targeted tissues and cells, aiming to optimize therapeutic efficacy while minimizing potential adverse effects. Furthermore, nanopharmaceuticals offer advantages, such as expedited therapeutic onset, reduced dosages, minimized variability between fed and fasted states, and enhanced patient compliance. The increasing interest in nanopharmaceuticals research among scientists and industry stakeholders highlights their potential for various medical applications from disease management to cancer treatment. This review examines the distinctive characteristics of ideal nanoparticles for efficient drug delivery, explores the current types of nanoparticles utilized in medicine, and delves into the applications of nanopharmaceuticals, including drug and gene delivery, as well as transdermal drug administration. This review provides insights into the nanopharmaceuticals field, contributing to the development of novel drug delivery systems and enhancing the potential of nanotechnology in healthcare.
Collapse
Affiliation(s)
- Kai Bin Liew
- Faculty of Pharmacy, University of Cyberjaya, Persiaran Bestari, Cyberjaya,63000, Selangor, Malaysia
| | - Ee Va Koh
- Faculty of Pharmacy, University of Cyberjaya, Persiaran Bestari, Cyberjaya,63000, Selangor, Malaysia
| | - Xue Er Kong
- Faculty of Pharmacy, University of Cyberjaya, Persiaran Bestari, Cyberjaya,63000, Selangor, Malaysia
| | - Nurdina Aleyah Ismail
- Faculty of Pharmacy, University of Cyberjaya, Persiaran Bestari, Cyberjaya,63000, Selangor, Malaysia
| | | | - Phei Er Kee
- Biorefinery and Bioprocessing Engineering Laboratory, Department of Chemical Engineering and Materials Science, Yuan Ze University, Chungli, Taoyuan, 320, Taiwan
| | - Syed Haroon Khalid
- Department of Pharmaceutics, Faculty of Pharmacy, Universiti Teknologi Mara, Puncak Alam, Selangor, 42300, Malaysia
| | - Hiu Ching Phang
- Faculty of Pharmacy, University of Cyberjaya, Persiaran Bestari, Cyberjaya,63000, Selangor, Malaysia
| |
Collapse
|
3
|
Zarrinnahad H, Dehdast SA, Fard GC, Nourbakhsh M, Koohi MK, Panahi G, Karimpour A, Rezayat SM, Shabani M. The effect of biosynthesized zinc oxide nanoparticles on gene expression and apoptosis in triple-negative breast cancer cells. Daru 2024; 33:10. [PMID: 39731629 PMCID: PMC11682029 DOI: 10.1007/s40199-024-00553-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 10/25/2024] [Indexed: 12/30/2024] Open
Abstract
OBJECTIVE(S) Some forms of breast cancer such as triple-negative phenotype, are serious challenge because of high metastatic cases, high mortality and resistance to conventional therapy motivated the search for alternative treatment approaches. Nanomaterials are promising candidates and suitable alternatives for improving tumor and cancer cell treatments. MATERIALS AND METHODS Biosynthesis of ZnO NPs by help of Berberis integerrima fruit extract, has been done. Analysis of Zinc Oxide NPs using DLS, FTIR, SEM, and EDS techniques have been performed. Moreover, biological activities of ZnO NPs evaluated through MTT method, Flow cytometry, and real time PCR methods. Biocatalytic and apoptotic activity of ZnO NPs on healthy HFF (human fibroblast cell line), MDA-MB 231, and MDA-MB 468 (triple negative breast cancer cell lines, (TNBC)) evaluated. Furthermore, Bax, Bcl-2 and caspase-3 apoptotic genes expression changes in cancer cells assessed in compare to GAPDH as a house keeping gene. RESULTS Physico-chemical investigation demonstrated ZnO NPs were confirmed by Berberis integerrima fruit extract for the first time. The MTT assay and Flow cytometry results indicated biocompatibility of the ZnO NPs in normal cell line and high anticancer potential against TNBC MDA-MB-231 and MDA-MB-468 cell lines. The IC50 of ZnO NPs were 104.4 and 44.86, 20.96 after 24 hours for HFF, MDA-MB-231 and MDA-MB-468 cells, respectively. CONCLUSION The current research showed a fast, cost effective and ecofriendly method for ZnO NPs nanoparticle synthesis. Furthermore, In vitro data analysis demonstrated biocompatibility and highly anticancer effects of biosynthesized ZnO NPs against TNBC cancerous cells.
Collapse
Affiliation(s)
- Hannaneh Zarrinnahad
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - S Ahmad Dehdast
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ghazaleh Chizari Fard
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Clothing and Fabric Design Department, Art Faculty, Imam Javad University College, Yazd, Iran
| | - Mitra Nourbakhsh
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Kazem Koohi
- Department of Comparative Biosciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Ghodratollah Panahi
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amin Karimpour
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - S Mehdi Rezayat
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mohammad Shabani
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Bennani I, Cherif Chefchaouni A, Hafidi Y, Moukafih B, El Marrakchi S, Bandadi FZ, Rahali Y, El Kartouti A. Advancements in the use of nanopharmaceuticals for cancer treatment. J Oncol Pharm Pract 2024; 30:1078-1083. [PMID: 38706188 DOI: 10.1177/10781552241251757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2024]
Abstract
OBJECTIVE Advances in nanotechnology make it possible to specifically target therapies to cancer cells and neoplasms, guide the surgical resection of tumors, and optimize the effectiveness of radiological treatments. This research article provides a concise synthesis of current knowledge in the field of galenic pharmacy focused on targeted drug delivery in oncology. This research article synthesizes current knowledge in galenic pharmacy, focusing on targeted drug delivery in oncology and reviewing recent advancements in nanopharmaceuticals for cancer treatment. DATA SOURCE The data for this review are derived from a comprehensive analysis of the most cited scientific literature (Pubmed). Recent studies, clinical trials, and technological breakthroughs related to nanopharmaceuticals have been rigorously examined. This diverse source ensures a comprehensive representation of the latest developments in the field. SUMMARY OF DATA The results highlight the emergence of nanopharmaceuticals as a promising approach to cancer treatment. The most common in oncology remain liposomes, nanopolymers, and nanocrystals. From a galenic point of view, these three forms offer a wide range of improvements compared to conventional forms such as improvement in solubility as well as stability. The same observation is in the clinic where treatment response rates are significantly improved. The most advantageous form will depend on the specific characteristics of each patient and each type of cancer. The precise design of nanocarriers allows for targeted drug delivery, enhancing therapeutic efficacy while reducing side effects. Concrete examples of clinical applications are presented, illustrating the practical potential of these advancements. CONCLUSION In conclusion, this review provides a holistic overview of recent developments in galenic pharmacy for targeted drug delivery in oncology. The stability of nanocarriers is a crucial challenge because it conditions the effectiveness and safety of the drugs transported. Environmental and biological variations encountered in the body can compromise this stability, jeopardizing the therapeutic effectiveness and safety of treatments. Likewise, personalized approaches are essential to address interindividual variations in treatment response, as well as patients' pharmacogenomic profiles, in order to optimize therapeutic effectiveness and minimize adverse effects.
Collapse
Affiliation(s)
- Ismail Bennani
- Department of Pharmacy, Faculty of Medicine, Pharmacy, and Dental Medicine of Fez, Sidi Mohamed Ben Abdellah University, Fez, Morocco
- Department of Pharmacy, Hassan II University Hospital of Fez, Fez, Morocco
| | - Ali Cherif Chefchaouni
- Team of Formulation and Quality Control of Health Products, Faculty of Medicine and Pharmacy, Mohammed V University of Rabat, Rabat, Morocco
| | - Youssef Hafidi
- Department of Pharmacy, Hassan II University Hospital of Fez, Fez, Morocco
| | - Badreddine Moukafih
- Department of Pharmacy, Faculty of Medicine, Pharmacy, and Dental Medicine of Fez, Sidi Mohamed Ben Abdellah University, Fez, Morocco
- Department of Pharmacy, Hassan II University Hospital of Fez, Fez, Morocco
| | - Soufiane El Marrakchi
- Department of Pharmacy, Faculty of Medicine, Pharmacy, and Dental Medicine of Fez, Sidi Mohamed Ben Abdellah University, Fez, Morocco
- Department of Pharmacy, Hassan II University Hospital of Fez, Fez, Morocco
| | - Fatima-Zahra Bandadi
- Department of Pharmacy, Faculty of Medicine, Pharmacy, and Dental Medicine of Fez, Sidi Mohamed Ben Abdellah University, Fez, Morocco
- Department of Pharmacy, Hassan II University Hospital of Fez, Fez, Morocco
| | - Younes Rahali
- Team of Formulation and Quality Control of Health Products, Faculty of Medicine and Pharmacy, Mohammed V University of Rabat, Rabat, Morocco
| | - Abdeslam El Kartouti
- Department of Pharmacy, Faculty of Medicine, Pharmacy, and Dental Medicine of Fez, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| |
Collapse
|
5
|
Gomte SS, Agnihotri TG, Khopade S, Jain A. Exploring the potential of pH-sensitive polymers in targeted drug delivery. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2024; 35:228-268. [PMID: 37927045 DOI: 10.1080/09205063.2023.2279792] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 11/01/2023] [Indexed: 11/07/2023]
Abstract
The pH-sensitive polymers have attained significant attention in the arena of targeted drug delivery (TDD) because of their exceptional capability to respond to alteration in pH in various physiological environments. This attribute aids pH-sensitive polymers to act as smart carriers for therapeutic agents, transporting them precisely to target locations while curtailing the release of drugs in off-targeted sites, thereby diminishing side effects. Many pH-responsive polymers in TDD have revealed promising results, with increased therapeutic efficacy and decreased toxic effects. Several pH-sensitive polymers, including, hydroxy-propyl-methyl cellulose, poly (methacrylic acid) (Eudragit series), poly (acrylic acid), and chitosan, have been broadly studied for their myriad applications in the management of various types of diseases. Additionally, the amalgamation of pH-sensitive polymers with, additive manufacturing techniques like 3D printing, has resulted in the progression of novel drug delivery systems that regulate drug release in a controlled manner. Herein, types of pH-sensitive polymers in TDD are systemically reviewed. We have briefly discussed the nanocarriers employed for the delivery of various pH-sensitive polymers in TDD. Finally, miscellaneous applications of pH-sensitive polymers are discussed thoroughly with special attention to the implication of 3D printing in pH-sensitive polymers.
Collapse
Affiliation(s)
- Shyam Sudhakar Gomte
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Palaj, Gandhinagar, Gujarat, India
| | - Tejas Girish Agnihotri
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Palaj, Gandhinagar, Gujarat, India
| | - Shivani Khopade
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Palaj, Gandhinagar, Gujarat, India
| | - Aakanchha Jain
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Palaj, Gandhinagar, Gujarat, India
| |
Collapse
|
6
|
Ramos INDF, da Silva MF, Lopes JMS, Cruz JN, Alves FS, do Rego JDAR, Costa MLD, Assumpção PPD, Barros Brasil DDS, Khayat AS. Extraction, Characterization, and Evaluation of the Cytotoxic Activity of Piperine in Its Isolated form and in Combination with Chemotherapeutics against Gastric Cancer. Molecules 2023; 28:5587. [PMID: 37513459 PMCID: PMC10385350 DOI: 10.3390/molecules28145587] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 06/30/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
Gastric cancer is one of the most frequent types of neoplasms worldwide, usually presenting as aggressive and difficult-to-manage tumors. The search for new structures with anticancer potential encompasses a vast research field in which natural products arise as promising alternatives. In this scenario, piperine, an alkaloid of the Piper species, has received attention due to its biological activity, including anticancer attributes. The present work proposes three heating-independent, reliable, low-cost, and selective methods for obtaining piperine from Piper nigrum L. (Black pepper). Electronic (SEM) and optical microscopies, X-ray diffraction, nuclear magnetic resonance spectroscopies (13C and 1H NMR), and optical spectroscopies (UV-Vis, photoluminescence, and FTIR) confirm the obtention of piperine crystals. The MTT assay reveals that the piperine samples exhibit good cytotoxic activity against primary and metastasis models of gastric cancer cell lines from the Brazilian Amazon. The samples showed selective cytotoxicity on the evaluated models, revealing higher effectiveness in cells bearing a higher degree of aggressiveness. Moreover, the investigated piperine crystals demonstrated the ability to act as a good cytotoxicity enhancer when combined with traditional chemotherapeutics (5-FU and GEM), allowing the drugs to achieve the same cytotoxic effect in cells employing lower concentrations. These results establish piperine as a promising molecule for therapy investigations in aggressive gastric cancer, both in its isolated form or as a bioenhancer.
Collapse
Affiliation(s)
| | | | | | - Jordy Neves Cruz
- Institute of Technology, Federal University of Pará, Belém 66075-110, PA, Brazil
| | - Fabrine Silva Alves
- Graduate Program in Pharmaceutical Innovation, Federal University of Pará, Belém 66075-110, PA, Brazil
| | | | | | | | - Davi do Socorro Barros Brasil
- Institute of Technology, Federal University of Pará, Belém 66075-110, PA, Brazil
- Graduate Program in Pharmaceutical Innovation, Federal University of Pará, Belém 66075-110, PA, Brazil
- Graduate Program in Science and Environment, Federal University of Pará, Belém 66075-110, PA, Brazil
| | - André Salim Khayat
- Oncology Research Center, Federal University of Pará, Belém 66075-110, PA, Brazil
- Institute of Biological Science, Federal University of Pará, Belém 66075-110, PA, Brazil
| |
Collapse
|
7
|
Alfonso-Triguero P, Lorenzo J, Candiota AP, Arús C, Ruiz-Molina D, Novio F. Platinum-Based Nanoformulations for Glioblastoma Treatment: The Resurgence of Platinum Drugs? NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:1619. [PMID: 37242036 PMCID: PMC10223043 DOI: 10.3390/nano13101619] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/06/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023]
Abstract
Current therapies for treating Glioblastoma (GB), and brain tumours in general, are inefficient and represent numerous challenges. In addition to surgical resection, chemotherapy and radiotherapy are presently used as standards of care. However, treated patients still face a dismal prognosis with a median survival below 15-18 months. Temozolomide (TMZ) is the main chemotherapeutic agent administered; however, intrinsic or acquired resistance to TMZ contributes to the limited efficacy of this drug. To circumvent the current drawbacks in GB treatment, a large number of classical and non-classical platinum complexes have been prepared and tested for anticancer activity, especially platinum (IV)-based prodrugs. Platinum complexes, used as alkylating agents in the anticancer chemotherapy of some malignancies, are though often associated with severe systemic toxicity (i.e., neurotoxicity), especially after long-term treatments. The objective of the current developments is to produce novel nanoformulations with improved lipophilicity and passive diffusion, promoting intracellular accumulation, while reducing toxicity and optimizing the concomitant treatment of chemo-/radiotherapy. Moreover, the blood-brain barrier (BBB) prevents the access of the drugs to the brain and accumulation in tumour cells, so it represents a key challenge for GB management. The development of novel nanomedicines with the ability to (i) encapsulate Pt-based drugs and pro-drugs, (ii) cross the BBB, and (iii) specifically target cancer cells represents a promising approach to increase the therapeutic effect of the anticancer drugs and reduce undesired side effects. In this review, a critical discussion is presented concerning different families of nanoparticles able to encapsulate platinum anticancer drugs and their application for GB treatment, emphasizing their potential for increasing the effectiveness of platinum-based drugs.
Collapse
Affiliation(s)
- Paula Alfonso-Triguero
- Institut de Biotecnologia i de Biomedicina, Departament de Bioquimica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain; (P.A.-T.); (J.L.); (A.P.C.); (C.A.)
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Campus UAB, Bellaterra, 08193 Barcelona, Spain;
| | - Julia Lorenzo
- Institut de Biotecnologia i de Biomedicina, Departament de Bioquimica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain; (P.A.-T.); (J.L.); (A.P.C.); (C.A.)
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
| | - Ana Paula Candiota
- Institut de Biotecnologia i de Biomedicina, Departament de Bioquimica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain; (P.A.-T.); (J.L.); (A.P.C.); (C.A.)
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
- Centro de Investigación Biomédica en Red, Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 08193 Cerdanyola del Vallès, Spain
| | - Carles Arús
- Institut de Biotecnologia i de Biomedicina, Departament de Bioquimica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain; (P.A.-T.); (J.L.); (A.P.C.); (C.A.)
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
- Centro de Investigación Biomédica en Red, Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 08193 Cerdanyola del Vallès, Spain
| | - Daniel Ruiz-Molina
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Campus UAB, Bellaterra, 08193 Barcelona, Spain;
| | - Fernando Novio
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Campus UAB, Bellaterra, 08193 Barcelona, Spain;
- Departament de Química, Universitat Autònoma de Barcelona (UAB), Campus UAB, 08193 Cerdanyola del Vallès, Spain
| |
Collapse
|
8
|
Mitrevska K, Cernei N, Michalkova H, Rodrigo MAM, Sivak L, Heger Z, Zitka O, Kopel P, Adam V, Milosavljevic V. Platinum-based drug-induced depletion of amino acids in the kidneys and liver. Front Oncol 2022; 12:986045. [PMID: 36212465 PMCID: PMC9535364 DOI: 10.3389/fonc.2022.986045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Accepted: 08/31/2022] [Indexed: 11/13/2022] Open
Abstract
Cisplatin (cis-diamminedichloroplatinum II; CDDP) is a widely used cytostatic agent; however, it tends to promote kidney and liver disease, which are a major signs of drug-induced toxicity. Platinum compounds are often presented as alternative therapeutics and subsequently easily dispersed in the environment as contaminants. Due to the major roles of the liver and kidneys in removing toxic materials from the human body, we performed a comparative study of the amino acid profiles in chicken liver and kidneys before and after the application of CDDP and platinum nanoparticles (PtNPs-10 and PtNPs-40). The treatment of the liver with the selected drugs affected different amino acids; however, Leu and Arg were decreased after all treatments. The treatment of the kidneys with CDDP mostly affected Val; PtNPs-10 decreased Val, Ile and Thr; and PtNPs-40 affected only Pro. In addition, we tested the same drugs on two healthy cell lines, HaCaT and HEK-293, and ultimately explored the amino acid profiles in relation to the tricarboxylic acid cycle (TCA) and methionine cycle, which revealed that in both cell lines, there was a general increase in amino acid concentrations associated with changes in the concentrations of the metabolites of these cycles.
Collapse
Affiliation(s)
- Katerina Mitrevska
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, Czechia
| | - Natalia Cernei
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, Czechia
| | - Hana Michalkova
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, Czechia
- Central European Institute of Technology, Brno University of Technology, Brno, Czechia
| | | | - Ladislav Sivak
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, Czechia
| | - Zbynek Heger
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, Czechia
- Central European Institute of Technology, Brno University of Technology, Brno, Czechia
| | - Ondrej Zitka
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, Czechia
- Central European Institute of Technology, Brno University of Technology, Brno, Czechia
| | - Pavel Kopel
- Department of Inorganic Chemistry, Faculty of Science, Palacky University, Olomouc, Czechia
| | - Vojtech Adam
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, Czechia
- Central European Institute of Technology, Brno University of Technology, Brno, Czechia
| | - Vedran Milosavljevic
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, Czechia
- Central European Institute of Technology, Brno University of Technology, Brno, Czechia
- *Correspondence: Vedran Milosavljevic,
| |
Collapse
|
9
|
Approaches to Improve Macromolecule and Nanoparticle Accumulation in the Tumor Microenvironment by the Enhanced Permeability and Retention Effect. Polymers (Basel) 2022; 14:polym14132601. [PMID: 35808648 PMCID: PMC9268820 DOI: 10.3390/polym14132601] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/17/2022] [Accepted: 06/18/2022] [Indexed: 12/17/2022] Open
Abstract
Passive targeting is the foremost mechanism by which nanocarriers and drug-bearing macromolecules deliver their payload selectively to solid tumors. An important driver of passive targeting is the enhanced permeability and retention (EPR) effect, which is the cornerstone of most carrier-based tumor-targeted drug delivery efforts. Despite the huge number of publications showcasing successes in preclinical animal models, translation to the clinic has been poor, with only a few nano-based drugs currently being used for the treatment of cancers. Several barriers and factors have been adduced for the low delivery efficiency to solid tumors and poor clinical translation, including the characteristics of the nanocarriers and macromolecules, vascular and physiological barriers, the heterogeneity of tumor blood supply which affects the homogenous distribution of nanocarriers within tumors, and the transport and penetration depth of macromolecules and nanoparticles in the tumor matrix. To address the challenges associated with poor tumor targeting and therapeutic efficacy in humans, the identified barriers that affect the efficiency of the enhanced permeability and retention (EPR) effect for macromolecular therapeutics and nanoparticle delivery systems need to be overcome. In this review, approaches to facilitate improved EPR delivery outcomes and the clinical translation of novel macromolecular therapeutics and nanoparticle drug delivery systems are discussed.
Collapse
|
10
|
Gabano E, Ferraris C, Osella D, Battaglia LS, Ravera M. Formulations of highly antiproliferative hydrophobic Pt(IV) complexes into lipidic nanoemulsions as delivery vehicles. Inorganica Chim Acta 2022. [DOI: 10.1016/j.ica.2022.120859] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
11
|
The development and progress of nanomedicine for esophageal cancer diagnosis and treatment. Semin Cancer Biol 2022; 86:873-885. [DOI: 10.1016/j.semcancer.2022.01.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/22/2021] [Accepted: 01/20/2022] [Indexed: 02/07/2023]
|
12
|
Pérez Quiñones J, Roschger C, Zierer A, Peniche-Covas C, Brüggemann O. Self-Assembled Silk Fibroin-Based Aggregates for Delivery of Camptothecin. Polymers (Basel) 2021; 13:polym13213804. [PMID: 34771362 PMCID: PMC8587969 DOI: 10.3390/polym13213804] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 10/27/2021] [Accepted: 10/28/2021] [Indexed: 12/24/2022] Open
Abstract
A water-soluble hydrolysate of silk fibroin (SF) (~30 kDa) was esterified with tocopherol, ergocalciferol, and testosterone to form SF aggregates for the controlled delivery of the anticancer drug camptothecin (CPT). Elemental analysis and 1H NMR spectroscopy showed a degree of substitution (DS) on SF of 0.4 to 3.8 mol %. Yields of 58 to 71% on vitamins- and testosterone-grafted SF conjugates were achieved. CPT was efficiently incorporated into the lipophilic core of SF aggregates using a dialysis-precipitation method, achieving drug contents of 6.3-8.5 wt %. FTIR spectra and DSC thermograms showed that tocopherol- and testosterone-grafted SF conjugates predominantly adopted a β-sheet conformation. After the esterification of tyrosine residues on SF chains with the vitamin or testosterone, the hydrodynamic diameters almost doubled or tripled that of SF. The zeta potential values after esterification increased to about -30 mV, which favors the stability of aggregates in aqueous medium. Controlled and almost quantitative release of CPT was achieved after 6 days in PBS at 37 °C, with almost linear release during the first 8 h. MCF-7 cancer cells exhibited good uptake of CPT-loaded SF aggregates after 6 h, causing cell death and cell cycle arrest in the G2/M phase. Substantial uptake of the CPT-loaded aggregates into MCF-7 spheroids was shown after 3 days. Furthermore, all CPT-loaded SF aggregates demonstrated superior toxicity to MCF-7 spheroids compared with parent CPT. Blank SF aggregates induced no hemolysis at pH 6.2 and 7.4, while CPT-loaded SF aggregates provoked hemolysis at pH 6.2 but not at pH 7.4. In contrast, parent CPT caused hemolysis at both pH tested. Therefore, CPT-loaded SF aggregates are promising candidates for chemotherapy.
Collapse
Affiliation(s)
- Javier Pérez Quiñones
- Institute of Polymer Chemistry, Johannes Kepler University Linz, Altenberger Straße 69, 4040 Linz, Austria;
- Correspondence: or ; Tel.: +43-670-4039820
| | - Cornelia Roschger
- Department for Cardiac-, Vascular- and Thoracic Surgery, Johannes Kepler University Linz, Kepler University Hospital GmBH, Altenberger Straße 69, 4040 Linz and Krankenhausstraße 7a, 4020 Linz, Austria; (C.R.); or (A.Z.)
| | - Andreas Zierer
- Department for Cardiac-, Vascular- and Thoracic Surgery, Johannes Kepler University Linz, Kepler University Hospital GmBH, Altenberger Straße 69, 4040 Linz and Krankenhausstraße 7a, 4020 Linz, Austria; (C.R.); or (A.Z.)
| | - Carlos Peniche-Covas
- Facultad de Química, Universidad de La Habana, Zapata S/N entre G y Carlitos Aguirre, La Habana 10400, Cuba;
| | - Oliver Brüggemann
- Institute of Polymer Chemistry, Johannes Kepler University Linz, Altenberger Straße 69, 4040 Linz, Austria;
| |
Collapse
|
13
|
Sharma A, Sah N, Kannan S, Kannan RM. Targeted drug delivery for maternal and perinatal health: Challenges and opportunities. Adv Drug Deliv Rev 2021; 177:113950. [PMID: 34454979 PMCID: PMC8544131 DOI: 10.1016/j.addr.2021.113950] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 08/06/2021] [Accepted: 08/24/2021] [Indexed: 12/18/2022]
Abstract
Pre-existing conditions at reproductive age, and complications arising during pregnancy can be detrimental to maternal and fetal health. Current therapies to combat obstetric disorders are limited due to the inherent complexity of pregnancy, and can have harmful effects on developing fetus. Emerging research shows intricate signaling between the cells from mother and fetus at maternal-fetal interface, providing unique opportunities for interventions specifically targeted to the mother, fetus, or placenta. Advancements in nanotechnology, stem-cell biology and gene therapy have resulted in target-specific treatments with promising results in pre-clinical maternal and fetal disorder models. Comprehensive understanding of the effect of physicochemical properties of delivery systems on their uptake, retention and accumulation across placenta will help in the better diagnosis and treatment of perinatal disorders. This review describes the factors leading to obstetric complications along with their effect on pregnancy outcomes, and discusses key targeted therapeutic strategies for addressing conditions related to maternal and fetal health.
Collapse
Affiliation(s)
- Anjali Sharma
- Center for Nanomedicine, Department of Ophthalmology, Wilmer Eye Institute Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Nirnath Sah
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Sujatha Kannan
- Center for Nanomedicine, Department of Ophthalmology, Wilmer Eye Institute Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Hugo W. Moser Research Institute at Kennedy Krieger, Inc., Baltimore MD, 21205, USA
| | - Rangaramanujam M Kannan
- Center for Nanomedicine, Department of Ophthalmology, Wilmer Eye Institute Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Hugo W. Moser Research Institute at Kennedy Krieger, Inc., Baltimore MD, 21205, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore MD, 21218, USA.
| |
Collapse
|
14
|
Singh V, Kesharwani P. Dendrimer as a promising nanocarrier for the delivery of doxorubicin as an anticancer therapeutics. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2021; 32:1882-1909. [PMID: 34078252 DOI: 10.1080/09205063.2021.1938859] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Dendrimers are macromolecules with high-polymeric branching capable of undergoing major modifications. These characteristics make them an efficient nanocarrier capable of encapsulating and delivering drug, antibodies, or any therapeutic gene. The failure of conventional techniques to deliver drug with higher efficacy and reduced side effects has led to the use of nanomedicines including dendrimers. Dendrimers are novel drug carriers that are modified, complexed, and conjugated with different ligands and receptors to target the delivery of drug at the specific site without impacting any of the normal cells in surrounding. Moreover, the biocompatibility and safety of the dendrimers can be altered accordingly by the process of functionalization by PEGylation, acetylation, or amination. Various dendrimers have been designed to incorporate and deliver anticancer drug either in free form or as codelivery in conjugation with other drugs or therapeutic siRNA/DNA. Doxorubicin (DOX) is one such chemotherapeutic drug that acts by disrupting the process of DNA repair in tumor cells and hence is, since long been used for anticancer therapy. Certain adverse effects such as cardiotoxicity has limited the use of conventional DOX and has shifted the focus on use of safe nanodelivery systems viz dendrimers. DOX either in free or salt form can be loaded or encapsulated accordingly within the core of the dendrimers and linked with different receptors expressed over tumor cells to improve targeting in any cancerous organ site. Positive results obtained after cytotoxicity assay and in vivo/in vitro studies on different cancerous cell lines, and grafted models suggested the potential use of multifunctional DOX-dendrimers characterized with controlled release, better penetration, improved bioavailability, and reduced organ toxicity. This review consolidates studies on different types of DOX-loaded dendrimers that were synthesized, investigated, and are currently being explored for better cancer targeting. Foreseeing the prospects of dendrimers and their compatibility with DOX (free/salt), the article was updated with all current insights.
Collapse
Affiliation(s)
- Vanshikha Singh
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| |
Collapse
|
15
|
Jeon J, Lee S, Kim H, Kang H, Youn H, Jo S, Youn B, Kim HY. Revisiting Platinum-Based Anticancer Drugs to Overcome Gliomas. Int J Mol Sci 2021; 22:ijms22105111. [PMID: 34065991 PMCID: PMC8151298 DOI: 10.3390/ijms22105111] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/08/2021] [Accepted: 05/10/2021] [Indexed: 12/12/2022] Open
Abstract
Although there are many patients with brain tumors worldwide, there are numerous difficulties in overcoming brain tumors. Among brain tumors, glioblastoma, with a 5-year survival rate of 5.1%, is the most malignant. In addition to surgical operations, chemotherapy and radiotherapy are generally performed, but the patients have very limited options. Temozolomide is the most commonly prescribed drug for patients with glioblastoma. However, it is difficult to completely remove the tumor with this drug alone. Therefore, it is necessary to discuss the potential of anticancer drugs, other than temozolomide, against glioblastomas. Since the discovery of cisplatin, platinum-based drugs have become one of the leading chemotherapeutic drugs. Although many studies have reported the efficacy of platinum-based anticancer drugs against various carcinomas, studies on their effectiveness against brain tumors are insufficient. In this review, we elucidated the anticancer effects and advantages of platinum-based drugs used in brain tumors. In addition, the cases and limitations of the clinical application of platinum-based drugs are summarized. As a solution to overcome these obstacles, we emphasized the potential of a novel approach to increase the effectiveness of platinum-based drugs.
Collapse
Affiliation(s)
- Jaewan Jeon
- Department of Radiation Oncology, Haeundae Paik Hospital, Inje University School of Medicine, Busan 48108, Korea; (J.J.); (S.J.)
| | - Sungmin Lee
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea; (S.L.); (H.K.); (H.K.)
| | - Hyunwoo Kim
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea; (S.L.); (H.K.); (H.K.)
| | - Hyunkoo Kang
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea; (S.L.); (H.K.); (H.K.)
| | - HyeSook Youn
- Department of Integrative Bioscience and Biotechnology, Sejong University, Seoul 05006, Korea;
| | - Sunmi Jo
- Department of Radiation Oncology, Haeundae Paik Hospital, Inje University School of Medicine, Busan 48108, Korea; (J.J.); (S.J.)
| | - BuHyun Youn
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea; (S.L.); (H.K.); (H.K.)
- Department of Biological Sciences, Pusan National University, Busan 46241, Korea
- Correspondence: (B.Y.); (H.Y.K.); Tel.: +82-51-510-2264 (B.Y.); +82-51-797-3923 (H.Y.K.)
| | - Hae Yu Kim
- Department of Neurosurgery, Haeundae Paik Hospital, Inje University School of Medicine, Busan 48108, Korea
- Correspondence: (B.Y.); (H.Y.K.); Tel.: +82-51-510-2264 (B.Y.); +82-51-797-3923 (H.Y.K.)
| |
Collapse
|
16
|
Varahachalam SP, Lahooti B, Chamaneh M, Bagchi S, Chhibber T, Morris K, Bolanos JF, Kim NY, Kaushik A. Nanomedicine for the SARS-CoV-2: State-of-the-Art and Future Prospects. Int J Nanomedicine 2021; 16:539-560. [PMID: 33519200 PMCID: PMC7837559 DOI: 10.2147/ijn.s283686] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 12/25/2020] [Indexed: 01/08/2023] Open
Abstract
The newly emerged ribonucleic acid (RNA) enveloped human beta-coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection caused the COVID-19 pandemic, severely affects the respiratory system, and may lead to death. Lacking effective diagnostics and therapies made this pandemic challenging to manage since the SARS-CoV-2 transmits via human-to-human, enters via ACE2 and TMPSSR2 receptors, and damages organs rich in host cells, spreads via symptomatic carriers and is prominent in an immune-compromised population. New SARS-CoV-2 informatics (structure, strains, like-cycles, functional sites) motivated bio-pharma experts to investigate novel therapeutic agents that act to recognize, inhibit, and knockdown combinations of drugs, vaccines, and antibodies, have been optimized to manage COVID-19. However, successful targeted delivery of these agents to avoid off-targeting and unnecessary drug ingestion is very challenging. To overcome these obstacles, this mini-review projects nanomedicine technology, a pharmacologically relevant cargo of size within 10 to 200 nm, for site-specific delivery of a therapeutic agent to recognize and eradicate the SARS-CoV-2, and improving the human immune system. Such combinational therapy based on compartmentalization controls the delivery and releases of a drug optimized based on patient genomic profile and medical history. Nanotechnology could help combat COVID-19 via various methods such as avoiding viral contamination and spraying by developing personal protective equipment (PPE) to increase the protection of healthcare workers and produce effective antiviral disinfectants surface coatings capable of inactivating and preventing the virus from spreading. To quickly recognize the infection or immunological response, design highly accurate and sensitive nano-based sensors. Development of new drugs with improved activity, reduced toxicity, and sustained release to the lungs, as well as tissue targets; and development of nano-based immunizations to improve humoral and cellular immune responses. The desired and controlled features of suggested personalized therapeutics, nanomedicine, is a potential therapy to manage COVID-19 successfully. The state-of-the-art nanomedicine, challenges, and prospects of nanomedicine are carefully and critically discussed in this report, which may serve as a key platform for scholars to investigate the role of nanomedicine for higher efficacy to manage the COVID-19 pandemic.
Collapse
Affiliation(s)
- Sree Pooja Varahachalam
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center (TTUHSC), Amarillo, TX79106, USA
| | - Behnaz Lahooti
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center (TTUHSC), Amarillo, TX79106, USA
| | - Masoumeh Chamaneh
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center (TTUHSC), Amarillo, TX79106, USA
| | - Sounak Bagchi
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center (TTUHSC), Amarillo, TX79106, USA
| | - Tanya Chhibber
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center (TTUHSC), Amarillo, TX79106, USA
| | - Kevin Morris
- Maharashtra University of Health Sciences (MUHS), Nashik, Maharashtra422004, India
| | - Joe F Bolanos
- Facultad De Ciencias De La Salud “Dr.Luis Edmundo Vasquez” Santa Tecla, Universidad Dr. Jose Matias Delgado, Cd Merliot, El Salvador
| | - Nam-Young Kim
- RFIC Bio Center, Department of Electronics Engineering, Kwangwoon University, Seoul01897, South Korea
| | - Ajeet Kaushik
- NanoBioTech Laboratory, Department of Natural Sciences, Division of Sciences, Art, and Mathematics, Florida Polytechnic University, Lakeland, FL3385, USA
| |
Collapse
|
17
|
Targeting of radioactive platinum-bisphosphonate anticancer drugs to bone of high metabolic activity. Sci Rep 2020; 10:5889. [PMID: 32246003 PMCID: PMC7125202 DOI: 10.1038/s41598-020-62039-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 03/06/2020] [Indexed: 11/08/2022] Open
Abstract
Platinum-based chemotherapeutics exhibit excellent antitumor properties. However, these drugs cause severe side effects including toxicity, drug resistance, and lack of tumor selectivity. Tumor-targeted drug delivery has demonstrated great potential to overcome these drawbacks. Herein, we aimed to design radioactive bisphosphonate-functionalized platinum (195mPt-BP) complexes to confirm preferential accumulation of these Pt-based drugs in metabolically active bone. In vitro NMR studies revealed that release of Pt from Pt BP complexes increased with decreasing pH. Upon systemic administration to mice, Pt-BP exhibited a 4.5-fold higher affinity to bone compared to platinum complexes lacking the bone-seeking bisphosphonate moiety. These Pt-BP complexes formed less Pt-DNA adducts compared to bisphosphonate-free platinum complexes, indicating that in vivo release of Pt from Pt-BP complexes proceeded relatively slow. Subsequently, radioactive 195mPt-BP complexes were synthesized using 195mPt(NO3)2(en) as precursor and injected intravenously into mice. Specific accumulation of 195mPt-BP was observed at skeletal sites with high metabolic activity using micro-SPECT/CT imaging. Furthermore, laser ablation-ICP-MS imaging of proximal tibia sections confirmed that 195mPt BP co-localized with calcium in the trabeculae of mice tibia.
Collapse
|
18
|
Chen YJ, Tsai CY, Cheng YM, Nieh SW, Yeh TK, Chen CP, Wang MH, Chou LH, Chiu TY, Liu L, Ho C, Chen CT, Liu TW. Impacts of Intralipid on Nanodrug Abraxane Therapy and on the Innate Immune System. Sci Rep 2020; 10:2838. [PMID: 32071352 PMCID: PMC7028991 DOI: 10.1038/s41598-020-59813-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 02/04/2020] [Indexed: 12/09/2022] Open
Abstract
A major obstacle to nanodrugs-mediated cancer therapy is their rapid uptake by the reticuloendothelial system that decreases the systemic exposure of the nanodrugs to tumors and also increases toxicities. Intralipid has been shown to reduce nano-oxaliplatin-mediated toxicity while improving bioavailability. Here, we have found that Intralipid reduces the cytotoxicity of paclitaxel for human monocytic cells, but not for breast, lung, or pancreatic cancer cells. Intralipid also promotes the polarization of macrophages to the anti-cancer M1-like phenotype. Using a xenograft breast cancer mouse model, we have found that Intralipid pre-treatment significantly increases the amount of paclitaxel reaching the tumor and promotes tumor apoptosis. The combination of Intralipid with half the standard clinical dose of Abraxane reduces the tumor growth rate as effectively as the standard clinical dose. Our findings suggest that pre-treatment of Intralipid has the potential to be a powerful agent to enhance the tumor cytotoxic effects of Abraxane and to reduce its off-target toxicities.
Collapse
Affiliation(s)
- Yen-Ju Chen
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, County, Taiwan
| | - Chin-Yi Tsai
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, County, Taiwan
| | - Ying-Min Cheng
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, County, Taiwan
| | - Su-Wen Nieh
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, County, Taiwan
| | - Teng-Kuang Yeh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, County, Taiwan
| | - Ching- Ping Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, County, Taiwan
| | - Min-Hsien Wang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, County, Taiwan
| | - Ling-Hui Chou
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, County, Taiwan
| | - Tai-Yu Chiu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, County, Taiwan
| | - Li Liu
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Chien Ho
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, USA.
| | - Chiung- Tong Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, County, Taiwan.
| | - Tsang-Wu Liu
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, County, Taiwan.
| |
Collapse
|
19
|
Yang F, Medik Y, Li L, Tian X, Fu D, Brouwer KL, Wagner K, Sun B, Sendi H, Mi Y, Wang AZ. Nanoparticle Drug Delivery Can Reduce the Hepatotoxicity of Therapeutic Cargo. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e1906360. [PMID: 31972070 PMCID: PMC7873714 DOI: 10.1002/smll.201906360] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 12/30/2019] [Indexed: 05/09/2023]
Abstract
Hepatotoxicity is a key concern in the clinical translation of nanotherapeutics because preclinical studies have consistently shown that nanotherapeutics accumulates extensively in the liver. However, clinical-stage nanotherapeutics have not shown increased hepatotoxicity. Factors that can contribute to the hepatotoxicity of nanotherapeutics beyond the intrinsic hepatotoxicity of nanoparticles (NPs) are poorly understood. Because of this knowledge gap, clinical translation efforts have avoided hepatotoxic molecules. By examining the hepatotoxicity of nanoformulations of known hepatotoxic compounds, it is demonstrated that nanotherapeutics are associated with lower hepatotoxicity than their small-molecule counterparts. It is also found that the reduced hepatotoxicity is related to the uptake of nanotherapeutics by macrophages in the liver. These findings can facilitate further development and clinical translation of nanotherapeutics.
Collapse
Affiliation(s)
- Feifei Yang
- Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences & Peking Union Medical College, Haidian District, Beijing, 100193, P.R. China; Laboratory of Nano- and Translational Medicine, Lineberger Comprehensive Cancer Center, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA; Department of Radiation Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Yusra Medik
- Laboratory of Nano- and Translational Medicine, Lineberger Comprehensive Cancer Center, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA; Department of Radiation Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Liantao Li
- Laboratory of Nano- and Translational Medicine, Lineberger Comprehensive Cancer Center, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA; Department of Radiation Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Xi Tian
- Laboratory of Nano- and Translational Medicine, Lineberger Comprehensive Cancer Center, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA; Department of Radiation Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Dong Fu
- UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Kim L.R. Brouwer
- UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Kyle Wagner
- Laboratory of Nano- and Translational Medicine, Lineberger Comprehensive Cancer Center, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA; Department of Radiation Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Bo Sun
- Laboratory of Nano- and Translational Medicine, Lineberger Comprehensive Cancer Center, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA; Department of Radiation Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Hossein Sendi
- Laboratory of Nano- and Translational Medicine, Lineberger Comprehensive Cancer Center, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA; Department of Radiation Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Yu Mi
- Laboratory of Nano- and Translational Medicine, Lineberger Comprehensive Cancer Center, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA; Department of Radiation Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Andrew Z. Wang
- Laboratory of Nano- and Translational Medicine, Lineberger Comprehensive Cancer Center, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA; Department of Radiation Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| |
Collapse
|
20
|
He Y, Qin L, Huang Y, Ma C. Advances of Nano-Structured Extended-Release Local Anesthetics. NANOSCALE RESEARCH LETTERS 2020; 15:13. [PMID: 31950284 PMCID: PMC6965527 DOI: 10.1186/s11671-019-3241-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 12/26/2019] [Indexed: 05/08/2023]
Abstract
Extended-release local anesthetics (LAs) have drawn increasing attention with their promising role in improving analgesia and reducing adverse events of LAs. Nano-structured carriers such as liposomes and polymersomes optimally meet the demands of/for extended-release, and have been utilized in drug delivery over decades and showed satisfactory results with extended-release. Based on mature technology of liposomes, EXPAREL, the first approved liposomal LA loaded with bupivacaine, has seen its success in an extended-release form. At the same time, polymersomes has advances over liposomes with complementary profiles, which inspires the emergence of hybrid carriers. This article summarized the recent research successes on nano-structured extended-release LAs, of which liposomal and polymeric are mainstream systems. Furthermore, with continual optimization, drug delivery systems carry properties beyond simple transportation, such as specificity and responsiveness. In the near future, we may achieve targeted delivery and controlled-release properties to satisfy various analgesic requirements.
Collapse
Affiliation(s)
- Yumiao He
- Department of Anesthesiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, 100730, China
- Joint Laboratory of Anesthesia and Pain, Peking Union Medical College, Beijing, 100730, China
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
| | - Linan Qin
- Joint Laboratory of Anesthesia and Pain, Peking Union Medical College, Beijing, 100730, China
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
| | - Yuguang Huang
- Department of Anesthesiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, 100730, China.
- Joint Laboratory of Anesthesia and Pain, Peking Union Medical College, Beijing, 100730, China.
| | - Chao Ma
- Joint Laboratory of Anesthesia and Pain, Peking Union Medical College, Beijing, 100730, China.
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China.
| |
Collapse
|
21
|
Chen Q, Bao Y, Burner D, Kaushal S, Zhang Y, Mendoza T, Bouvet M, Ozkan C, Minev B, Ma W. Tumor growth inhibition by mSTEAP peptide nanovaccine inducing augmented CD8 + T cell immune responses. Drug Deliv Transl Res 2019; 9:1095-1105. [PMID: 31228097 DOI: 10.1007/s13346-019-00652-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Poly(lactic-co-glycolic) acid (PLGA) has been successfully used in drug delivery and biomaterial applications, but very little attention has been directed towards the potential in vivo effects of peptide-loaded PLGA nanoparticles (NPs), specifically the potency of intravenous (IV) STEAP peptide-loaded PLGA-NP (nanovaccine) dosing and whether STEAP-specific CD8+ T cells directly play a key role in tumor inhibition. To address these concerns, syngeneic prostate cancer mouse models were established and treated with either mSTEAP peptide emulsified in incomplete Freund's adjuvant (IFA) via subcutaneous (SC) injection or mSTEAP peptide nanovaccine containing the same amount of peptide via IV or SC injection. Meanwhile, mice were treated with either CD8b mAb followed by nanovaccine treatment, free mSTEAP peptide, or empty PLGA-NPs. Immune responses in these mice were examined using cytotoxicity assays at 14 days after treatment. Tumor size and survival in various treatment groups were measured and monitored. The results demonstrated that mSTEAP peptide nanovaccine resulted in tumor inhibition by eliciting a significantly stronger CD8+ T cell immune response when compared with the controls. Moreover, the survival periods of mice treated with mSTEAP nanovaccine were significantly longer than those of mice treated with mSTEAP peptide emulsified in IFA or the treatment controls. Additionally, it was observed that the peptide nanovaccine was mainly distributed in the mouse liver and lungs after IV injection. These findings suggest that the peptide nanovaccine is a promising immunotherapeutic approach and offers a new opportunity for prostate cancer therapies.
Collapse
Affiliation(s)
- Qiuqiang Chen
- Key Laboratory for Translational Medicine, The First Affiliated Hospital of Huzhou University School of Medicine, Huzhou, 313000, Zhejiang, China
- Department of Clinical Medicine, Huzhou University School of Medicine, Huzhou, 313000, Zhejiang, China
| | - Ying Bao
- Key Laboratory for Translational Medicine, The First Affiliated Hospital of Huzhou University School of Medicine, Huzhou, 313000, Zhejiang, China
| | - Danielle Burner
- Department of Medicine and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Sharmeela Kaushal
- Department of Medicine and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Yu Zhang
- Materials Science and Engineering Program, Department of Mechanical Engineering, University of California Riverside, Riverside, CA, 92521, USA
- Mechanical and Automotive Engineering, School of Engineering, RMIT University, Melbourne, Victoria, 3083, Australia
| | - Theresa Mendoza
- Department of Medicine and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Michael Bouvet
- Department of Medicine and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Cengiz Ozkan
- Materials Science and Engineering Program, Department of Mechanical Engineering, University of California Riverside, Riverside, CA, 92521, USA
| | - Boris Minev
- Department of Medicine and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA.
- Calidi Biotherapeutics, San Diego, CA, 92121, USA.
| | - Wenxue Ma
- Department of Clinical Medicine, Huzhou University School of Medicine, Huzhou, 313000, Zhejiang, China.
- Department of Medicine and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
22
|
Sakurai Y, Harashima H. Hyaluronan-modified nanoparticles for tumor-targeting. Expert Opin Drug Deliv 2019; 16:915-936. [DOI: 10.1080/17425247.2019.1645115] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Yu Sakurai
- Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | | |
Collapse
|
23
|
In vivo anti-proliferative activity of silver nanoparticles against Pseudomonas aeruginosa in freshwater Labeo rohita. APPLIED NANOSCIENCE 2019. [DOI: 10.1007/s13204-019-01053-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
24
|
Farjadian F, Ghasemi A, Gohari O, Roointan A, Karimi M, Hamblin MR. Nanopharmaceuticals and nanomedicines currently on the market: challenges and opportunities. Nanomedicine (Lond) 2019; 14:93-126. [PMID: 30451076 PMCID: PMC6391637 DOI: 10.2217/nnm-2018-0120] [Citation(s) in RCA: 327] [Impact Index Per Article: 54.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 10/15/2018] [Indexed: 12/23/2022] Open
Abstract
There has been a revolution in nanotechnology and nanomedicine. Since 1980, there has been a remarkable increase in approved nano-based pharmaceutical products. These novel nano-based systems can either be therapeutic agents themselves, or else act as vehicles to carry different active pharmaceutical agents into specific parts of the body. Currently marketed nanostructures include nanocrystals, liposomes and lipid nanoparticles, PEGylated polymeric nanodrugs, other polymers, protein-based nanoparticles and metal-based nanoparticles. A range of issues must be addressed in the development of these nanostructures. Ethics, market size, possibility of market failure, costs and commercial development, are some topics which are on the table to be discussed. After passing all the ethical and biological assessments, and satisfying the investors as to future profitability, only a handful of these nanoformulations, successfully obtained marketing approval. We survey the range of nanomedicines that have received regulatory approval and are marketed. We discuss ethics, costs, commercial development and possible market failure. We estimate the global nanomedicine market size and future growth. Our goal is to summarize the different approved nanoformulations on the market, and briefly cover the challenges and future outlook.
Collapse
Affiliation(s)
- Fatemeh Farjadian
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz 71468-64685, Iran
| | - Amir Ghasemi
- Department of Materials Science & Engineering, Sharif University of Technology, Tehran 11365-9466, Iran
- Advances Nanobiotechnology & Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran 14496-4535, Iran
| | - Omid Gohari
- Department of Materials Science & Engineering, Sharif University of Technology, Tehran 11365-9466, Iran
| | - Amir Roointan
- Department of Medical Biotechnology, School of Advanced Medical Sciences & Technologies, Shiraz University of Medical Science, Shiraz 71348-14336, Iran
| | - Mahdi Karimi
- Cellular & Molecular Research Center, Iran University of Medical Sciences, Tehran 14496-14535, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 14496-14535, Iran
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Department of Dermatology, Harvard Medical School, Boston, MA 02115, USA
- Harvard – MIT Division of Health Sciences & Technology, Cambridge, MA 02139, USA
| |
Collapse
|
25
|
Miller MA, Mikula H, Luthria G, Li R, Kronister S, Prytyskach M, Kohler RH, Mitchison T, Weissleder R. Modular Nanoparticulate Prodrug Design Enables Efficient Treatment of Solid Tumors Using Bioorthogonal Activation. ACS NANO 2018; 12:12814-12826. [PMID: 30550257 PMCID: PMC6307086 DOI: 10.1021/acsnano.8b07954] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 12/04/2018] [Indexed: 05/18/2023]
Abstract
Prodrug strategies that facilitate localized and controlled activity of small-molecule therapeutics can reduce systemic exposure and improve pharmacokinetics, yet limitations in activation chemistry have made it difficult to assign tunable multifunctionality to prodrugs. Here, we present the design and application of a modular small-molecule caging strategy that couples bioorthogonal cleavage with a self-immolative linker and an aliphatic anchor. This strategy leverages recently discovered in vivo catalysis by a nanoencapsulated palladium compound (Pd-NP), which mediates alloxylcarbamate cleavage and triggers release of the activated drug. The aliphatic anchor enables >90% nanoencapsulation efficiency of the prodrug, while also allowing >104-fold increased cytotoxicity upon prodrug activation. We apply the strategy to a prodrug formulation of monomethyl auristatin E (MMAE), demonstrating its ability to target microtubules and kill cancer cells only after selective activation by Pd-NP. Computational pharmacokinetic modeling provides a mechanistic basis for the observation that the nanotherapeutic prodrug strategy can lead to more selective activation in the tumor, yet in a manner that is more sensitive to variable enhanced permeability and retention (EPR) effects. Combination treatment with the nanoencapsulated MMAE prodrug and Pd-NP safely blocks tumor growth, especially when combined with a local radiation therapy regimen that is known to improve EPR effects, and represents a conceptual step forward in prodrug design.
Collapse
Affiliation(s)
- Miles A. Miller
- Center
for Systems Biology, Massachusetts General
Hospital, Boston, Massachusetts 02114, United States
- Department
of Radiology, Massachusetts General Hospital
and Harvard Medical School, Boston, Massachusetts 02114, United States
- E-mail:
| | - Hannes Mikula
- Center
for Systems Biology, Massachusetts General
Hospital, Boston, Massachusetts 02114, United States
- Institute
of Applied Synthetic Chemistry, Vienna University
of Technology (TU Wien), Vienna 1060, Austria
| | - Gaurav Luthria
- Center
for Systems Biology, Massachusetts General
Hospital, Boston, Massachusetts 02114, United States
- Department
of Biomedical Informatics, Harvard Medical
School, Boston, Massachusetts 02115, United States
| | - Ran Li
- Center
for Systems Biology, Massachusetts General
Hospital, Boston, Massachusetts 02114, United States
| | - Stefan Kronister
- Institute
of Applied Synthetic Chemistry, Vienna University
of Technology (TU Wien), Vienna 1060, Austria
| | - Mark Prytyskach
- Center
for Systems Biology, Massachusetts General
Hospital, Boston, Massachusetts 02114, United States
| | - Rainer H. Kohler
- Center
for Systems Biology, Massachusetts General
Hospital, Boston, Massachusetts 02114, United States
| | - Timothy Mitchison
- Department
of Systems Biology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Ralph Weissleder
- Center
for Systems Biology, Massachusetts General
Hospital, Boston, Massachusetts 02114, United States
- Department
of Radiology, Massachusetts General Hospital
and Harvard Medical School, Boston, Massachusetts 02114, United States
- Department
of Systems Biology, Harvard Medical School, Boston, Massachusetts 02115, United States
| |
Collapse
|
26
|
Kim HY, Li R, Ng TS, Courties G, Rodell CB, Prytyskach M, Kohler RH, Pittet MJ, Nahrendorf M, Weissleder R, Miller MA. Quantitative Imaging of Tumor-Associated Macrophages and Their Response to Therapy Using 64Cu-Labeled Macrin. ACS NANO 2018; 12:12015-12029. [PMID: 30508377 PMCID: PMC6482841 DOI: 10.1021/acsnano.8b04338] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Tumor-associated macrophages (TAMs) are widely implicated in cancer progression, and TAM levels can influence drug responses, particularly to immunotherapy and nanomedicines. However, it has been difficult to quantify total TAM numbers and their dynamic spatiotemporal distribution in a non-invasive and translationally relevant manner. Here, we address this need by developing a pharmacokinetically optimized, 64Cu-labeled polyglucose nanoparticle (Macrin) for quantitative positron emission tomography (PET) imaging of macrophages in tumors. By combining PET with high-resolution in vivo confocal microscopy and ex vivo imaging of optically cleared tissue, we found that Macrin was taken up by macrophages with >90% selectivity. Uptake correlated with the content of macrophages in both healthy tissue and tumors ( R2 > 0.9) and showed striking heterogeneity in the TAM content of an orthotopic and immunocompetent mouse model of lung carcinoma. In a proof-of-principle application, we imaged Macrin to monitor the macrophage response to neo-adjuvant therapy, using a panel of chemotherapeutic and γ-irradiation regimens. Multiple treatments elicited 180-650% increase in TAMs. Imaging identified especially TAM-rich tumors thought to exhibit enhanced permeability and retention of nanotherapeutics. Indeed, these TAM-rich tumors accumulated >700% higher amounts of a model poly(d,l-lactic- co-glycolic acid)- b-polyethylene glycol (PLGA-PEG) therapeutic nanoparticle compared to TAM-deficient tumors, suggesting that imaging may guide patient selection into nanomedicine trials. In an orthotopic breast cancer model, chemoradiation enhanced TAM and Macrin accumulation in tumors, which corresponded to the improved delivery and efficacy of two model nanotherapies, PEGylated liposomal doxorubicin and a TAM-targeted nanoformulation of the toll-like receptor 7/8 agonist resiquimod (R848). Thus, Macrin imaging offers a selective and translational means to quantify TAMs and inform therapeutic decisions.
Collapse
Affiliation(s)
- Hye-Yeong Kim
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA 02114
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | - Ran Li
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA 02114
| | - Thomas S.C. Ng
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA 02114
| | - Gabriel Courties
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA 02114
| | - Christopher B. Rodell
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA 02114
| | - Mark Prytyskach
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA 02114
| | - Rainer H. Kohler
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA 02114
| | - Mikael J. Pittet
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA 02114
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | - Matthias Nahrendorf
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA 02114
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA 02114
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115
| | - Miles A. Miller
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA 02114
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| |
Collapse
|
27
|
Fabbrini MG, Cirri D, Pratesi A, Ciofi L, Marzo T, Guerri A, Nistri S, Dell'Accio A, Gamberi T, Severi M, Bencini A, Messori L. A Fluorescent Silver(I) Carbene Complex with Anticancer Properties: Synthesis, Characterization, and Biological Studies. ChemMedChem 2018; 14:182-188. [PMID: 30444581 DOI: 10.1002/cmdc.201800672] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 11/14/2018] [Indexed: 12/20/2022]
Abstract
The silver(I) N-heterocyclic carbene (NHC) complex bis(1-(anthracen-9-ylmethyl)-3-ethylimidazol-2-ylidene) silver chloride ([Ag(EIA)2 ]Cl), bearing two anthracenyl fluorescent probes, has been synthesized and characterized. [Ag(EIA)2 ]Cl is stable in organic solvents and under physiological conditions, and shows potent cytotoxic effects in vitro toward human SH-SY5Y neuroblastoma cells. The interactions of [Ag(EIA)2 ]Cl with a few model biological targets have been studied as well as its ability to be internalized in cells. The in vitro anticancer activity is apparently related to the level of drug internalization. Notably, [Ag(EIA)2 ]Cl does not react with a few model proteins, but is capable of binding the C-terminal dodecapeptide of thioredoxin reductase hTrxR(488-499) and to strongly inhibit the activity of this enzyme. Binding occurs through an unconventional process leading to covalent binding of one or two carbene ligands to the C-terminal dodecapeptide with concomitant release of the silver cation. To the best of our knowledge, this mode of interaction is reported here for the first time for Ag(NHC)2 complexes.
Collapse
Affiliation(s)
- Maria Giulia Fabbrini
- Department of Chemistry "U. Schiff", University of Florence, Via della Lastruccia 3, 50019, Sesto Fiorentino, Italy
| | - Damiano Cirri
- Laboratory of Metals in Medicine (MetMed), Department of Chemistry "U. Schiff", University of Florence, Via della Lastruccia 3, 50019, Sesto Fiorentino, Italy
| | - Alessandro Pratesi
- Laboratory of Metals in Medicine (MetMed), Department of Chemistry "U. Schiff", University of Florence, Via della Lastruccia 3, 50019, Sesto Fiorentino, Italy
| | - Lorenzo Ciofi
- Laboratory of Metals in Medicine (MetMed), Department of Chemistry "U. Schiff", University of Florence, Via della Lastruccia 3, 50019, Sesto Fiorentino, Italy
| | - Tiziano Marzo
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126, Pisa, Italy
| | - Annalisa Guerri
- Department of Chemistry "U. Schiff", University of Florence, Via della Lastruccia 3, 50019, Sesto Fiorentino, Italy
| | - Silvia Nistri
- Department of Experimental and Clinical Medicine, Largo Brambilla 3, 50134, Florence, Italy
| | - Alfonso Dell'Accio
- Department of Experimental and Clinical Medicine, Largo Brambilla 3, 50134, Florence, Italy
| | - Tania Gamberi
- Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale GB Morgagni 50, 50134, Florence, Italy
| | - Mirko Severi
- Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale GB Morgagni 50, 50134, Florence, Italy
| | - Andrea Bencini
- Department of Chemistry "U. Schiff", University of Florence, Via della Lastruccia 3, 50019, Sesto Fiorentino, Italy
| | - Luigi Messori
- Laboratory of Metals in Medicine (MetMed), Department of Chemistry "U. Schiff", University of Florence, Via della Lastruccia 3, 50019, Sesto Fiorentino, Italy
| |
Collapse
|
28
|
Nanotherapeutic Anti-influenza Solutions: Current Knowledge and Future Challenges. J CLUST SCI 2018. [DOI: 10.1007/s10876-018-1417-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
29
|
Nair RR, Piktel D, Geldenhuys WJ, Gibson LF. Combination of cabazitaxel and plicamycin induces cell death in drug resistant B-cell acute lymphoblastic leukemia. Leuk Res 2018; 72:59-66. [PMID: 30103201 PMCID: PMC6414069 DOI: 10.1016/j.leukres.2018.08.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 07/31/2018] [Accepted: 08/05/2018] [Indexed: 01/17/2023]
Abstract
Bone marrow microenvironment mediated downregulation of BCL6 is critical for maintaining cell quiescence and modulating therapeutic response in B-cell acute lymphoblastic leukemia (ALL). In the present study, we have performed a high throughput cell death assay using BCL6 knockdown REH ALL cell line to screen a library of FDA-approved oncology drugs. In the process, we have identified a microtubule inhibitor, cabazitaxel (CAB), and a RNA synthesis inhibitor, plicamycin (PLI) as potential anti-leukemic agents. CAB and PLI inhibited cell proliferation in not only the BCL6 knockdown REH cell line, but also six other ALL cell lines. Furthermore, combination of CAB and PLI had a synergistic effect in inhibiting proliferation in a cytarabine-resistant (REH/Ara-C) ALL cell line. Use of nanoparticles for delivery of CAB and PLI demonstrated that the combination was very effective when tested in a co-culture model that mimics the in vivo bone marrow microenvironment that typically supports ALL cell survival and migration into protective niches. Furthermore, exposure to PLI inhibited SOX2 transcription and exposure to CAB inhibited not only Mcl-1 expression but also chemotaxis in ALL cells. Taken together, our study demonstrates the utility of concomitantly targeting different critical regulatory pathways to induce cell death in drug resistant ALL cells.
Collapse
Affiliation(s)
- Rajesh R Nair
- Department of Microbiology, Immunology and Cell Biology, School of Medicine, West Virginia University, Morgantown, WV, United States
| | - Debbie Piktel
- Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, United States
| | - Werner J Geldenhuys
- Department of Pharmaceutical Sciences, School of Pharmacy, and WVU Cancer Institute, West Virginia University, Morgantown, WV, United States
| | - Laura F Gibson
- Department of Microbiology, Immunology and Cell Biology, School of Medicine, West Virginia University, Morgantown, WV, United States; Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, United States; WVU Cancer Institute, West Virginia University, Morgantown, WV, United States.
| |
Collapse
|
30
|
Simon J, Müller LK, Kokkinopoulou M, Lieberwirth I, Morsbach S, Landfester K, Mailänder V. Exploiting the biomolecular corona: pre-coating of nanoparticles enables controlled cellular interactions. NANOSCALE 2018; 10:10731-10739. [PMID: 29845991 DOI: 10.1039/c8nr03331e] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Formation of the biomolecular corona ultimately determines the successful application of nanoparticles in vivo. Adsorption of biomolecules such as proteins is an inevitable process that takes place instantaneously upon contact with physiological fluid (e.g. blood). Therefore, strategies are needed to control this process in order to improve the properties of the nanoparticles and to allow targeted drug delivery. Here, we show that the design of the protein corona by a pre-formed protein corona with tailored properties enables targeted cellular interactions. Nanoparticles were pre-coated with immunoglobulin depleted plasma to create and design a protein corona that reduces cellular uptake by immune cells. It was proven that a pre-formed protein corona remains stable even after nanoparticles were re-introduced to plasma. This opens up the great potential to exploit protein corona formation, which will significantly influence the development of novel nanomaterials.
Collapse
Affiliation(s)
- Johanna Simon
- Dermatology Clinic, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany. and Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Laura K Müller
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Maria Kokkinopoulou
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Ingo Lieberwirth
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Svenja Morsbach
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Katharina Landfester
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Volker Mailänder
- Dermatology Clinic, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany. and Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| |
Collapse
|
31
|
Shin SH, Park SH, Kim SW, Kim M, Kim D. Fluorine MR Imaging Monitoring of Tumor Inflammation after High-Intensity Focused Ultrasound Ablation. Radiology 2018; 287:476-484. [PMID: 29369752 DOI: 10.1148/radiol.2017171603] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Purpose To investigate whether high-intensity focused ultrasound (HIFU)-induced macrophage infiltration could be longitudinally monitored with fluorine 19 (19F) magnetic resonance (MR) imaging in a quantitative manner. Materials and Methods BALB/c mice were subcutaneously inoculated with 4T1 cells and were separated into three groups: untreated mice (control, n = 9), HIFU-treated mice (HIFU, n = 9), and HIFU- and clodronate-treated mice (HIFU+Clod, n = 9). Immediately after HIFU treatment, all mice were intravenously given perfluorocarbon (PFC) emulsion. MR imaging examinations were performed 2, 4, 7, 10, and 14 days after HIFU treatment. Two-way repeated measures analysis of variance was used to analyze the changes in 19F signal over time and differences between groups. Histologic examinations were performed to confirm in vivo data. Results Fluorine 19 signals were detected at the rims of tumors and the peripheries of ablated lesions. Mean 19F signal in tumors was significantly higher in HIFU-treated mice than in control mice up to day 4 (0.82 ± 0.26 vs 0.42 ± 0.17, P < .001). Fluorine 19 signals were higher in the HIFU+Clod group than in the control group from day 4 (0.82 ± 0.23, P < .001) to day 14 (0.55 ± 0.16 vs 0.28 ± 0.06, P < .05). Histologic examination revealed macrophage infiltration around ablated lesions. Immunofluorescence staining confirmed PFC labeling of macrophages. Conclusion Fluorine 19 MR imaging can longitudinally capture and quantify HIFU-induced macrophage infiltration in preclinical tumor models. © RSNA, 2018 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Soo Hyun Shin
- From the Molecular Imaging Branch, Division of Convergence Technology, National Cancer Center, Research Building, Ilsanro-323, Ilsandong-gu, Goyang 10408, Korea
| | - Sang Hyun Park
- From the Molecular Imaging Branch, Division of Convergence Technology, National Cancer Center, Research Building, Ilsanro-323, Ilsandong-gu, Goyang 10408, Korea
| | - Seung Won Kim
- From the Molecular Imaging Branch, Division of Convergence Technology, National Cancer Center, Research Building, Ilsanro-323, Ilsandong-gu, Goyang 10408, Korea
| | - Minsun Kim
- From the Molecular Imaging Branch, Division of Convergence Technology, National Cancer Center, Research Building, Ilsanro-323, Ilsandong-gu, Goyang 10408, Korea
| | - Daehong Kim
- From the Molecular Imaging Branch, Division of Convergence Technology, National Cancer Center, Research Building, Ilsanro-323, Ilsandong-gu, Goyang 10408, Korea
| |
Collapse
|
32
|
A New Approach to Deliver Anti-cancer Nanodrugs with Reduced Off-target Toxicities and Improved Efficiency by Temporarily Blunting the Reticuloendothelial System with Intralipid. Sci Rep 2017; 7:16106. [PMID: 29170482 PMCID: PMC5701028 DOI: 10.1038/s41598-017-16293-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 11/08/2017] [Indexed: 12/19/2022] Open
Abstract
We have developed a new strategy to temporarily blunt the reticuloendothelial system uptake of nanodrugs, a major challenge for nanodrug delivery and causing off-target toxicities, using an FDA approved nutrition supplement, Intralipid. We have tested our methodology in rats using an experimental platinum-containing anti-cancer nanodrug and three FDA approved nanodrugs, Abraxane, Marqibo, and Onivyde, to determine their toxicities in liver, spleen, and kidney, with and without the addition of Intralipid. Our method illustrates its potentials to deliver nanodrugs with an increase in the bioavailability and a decrease in toxicities. Our study shows that Intralipid treatment exhibits no harmful effect on tumor growing and no negative effect on the anti-tumor efficacy of the platinum-containing nanodrug, as well as animal survival rate in a HT-29 xenograft mouse model. Our methodology could also be a valuable complement/supplement to the “stealth” strategies. Our approach is a general one applicable to any approved and in-development nanodrugs without additional modification of the nanodrugs, thus facilitating its translation to clinical settings.
Collapse
|
33
|
Lee KY, Jang GH, Byun CH, Jeun M, Searson PC, Lee KH. Zebrafish models for functional and toxicological screening of nanoscale drug delivery systems: promoting preclinical applications. Biosci Rep 2017; 37:BSR20170199. [PMID: 28515222 PMCID: PMC5463258 DOI: 10.1042/bsr20170199] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 04/27/2017] [Accepted: 05/16/2017] [Indexed: 12/16/2022] Open
Abstract
Preclinical screening with animal models is an important initial step in clinical translation of new drug delivery systems. However, establishing efficacy, biodistribution, and biotoxicity of complex, multicomponent systems in small animal models can be expensive and time-consuming. Zebrafish models represent an alternative for preclinical studies for nanoscale drug delivery systems. These models allow easy optical imaging, large sample size, and organ-specific studies, and hence an increasing number of preclinical studies are employing zebrafish models. In this review, we introduce various models and discuss recent studies of nanoscale drug delivery systems in zebrafish models. Also in the end, we proposed a guideline for the preclinical trials to accelerate the progress in this field.
Collapse
Affiliation(s)
- Keon Yong Lee
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Gun Hyuk Jang
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- Department of Biomedical Engineering, Korea University of Science and Technology (UST), Daejeon 02792, Republic of Korea
| | - Cho Hyun Byun
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- Division of Life Sciences, School of Life Science and Biotechnology, Korea University, Seoul 02792, Republic of Korea
| | - Minhong Jeun
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Peter C Searson
- Institute for Nanobiotechnology (INBT), Johns Hopkins University, Baltimore, MD 21218, U.S.A.
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, U.S.A
| | - Kwan Hyi Lee
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- Department of Biomedical Engineering, Korea University of Science and Technology (UST), Daejeon 02792, Republic of Korea
| |
Collapse
|
34
|
Özeş AR, Wang Y, Zong X, Fang F, Pilrose J, Nephew KP. Therapeutic targeting using tumor specific peptides inhibits long non-coding RNA HOTAIR activity in ovarian and breast cancer. Sci Rep 2017; 7:894. [PMID: 28420874 PMCID: PMC5429858 DOI: 10.1038/s41598-017-00966-3] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 03/21/2017] [Indexed: 12/16/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) play key roles in human diseases, including cancer. Functional studies of the lncRNA HOTAIR (HOX transcript antisense RNA) provide compelling evidence for therapeutic targeting of HOTAIR in cancer, but targeting lncRNAs in vivo has proven to be difficult. In the current study, we describe a peptide nucleic acids (PNA)-based approach to block the ability of HOTAIR to interact with EZH2 and subsequently inhibit HOTAIR-EZH2 activity and resensitize resistant ovarian tumors to platinum. Treatment of HOTAIR-overexpressing ovarian and breast cancer cell lines with PNAs decreased invasion and increased chemotherapy sensitivity. Furthermore, the mechanism of action correlated with reduced nuclear factor-kappaB (NF-κB) activation and decreased expression of NF-κB target genes matrix metalloprotease 9 and interleukin 6. To deliver the anti-lncRNA to the acidic (pH approximately 6) tumor microenvironment, PNAs were conjugated to pH-low insertion peptide (pHLIP). Treatment of mice harboring platinum-resistant ovarian tumor xenografts with pHLIP-PNA constructs suppressed HOTAIR activity, reduced tumor formation and improved survival. This first report on pHLIP-PNA lncRNA targeting solid tumors in vivo suggests a novel cancer therapeutic approach.
Collapse
Affiliation(s)
- Ali R Özeş
- Molecular and Cellular Biochemistry Department, Indiana University, Bloomington, IN, 47405, USA
| | - Yinu Wang
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN, 47405, USA
| | - Xingyue Zong
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN, 47405, USA
| | - Fang Fang
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN, 47405, USA
| | - Jay Pilrose
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN, 47405, USA
| | - Kenneth P Nephew
- Molecular and Cellular Biochemistry Department, Indiana University, Bloomington, IN, 47405, USA.
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN, 47405, USA.
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana, 46202, USA.
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
35
|
Ye Q, Liu L, Wu Y, Yeh F, Li W, Tseng L, Ho C. Intralipid ® attenuates acute cardiac allograft rejection in relation to promoting CD4 + CD25 + Foxp3 + regulatory T-cells and inhibiting toll-like receptor 4 expression. TRANSPLANTATION REPORTS 2017. [DOI: 10.1016/j.tpr.2017.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
36
|
Abstract
Cancer is one of the leading causes of death worldwide. Curcumin is a well-established anticancer agent in vitro but its efficacy is yet to be proven in clinical trials. Poor bioavailability of curcumin is the principal reason behind the lack of efficiency of curcumin in clinical trials. Many studies prove that the bioavailability of curcumin can be improved by administering it through nanoparticle drug carriers. This review focuses on the efforts made in the field of nanotechnology to improve the bioavailability of curcumin. Nanotechnologies of curcumin come in various shapes and sizes. The simplest curcumin nanoparticle that increased the bioavailability of curcumin is the curcumin-metal complex. On the other hand, we have intricate thermoresponsive nanoparticles that can release curcumin upon stimulation (analogous to a remote control). Future research required for developing potent curcumin nanoparticles is also discussed.
Collapse
Affiliation(s)
- Parasuraman Aiya Subramani
- a Department of Zoology , Yogi Vemana University , Kadapa , India.,b Centre for Fish Immunology, School of Life Sciences , Vels Institute of Science Technology and Advanced Studies , Chennai , India
| | - Kalpana Panati
- c Department of Biotechnology , Govt. College for Men , Kadapa , India
| | | |
Collapse
|
37
|
Geldenhuys WJ, Khayat MT, Yun J, Nayeem MA. Drug Delivery and Nanoformulations for the Cardiovascular System. RESEARCH & REVIEWS. DRUG DELIVERY 2017; 1:32-40. [PMID: 28713881 PMCID: PMC5507069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Therapeutic delivery to the cardiovascular system may play an important role in the successful treatment of a variety of disease state, including atherosclerosis, ischemic-reperfusion injury and other types of microvascular diseases including hypertension. In this review we evaluate the different options available for the development of suitable delivery systems that include the delivery of small organic compounds [adenosin A2A receptor agonist (CGS 21680), CYP-epoxygenases inhibitor (N-(methylsulfonyl)-2-(2-propynyloxy)-benzenehexanamide, trans-4-[4-(3-adamantan-1-ylureido)cyclohexyloxy] benzoic acid), soluble epoxide hydrolase inhibitor (N-methylsulfonyl-12,12-dibromododec-11-enamide), PPARγ agonist (rosiglitazone) and PPARγ antagonist (T0070907)], nanoparticles, peptides, and siRNA to the cardiovascular system. Effective formulations of nanoproducts have significant potential to overcome physiological barriers and improve therapeutic outcomes in patients. As per the literature covering targeted delivery to the cardiovascular system, we found that this area is still at infancy stage, as compare to the more mature fields of tumor cancer or brain delivery (e.g. blood-brain barrier permeability) with fewer publications focused on the targeted drug delivery technologies. Additionally, we show how pharmacology needs to be well understood when considering the cardiovascular system. Therefore, we discussed in this review various receptors agonists, antagonists, activators and inhibitors which will have effects on cardiovascular system.
Collapse
Affiliation(s)
- WJ Geldenhuys
- Department of Pharmaceutical Sciences, West Virginia University, School of Pharmacy, Morgantown WV 26506 USA
| | - MT Khayat
- Department of Pharmaceutical Sciences, West Virginia University, School of Pharmacy, Morgantown WV 26506 USA
- Deparment of Pharmaceutical Chemistry, King Abdulaziz University, School of Pharmacy, Jeddah, Saudi Arabia
| | - J Yun
- Department of Integrative Medical Sciences, Northeast Ohio Medical University College of Medicine, Rootstown OH 44272 USA
| | - MA Nayeem
- Department of Pharmaceutical Sciences, West Virginia University, School of Pharmacy, Morgantown WV 26506 USA
| |
Collapse
|
38
|
Balzaretti R, Meder F, Monopoli MP, Boselli L, Armenia I, Pollegioni L, Bernardini G, Gornati R. Synthesis, characterization and programmable toxicity of iron oxide nanoparticles conjugated withd-amino acid oxidase. RSC Adv 2017. [DOI: 10.1039/c6ra25349k] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
RgDAAO conjugated to γ-Fe2O3NPs generates a low toxic NP-DAAO system, which kills cancer cells through ROS production.
Collapse
Affiliation(s)
- Riccardo Balzaretti
- Department of Biotechnology and Life Sciences
- University of Insubria
- Varese
- Italy
| | - Fabian Meder
- Centre for Bionano Interactions
- University College Dublin
- Dublin
- Ireland
| | - Marco P. Monopoli
- Centre for Bionano Interactions
- University College Dublin
- Dublin
- Ireland
- Department of Pharmacy and Medical Chemistry
| | - Luca Boselli
- Centre for Bionano Interactions
- University College Dublin
- Dublin
- Ireland
| | - Ilaria Armenia
- Department of Biotechnology and Life Sciences
- University of Insubria
- Varese
- Italy
| | - Loredano Pollegioni
- Department of Biotechnology and Life Sciences
- University of Insubria
- Varese
- Italy
- The Protein Factory Research Center
| | - Giovanni Bernardini
- Department of Biotechnology and Life Sciences
- University of Insubria
- Varese
- Italy
- The Protein Factory Research Center
| | - Rosalba Gornati
- Department of Biotechnology and Life Sciences
- University of Insubria
- Varese
- Italy
- The Protein Factory Research Center
| |
Collapse
|
39
|
Wu CH, Liu IJ, Lu RM, Wu HC. Advancement and applications of peptide phage display technology in biomedical science. J Biomed Sci 2016; 23:8. [PMID: 26786672 PMCID: PMC4717660 DOI: 10.1186/s12929-016-0223-x] [Citation(s) in RCA: 222] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Accepted: 01/11/2016] [Indexed: 12/25/2022] Open
Abstract
Combinatorial phage library is a powerful research tool for high-throughput screening of protein interactions. Of all available molecular display techniques, phage display has proven to be the most popular approach. Screening phage-displayed random peptide libraries is an effective means of identifying peptides that can bind target molecules and regulate their function. Phage-displayed peptide libraries can be used for (i) B-cell and T-cell epitope mapping, (ii) selection of bioactive peptides bound to receptors or proteins, disease-specific antigen mimics, peptides bound to non-protein targets, cell-specific peptides, or organ-specific peptides, and (iii) development of peptide-mediated drug delivery systems and other applications. Targeting peptides identified using phage display technology may be useful for basic research and translational medicine. In this review article, we summarize the latest technological advancements in the application of phage-displayed peptide libraries to applied biomedical sciences.
Collapse
Affiliation(s)
- Chien-Hsun Wu
- Institute of Cellular and Organismic Biology, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei, 11529, Taiwan
| | - I-Ju Liu
- Institute of Cellular and Organismic Biology, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei, 11529, Taiwan
| | - Ruei-Min Lu
- Institute of Cellular and Organismic Biology, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei, 11529, Taiwan
| | - Han-Chung Wu
- Institute of Cellular and Organismic Biology, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei, 11529, Taiwan.
| |
Collapse
|
40
|
Raza MK, Mitra K, Shettar A, Basu U, Kondaiah P, Chakravarty AR. Photoactive platinum(ii) β-diketonates as dual action anticancer agents. Dalton Trans 2016; 45:13234-43. [DOI: 10.1039/c6dt02590k] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cyclometallated platinum(ii) β-diketonates show significant photocytotoxicity in skin-keratinocyte HaCaT cells [IC50: ∼10 μM (visible light, 400–700 nm), ≥60 μM (dark)].
Collapse
Affiliation(s)
- Md Kausar Raza
- Department of Inorganic and Physical Chemistry
- Indian Institute of Science
- Bangalore 560012
- India
| | - Koushambi Mitra
- Department of Inorganic and Physical Chemistry
- Indian Institute of Science
- Bangalore 560012
- India
| | - Abhijith Shettar
- Department of Molecular Reproduction
- Development and Genetics
- Indian Institute of Science
- Bangalore 560012
- India
| | - Uttara Basu
- Department of Inorganic and Physical Chemistry
- Indian Institute of Science
- Bangalore 560012
- India
| | - Paturu Kondaiah
- Department of Molecular Reproduction
- Development and Genetics
- Indian Institute of Science
- Bangalore 560012
- India
| | - Akhil R. Chakravarty
- Department of Inorganic and Physical Chemistry
- Indian Institute of Science
- Bangalore 560012
- India
| |
Collapse
|
41
|
Abstract
Platinum-based anticancer drugs are the mainstay of chemotherapy regimens in clinic. Nevertheless, the efficacy of platinum drugs is badly affected by serious systemic toxicities and drug resistance, and the pharmacokinetics of most platinum drugs is largely unknown. In recent years, a keen interest in functionalizing platinum complexes with bioactive molecules, targeting groups, photosensitizers, fluorophores, or nanomaterials has been sparked among chemical and biomedical researchers. The motivation for functionalization comes from some of the following demands: to improve the tumor selectivity or minimize the systemic toxicity of the drugs, to enhance the cellular accumulation of the drugs, to overcome the tumor resistance to the drugs, to visualize the drug molecules in vitro or in vivo, to achieve a synergistic anticancer effect between different therapeutic modalities, or to add extra functionality to the drugs. In this Account, we present different strategies being used for functionalizing platinum complexes, including conjugation with bisphosphonates, peptides, receptor-specific ligands, polymers, nanoparticles, magnetic resonance imaging contrast agents, metal chelators, or photosensitizers. Among them, bisphosphonates, peptides, and receptor-specific ligands are used for actively targeted drug delivery, polymers and nanoparticles are for passively targeted drug delivery, magnetic resonance imaging contrast agents are for theranostic purposes, metal chelators are for the treatment or prevention of Alzheimer's disease (AD), and photosensitizers are for photodynamic therapy of cancers. The rationales behind these designs are explained and justified at the molecular or cellular level, associating with the requirements for diagnosis, therapy, and visualization of biological processes. To illustrate the wide range of opportunities and challenges that are emerging in this realm, representative examples of targeted drug delivery systems, anticancer conjugates, anticancer theranostic agents, and anti-AD compounds relevant to functionalized platinum complexes are provided. All the examples exhibit new potential of platinum complexes for future applications in biomedical areas. The emphases of this Account are placed on the functionalization for targeted drug delivery and theranostic agents. In the end, a general assessment of various strategies has been made according to their major shortcomings and defects. The original information in this Account comes entirely from literature appearing since 2010.
Collapse
Affiliation(s)
- Xiaoyong Wang
- State
Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences,
State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing 210023, P. R. China
- Collaborative
Innovation Center of Chemistry for Life Sciences, Nanjing University, Nanjing 210093, P. R. China
| | - Xiaohui Wang
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Nanjing University, Nanjing 210093, P. R. China
- College
of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing 211816, P. R. China
| | - Zijian Guo
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Nanjing University, Nanjing 210093, P. R. China
- Collaborative
Innovation Center of Chemistry for Life Sciences, Nanjing University, Nanjing 210093, P. R. China
| |
Collapse
|