1
|
Sahoo GP, Rai VK, Pradhan D, Halder J, Rajwar TK, Mahanty R, Saha I, Mishra A, Dash P, Dash C, Al-Tamimi J, Manoharadas S, Kar B, Ghosh G, Rath G. A doxorubicin loaded chitosan-poloxamer in situ implant for the treatment of breast cancer. RSC Adv 2024; 14:33952-33967. [PMID: 39463476 PMCID: PMC11503159 DOI: 10.1039/d4ra06253a] [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: 08/29/2024] [Accepted: 10/14/2024] [Indexed: 10/29/2024] Open
Abstract
Breast cancer is a serious concern for many women worldwide. Drug-loaded implants have shown several benefits over systemic administrations. To provide anti-cancer drugs with controlled release and reduced systemic toxicity, biodegradable in situ implants have attracted a lot of attention. In the present study, we aimed to design and optimize a doxorubicin-loaded chitosan-poloxamer in situ implant for breast cancer treatment. Utilizing Box-Behnken Design and a Quality-by-Design (QbD) methodology, the in situ implant was prepared with chitosan (X1), poloxamer 407 concentration (X2), and stirring time (X3) as the independent variables. It was characterized for its in vitro gelation time, pH, rheology, and morphology, and evaluated based on drug release profile, in vitro cytotoxicity activities, in vitro anti-inflammatory potential, in vitro cellular uptake, and in vivo anti-inflammatory and pharmacokinetics to ensure their therapeutic outcomes. The results revealed that the prepared formulation showed a gelation time of 26 ± 0.2 s with a viscosity of 8312.6 ± 114.2 cPs at 37 °C. The developed formulation showed better cytotoxic activity in MCF-7 cell lines compared to the free drug solution. It demonstrated reduced levels of pro-inflammatory cytokines in RAW 264.7 macrophages. Further, the prepared in situ implant increases the intracellular accumulation of DOX in the MCF-7 cells. The in vivo pharmacokinetic investigations depicted an increase in t 1/2 and a decrease in AUC of the developed formulation resulting in prolonged drug release and there could be a lower drug concentration in the bloodstream than for the free drug. Therefore, the developed in situ implant may offer a viable option for breast cancer treatment.
Collapse
Affiliation(s)
- Guru Prasanna Sahoo
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University) Bhubaneswar Odisha India +91-9888206383
| | - Vineet Kumar Rai
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University) Bhubaneswar Odisha India +91-9888206383
| | - Deepak Pradhan
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University) Bhubaneswar Odisha India +91-9888206383
| | - Jitu Halder
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University) Bhubaneswar Odisha India +91-9888206383
| | - Tushar Kanti Rajwar
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University) Bhubaneswar Odisha India +91-9888206383
| | - Ritu Mahanty
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University) Bhubaneswar Odisha India +91-9888206383
| | - Ivy Saha
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University) Bhubaneswar Odisha India +91-9888206383
| | - Ajit Mishra
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University) Bhubaneswar Odisha India +91-9888206383
| | - Priyanka Dash
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University) Bhubaneswar Odisha India +91-9888206383
| | - Chandan Dash
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University) Bhubaneswar Odisha India +91-9888206383
| | - Jameel Al-Tamimi
- Zoology Department, College of Science, King Saud University P. O. Box 2455 Riyadh Saudi Arabia
| | - Salim Manoharadas
- Department of Botany and Microbiology, College of Science, King Saud University P. O. Box. 2454 Riyadh 11451 Saudi Arabia
| | - Biswakanth Kar
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University) Bhubaneswar Odisha India +91-9888206383
| | - Goutam Ghosh
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University) Bhubaneswar Odisha India +91-9888206383
| | - Goutam Rath
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University) Bhubaneswar Odisha India +91-9888206383
| |
Collapse
|
2
|
McConville C, Lastakchi S, Al Amri A, Ngoga D, Fayeye O, Cruickshank G. Local Delivery of Irinotecan to Recurrent GBM Patients at Reoperation Offers a Safe Route of Administration. Cancers (Basel) 2024; 16:3008. [PMID: 39272866 PMCID: PMC11393903 DOI: 10.3390/cancers16173008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 08/14/2024] [Accepted: 08/21/2024] [Indexed: 09/15/2024] Open
Abstract
Glioblastomas are impossible to completely resect and almost always recur at the borders of the resection margin. There is no established chemotherapy regimen available to patients who recur, while systemic treatment is hampered by the blood-brain barrier. Here, we report on the first evaluation in humans of the intraparenchymal injection of irinotecan into the resection cavity after surgical resection of recurrent glioblastoma patients. The cytotoxicity of irinotecan was compared to SN-38 in primary cells from recurrent glioblastoma patients. Irinotecan was injected at multiple (~30) sites of the resection cavity wall at a depth of 3 to 5 mm. SN-38 was more cytotoxic than irinotecan at concentrations below 1 µM due to enzyme kinetics. The intraparenchymal administration of irinotecan was safe, with good wound healing and an absence of swelling, inflammation, or pseudo-abscess formation. The median survival post irinotecan administration was 32.6 weeks. The median overall survival was 30.5 months, with a two-year survival rate of 56%. This study demonstrates that local delivery of irinotecan into the brain parenchyma offers a safe route of administration over systemic delivery in the treatment of recurrent glioblastoma.
Collapse
Affiliation(s)
- Christopher McConville
- School of Pharmacy, Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Sarah Lastakchi
- School of Pharmacy, Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Ali Al Amri
- School of Pharmacy, Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Desire Ngoga
- Pediatric Neurosurgery, The Bristol Royal Hospital for Children, Bristol BS2 8BJ, UK
| | - Oluwafikayo Fayeye
- Department of Neurosurgery, University Hospitals Birmingham, NHS Foundation Trust, Birmingham B15 2GW, UK
| | - Garth Cruickshank
- Department of Neurosurgery, University Hospitals Birmingham, NHS Foundation Trust, Birmingham B15 2GW, UK
| |
Collapse
|
3
|
Agiba AM, Elsayyad N, ElShagea HN, Metwalli MA, Mahmoudsalehi AO, Beigi-Boroujeni S, Lozano O, Aguirre-Soto A, Arreola-Ramirez JL, Segura-Medina P, Hamed RR. Advances in Light-Responsive Smart Multifunctional Nanofibers: Implications for Targeted Drug Delivery and Cancer Therapy. Pharmaceutics 2024; 16:1017. [PMID: 39204362 PMCID: PMC11359459 DOI: 10.3390/pharmaceutics16081017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 07/28/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
Over the last decade, scientists have shifted their focus to the development of smart carriers for the delivery of chemotherapeutics in order to overcome the problems associated with traditional chemotherapy, such as poor aqueous solubility and bioavailability, low selectivity and targeting specificity, off-target drug side effects, and damage to surrounding healthy tissues. Nanofiber-based drug delivery systems have recently emerged as a promising drug delivery system in cancer therapy owing to their unique structural and functional properties, including tunable interconnected porosity, a high surface-to-volume ratio associated with high entrapment efficiency and drug loading capacity, and high mass transport properties, which allow for controlled and targeted drug delivery. In addition, they are biocompatible, biodegradable, and capable of surface functionalization, allowing for target-specific delivery and drug release. One of the most common fiber production methods is electrospinning, even though the relatively two-dimensional (2D) tightly packed fiber structures and low production rates have limited its performance. Forcespinning is an alternative spinning technology that generates high-throughput, continuous polymeric nanofibers with 3D structures. Unlike electrospinning, forcespinning generates fibers by centrifugal forces rather than electrostatic forces, resulting in significantly higher fiber production. The functionalization of nanocarriers on nanofibers can result in smart nanofibers with anticancer capabilities that can be activated by external stimuli, such as light. This review addresses current trends and potential applications of light-responsive and dual-stimuli-responsive electro- and forcespun smart nanofibers in cancer therapy, with a particular emphasis on functionalizing nanofiber surfaces and developing nano-in-nanofiber emerging delivery systems for dual-controlled drug release and high-precision tumor targeting. In addition, the progress and prospective diagnostic and therapeutic applications of light-responsive and dual-stimuli-responsive smart nanofibers are discussed in the context of combination cancer therapy.
Collapse
Affiliation(s)
- Ahmed M. Agiba
- School of Engineering and Sciences, Tecnologico de Monterrey, Monterrey 64849, Mexico; (A.M.A.); (A.O.M.); (A.A.-S.)
| | - Nihal Elsayyad
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, October for Modern Sciences and Arts University, Cairo 12451, Egypt;
| | - Hala N. ElShagea
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ahram Canadian University, Cairo 12451, Egypt;
| | - Mahmoud A. Metwalli
- El Demerdash Hospital, Faculty of Medicine, Ain Shams University, Cairo 11591, Egypt;
| | - Amin Orash Mahmoudsalehi
- School of Engineering and Sciences, Tecnologico de Monterrey, Monterrey 64849, Mexico; (A.M.A.); (A.O.M.); (A.A.-S.)
| | - Saeed Beigi-Boroujeni
- School of Engineering and Sciences, Tecnologico de Monterrey, Monterrey 64849, Mexico; (A.M.A.); (A.O.M.); (A.A.-S.)
| | - Omar Lozano
- School of Medicine and Health Sciences, Tecnológico de Monterrey, Monterrey 64849, Mexico;
- Institute for Obesity Research, Tecnológico de Monterrey, Monterrey 64849, Mexico
| | - Alan Aguirre-Soto
- School of Engineering and Sciences, Tecnologico de Monterrey, Monterrey 64849, Mexico; (A.M.A.); (A.O.M.); (A.A.-S.)
| | - Jose Luis Arreola-Ramirez
- Department of Bronchial Hyperresponsiveness, National Institute of Respiratory Diseases “Ismael Cosío Villegas”, Mexico City 14080, Mexico;
| | - Patricia Segura-Medina
- Department of Bronchial Hyperresponsiveness, National Institute of Respiratory Diseases “Ismael Cosío Villegas”, Mexico City 14080, Mexico;
- School of Medicine and Health Sciences, Tecnológico de Monterrey, Mexico City 14380, Mexico
| | - Raghda Rabe Hamed
- Department of Industrial Pharmacy, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, Cairo 12566, Egypt;
| |
Collapse
|
4
|
Fang Q. The Versatile Attributes of MGMT: Its Repair Mechanism, Crosstalk with Other DNA Repair Pathways, and Its Role in Cancer. Cancers (Basel) 2024; 16:331. [PMID: 38254819 PMCID: PMC10814553 DOI: 10.3390/cancers16020331] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/09/2024] [Accepted: 01/10/2024] [Indexed: 01/24/2024] Open
Abstract
O6-methylguanine-DNA methyltransferase (MGMT or AGT) is a DNA repair protein with the capability to remove alkyl groups from O6-AlkylG adducts. Moreover, MGMT plays a crucial role in repairing DNA damage induced by methylating agents like temozolomide and chloroethylating agents such as carmustine, and thereby contributes to chemotherapeutic resistance when these agents are used. This review delves into the structural roles and repair mechanisms of MGMT, with emphasis on the potential structural and functional roles of the N-terminal domain of MGMT. It also explores the development of cancer therapeutic strategies that target MGMT. Finally, it discusses the intriguing crosstalk between MGMT and other DNA repair pathways.
Collapse
Affiliation(s)
- Qingming Fang
- Department of Biochemistry and Structural Biology, Greehey Children's Cancer Research Institute, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
5
|
Shikalov A, Koman I, Kogan NM. Targeted Glioma Therapy-Clinical Trials and Future Directions. Pharmaceutics 2024; 16:100. [PMID: 38258110 PMCID: PMC10820492 DOI: 10.3390/pharmaceutics16010100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/05/2024] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
Glioblastoma multiforme (GBM) is the most common type of glioma, with a median survival of 14.6 months post-diagnosis. Understanding the molecular profile of such tumors allowed the development of specific targeted therapies toward GBM, with a major role attributed to tyrosine kinase receptor inhibitors and immune checkpoint inhibitors. Targeted therapeutics are drugs that work by specific binding to GBM-specific or overexpressed markers on the tumor cellular surface and therefore contain a recognition moiety linked to a cytotoxic agent, which produces an antiproliferative effect. In this review, we have summarized the available information on the targeted therapeutics used in clinical trials of GBM and summarized current obstacles and advances in targeted therapy concerning specific targets present in GBM tumor cells, outlined efficacy endpoints for major classes of investigational drugs, and discussed promising strategies towards an increase in drug efficacy in GBM.
Collapse
Affiliation(s)
| | | | - Natalya M. Kogan
- Department of Molecular Biology, Institute of Personalized and Translational Medicine, Ariel University, Ariel 40700, Israel; (A.S.); (I.K.)
| |
Collapse
|
6
|
Bakhrushina EO, Mikhel IB, Buraya LM, Moiseev ED, Zubareva IM, Belyatskaya AV, Evzikov GY, Bondarenko AP, Krasnyuk II, Krasnyuk II. Implantation of In Situ Gelling Systems for the Delivery of Chemotherapeutic Agents. Gels 2024; 10:44. [PMID: 38247767 PMCID: PMC10815592 DOI: 10.3390/gels10010044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/30/2023] [Accepted: 01/02/2024] [Indexed: 01/23/2024] Open
Abstract
Implantation is a modern method of administering chemotherapeutic agents, with a highly targeted effect and better patient tolerance due to the low frequency of administration. Implants are capable of controlled release, which makes them a viable alternative to infusional chemotherapy, allowing patients to enjoy a better quality of life without the need for prolonged hospitalization. Compared to subcutaneous implantation, intratumoral implantation has a number of significant advantages in terms of targeting and side effects, but this area of chemotherapy is still poorly understood in terms of clinical trials. At the same time, there are more known developments of drugs in the form of implants and injections for intratumoral administration. The disadvantages of classical intratumoral implants are the need for surgical intervention to install the system and the increased risk of tumor rupture noted by some specialists. The new generation of implants are in situ implants-systems formed in the tumor due to a phase transition (sol-gel transition) under the influence of various stimuli. Among this systems some are highly selective for a certain type of malignant neoplasm. Such systems are injected and have all the advantages of intratumoral injections, but due to the phase transition occurring in situ, they form depot forms that allow the long-term release of chemotherapeutic agents.
Collapse
Affiliation(s)
- Elena O. Bakhrushina
- Department of Pharmaceutical Technology, A.P. Nelyubin Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119048, Russia; (E.O.B.); (L.M.B.); (E.D.M.); (I.M.Z.); (A.V.B.); (I.I.K.)
| | - Iosif B. Mikhel
- Department of Pharmaceutical Technology, A.P. Nelyubin Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119048, Russia; (E.O.B.); (L.M.B.); (E.D.M.); (I.M.Z.); (A.V.B.); (I.I.K.)
| | - Liliya M. Buraya
- Department of Pharmaceutical Technology, A.P. Nelyubin Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119048, Russia; (E.O.B.); (L.M.B.); (E.D.M.); (I.M.Z.); (A.V.B.); (I.I.K.)
| | - Egor D. Moiseev
- Department of Pharmaceutical Technology, A.P. Nelyubin Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119048, Russia; (E.O.B.); (L.M.B.); (E.D.M.); (I.M.Z.); (A.V.B.); (I.I.K.)
| | - Irina M. Zubareva
- Department of Pharmaceutical Technology, A.P. Nelyubin Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119048, Russia; (E.O.B.); (L.M.B.); (E.D.M.); (I.M.Z.); (A.V.B.); (I.I.K.)
- Department of Pharmacology, A.P. Nelyubin Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119048, Russia
| | - Anastasia V. Belyatskaya
- Department of Pharmaceutical Technology, A.P. Nelyubin Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119048, Russia; (E.O.B.); (L.M.B.); (E.D.M.); (I.M.Z.); (A.V.B.); (I.I.K.)
| | - Grigory Y. Evzikov
- Department of Nervous Diseases and Neurosurgery, N.V. Sklifosovsky Institute of Clinical Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119048, Russia;
| | | | - Ivan I. Krasnyuk
- Department of Analytical, Physical and Colloidal Chemistry, A.P. Nelyubin Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119048, Russia;
| | - Ivan I. Krasnyuk
- Department of Pharmaceutical Technology, A.P. Nelyubin Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119048, Russia; (E.O.B.); (L.M.B.); (E.D.M.); (I.M.Z.); (A.V.B.); (I.I.K.)
| |
Collapse
|
7
|
Qing X, Dou R, Wang P, Zhou M, Cao C, Zhang H, Qiu G, Yang Z, Zhang J, Liu H, Zhu S, Liu X. Ropivacaine-loaded hydrogels for prolonged relief of chemotherapy-induced peripheral neuropathic pain and potentiated chemotherapy. J Nanobiotechnology 2023; 21:462. [PMID: 38041074 PMCID: PMC10693114 DOI: 10.1186/s12951-023-02230-5] [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: 07/20/2023] [Accepted: 11/20/2023] [Indexed: 12/03/2023] Open
Abstract
Chemotherapy can cause severe pain for patients, but there are currently no satisfactory methods of pain relief. Enhancing the efficacy of chemotherapy to reduce the side effects of high-dose chemotherapeutic drugs remains a major challenge. Moreover, the treatment of chemotherapy-induced peripheral neuropathic pain (CIPNP) is separate from chemotherapy in the clinical setting, causing inconvenience to cancer patients. In view of the many obstacles mentioned above, we developed a strategy to incorporate local anesthetic (LA) into a cisplatin-loaded PF127 hydrogel for painless potentiated chemotherapy. We found that multiple administrations of cisplatin-loaded PF127 hydrogels (PFC) evoked severe CIPNP, which correlated with increased pERK-positive neurons in the dorsal root ganglion (DRG). However, incorporating ropivacaine into the PFC relieved PFC-induced CIPNP for more than ten hours and decreased the number of pERK-positive neurons in the DRG. Moreover, incorporating ropivacaine into the PFC for chemotherapy is found to upregulate major histocompatibility complex class I (MHC-I) expression in tumor cells and promote the infiltration of cytotoxic T lymphocytes (CD8+ T cells) in tumors, thereby potentiating chemotherapy efficacy. This study proposes that LA can be used as an immunemodulator to enhance the effectiveness of chemotherapy, providing new ideas for painless cancer treatment.
Collapse
Affiliation(s)
- Xin Qing
- Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Renbin Dou
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Peng Wang
- Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Mengni Zhou
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Chenchen Cao
- Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Huiwen Zhang
- Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Gaolin Qiu
- Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Zhilai Yang
- Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Jiqian Zhang
- Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230032, China.
| | - Hu Liu
- Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230032, China.
| | - Shasha Zhu
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China.
| | - Xuesheng Liu
- Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
8
|
Huang J, Yu P, Liao M, Dong X, Xu J, Ming J, Bin D, Wang Y, Zhang F, Xia Y. A self-charging salt water battery for antitumor therapy. SCIENCE ADVANCES 2023; 9:eadf3992. [PMID: 37000876 PMCID: PMC10065443 DOI: 10.1126/sciadv.adf3992] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 02/27/2023] [Indexed: 06/19/2023]
Abstract
Implantable devices on the tumor tissue as a local treatment are able to work in situ, which minimizes systemic toxicities and adverse effects. Here, we demonstrated an implantable self-charging battery that can regulate tumor microenvironment persistently by the well-designed electrode redox reaction. The battery consists of biocompatible polyimide electrode and zinc electrode, which can consume oxygen sustainably during battery discharge/self-charge cycle, thus modulating hypoxia level in tumor microenvironment. The oxygen reduction in battery leads to the formation of reactive oxygen species, showing 100% prevention on tumor formation. Sustainable consumption of oxygen causes adequate intratumoral hypoxic conditions over the course of 14 days, which is helpful for the hypoxia-activated prodrugs (HAPs) to kill tumor cells. The synergistic effect of the battery/HAPs can deliver more than 90% antitumor rate. Using redox reactions in electrochemical battery provides a potential approach for the tumor inhibition and regulation of tumor microenvironment.
Collapse
Affiliation(s)
- Jianhang Huang
- Department of Chemistry and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, iChEM (Collaborative Innovation Center of Chemistry for Energy Materials), Fudan University, Shanghai 200433, China
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, College of Chemistry and Materials Science, Zhejiang Normal University, Jinhua 321004, China
| | - Peng Yu
- Department of Chemistry and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, iChEM (Collaborative Innovation Center of Chemistry for Energy Materials), Fudan University, Shanghai 200433, China
| | - Mochou Liao
- Department of Chemistry and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, iChEM (Collaborative Innovation Center of Chemistry for Energy Materials), Fudan University, Shanghai 200433, China
| | - Xiaoli Dong
- Department of Chemistry and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, iChEM (Collaborative Innovation Center of Chemistry for Energy Materials), Fudan University, Shanghai 200433, China
| | - Jie Xu
- Department of Chemistry and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, iChEM (Collaborative Innovation Center of Chemistry for Energy Materials), Fudan University, Shanghai 200433, China
| | - Jiang Ming
- Department of Chemistry and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, iChEM (Collaborative Innovation Center of Chemistry for Energy Materials), Fudan University, Shanghai 200433, China
| | - Duan Bin
- Department of Chemistry and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, iChEM (Collaborative Innovation Center of Chemistry for Energy Materials), Fudan University, Shanghai 200433, China
| | - Yonggang Wang
- Department of Chemistry and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, iChEM (Collaborative Innovation Center of Chemistry for Energy Materials), Fudan University, Shanghai 200433, China
| | - Fan Zhang
- Department of Chemistry and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, iChEM (Collaborative Innovation Center of Chemistry for Energy Materials), Fudan University, Shanghai 200433, China
| | - Yongyao Xia
- Department of Chemistry and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, iChEM (Collaborative Innovation Center of Chemistry for Energy Materials), Fudan University, Shanghai 200433, China
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, College of Chemistry and Materials Science, Zhejiang Normal University, Jinhua 321004, China
| |
Collapse
|
9
|
Bai P, Fan T, Sun G, Wang X, Zhao L, Zhong R. The dual role of DNA repair protein MGMT in cancer prevention and treatment. DNA Repair (Amst) 2023; 123:103449. [PMID: 36680944 DOI: 10.1016/j.dnarep.2023.103449] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 12/21/2022] [Accepted: 01/09/2023] [Indexed: 01/15/2023]
Abstract
Alkylating agents are genotoxic chemicals that can induce and treat various types of cancer. This occurs through covalent bonding with cellular macromolecules, in particular DNA, leading to the loss of functional integrity under the persistence of modifications upon replication. O6-alkylguanine (O6-AlkylG) adducts are proposed to be the most potent DNA lesions induced by alkylating agents. If not repaired correctly, these adducts can result, at the molecular level, in DNA point mutations, chromosome aberrations, recombination, crosslinking, and single- and double-strand breaks (SSB/DSBs). At the cellular level, these lesions can result in malignant transformation, senescence, or cell death. O6-methylguanine-DNA methyltransferase (MGMT) is a DNA repair protein capable of removing the alkyl groups from O6-AlkylG adducts in a damage reversal process that can prevent the adverse biological effects of DNA damage caused by guanine O6-alkylation. MGMT can thereby defend normal cells against tumor initiation, however it can also protect tumor cells against the beneficial effects of chemotherapy. Hence, MGMT can play an important role in both the prevention and treatment of cancer; thus, it can be considered as a double-edged sword. From a clinical perspective, MGMT is a therapeutic target, and it is important to explore the rational development of its clinical exploitation.
Collapse
Affiliation(s)
- Peiying Bai
- Beijing Key Laboratory of Environmental and Viral Oncology, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China
| | - Tengjiao Fan
- Beijing Key Laboratory of Environmental and Viral Oncology, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China; Department of Medical Technology, Beijing Pharmaceutical University of Staff and Workers, Beijing 100079, China
| | - Guohui Sun
- Beijing Key Laboratory of Environmental and Viral Oncology, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China.
| | - Xin Wang
- Department of Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100029, China
| | - Lijiao Zhao
- Beijing Key Laboratory of Environmental and Viral Oncology, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China
| | - Rugang Zhong
- Beijing Key Laboratory of Environmental and Viral Oncology, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China
| |
Collapse
|
10
|
He P, Xu S, Guo Z, Yuan P, Liu Y, Chen Y, Zhang T, Que Y, Hu Y. Pharmacodynamics and pharmacokinetics of PLGA-based doxorubicin-loaded implants for tumor therapy. Drug Deliv 2022; 29:478-488. [PMID: 35147071 PMCID: PMC8843208 DOI: 10.1080/10717544.2022.2032878] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 01/14/2022] [Accepted: 01/17/2022] [Indexed: 02/08/2023] Open
Abstract
The traditional systemic chemotherapy through intravenous infusion of doxorubicin (DOX) has many side effects. The aim of this study was to develop a PLGA-based DOX-loaded implant and to evaluate the efficacy and drug metabolism distribution of the implant in intratumoral chemotherapy for osteosarcoma (OS). In this study, implants containing DOX, poly(d,l-lactide-co-glycolide), and polyethylene glycol 4000 were prepared by melt-molding method. Then, the antitumor activity and systemic drug distribution of the implants were tested in a K7M2 OS bearing mouse model. The scanning electron microscope images showed that DOX was uniformly dispersed in the polymer matrix. Both the in vitro and in vivo release profiles of implants are characterized by three-phase release. Implantation of DOX-loaded implants into tumors can inhibit tumor growth in a dose-dependent manner. The pharmacokinetic behavior shows that intratumor chemotherapy through implants has a much higher drug concentration in tumors than in normal tissues, which may be the reason for improving antitumor activity and reducing systemic side effects. In summary, the drug release of the implants prepared in this study is sustained and stable, which promotes long-term local accumulation of drugs in tumors, improves the efficacy of chemotherapy and has low toxicity to normal tissues.
Collapse
Affiliation(s)
- Peng He
- Department of Orthopedics, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Shenglin Xu
- Department of Orthopedics, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zehao Guo
- Department of Orthopedics, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Peng Yuan
- Department of Orthopedics, Fuyang Hospital of Anhui Medical University, Fuyang, China
| | - Yulei Liu
- Department of Orthopedics, Fuyang Hospital of Anhui Medical University, Fuyang, China
| | - Yu Chen
- Department of Pharmacy, Anqing Medical College, Anqing, China
| | - Tiantian Zhang
- Laboratory of Pharmaceutical Research, Anhui Zhongren Science and Technology Co., Ltd, Hefei, China
| | - Yukang Que
- Department of Orthopedics, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yong Hu
- Department of Orthopedics, First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
11
|
Rakib Hasan Khan M, Shankar Hazra R, Nair G, Mohammad J, Jiang L, Reindl K, Khalid Jawed M, Ganai S, Quadir M. Cellulose nanofibers as Scaffold-forming materials for thin film drug delivery systems. Int J Pharm 2022; 627:122189. [PMID: 36100147 DOI: 10.1016/j.ijpharm.2022.122189] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 09/02/2022] [Accepted: 09/05/2022] [Indexed: 10/31/2022]
Abstract
We explored the potential of cellulose nanofiber (CNF) for designing prolonged-release, thin-film drug delivery systems (TF-DDS). These delivery systems can be used as locally deployable drug-releasing scaffolds for achieving spatial and temporal control over therapeutic concentration in target tissues. Using doxorubicin (DOX) as a model anticancer drug, CNF-based TF-DDS were prepared using different film-formation processes, such as solvent casting and lyophilization. Formulations were prepared with or without the incorporation of additional macromolecular additives, such as gelatin, to include further biomechanical functionality. We studied the films for their mechanical properties, thermal stability, wettability, porosity and in vitro drug release properties. Our experimental results showed that CNF-based films, when prepared via solvent casting method, showed optimized performance in terms of DOX loading, and prolonged-release than those prepared via lyophilization-based fabrication processes. Scanning electron microscopy (SEM) analysis of the CNF-based films showed uniform distribution of fiber entanglement, which provided the scaffolds with sufficient porosity and tortuosity contributing to the sustained release of the drug from the delivery system. We also observed that surface layering of gelatin on CNF films via dip-coating significantly increased the mechanical strength and reduced the wettability of the films, and as such, affected drug release kinetics. The performance of the TF-DDS was evaluated in-vitro against two pancreatic cancer cell lines, i.e. MIA PaCa-2 and PANC-1. We observed that, along with the enhancement of mean dissolution time (MDT) of DOX, CNF-based TF-DDS were able to suppress the proliferation of pancreatic cancer cells in a time-dependent fashion, indicating that the drug liberated from the films were therapeutically active against cancer cells. Additionally, TF-DDS were also tested ex-vivo on patient-derived xenograft (PDX) model of pancreatic ductal adenocarcinoma (PDAC). We observed that DOX released from the TF-DDS was able to reduce Ki-67 positive, pancreatic cancer cells in these models.
Collapse
Affiliation(s)
- Md Rakib Hasan Khan
- Biomedical Engineering Program, North Dakota State University, Fargo, ND 58105, USA
| | - Raj Shankar Hazra
- Materials and Nanotechnology Program, North Dakota State University, Fargo, ND 58105, USA
| | - Gauthami Nair
- Department of Biological Sciences, North Dakota State University, Fargo, ND 58102, USA
| | - Jiyan Mohammad
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58108, USA
| | - Long Jiang
- Department of Mechanical Engineering, North Dakota State University, Fargo, ND 58105, USA
| | - Katie Reindl
- Department of Biological Sciences, North Dakota State University, Fargo, ND 58102, USA
| | - Mohammad Khalid Jawed
- Department of Mechanical & Aerospace Engineering, University of California, Los Angeles, CA 90095, USA
| | - Sabha Ganai
- Division of Surgical Oncology, Sanford Research, Fargo, ND 58122, USA
| | - Mohiuddin Quadir
- Biomedical Engineering Program, North Dakota State University, Fargo, ND 58105, USA; Materials and Nanotechnology Program, North Dakota State University, Fargo, ND 58105, USA; Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, ND 58102, USA.
| |
Collapse
|
12
|
Sarkar K, Torregrossa-Allen SE, Elzey BD, Narayanan S, Langer MP, Durm GA, Won YY. Effect of Paclitaxel Stereochemistry on X-ray-Triggered Release of Paclitaxel from CaWO 4/Paclitaxel-Coloaded PEG-PLA Nanoparticles. Mol Pharm 2022; 19:2776-2794. [PMID: 35834797 PMCID: PMC11975462 DOI: 10.1021/acs.molpharmaceut.2c00148] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
For many locally advanced tumors, the chemotherapy-radiotherapy (CT-RT) combination ("chemoradiation") is currently the standard of care. Intratumoral (IT) CT-based chemoradiation has the potential to overcome the limitations of conventional systemic CT-RT (side effects). For maximizing the benefits of IT CT-RT, our laboratory has previously developed a radiation-controlled drug release formulation, in which anticancer drug paclitaxel (PTX) and radioluminescent CaWO4 (CWO) nanoparticles (NPs) are co-encapsulated with poly(ethylene glycol)-poly(lactic acid) (PEG-PLA) block copolymers ("PEG-PLA/CWO/PTX NPs"). These PEG-PLA/CWO/PTX NPs enable radiation-controlled release of PTX and are capable of producing sustained therapeutic effects lasting for at least one month following a single IT injection. The present article focuses on discussing our recent finding about the effect of the stereochemical structure of PTX on the efficacy of this PEG-PLA/CWO/PTX NP formulation. Stereochemical differences in two different PTX compounds ("PTX-S" from Samyang Biopharmaceuticals and "PTX-B" from Biotang) were characterized by 2D heteronuclear/homonuclear NMR, Raman spectroscopy, and circular dichroism measurements. The difference in PTX stereochemistry was found to significantly influence their water solubility (WS); PTX-S (WS ≈ 4.69 μg/mL) is about 19 times more water soluble than PTX-B (WS ≈ 0.25 μg/mL). The two PTX compounds showed similar cancer cell-killing performances in vitro when used as free drugs. However, the subtle stereochemical difference significantly influenced their X-ray-triggered release kinetics from the PEG-PLA/CWO/PTX NPs; the more water-soluble PTX-S was released faster than the less water-soluble PTX-B. This difference was manifested in the IT pharmacokinetics and eventually in the survival percentages of test animals (mice) treated with PEG-PLA/CWO/PTX NPs + X-rays in an in vivo human tumor xenograft study; at short times (<1 month), concurrent PEG-PLA/CWO/PTX-S NPs produced a greater tumor-suppression effect, whereas PEG-PLA/CWO/PTX-B NPs had a longer-lasting radio-sensitizing effect. This study demonstrates the importance of the stereochemistry of a drug in a therapy based on a controlled release formulation.
Collapse
Affiliation(s)
- Kaustabh Sarkar
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN 47907, USA
| | | | - Bennett D. Elzey
- Purdue University Center of Cancer Research, West Lafayette, IN 47907, USA
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN 47907, USA
| | - Sanjeev Narayanan
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN 47907, USA
| | - Mark P. Langer
- Department of Radiation Oncology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Gregory A. Durm
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - You-Yeon Won
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN 47907, USA
- Purdue University Center of Cancer Research, West Lafayette, IN 47907, USA
| |
Collapse
|
13
|
Kefayat A, Hosseini M, Ghahremani F, Jolfaie NA, Rafienia M. Biodegradable and biocompatible subcutaneous implants consisted of pH-sensitive mebendazole-loaded/folic acid-targeted chitosan nanoparticles for murine triple-negative breast cancer treatment. J Nanobiotechnology 2022; 20:169. [PMID: 35361226 PMCID: PMC8973744 DOI: 10.1186/s12951-022-01380-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 03/17/2022] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Mebendazole (MBZ) is a well-known anti-parasite drug with significant anti-cancer properties. However, MBZ exhibits low solubility, limited absorption efficacy, extensive first-pass effect, and low bioavailability. Therefore, multiple oral administration of high dose MBZ is required daily for achieving the therapeutic serum level which can cause severe side effects and patients' non-compliance. METHOD In the present study, MBZ-loaded/folic acid-targeted chitosan nanoparticles (CS-FA-MBZ) were synthesized, characterized, and used to form cylindrical subcutaneous implants for 4T1 triple-negative breast tumor (TNBC) treatment in BALB/c mice. The therapeutic efficacy of the CS-FA-MBZ implants was investigated after subcutaneous implantation in comparison with Control, MBZ (40 mg/kg, oral administration, twice a week for 2 weeks), and CS-FA implants, according to 4T1 tumors' growth progression, metastasis, and tumor-bearing mice survival time. Also, their biocompatibility was evaluated by blood biochemical analyzes and histopathological investigation of vital organs. RESULTS The CS-FA-MBZ implants were completely degraded 15 days after implantation and caused about 73.3%, 49.2%, 57.4% decrease in the mean tumors' volume in comparison with the Control (1050.5 ± 120.7 mm3), MBZ (552.4 ± 76.1 mm3), and CS-FA (658.3 ± 88.1 mm3) groups, respectively. Average liver metastatic colonies' number per microscope field at the CS-FA-MBZ group (2.3 ± 0.7) was significantly (P < 0.05) lower than the Control (9.6 ± 1.7), MBZ (5.0 ± 1.5), and CS-FA (5.2 ± 1) groups. In addition, the CS-FA-MBZ treated mice exhibited about 52.1%, 27.3%, and 17% more survival days after the cancer cells injection in comparison with the Control, MBZ, and CS-FA groups, respectively. Moreover, the CS-FA-MBZ implants were completely biocompatible based on histopathology and blood biochemical analyzes. CONCLUSION Taking together, CS-FA-MBZ implants were completely biodegradable and biocompatible with high therapeutic efficacy in a murine TNBC model.
Collapse
Affiliation(s)
- Amirhosein Kefayat
- Cancer Prevention Research Center, Department of Oncology, Isfahan University of Medical Sciences, 81746-73461, Isfahan, Iran.,Biosensor Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Maryam Hosseini
- Department of Chemistry, Amirkabir University of Technology (Tehran Polytechnic), 1591634311, Tehran, Iran
| | - Fatemeh Ghahremani
- Department of Medical Physics and Radiotherapy, School of Paramedicine, Arak University of Medical Sciences, Arak, Iran
| | - Nafise Arbab Jolfaie
- Biosensor Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Rafienia
- Biosensor Research Center, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
14
|
Recent advancements of electrospun nanofibers for cancer therapy. Polym Bull (Berl) 2022. [DOI: 10.1007/s00289-022-04153-x] [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]
|
15
|
Response to the Comment on "Does Thermal Ablation Increase or Decrease the Risk of Tumor Local Recurrence?". Ann Surg 2021; 274:e692-e693. [PMID: 32427765 DOI: 10.1097/sla.0000000000003873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
16
|
Anisimova N, Kiselevskiy M, Martynenko N, Willumeit-Römer R, Kornyushenkov E, Rodionov M, Dobatkin S, Estrin Y. Anti-tumour activity of Mg-6%Ag and Mg-10%Gd alloys in mice with inoculated melanoma. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 130:112464. [PMID: 34702539 DOI: 10.1016/j.msec.2021.112464] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 09/04/2021] [Accepted: 09/24/2021] [Indexed: 10/20/2022]
Abstract
Cytotoxicity in vitro and anti-tumour activity in vivo of magnesium alloys Mg-6%Ag and Mg-10%Gd was studied. It was shown that specifically designed and thermomechanically processed Mg alloys produced a sizeable cytotoxic effect on PC-3 line tumour cells in vitro through inhibition of their proliferation. A pronounced grain refinement in combination with the formation of second phases and precipitates attained by means of equal-channel angular pressing (ECAP) accellerated the degradation and gave rise to enhanced anti-tumour activity of both alloys. The gadolinium-containing alloy outperformed the silver-containing one substantially. In an in vivo assay, intratumoural implantation of pins made from both alloys in the homogenised and the ECAP states in mice with inoculated B16 melanoma gave rise to an indubitable anti-tumour effect. It was documented in a decrease of Ki-67(+) cells and the occurrence of regions of destruction of tumoural tissue that were filled with gas evolving in the course of biodegradation of the implants. The data obtained suggest that intratumoural implantation of gadolinium-containing magnesium alloys can be considered for therapy of inoperable or chemoresistant forms of cancer.
Collapse
Affiliation(s)
- Natalia Anisimova
- N.N. Blokhin National Medical Research Center of Oncology of the Ministry of Health of the Russian Federation (N.N. Blokhin NMRCO), Moscow, Russia; National University of Science and Technology "MISIS", Moscow, Russia
| | - Mikhail Kiselevskiy
- N.N. Blokhin National Medical Research Center of Oncology of the Ministry of Health of the Russian Federation (N.N. Blokhin NMRCO), Moscow, Russia; National University of Science and Technology "MISIS", Moscow, Russia
| | - Natalia Martynenko
- A.A. Baikov Institute of Metallurgy and Materials Science of the RAS, Moscow, Russia.
| | - Regine Willumeit-Römer
- Institute of Metallic Biomaterials, Helmholtz-Zentrum Geesthacht (HZG), Geesthacht, Germany
| | - Evgeniy Kornyushenkov
- N.N. Blokhin National Medical Research Center of Oncology of the Ministry of Health of the Russian Federation (N.N. Blokhin NMRCO), Moscow, Russia
| | - Maxim Rodionov
- N.N. Blokhin National Medical Research Center of Oncology of the Ministry of Health of the Russian Federation (N.N. Blokhin NMRCO), Moscow, Russia
| | - Sergey Dobatkin
- National University of Science and Technology "MISIS", Moscow, Russia; A.A. Baikov Institute of Metallurgy and Materials Science of the RAS, Moscow, Russia
| | - Yuri Estrin
- Department of Materials Science and Engineering, Monash University, Clayton, Australia; Department of Mechanical Engineering, The University of Western Australia, Nedlands, Australia
| |
Collapse
|
17
|
Hazra RS, Dutta D, Mamnoon B, Nair G, Knight A, Mallik S, Ganai S, Reindl K, Jiang L, Quadir M. Polymeric Composite Matrix with High Biobased Content as Pharmaceutically Relevant Molecular Encapsulation and Release Platform. ACS APPLIED MATERIALS & INTERFACES 2021; 13:40229-40248. [PMID: 34423963 DOI: 10.1021/acsami.1c03805] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Drug delivery systems (DDS) that can temporally control the rate and extent of release of therapeutically active molecules find applications in many clinical settings, ranging from infection control to cancer therapy. With an aim to design a locally implantable, controlled-release DDS, we demonstrated the feasibility of using cellulose nanocrystal (CNC)-reinforced poly (l-lactic acid) (PLA) composite beads. The performance of the platform was evaluated using doxorubicin (DOX) as a model drug for applications in triple-negative breast cancer. A facile, nonsolvent-induced phase separation (NIPS) method was adopted to form composite beads. We observed that CNC loading within these beads played a critical role in the mechanical stability, porosity, water uptake, diffusion, release, and pharmacological activity of the drug from the delivery system. When loaded with DOX, composite beads significantly controlled the release of the drug in a pH-dependent pattern. For example, PLA/CNC beads containing 37.5 wt % of CNCs showed a biphasic release of DOX, where 41 and 82% of the loaded drug were released at pH 7.4 and pH 5.5, respectively, over 7 days. Drug release followed Korsmeyer's kinetics, indicating that the release mechanism was mostly diffusion and swelling-controlled. We showed that DOX released from drug-loaded PLA/CNC composite beads locally suppressed the growth and proliferation of triple-negative breast cancer cells, MBA-MB-231, via the apoptotic pathway. The efficacy of the DDS was evaluated in human tissue explants. We envision that such systems will find applications for designing biobased platforms with programmed stability and drug delivery functions.
Collapse
Affiliation(s)
- Raj Shankar Hazra
- Department of Mechanical Engineering, North Dakota State University, Fargo, North Dakota 58108, United States
- Materials and Nanotechnology Program, North Dakota State University, Fargo, North Dakota 58108, United States
| | - Debasmita Dutta
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, North Dakota 58108, United States
| | - Babak Mamnoon
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58108, United States
| | - Gauthami Nair
- Department of Biological Sciences, North Dakota State University, Fargo, North Dakota 58108, United States
| | - Austin Knight
- Department of Mechanical Engineering, North Dakota State University, Fargo, North Dakota 58108, United States
| | - Sanku Mallik
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58108, United States
| | - Sabha Ganai
- Division of Surgical Oncology, Sanford Research, Fargo, North Dakota 58122, United States
| | - Katie Reindl
- Department of Biological Sciences, North Dakota State University, Fargo, North Dakota 58108, United States
| | - Long Jiang
- Department of Mechanical Engineering, North Dakota State University, Fargo, North Dakota 58108, United States
- Materials and Nanotechnology Program, North Dakota State University, Fargo, North Dakota 58108, United States
| | - Mohiuddin Quadir
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, North Dakota 58108, United States
- Materials and Nanotechnology Program, North Dakota State University, Fargo, North Dakota 58108, United States
| |
Collapse
|
18
|
Shape Memory Alloy Capsule Micropump for Drug Delivery Applications. MICROMACHINES 2021; 12:mi12050520. [PMID: 34066315 PMCID: PMC8148152 DOI: 10.3390/mi12050520] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/17/2021] [Accepted: 04/27/2021] [Indexed: 11/17/2022]
Abstract
We introduce a shape memory alloy (SMA) actuated micropump optimized for drug delivery applications. The proposed novel design integrates a built-in replaceable drug reservoir within the pump package forming a self-contained preloaded capsule pump with an overall pump volume of 424.7 μL. The new design results in a compact, simple, and inexpensive micropump and reduces the probability of contamination with attained almost zero dead volume values. The pump consists of NiTi-alloy SMA wires coiled on a flexible polymeric enclosure and actuated by joule heating. Unlike diaphragm and peristaltic SMA micropump designs that actuate transversely, our design is actuated longitudinally along the direction of the highest mechanical compliance resulting in large strokes in the order of 5.6 mm at 27% deflection ratio, actuation speed up to 11 mm/s, and static head pressures up to 14 kPa (105 mmHg) at 7.1 W input power; thus, high throughputs exceeding 2524 μL/min under free convention conditions could be achieved. A model was developed to optimize the pump’s geometrical parameters and the enclosure material. The model concluded that low stiffness enclosure material combined with thinner SMA wire diameter would result in the maximum deflection at the lowest power rating. To prove its viability for drug delivery applications, the pump was operated at a constant discharge volume at a relatively constant static head pressure. Furthermore, a design of bicuspid-inspired polymeric check-valves is presented and integrated onto the pump to regulate the flow. Since the built-in reservoir is replaceable, the pump capsule can be reused multiple times and for multiple drug types.
Collapse
|
19
|
Wu C, Yi X, Xu R, Zhang M, Xu Y, Ma Y, Gao L, Zha Z. Biodistribution of etoposide via intratumoral chemotherapy with etoposide-loaded implants. Drug Deliv 2021; 27:974-982. [PMID: 32611260 PMCID: PMC8216434 DOI: 10.1080/10717544.2020.1787558] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Etoposide (VP16) is the traditional antitumor agent which has been widely used in a variety of cancers. However, intravenous administration of VP16 was limited in clinical application because of its low aqueous solubility, poor bioavailability and dose-limiting adverse effects. Local chemotherapy with VP16-loaded drug delivery systems could provide a continuous release of drug at the target site, while minimizing the systemic toxicity. In this study, we prepared the poly-l-lactic acid (PLLA) based VP16-loaded implants (VP16 implants) by the direct compression method. The VP16 implants were characterized with regards to drug content, micromorphology, drug release profiles, differential scanning calorimetry (DSC) and Fourier transform infrared spectroscopy (FTIR) analyses. Furthermore, the biodistribution of VP16 via intratumoral chemotherapy with VP16 implants was investigated using the murine Lewis lung carcinoma model. Our results showed that VP16 dispersed homogenously in the polymeric matrix. Both in vitro and in vivo drug release profiles of the implants were characterized by high initial burst release followed by sustained release of VP16. The VP16 implants showed good compatibility between VP16 and the excipients. Intratumoral chemotherapy with VP16 implants resulted in significantly higher concentration and longer duration of VP16 in tumor tissues compared with single intraperitoneal injection of VP16 solution. Moreover, we found the low level of VP16 in plasma and normal organ tissues. These results suggested that intratumoral chemotherapy with VP16 implants enabled high drug concentration at the target site and has the potential to be used as a novel method to treat cancer.
Collapse
Affiliation(s)
- Chunsheng Wu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, PR China
| | - Xiangting Yi
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, PR China
| | - Renzhi Xu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, PR China
| | - Maokuan Zhang
- Laboratory of Pharmaceutical Research, Anhui Zhongren Science and Technology Co., Ltd, Hefei, PR China
| | - Yan Xu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, PR China
| | - Yan Ma
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, PR China
| | - Li Gao
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, PR China
| | - Zhengbao Zha
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, PR China
| |
Collapse
|
20
|
Ray P, Kale N, Quadir M. New side chain design for pH-responsive block copolymers for drug delivery. Colloids Surf B Biointerfaces 2021; 200:111563. [DOI: 10.1016/j.colsurfb.2021.111563] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 12/30/2020] [Accepted: 01/02/2021] [Indexed: 01/12/2023]
|
21
|
Saw WS, Anasamy T, Foo YY, Kwa YC, Kue CS, Yeong CH, Kiew LV, Lee HB, Chung LY. Delivery of Nanoconstructs in Cancer Therapy: Challenges and Therapeutic Opportunities. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202000206] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Wen Shang Saw
- Department of Pharmaceutical Chemistry Faculty of Pharmacy University of Malaya Kuala Lumpur 50603 Malaysia
| | - Theebaa Anasamy
- Department of Pharmaceutical Chemistry Faculty of Pharmacy University of Malaya Kuala Lumpur 50603 Malaysia
| | - Yiing Yee Foo
- Department of Pharmacology Faculty of Medicine University of Malaya Kuala Lumpur 50603 Malaysia
| | - Yee Chu Kwa
- Department of Pharmaceutical Chemistry Faculty of Pharmacy University of Malaya Kuala Lumpur 50603 Malaysia
| | - Chin Siang Kue
- Department of Diagnostic and Allied Health Sciences Faculty of Health and Life Sciences Management and Science University Shah Alam Selangor 40100 Malaysia
| | - Chai Hong Yeong
- School of Medicine Faculty of Health and Medical Sciences Taylor's University Subang Jaya Selangor 47500 Malaysia
| | - Lik Voon Kiew
- Department of Pharmacology Faculty of Medicine University of Malaya Kuala Lumpur 50603 Malaysia
| | - Hong Boon Lee
- Department of Pharmaceutical Chemistry Faculty of Pharmacy University of Malaya Kuala Lumpur 50603 Malaysia
- School of Biosciences Faculty of Health and Medical Sciences Taylor's University Subang Jaya Selangor 47500 Malaysia
| | - Lip Yong Chung
- Department of Pharmaceutical Chemistry Faculty of Pharmacy University of Malaya Kuala Lumpur 50603 Malaysia
| |
Collapse
|
22
|
Modelling of combination therapy using implantable anticancer drug delivery with thermal ablation in solid tumor. Sci Rep 2020; 10:19366. [PMID: 33168846 PMCID: PMC7653950 DOI: 10.1038/s41598-020-76123-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 10/22/2020] [Indexed: 12/20/2022] Open
Abstract
Local implantable drug delivery system (IDDS) can be used as an effective adjunctive therapy for solid tumor following thermal ablation for destroying the residual cancer cells and preventing the tumor recurrence. In this paper, we develop comprehensive mathematical pharmacokinetic/pharmacodynamic (PK/PD) models for combination therapy using implantable drug delivery system following thermal ablation inside solid tumors with the help of molecular communication paradigm. In this model, doxorubicin (DOX)-loaded implant (act as a transmitter) is assumed to be inserted inside solid tumor (acts as a channel) after thermal ablation. Using this model, we can predict the extracellular and intracellular concentration of both free and bound drugs. Also, Impact of the anticancer drug on both cancer and normal cells is evaluated using a pharmacodynamic (PD) model that depends on both the spatiotemporal intracellular concentration as well as characteristics of anticancer drug and cells. Accuracy and validity of the proposed drug transport model is verified with published experimental data in the literature. The results show that this combination therapy results in high therapeutic efficacy with negligible toxicity effect on the normal tissue. The proposed model can help in optimize development of this combination treatment for solid tumors, particularly, the design parameters of the implant.
Collapse
|
23
|
Pizzuti VJ, Viswanath D, Torregrosa-Allen SE, Currie MP, Elzey BD, Won YY. Bilirubin-Coated Radioluminescent Particles for Radiation-Induced Photodynamic Therapy. ACS APPLIED BIO MATERIALS 2020; 3:4858-4872. [PMID: 35021730 DOI: 10.1021/acsabm.0c00354] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Vincenzo J. Pizzuti
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Dhushyanth Viswanath
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Sandra E. Torregrosa-Allen
- Purdue University Center for Cancer Research, West Lafayette, Indiana 47906, United States
- Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana 47907, United States
| | - Melanie P. Currie
- Purdue University Center for Cancer Research, West Lafayette, Indiana 47906, United States
- Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana 47907, United States
| | - Bennett D. Elzey
- Purdue University Center for Cancer Research, West Lafayette, Indiana 47906, United States
- Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana 47907, United States
| | - You-Yeon Won
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
- Purdue University Center for Cancer Research, West Lafayette, Indiana 47906, United States
| |
Collapse
|
24
|
Bender LH, Abbate F, Walters IB. Intratumoral Administration of a Novel Cytotoxic Formulation with Strong Tissue Dispersive Properties Regresses Tumor Growth and Elicits Systemic Adaptive Immunity in In Vivo Models. Int J Mol Sci 2020; 21:E4493. [PMID: 32599852 PMCID: PMC7349938 DOI: 10.3390/ijms21124493] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/10/2020] [Accepted: 06/21/2020] [Indexed: 02/06/2023] Open
Abstract
The recent development of immune-based therapies has improved the outcome for cancer patients; however, adjuvant therapies remain an important line of treatment for several cancer types. To maximize efficacy, checkpoint inhibitors are often combined with cytotoxic agents. While this approach often leads to increased tumor regression, higher off target toxicity often results in certain patients. This report describes a novel formulation comprising a unique amphiphilic molecule, 8-((2-hydroxybenzoyl)amino)octanoate (SHAO), that non-covalently interacts with payloads to increase drug dispersion and diffusion when dosed intratumorally (IT) into solid tumors. SHAO is co-formulated with cisplatin and vinblastine (referred to as INT230-6). IT dosing of the novel formulation achieved greater tumor growth inhibition and improved survival in in vivo tumor models compared to the same drugs without enhancer given intravenously or IT. INT230-6 treatment increased immune infiltrating cells in injected tumors with 10% to 20% of the animals having complete responses and developing systemic immunity to the cancer. INT230-6 was also shown to be synergistic with programmed cell death protein 1 (PD-1) antibodies at improving survival and increasing complete responses. INT230-6 induced significant tumor necrosis potentially releasing antigens to induce the systemic immune-based anti-cancer attack. This research demonstrates a novel, local treatment approach for cancer that minimizes systemic toxicity while stimulating adaptive immunity.
Collapse
Affiliation(s)
- Lewis H. Bender
- Intensity Therapeutics, Inc., Westport, CT 06880, USA; (F.A.); (I.B.W.)
| | | | | |
Collapse
|
25
|
Gawley M, Almond L, Daniel S, Lastakchi S, Kaur S, Detta A, Cruickshank G, Miller R, Hingtgen S, Sheets K, McConville C. Development and in vivo evaluation of Irinotecan-loaded Drug Eluting Seeds (iDES) for the localised treatment of recurrent glioblastoma multiforme. J Control Release 2020; 324:1-16. [PMID: 32407745 DOI: 10.1016/j.jconrel.2020.05.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 04/01/2020] [Accepted: 05/06/2020] [Indexed: 12/12/2022]
Abstract
Glioblastoma multiforme (GBM) is impossible to fully remove surgically and almost always recurs at the borders of the resection cavity, while systemic delivery of therapeutic drug levels to the brain tumour is limited by the blood-brain barrier. This research describes the development of a novel formulation of Irinotecan-loaded Drug Eluting Seeds (iDES) for insertion into the margin of the GBM resection cavity to provide a sustained high local dose with reduced systemic toxicities. We used primary GBM cells from both the tumour core and Brain Around the Tumour tissue from recurrent GBM patients to demonstrate that irinotecan is more effective than temozolomide. Irinotecan had a 75% response rate, while only 50% responded to temozolomide. With temozolomide the cell viability was never below 80% whereas irinotecan achieved cell viabilities of less than 44%. The iDES were manufactured using a hot melt extrusion process with accurate irinotecan drug loadings and the same cytotoxicity as unformulated irinotecan. The iDES released irinotecan in a sustained fashion for up to 7 days. However, only the 30, 40 and 50% w/w loaded iDES formulations released the 300 to 1000 μg of irinotecan needed to be effective in vivo. The 30 and 40% w/w iDES formulations containing 10% plasticizer and either 60 or 50% PLGA prolonged survival from 27 to 70 days in a GBM xenograft mouse resection model with no sign of tumour recurrence. The 30% w/w iDES formulations showed equivalent toxicity to a placebo in non-tumour bearing mice. This innovative drug delivery approach could transform the treatment of recurrent GBM patients by improving survival and reducing toxicity.
Collapse
Affiliation(s)
- Matthew Gawley
- School of Pharmacy, Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston B15 2TT, United Kingdom
| | - Lorna Almond
- School of Pharmacy, Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston B15 2TT, United Kingdom
| | - Senam Daniel
- School of Pharmacy, Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston B15 2TT, United Kingdom
| | - Sarah Lastakchi
- School of Pharmacy, Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston B15 2TT, United Kingdom
| | - Sharnjit Kaur
- School of Pharmacy, Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston B15 2TT, United Kingdom
| | - Allah Detta
- Department of Neurosurgery, University Hospitals Birmingham, NHS Foundation Trust, United Kingdom
| | - Garth Cruickshank
- Department of Neurosurgery, University Hospitals Birmingham, NHS Foundation Trust, United Kingdom
| | - Ryan Miller
- Division of Neuropathology, Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America; Departments of Neurology and Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States; Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Shawn Hingtgen
- Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kevin Sheets
- Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Christopher McConville
- School of Pharmacy, Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston B15 2TT, United Kingdom.
| |
Collapse
|
26
|
Padmakumar S, Menon D. Nanofibrous Polydioxanone Depots for Prolonged Intraperitoneal Paclitaxel Delivery. Curr Drug Deliv 2020; 16:654-662. [PMID: 31418659 DOI: 10.2174/1567201816666190816102949] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 01/11/2019] [Accepted: 07/19/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND Prolonged chemodrug delivery to the tumor site is a prerequisite to maintaining its localised therapeutic concentrations for effective treatment of malignant solid tumors. OBJECTIVE The current study aims to develop implantable polymeric depots through conventional electrospinning for sustained drug delivery, specifically to the peritoneum. METHODS Non-woven electrospun mats were fabricated by simple electrospinning of Polydioxanone solution loaded with the chemodrug, Paclitaxel. The implants were subjected to the analysis of morphology, mechanical properties, degradation and drug release in phosphate buffer and patient-derived peritoneal drain fluid samples. In vivo studies were conducted by surgical knotting of these implants to the peritoneal wall of healthy mice. RESULTS Non-woven electrospun mats with a thickness of 0.65±0.07 mm, weighing ~ 20 mg were fabricated by electrospinning 15 w/v% polymer loaded with 10 w/w% drug. These implants possessing good mechanical integrity showed a drug entrapment efficiency of 87.82±2.54 %. In vitro drug release studies in phosphate buffer showed a sustained profile for ~4 weeks with a burst of 10 % of total drug content, whereas this amounted to >60% in patient samples. Mice implanted with these depots remained healthy during the study period. The biphasic drug release profile obtained in vivo showed a slow trend, with peritoneal lavage and tissues retaining good drug concentrations for a sustained period. CONCLUSION The results indicate that non-woven electrospun mats developed from biodegradable Polydioxanone polymer can serve as ideal candidates for easily implantable drug depots to address the challenges of peritoneal metastasis in ovarian cancer.
Collapse
Affiliation(s)
- Smrithi Padmakumar
- Centre for Nanosciences & Molecular Medicine, Amrita Institute of Medical Sciences & Research Centre, Amrita Vishwa Vidyapeetham, Kochi, Kerala-682041, India
| | - Deepthy Menon
- Centre for Nanosciences & Molecular Medicine, Amrita Institute of Medical Sciences & Research Centre, Amrita Vishwa Vidyapeetham, Kochi, Kerala-682041, India
| |
Collapse
|
27
|
Wen A, Mei X, Feng C, Shen C, Wang B, Zhang X. Electrosprayed nanoparticles of poly(p-dioxanone-co-melphalan) macromolecular prodrugs for treatment of xenograft ovarian carcinoma. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 111:110759. [PMID: 32279799 DOI: 10.1016/j.msec.2020.110759] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 02/16/2020] [Accepted: 02/16/2020] [Indexed: 01/18/2023]
Abstract
Ovarian cancer is considered to be the most fatal reproductive cancers. Melphalan is used to treat ovarian cancer as an intraperitoneal chemotherapy agent. However, elucidating its pharmacokinetic behavior and preparing it for administration are challenging since it undergoes spontaneous hydrolysis. In this study, melphalan is transformed into a macromolecular prodrug by copolymerizing with p-dioxanone. The hydrophobicity of copolymer chains protects melphalan from hydrolysis. Poly(p-dioxanone-co-melphalan; PDCM) is electrosprayed and converted into nanoparticles (PDCM NPs) with diameters of ~300-350 nm to facilitate its intracellular delivery. UPLC-MS and HPLC are applied to verify and monitor the release of melphalan from PDCM NPs. PDCM NPs could suppress the proliferation of SKOV-3 cells. The IC50 of 4.3% melphalan-containing PDCM-3 NP was 70 mg/L, 72 h post administration. These suppression characteristics not only affected by the degradation and then the extracellular release of melphalan from PDCM NPs, but also the uptake via phagocytosis phenomenon in SKOV-3 cells. As revealed by flow cytometry, phagocytosis is a first-order process. Once phagocytosed, PDCM NPs are digested by lysosomes, causing a rapid release of melphalan into the cytoplasm, which ultimately causes suppression of SKOV-3 cell proliferation. Finally, the in vivo antitumor effects of PDCM NPs are verified in xenograft ovarian carcinoma. After a 20-day treatment, the tumor growth rate of the PDCM-3 NP group was (266 ± 178%) which was lower than those in the free melphalan group (367 ± 150%) and control group (648 ± 149%). Besides, significant tissue necrosis and growth suppression were observed in animals administered injections of PDCM NPs. Furthermore, the in vivo tracing results of Nile red-labeled PDCM NPs demonstrated that PDCM-3 NPs might be phagocytosed by macrophages and then taken to adjacent lymph nodes, which is a way of prevention or early treatment of lymphatic metastasis of tumors.
Collapse
Affiliation(s)
- Aiping Wen
- Department of Gynecology and Obstetrics, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xue Mei
- School of Pharmacy, North Sichuan Medical College, Nanchong, China
| | - Chengmin Feng
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, China
| | - Chengyi Shen
- Sichuan Key Laboratory of Medical Imaging & Institute of Morphological Research, North Sichuan Medical College, Nanchong, China
| | - Bing Wang
- Sichuan Key Laboratory of Medical Imaging & Department of Chemistry, School of Preclinical Medicine, North Sichuan Medical College, Nanchong 637000, China.
| | - Xiaoming Zhang
- Sichuan Key Laboratory of Medical Imaging & Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| |
Collapse
|
28
|
Gao L, Li Q, Zhang J, Huang Y, Deng L, Li C, Tai G, Ruan B. Local penetration of doxorubicin via intrahepatic implantation of PLGA based doxorubicin-loaded implants. Drug Deliv 2020; 26:1049-1057. [PMID: 31691602 PMCID: PMC6844384 DOI: 10.1080/10717544.2019.1676842] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Doxorubicin (DOX) is widely used in the chemotherapy of a wide range of cancers. However, intravenous administration of DOX causes toxicity to most major organs which limits its clinical application. DOX-loaded drug delivery system could provide a continuous sustained-release of drugs and enables high drug concentrations at the target site, while reducing systemic toxicity. Additionally, local chemotherapy with DOX may be a promising approach for lowering post-surgical recurrence of cancer. In this study, the sustained-release DOX-loaded implants were prepared by melt-molding method. The implants were characterized with regards to drug content uniformity, micromorphology and drug release profiles. Furthermore, differential scanning calorimetry (DSC) and Fourier transform infrared spectroscopy (FTIR) analyses were carried out to investigate the drug-excipient compatibility. To determine the local penetration of DOX in liver, the minipigs received intrahepatic implantation of DOX-loaded implants by abdominal surgery. UPLC-MS/MS method was used to detect the concentration of DOX in liver tissues. Our results suggested that DOX-loaded implants delivered high doses of drug at the implantation site for a prolonged period and provided valuable information for the future clinical applications of the DOX-loaded implants.
Collapse
Affiliation(s)
- Li Gao
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, People's Republic of China
| | - Qingshan Li
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, People's Republic of China
| | - Jie Zhang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, People's Republic of China
| | - Yixin Huang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, People's Republic of China
| | - Lin Deng
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, People's Republic of China
| | - Chenyang Li
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, People's Republic of China
| | - Guangping Tai
- Key Lab of Biofabrication of Anhui Higher Education Institution Centre for Advanced Biofabrication, Hefei University, Hefei, People's Republic of China
| | - Banfeng Ruan
- Key Lab of Biofabrication of Anhui Higher Education Institution Centre for Advanced Biofabrication, Hefei University, Hefei, People's Republic of China
| |
Collapse
|
29
|
Gao J, Xu Y, Zheng Y, Wang X, Li S, Yan G, Wang J, Tang R. pH-sensitive carboxymethyl chitosan hydrogels via acid-labile ortho ester linkage as an implantable drug delivery system. Carbohydr Polym 2019; 225:115237. [DOI: 10.1016/j.carbpol.2019.115237] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 07/30/2019] [Accepted: 08/21/2019] [Indexed: 12/18/2022]
|
30
|
Zheng Y, Wang W, Zhao J, Wu C, Ye C, Huang M, Wang S. Preparation of injectable temperature-sensitive chitosan-based hydrogel for combined hyperthermia and chemotherapy of colon cancer. Carbohydr Polym 2019; 222:115039. [PMID: 31320053 DOI: 10.1016/j.carbpol.2019.115039] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 06/27/2019] [Accepted: 06/28/2019] [Indexed: 12/20/2022]
Abstract
The purpose of this study was to design an injectable hydrogel with temperature-sensitive property for safe and high efficient in vivo colon cancer hyperthermia and chemotherapy. Chitosan (CS) solution was injected into the tumor at room temperature and automatically gelled after warming to body temperature in the present of β-glycerophosphate (β-GP). Combined localized tumor photothermal and chemotherapy were achieved by dissolving photothermal material MoS2/Bi2S3-PEG (MBP) nanosheets and drug molecule doxorubicin (DOX) into the hydrogel, and the gel system could encapsulate DOX and MBP nanosheets and prevent them from entering the blood circulation and damaging normal tissues and cells. More importantly, the CS/MBP/DOX (CMD) hydrogel exhibited a photothermal efficiency of 22.18% and 31.42% in the first and second near infrared light (NIR I and NIR II) biowindows respectively at a low MBP concentration (0.5 mg/mL). Besides, the release of the DOX from CMD hydrogel was controllable since the gel temperature could be governed by NIR laser irradiation. Moreover, the chitosan-based hydrogel had antibacterial effects. The designed composite hydrogel is anticipated to act as a platform for the high efficient treatment of tumors owing to the different penetration depths of NIR I and NIR II.
Collapse
Affiliation(s)
- Yuting Zheng
- College of Science, University of Shanghai for Science and Technology, No. 334 Jungong Road, Shanghai, 200093, China
| | - Weifan Wang
- Department of Allergy and Immunology, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, No. 1678 Dongfang Road, Shanghai, 200127, China
| | - Jiulong Zhao
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, No. 168 Dongfang Road, Shanghai, 200433, China
| | - Chenyao Wu
- College of Science, University of Shanghai for Science and Technology, No. 334 Jungong Road, Shanghai, 200093, China
| | - Changqing Ye
- College of Science, University of Shanghai for Science and Technology, No. 334 Jungong Road, Shanghai, 200093, China
| | - Mingxian Huang
- College of Science, University of Shanghai for Science and Technology, No. 334 Jungong Road, Shanghai, 200093, China
| | - Shige Wang
- College of Science, University of Shanghai for Science and Technology, No. 334 Jungong Road, Shanghai, 200093, China.
| |
Collapse
|
31
|
Kefayat A, Vaezifar S. Biodegradable PLGA implants containing doxorubicin-loaded chitosan nanoparticles for treatment of breast tumor-bearing mice. Int J Biol Macromol 2019; 136:48-56. [DOI: 10.1016/j.ijbiomac.2019.06.055] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 06/03/2019] [Accepted: 06/09/2019] [Indexed: 12/11/2022]
|
32
|
Gao L, Cai S, Cai A, Zhao Y, Xu T, Ma Y, Xu Y, Wang Y, Wang H, Hu Y. The improved antitumor efficacy of continuous intratumoral chemotherapy with cisplatin-loaded implants for the treatment of sarcoma 180 tumor-bearing mice. Drug Deliv 2019; 26:208-215. [PMID: 30835582 PMCID: PMC6407574 DOI: 10.1080/10717544.2019.1574938] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Cisplatin is the most commonly used antitumor drug in the chemotherapy of a variety of malignancies. However, the severe side effects and drug resistance limit its clinical application. The aim of this study was to develop PLGA-based cisplatin-loaded implants and evaluate the antitumor efficacy of continuous intratumoral chemotherapy with the implants. The cisplatin-loaded implants were prepared by the direct compression method and characterized regarding drug content, micromorphology, in vitro and in vivo drug release profiles. Furthermore, the antitumor activity of the implants was conducted in sarcoma 180 tumor-bearing mice. The SEM images showed smooth surface of the implants and the mean drug content of the tested implants was (37.7% ± 0.5%, w/w). Both in vitro and in vivo release profiles of the implants were characterized by initial burst release followed by the sustained-release of cisplatin. Intratumoral implantation of the cisplatin-loaded implants could effectively inhibit the tumor growth. Additionally, intratumoral chemotherapy with the implants significantly reduced the systemic toxicity compared with intravenous injection of cisplatin. It is worth noting that an increase in the dose of the implants led to a higher tumor suppression rate without additional systemic toxicity. These results demonstrated that cisplatin-loaded implants enhanced the antitumor efficacy and reduced the dose-related side effects in sarcoma 180 tumor-bearing mice.
Collapse
Affiliation(s)
- Li Gao
- a School of Food and Biological Engineering , Hefei University of Technology , Hefei , People's Republic of China
| | - Shang Cai
- b Department of Bone Disease and Bone Tumors Surgery , First Affiliated Hospital of Anhui Medical University , Hefei , People's Republic of China
| | - Awei Cai
- b Department of Bone Disease and Bone Tumors Surgery , First Affiliated Hospital of Anhui Medical University , Hefei , People's Republic of China
| | - Yang Zhao
- c Department of Pathology , The Second People's Hospital of Hefei , Hefei , People's Republic of China
| | - Tangbing Xu
- d Department of Orthopeadic Surgery , Fourth Affiliated Hospital of Anhui Medical University , Hefei , People's Republic of China
| | - Yan Ma
- a School of Food and Biological Engineering , Hefei University of Technology , Hefei , People's Republic of China
| | - Yan Xu
- a School of Food and Biological Engineering , Hefei University of Technology , Hefei , People's Republic of China
| | - Yuan Wang
- b Department of Bone Disease and Bone Tumors Surgery , First Affiliated Hospital of Anhui Medical University , Hefei , People's Republic of China
| | - Hao Wang
- b Department of Bone Disease and Bone Tumors Surgery , First Affiliated Hospital of Anhui Medical University , Hefei , People's Republic of China
| | - Yong Hu
- b Department of Bone Disease and Bone Tumors Surgery , First Affiliated Hospital of Anhui Medical University , Hefei , People's Republic of China
| |
Collapse
|
33
|
Misra R, Sarkar K, Lee J, Pizzuti VJ, Lee DS, Currie MP, Torregrosa-Allen SE, Long DE, Durm GA, Langer MP, Elzey BD, Won YY. Radioluminescent nanoparticles for radiation-controlled release of drugs. J Control Release 2019; 303:237-252. [PMID: 31026550 DOI: 10.1016/j.jconrel.2019.04.033] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 04/10/2019] [Accepted: 04/22/2019] [Indexed: 01/03/2023]
Abstract
The present work demonstrates a novel concept for intratumoral chemo-radio combination therapy for locally advanced solid tumors. For some locally advanced tumors, chemoradiation is currently standard of care. This combination treatment can cause acute and long term toxicity that can limit its use in older patients or those with multiple medical comorbidities. Intratumoral chemotherapy has the potential to address the problem of systemic toxicity that conventional chemotherapy suffers, and may, in our view, be a better strategy for treating certain locally advanced tumors. The present study proposes how intratumoral chemoradiation can be best implemented. The enabling concept is the use of a new chemotherapeutic formulation in which chemotherapy drugs (e.g., paclitaxel (PTX)) are co-encapsulated with radioluminecsnt nanoparticles (e.g., CaWO4 (CWO) nanoparticles (NPs)) within protective capsules formed by biocompatible/biodegradable polymers (e.g., poly(ethylene glycol)-poly(lactic acid) or PEG-PLA). This drug-loaded polymer-encapsulated radioluminescent nanoparticle system can be locally injected in solution form into the patient's tumor before the patient receives normal radiotherapy (e.g., 30-40 fractions of 2-3 Gy daily X-ray dose delivered over several weeks for locally advanced head and neck tumors). Under X-ray irradiation, the radioluminescent nanoparticles produce UV-A light that has a radio-sensitizing effect. These co-encapsulated radioluminescent nanoparticles also enable radiation-triggered release of chemo drugs from the polymer coating layer. The non-toxic nature (absence of dark toxicity) of this drug-loaded polymer-encapsulated radioluminescent nanoparticle ("PEG-PLA/CWO/PTX") formulation was confirmed by the MTT assay in cancer cell cultures. A clonogenic cell survival assay confirmed that these drug-loaded polymer-encapsulated radioluminescent nanoparticles significantly enhance the cancer cell killing effect of radiation therapy. In vivo study validated the efficacy of PEG-PLA/CWO/PTX-based intratumoral chemo-radio therapy in mouse tumor xenografts (in terms of tumor response and mouse survival). Results of a small-scale NP biodistribution (BD) study demonstrate that PEG-PLA/CWO/PTX NPs remained at the tumor sites for a long period of time (> 1 month) following direct intratumoral administration. A multi-compartmental pharmacokinetic model (with rate constants estimated from in vitro experiments) predicts that this radiation-controlled drug release technology enables significant improvements in the level and duration of drug availability within the tumor (throughout the typical length of radiation treatment, i.e., > 1 month) over conventional delivery systems (e.g., PEG-PLA micelles with no co-encapsulated CaWO4, or an organic liquid, e.g., a 50:50 mixture of Cremophor EL and ethanol, as in Taxol), while it is capable of maintaining the systemic level of the chemo drug far below the toxic threshold limit over the entire treatment period. This technology thus has the potential to offer a new therapeutic option that has not previously been available for patients excluded from conventional chemoradiation protocols.
Collapse
Affiliation(s)
- Rahul Misra
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN 47907, United States
| | - Kaustabh Sarkar
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN 47907, United States
| | - Jaewon Lee
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN 47907, United States
| | - Vincenzo J Pizzuti
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN 47907, United States
| | - Deborah S Lee
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN 47907, United States
| | - Melanie P Currie
- Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN 47907, United States
| | - Sandra E Torregrosa-Allen
- Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN 47907, United States
| | - David E Long
- Department of Radiation Oncology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Gregory A Durm
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Mark P Langer
- Department of Radiation Oncology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Bennett D Elzey
- Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN 47907, United States
| | - You-Yeon Won
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN 47907, United States; Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN 47907, United States.
| |
Collapse
|
34
|
Abid S, Hussain T, Raza ZA, Nazir A. Current applications of electrospun polymeric nanofibers in cancer therapy. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 97:966-977. [DOI: 10.1016/j.msec.2018.12.105] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 09/03/2018] [Accepted: 12/25/2018] [Indexed: 12/20/2022]
|
35
|
Li W, Li W, Kuang Y, Yang T, Zhu J, Xu Z, Yuan X, Li M, Zhang Z, Yang Y. Toxicity Assessment of PEG-PCCL Nanoparticles and Preliminary Investigation on Its Anti-tumor Effect of Paclitaxel-Loading. NANOSCALE RESEARCH LETTERS 2018; 13:253. [PMID: 30143890 PMCID: PMC6108981 DOI: 10.1186/s11671-018-2615-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 06/25/2018] [Indexed: 02/05/2023]
Abstract
The efficiency of single treatment of conventional chemotherapy drugs is unpleasantly reduced by the physiological barriers of tumors. In this regard, nanoparticles have become attractive for achieving such medical purpose of targeted cancer therapy by delivering anti-tumor agents to the needed area. A novel drug deliverer, poly (ethylene glycol) carboxyl-poly (ε-caprolactone) (PEG-PCCL), has been reported to be highly hydrophilic and stable, while little is known about its organic toxicity. This study focused on systemic toxicity assessments of PEG-PCCL. The pharmacokinetics of PTX-loaded PEG-PCCL (PEG-PCCL/PTX) and its anti-tumor effect were preliminarily investigated. In the present work, PEG-PCCL was characterized by laser particle size analyzer and transmission electron microscopy. The cytotoxicity was investigated by MTT test, LDH leakage assay, immunofluorescence, and transmission electron microscopy. Hemolysis, phlebitis, and organ toxicity tests were performed to demonstrate the biocompatibility and acute biotoxicity. H22 tumor-bearing mice were used to evaluate the pharmacokinetics of the micells of PEG-PCCL/PTX and its anti-tumor effect. The results showed that the size of PEG-PCCL nanospheres was 97 ± 2.6 nm. PEG-PCCL treatment showed little cytotoxicity and good biocompatibility, and did not exhibit organ toxicity. PTX-loading efficiency was 49.98%. The pharmacokinetic study on H22 tumor-bearing mice revealed that PEG-PCCL/PTX has higher stability and slower release than PTX alone. Together, these results suggest that PEG-PCCL nanosphere has little toxicity to organisms and is a potential candidate of biocompatible drug vehicle for hydrophobic drugs.
Collapse
Affiliation(s)
- Wei Li
- Department of Microbiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, China.,Department of Burns surgery, Sichuan Academy of Medical Sciences and Sichuan Province People's Hospital, Chengdu, 610072, China
| | - Wanyi Li
- Department of Microbiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Yu Kuang
- Department of Microbiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Ting Yang
- Department of Microbiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Jie Zhu
- Department of Microbiology, Guizhou Medical University, Guiyang, 55000, China
| | - Zilin Xu
- Department of Microbiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Xiang Yuan
- Department of Microbiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Mingyuan Li
- Department of Microbiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Zhongwei Zhang
- ICU, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yuan Yang
- Department of Microbiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
36
|
Salmoria GV, Ghizoni GB, Gindri IM, Marques MS, Kanis LA. Hot extrusion of PE/fluorouracil implantable rods for targeted drug delivery in cancer treatment. Polym Bull (Berl) 2018. [DOI: 10.1007/s00289-018-2451-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
37
|
Talebian S, Foroughi J, Wade SJ, Vine KL, Dolatshahi-Pirouz A, Mehrali M, Conde J, Wallace GG. Biopolymers for Antitumor Implantable Drug Delivery Systems: Recent Advances and Future Outlook. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1706665. [PMID: 29756237 DOI: 10.1002/adma.201706665] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 02/15/2018] [Indexed: 06/08/2023]
Abstract
In spite of remarkable improvements in cancer treatments and survivorship, cancer still remains as one of the major causes of death worldwide. Although current standards of care provide encouraging results, they still cause severe systemic toxicity and also fail in preventing recurrence of the disease. In order to address these issues, biomaterial-based implantable drug delivery systems (DDSs) have emerged as promising therapeutic platforms, which allow local administration of drugs directly to the tumor site. Owing to the unique properties of biopolymers, they have been used in a variety of ways to institute biodegradable implantable DDSs that exert precise spatiotemporal control over the release of therapeutic drug. Here, the most recent advances in biopolymer-based DDSs for suppressing tumor growth and preventing tumor recurrence are reviewed. Novel emerging biopolymers as well as cutting-edge polymeric microdevices deployed as implantable antitumor DDSs are discussed. Finally, a review of a new therapeutic modality within the field, which is based on implantable biopolymeric DDSs, is given.
Collapse
Affiliation(s)
- Sepehr Talebian
- Intelligent Polymer Research Institute, ARC Centre of Excellence for Electromaterials Science, AIIM Facility, University of Wollongong, NSW 2522, Australia
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Javad Foroughi
- Intelligent Polymer Research Institute, ARC Centre of Excellence for Electromaterials Science, AIIM Facility, University of Wollongong, NSW 2522, Australia
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Samantha J Wade
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, 2522, Australia
- School of Biological Sciences, University of Wollongong, NSW 2522, Australia
| | - Kara L Vine
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, 2522, Australia
- School of Biological Sciences, Centre for Medical and Molecular Bioscience, University of Wollongong, NSW 2522, Australia
| | - Alireza Dolatshahi-Pirouz
- Technical University of Denmark, DTU Nanotech, Center for Nanomedicine and Theranostics, 2800 Kongens Lyngby, Denmark
| | - Mehdi Mehrali
- Technical University of Denmark, DTU Nanotech, Center for Nanomedicine and Theranostics, 2800 Kongens Lyngby, Denmark
| | - João Conde
- Massachusetts Institute of Technology, Institute for Medical Engineering and Science, Harvard-MIT Division for Health Sciences and Technology, Cambridge, MA, 02139, USA
| | - Gordon G Wallace
- Intelligent Polymer Research Institute, ARC Centre of Excellence for Electromaterials Science, AIIM Facility, University of Wollongong, NSW 2522, Australia
| |
Collapse
|
38
|
Controlling the burst release of doxorubicin from polymeric depots via adjusting hydrophobic/hydrophilic properties. J Drug Deliv Sci Technol 2018. [DOI: 10.1016/j.jddst.2018.06.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
39
|
Manaspon C, Chaimongkolnukul K, Kengkoom K, Boongird A, Hongeng S, Chairoungdua A, Nasongkla N. Time-dependent distribution of SN-38 from injectable polymeric depots in brain tumor model. Biomed Phys Eng Express 2018. [DOI: 10.1088/2057-1976/aad396] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
40
|
Xu M, Mou Y, Hu M, Dong W, Su X, Wu R, Zhang P. Evaluation of micelles incorporated into thermosensitive hydrogels for intratumoral delivery and controlled release of docetaxel: A dual approach for in situ treatment of tumors. Asian J Pharm Sci 2018; 13:373-382. [PMID: 32104411 PMCID: PMC7032140 DOI: 10.1016/j.ajps.2018.05.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 04/02/2018] [Accepted: 05/13/2018] [Indexed: 12/19/2022] Open
Abstract
The in situ gelling hybrid hydrogel system has been reported to effectively concentrate chemotherapeutic drugs at the tumor site and sustain their release for a long period. DTX-micelles (docetaxel-loaded mixed micelles) are able to increase the solubility of DTX in water, and then a high drug loading rate of hydrogels can be achieved by encapsulating the docetaxel-loaded mixed micelles into the hydrogels. The thermosensitive nature of DTX-MM-hydrogels (thermosensitive hydrogels incorporated with docetaxel-loaded mixed micelles) can accelerate the formation of a depot of this drug-loaded system at the site of administration. Therefore, the hydrogels provide a much slower release compared with DTX-micelles and DTX-injection. An in vivo retention study has demonstrated that the DTX-MM-hydrogels can prolong the drug retention time and in vivo trials have shown that the DTX-MM-hydrogels have a higher antitumor efficacy and systemic safety. In conclusion, the DTX-MM-hydrogels prepared in this study have considerable potential as a drug delivery system, with higher tumor inhibition effects and are less toxic to normal tissues.
Collapse
Affiliation(s)
- Meng Xu
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Yanhua Mou
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Mingming Hu
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Wenxiang Dong
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Xitong Su
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Rongxia Wu
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Peng Zhang
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| |
Collapse
|
41
|
Agwa M, Elessawy FM, Hussein A, El Demellawy MA, Elzoghby AO, Abd El-Salam MH, Eldiwany AI. Development and validation of a robust analytical method to quantify both etoposide and prodigiosin in polymeric nanoparticles by reverse-phase high-performance liquid chromatography. ANALYTICAL METHODS 2018; 10:2272-2280. [DOI: 10.1039/c8ay00030a] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Preparation of the ETP-PRO loaded B-CN nanocarrier (1) and analysis of the % of ETP and PRO released (2) using the validated HPLC method (3).
Collapse
Affiliation(s)
- M. M. Agwa
- Department of Natural & Microbial Products
- National Research Centre
- Cairo
- Egypt
- Pharmaceutical & Fermentation Industries Development Center (PFIDC)
| | - F. M. Elessawy
- Pharmaceutical & Fermentation Industries Development Center (PFIDC)
- City for Scientific Research & Technology Applications
- Alexandria
- Egypt
- College of Pharmacy and Nutrition
| | - A. Hussein
- Department of Biotechnology
- Institute of Graduate Studies and Research
- Alexandria University
- Alexandria
- Egypt
| | - M. A. El Demellawy
- Pharmaceutical & Fermentation Industries Development Center (PFIDC)
- City for Scientific Research & Technology Applications
- Alexandria
- Egypt
| | - A. O. Elzoghby
- Cancer Nanotechnology Research Laboratory (CNRL)
- Faculty of Pharmacy
- Alexandria University
- Alexandria
- Egypt
| | | | - A. I. Eldiwany
- Department of Natural & Microbial Products
- National Research Centre
- Cairo
- Egypt
| |
Collapse
|
42
|
Functionalized diterpene parvifloron D-loaded hybrid nanoparticles for targeted delivery in melanoma therapy. Ther Deliv 2017; 7:521-44. [PMID: 27444493 DOI: 10.4155/tde-2016-0027] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
AIM Parvifloron D is a natural diterpene with a broad and not selective cytotoxicity toward human tumor cells. In order to develop a targeted antimelanoma drug delivery platform for Parvifloron D, hybrid nanoparticles were prepared with biopolymers and functionalized with α-melanocyte stimulating hormone. Results/methodology: Nanoparticles were produced according to a solvent displacement method and the physicochemical properties were assessed. It was shown that Parvifloron D is cytotoxic and can induce, both as free and as encapsulated drug, cell death in melanoma cells (human A375 and mouse B16V5). Parvifloron D-loaded nanoparticles showed a high encapsulation efficiency (87%) and a sustained release profile. In vitro experiments showed the nanoparticles' uptake and cell internalization. CONCLUSION Hybrid nanoparticles appear to be a promising platform for long-term drug release, presenting the desired structure and a robust performance for targeted anticancer therapy.
Collapse
|
43
|
Wu Y, Wang L, Zhang K, Zhou L, Zhang X, Jiang X, Zhu C. N-Butyl-2-cyanoacrylate-based injectable and in situ-forming implants for efficient intratumoral chemotherapy. Drug Deliv 2017; 24:729-736. [PMID: 28440691 PMCID: PMC8240990 DOI: 10.1080/10717544.2017.1309478] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The local delivery of chemotherapeutic drugs to tumor sites is an effective approach for achieving therapeutic drug concentrations in solid tumors. Injectable implants with the ability to form in situ represent one of the most promising technologies for intratumoral chemotherapy. However, many issues must be resolved before these implants can be applied in clinical practice. Herein, we report a novel injectable in situ-forming implant system composed of n-butyl-2-cyanoacrylate (NBCA) and ethyl oleate, and the sol–gel phase transition is activated by anions in body fluids or blood. This newly developed injectable NBCA ethyl oleate implant (INEI) is biodegradable, biocompatible, and non-toxic. INEI solidifies in several seconds after exposure to body fluids or blood, and the implant’s in vivo degradation time can be controlled. In addition, the pore sizes formed by the polymerization of NBCA can be decreased by increasing the NBCA concentration in the implants. Therefore, the drug retention/release time can be adjusted from a few weeks to several months by changing the concentration of NBCA in the implant formulation. Anti-tumor experiments in animal models showed that the average growth inhibition rate of xenografted human breast cancer cells by the paclitaxel-loaded INEI (40% NBCA) was 80%, and they also indicated that tumors in some of the mice were completely eliminated by just a single dosage injection. For the epirubicin-loaded INEI (50% NBCA), the average growth inhibition rate of xenografted human liver cancer cells was 58%. Thus, the chemotherapeutic drug-loaded INEIs exhibited excellent therapeutic efficacy for local chemotherapy.
Collapse
Affiliation(s)
- Yanpu Wu
- a College of Life Sciences, Zhejiang University , Hangzhou , Zhejiang , China and
| | - Luming Wang
- b Jiaxing Maternity and Child Health Care Hospital , Jiaxing , Zhejiang , China
| | - Kaili Zhang
- a College of Life Sciences, Zhejiang University , Hangzhou , Zhejiang , China and
| | - Lixiao Zhou
- a College of Life Sciences, Zhejiang University , Hangzhou , Zhejiang , China and
| | - Xiaobing Zhang
- a College of Life Sciences, Zhejiang University , Hangzhou , Zhejiang , China and
| | - Xuecheng Jiang
- a College of Life Sciences, Zhejiang University , Hangzhou , Zhejiang , China and
| | - Chenggang Zhu
- a College of Life Sciences, Zhejiang University , Hangzhou , Zhejiang , China and
| |
Collapse
|
44
|
Gao L, Xie C, Du Y, Wang X, Xuan E, Liu X, Zhao Y, Xu J, Luo L. Characterization and antitumor efficacy of poly(L-lactid acid)-based etoposide-loaded implants. Drug Deliv 2017; 24:765-774. [PMID: 28475414 PMCID: PMC8241189 DOI: 10.1080/10717544.2017.1321063] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Etoposide is widely used in the chemotherapy of a variety of malignancies. But the strong lipophilicity, poor bioavailability, and severe side effects of etoposide limit its clinical application. The aim of this study was to develop sustained-release etoposide-loaded implants and evaluate antitumor activity of the implants after intratumoral implantation. We prepared the implants containing etoposide, poly(L-lactid acid) and polyethylene glycol 4000 by the direct compression method. The implants were characterized regarding drug-excipient compatibility, content uniformity, morphology, sterility, in vitro, and in vivo release profiles. Then the antitumor activity of the implants was tested in xenograft model of A549 human non-small cell lung cancer. SEM images displayed smooth surface of the implant and indicated that etoposide was homogeneously dispersed in the polymeric matrix. The results of content uniformity met the requirements of the Chinese Pharmacopoeia. Both in vitro and in vivo release profiles of the implants were characterized by high burst release followed by sustained release of etoposide. Intratumoral implantation of etoposide-loaded implants could efficiently delay the tumor growth. Furthermore, increasing the dose of implants led to higher tumor suppression rate without adding systemic toxicity. These results indicated that etoposide-loaded implants have significant antitumor efficacy in xenograft model without dose-limiting side effects and they possess a strong potential to be used as an intratumoral chemotherapy option for lung cancer treatment.
Collapse
Affiliation(s)
- Li Gao
- a State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University , Nanjing , People's Republic of China.,b School of Biological and Medical Engineering, Hefei University of Technology , Hefei , People's Republic of China
| | - Chuanqi Xie
- c Laboratory of pharmaceutical research , Anhui Zhongren Science and Technology Co., Ltd , Hefei , People's Republic of China , and
| | - Yuzhi Du
- b School of Biological and Medical Engineering, Hefei University of Technology , Hefei , People's Republic of China
| | - Xiaodong Wang
- c Laboratory of pharmaceutical research , Anhui Zhongren Science and Technology Co., Ltd , Hefei , People's Republic of China , and
| | - Erkang Xuan
- c Laboratory of pharmaceutical research , Anhui Zhongren Science and Technology Co., Ltd , Hefei , People's Republic of China , and
| | - Xiuxiu Liu
- b School of Biological and Medical Engineering, Hefei University of Technology , Hefei , People's Republic of China
| | - Yang Zhao
- d Department of Pathology , The Second People's Hospital of Hefei , Hefei , People's Republic of China
| | - Jianjian Xu
- b School of Biological and Medical Engineering, Hefei University of Technology , Hefei , People's Republic of China
| | - Lan Luo
- a State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University , Nanjing , People's Republic of China
| |
Collapse
|
45
|
Affiliation(s)
- Kıvılcım Öztürk-Atar
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Hakan Eroğlu
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Sema Çalış
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| |
Collapse
|
46
|
Gao L, Xia L, Zhang R, Duan D, Liu X, Xu J, Luo L. Enhanced antitumor efficacy of poly(D,L-lactide-co-glycolide)-based methotrexate-loaded implants on sarcoma 180 tumor-bearing mice. DRUG DESIGN DEVELOPMENT AND THERAPY 2017; 11:3065-3075. [PMID: 29118572 PMCID: PMC5659257 DOI: 10.2147/dddt.s143942] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Purpose Methotrexate is widely used in chemotherapy for a variety of malignancies. However, severe toxicity, poor pharmacokinetics, and narrow safety margin of methotrexate limit its clinical application. The aim of this study was to develop sustained-release methotrexate-loaded implants and evaluate antitumor activity of the implants after intratumoral implantation. Materials and methods We prepared the implants containing methotrexate, poly(D,L-lactide-co-glycolide), and polyethylene glycol 4000 with the melt-molding technique. The implants were characterized with regards to drug content, morphology, in vitro, and in vivo release profiles. Differential scanning calorimetry (DSC) and Fourier transform infrared spectroscopy (FTIR) were carried out to investigate the physicochemical properties of the implants. Furthermore, the antitumor activity of the implants was tested in a sarcoma 180 mouse model. Results The implants were prepared as solid rods. Scanning electron microscopy images showed a smooth surface of the implant, suggesting that methotrexate was homogeneously dispersed in the polymeric matrix. The results of DSC and FTIR indicated that no significant interaction between methotrexate and the polymer was observed in the implants. Both in vitro and in vivo release profiles of the implants were characterized by burst release followed by sustained release of methotrexate. Intratumoral implantation of methotrexate-loaded implants could efficiently delay tumor growth. Moreover, an increase in the dose of implants led to a higher tumor suppression rate without additional systemic toxicity. Conclusion These results demonstrate that methotrexate-loaded implants had significant antitumor efficacy in a sarcoma 180 mouse model without dose-limiting side effects, and suggest that the implants could be potentially applied as an intratumoral delivery system to treat cancer.
Collapse
Affiliation(s)
- Li Gao
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing.,School of Biological and Medical Engineering, Hefei University of Technology, Hefei
| | - Lunyang Xia
- Laboratory of Pharmaceutical Research, Anhui Zhongren Science and Technology Co., Ltd., Hefei, People's Republic of China
| | - Ruhui Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing
| | - Dandan Duan
- Laboratory of Pharmaceutical Research, Anhui Zhongren Science and Technology Co., Ltd., Hefei, People's Republic of China
| | - Xiuxiu Liu
- School of Biological and Medical Engineering, Hefei University of Technology, Hefei
| | - Jianjian Xu
- School of Biological and Medical Engineering, Hefei University of Technology, Hefei
| | - Lan Luo
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing
| |
Collapse
|
47
|
Suman S, Mondal R, Kumar J, Dubey K, Kadam R, Melo J, Bhardwaj Y, Varshney L. Development of highly radiopaque flexible polymer composites for X-ray imaging applications and copolymer architecture-morphology-property correlations. Eur Polym J 2017. [DOI: 10.1016/j.eurpolymj.2017.07.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
48
|
Thermosensitive Gel–Based Formulation for Intratumoral Delivery of Toll-Like Receptor 7/8 Dual Agonist, MEDI9197. J Pharm Sci 2017; 106:2037-2045. [DOI: 10.1016/j.xphs.2017.04.041] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 04/06/2017] [Accepted: 04/19/2017] [Indexed: 11/20/2022]
|
49
|
Ramot Y, Rotkopf S, Gabai RM, Zorde Khvalevsky E, Muravnik S, Marzoli GA, Domb AJ, Shemi A, Nyska A. Preclinical Safety Evaluation in Rats of a Polymeric Matrix Containing an siRNA Drug Used as a Local and Prolonged Delivery System for Pancreatic Cancer Therapy. Toxicol Pathol 2016; 44:856-65. [PMID: 27147553 DOI: 10.1177/0192623316645860] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Conventional chemotherapy treatments for pancreatic cancer are mainly palliative. RNA interference (RNAi)-based drugs present the potential for a new targeted treatment. LOcal Drug EluteR (LODER(TM)) is a novel biodegradable polymeric matrix that shields drugs against enzymatic degradation and releases small interfering RNA (siRNA) against G12D-mutated KRAS (siG12D). siG12D-LODER has successfully passed a phase 1/2a clinical trial. Such a formulation necessitates biocompatibility and safety studies. We describe the safety and toxicity studies with siG12D-LODER in 192 Hsd:Sprague Dawley rats, after repeated subcutaneous administrations (days 1, 14, and 28). Animals were sacrificed on days 29 and 42 (recovery phase). In all groups, no adverse effects were noted, and all animals showed favorable local and systemic tolerability. Histopathologically, LODER implantation resulted in the expected capsule formation, composed of a thin fibrotic tissue. On the interface between the cavity and the capsule, a single layer composed of macrophages and multinucleated giant cells was observed. No difference was noted between the placebo and siG12D-LODER groups. These findings provide valuable information for future preclinical studies with siRNA-bearing biodegradable polymers and for the safety aspects of RNAi-based drugs as a targeted therapy.
Collapse
Affiliation(s)
- Yuval Ramot
- Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | | | | | | | | | | | - Abraham J Domb
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Ein Kerem, Jerusalem, Israel
| | | | - Abraham Nyska
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel Consultant in Toxicologic Pathology, Timrat, Israel
| |
Collapse
|
50
|
|