1
|
Wei W, Chen F, Wang Y. Effect of multidisciplinary team-style continuity of care and nutritional nursing on lung cancer: randomized study. Future Oncol 2024:1-10. [PMID: 39435906 DOI: 10.1080/14796694.2024.2407757] [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: 12/12/2023] [Accepted: 09/19/2024] [Indexed: 10/23/2024] Open
Abstract
Aim: The clinical efficacy of systemic chemotherapy is limited due to the nonspecific delivery of anticancer drugs and is associated with serious systemic adverse effects. Therefore, integrated treatment and comprehensive care are particularly important for postoperative chemotherapy patients with lung cancer.Materials & methods: This study aimed to ascertain the application effect of multidisciplinary team (MDT)-style continuity of care combined with whole-process nutritional nursing in postoperative chemotherapy patients with lung cancer. Nutritional indices, immune function, adverse emotions, self-efficacy, self-care ability, quality of life and toxic reactions during chemotherapy were recorded in postoperative chemotherapy patients with lung cancer receiving routine care (control group) and MDT-style continuity of care combined with whole-process nutritional care (intervention group).Results: After care, the intervention group performed higher BMI, PA, TP and ALB, CD3+, CD4+ and CD4+/CD8+, lower levels of CD8+, lower self-rating anxiety scale, self-rating depression scale and QLQ-C30 symptom domain scores and higher general self-efficacy scale, exercise of self-care agency scale and QLQ-C30 functional domain scores versus the control group (all p < 0.05).Conclusion: MDT-style continuity of care combined with whole-process nutritional care can improve the nutritional status of postoperative chemotherapy patients with lung cancer, and in turn enhance their quality of life.
Collapse
Affiliation(s)
- Wei Wei
- Tumor Center, Henan Provincial Key Medicine Laboratory of Nursing, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, Henan, 450003, China
| | - Fengxia Chen
- Tumor Center, Henan Provincial Key Medicine Laboratory of Nursing, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, Henan, 450003, China
| | - Yuxia Wang
- Tumor Center, Henan Provincial Key Medicine Laboratory of Nursing, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, Henan, 450003, China
| |
Collapse
|
2
|
Kumar A, Vaiphei KK, Gulbake A. A nanotechnology driven effectual localized lung cancer targeting approaches using tyrosine kinases inhibitors: Recent progress, preclinical assessment, challenges, and future perspectives. Int J Pharm 2024; 666:124745. [PMID: 39321904 DOI: 10.1016/j.ijpharm.2024.124745] [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: 07/09/2024] [Revised: 09/09/2024] [Accepted: 09/21/2024] [Indexed: 09/27/2024]
Abstract
The higher incidence and mortality rate among all populations worldwide explains the unmet solutions in the treatment of lung cancer. The evolution of targeted therapies using tyrosine kinase inhibitors (TKI) has encouraged anticancer therapies. However, on-target and off-target effects and the development of drug resistance limited the anticancer potential of such targeted biologics. The advances in nanotechnology-driven-TKI embedded carriers that offered a new path toward lung cancer treatment. It is the inhalation route of administration known for its specific, precise, and efficient drug delivery to the lungs. The development of numerous TKI-nanocarriers through inhalation is proof of TKI growth. The future scopes involve using potential lung cancer biomarkers to achieve localized active cancer-targeting strategies. The adequate knowledge of in vitro absorption models usually helps establish better in vitro - in vivo correlation/extrapolation (IVIVC/E) to successfully evaluate inhalable drugs and drug products. The advanced in vitro and ex vivo lung tissue/ organ models offered better tumor heterogeneity, etiology, and microenvironment heterogeneity. The involvement of lung cancer organoids (LCOs), human organ chip models, and genetically modified mouse models (GEMMs) has resolved the challenges associated with conventional in vitro and in vivo models. To access potential inhalation-based drugtherapies, biological barriers, drug delivery, device-based challenges, and regulatory challenges must be encountered associated with their development. A proper understanding of material toxicity, size-based particle deposition at active disease sites, mucociliary clearance, phagocytosis, and the presence of enzymes and surfactants are required to achieve successful inhalational drug delivery (IDD). This article summarizes the future of lung cancer therapy using targeted drug-mediated inhalation using TKI.
Collapse
Affiliation(s)
- Ankaj Kumar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research Guwahati, Assam 781101, India
| | - Klaudi K Vaiphei
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research Guwahati, Assam 781101, India
| | - Arvind Gulbake
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research Guwahati, Assam 781101, India.
| |
Collapse
|
3
|
Eid HM, Turkia TH, Ali AA, Aboud HM. A Novel Chitosan-coated Leciplex Loaded with Ambrisentan as a Possible Pulmonary Nanosystem: Optimization, Characterization, and Pharmacokinetics Assessments. J Pharm Sci 2024; 113:2320-2330. [PMID: 38582282 DOI: 10.1016/j.xphs.2024.03.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 03/29/2024] [Accepted: 03/29/2024] [Indexed: 04/08/2024]
Abstract
The purpose of this research was to formulate, optimize, and characterize ambrisentan chitosan-coated LeciPlex (AMS-CTS-LPX) to increase the therapeutic effectiveness and bioavailability of ambrisentan. A central composite design (CCD) was implemented to assess the impact of various factors on the production of AMS-CTS-LPX and to identify the optimum formulation via the use of Design Expert® software. The assembly of AMS-CTS-LPX was conducted using a single-step process. Subsequently, the optimal formulation was chosen and subjected to further assessments. Further, a comparative pharmacokinetic study was carried out using a rat model. The optimized formulation exhibited an entrapment efficiency of 82.39%, with a diameter of 137.53 nm and a surface charge of +43.65 mV. Additionally, it had a sustained cumulative release of 90.41% after 8 h and showed good stability. The safety of AMS-CTS-LPX administered intratracheally was confirmed by in vivo histopathological studies. The pharmacokinetic investigations revealed a 5.6-fold increase in the bioavailability of AMS from the optimal AMS-CTS-LPX formulation compared to the oral AMS solution. Collectively, the results of the current study suggest that CTS-LPX may be beneficial as a pulmonary nanosystem for the administration of AMS.
Collapse
Affiliation(s)
- Hussein M Eid
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, 62514, Egypt.
| | - Toqa H Turkia
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, 62514, Egypt
| | - Adel A Ali
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, 62514, Egypt
| | - Heba M Aboud
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, 62514, Egypt
| |
Collapse
|
4
|
Huang Y, Zhang J, Wang X, Jing H, Li H. Aerosol Inhalation of Gene Delivery Therapy for Pulmonary Diseases. Biomolecules 2024; 14:904. [PMID: 39199292 PMCID: PMC11352762 DOI: 10.3390/biom14080904] [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: 05/14/2024] [Revised: 06/27/2024] [Accepted: 07/20/2024] [Indexed: 09/01/2024] Open
Abstract
Gene delivery therapy has emerged as a popular approach for the treatment of various diseases. However, it still poses the challenges of accumulation in target sites and reducing off-target effects. Aerosol gene delivery for the treatment of pulmonary diseases has the advantages of high lung accumulation, specific targeting and fewer systemic side effects. However, the key challenge is selecting the appropriate formulation for aerosol gene delivery that can overcome physiological barriers. There are numerous existing gene carriers under study, including viral vectors and non-viral vectors. With the development of biomaterials, more biocompatible substances have applied gene delivery via inhalation. Furthermore, many types of genes can be delivered through aerosol inhalation, such as DNA, mRNA, siRNA and CRISPR/Cas9. Aerosol delivery of different types of genes has proven to be efficient in the treatment of many diseases such as SARS-CoV-2, cystic fibrosis and lung cancer. In this paper, we provide a comprehensive review of the ongoing research on aerosol gene delivery therapy, including the basic respiratory system, different types of gene carriers, different types of carried genes and clinical applications.
Collapse
Affiliation(s)
| | | | | | - Hui Jing
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (Y.H.); (J.Z.); (X.W.)
| | - Hecheng Li
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (Y.H.); (J.Z.); (X.W.)
| |
Collapse
|
5
|
Ediriweera GR, Butcher NJ, Kothapalli A, Zhao J, Blanchfield JT, Subasic CN, Grace JL, Fu C, Tan X, Quinn JF, Ascher DB, Whittaker MR, Whittaker AK, Kaminskas LM. Lipid sulfoxide polymers as potential inhalable drug delivery platforms with differential albumin binding affinity. Biomater Sci 2024; 12:2978-2992. [PMID: 38683548 DOI: 10.1039/d3bm02020g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2024]
Abstract
Inhalable nanomedicines are increasingly being developed to optimise the pharmaceutical treatment of respiratory diseases. Large lipid-based nanosystems at the forefront of the inhalable nanomedicines development pipeline, though, have a number of limitations. The objective of this study was, therefore, to investigate the utility of novel small lipidated sulfoxide polymers based on poly(2-(methylsulfinyl)ethyl acrylate) (PMSEA) as inhalable drug delivery platforms with tuneable membrane permeability imparted by differential albumin binding kinetics. Linear PMSEA (5 kDa) was used as a hydrophilic polymer backbone with excellent anti-fouling and stealth properties compared to poly(ethylene glycol). Terminal lipids comprising single (1C2, 1C12) or double (2C12) chain diglycerides were installed to provide differing affinities for albumin and, by extension, albumin trafficking pathways in the lungs. Albumin binding kinetics, cytotoxicity, lung mucus penetration and cellular uptake and permeability through key cellular barriers in the lungs were examined in vitro. The polymers showed good mucus penetration and no cytotoxicity over 24 h at up to 1 mg ml-1. While 1C2-showed no interaction with albumin, 1C12-PMSEA and 2C12-PMSEA bound albumin with KD values of approximately 76 and 10 μM, respectively. Despite binding to albumin, 2C12-PMSEA showed reduced cell uptake and membrane permeability compared to the smaller polymers and the presence of albumin had little effect on cell uptake and membrane permeability. While PMSEA strongly shielded these lipids from albumin, the data suggest that there is scope to tune the lipid component of these systems to control membrane permeability and cellular interactions in the lungs to tailor drug disposition in the lungs.
Collapse
Affiliation(s)
- Gayathri R Ediriweera
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia.
| | - Neville J Butcher
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia.
| | - Ashok Kothapalli
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia.
| | - Jiacheng Zhao
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia.
| | - Joanne T Blanchfield
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Christopher N Subasic
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia.
| | - James L Grace
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia
| | - Changkui Fu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia.
| | - Xiao Tan
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia.
| | - John F Quinn
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia
- Department of Chemical Engineering, Monash University, Clayton, VIC, Australia
| | - David B Ascher
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, 4072, Australia
- Computational Biology and Clinical Informatics, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Michael R Whittaker
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia
| | - Andrew K Whittaker
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia.
| | - Lisa M Kaminskas
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia.
| |
Collapse
|
6
|
Nia GE, Nikpayam E, Farrokhi M, Bolhassani A, Meuwissen R. Advances in cell-based delivery of oncolytic viruses as therapy for lung cancer. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200788. [PMID: 38596310 PMCID: PMC10976516 DOI: 10.1016/j.omton.2024.200788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
Lung cancer's intractability is enhanced by its frequent resistance to (chemo)therapy and often high relapse rates that make it the leading cause of cancer death worldwide. Improvement of therapy efficacy is a crucial issue that might lead to a significant advance in the treatment of lung cancer. Oncolytic viruses are desirable combination partners in the developing field of cancer immunotherapy due to their direct cytotoxic effects and ability to elicit an immune response. Systemic oncolytic virus administration through intravenous injection should ideally lead to the highest efficacy in oncolytic activity. However, this is often hampered by the prevalence of host-specific, anti-viral immune responses. One way to achieve more efficient systemic oncolytic virus delivery is through better protection against neutralization by several components of the host immune system. Carrier cells, which can even have innate tumor tropism, have shown their appropriateness as effective vehicles for systemic oncolytic virus infection through circumventing restrictive features of the immune system and can warrant oncolytic virus delivery to tumors. In this overview, we summarize promising results from studies in which carrier cells have shown their usefulness for improved systemic oncolytic virus delivery and better oncolytic virus therapy against lung cancer.
Collapse
Affiliation(s)
- Giti Esmail Nia
- Faculty of Allied Medicine, Cellular and Molecular Research Centre, Iran University of Medical Science, Tehran, Iran
- Department of Basic Oncology, Health Institute of Ege University, Izmir, Turkey
| | - Elahe Nikpayam
- Department of Regenerative and Cancer Biology, Albany Medical College, Albany, NY, USA
| | | | - Azam Bolhassani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | - Ralph Meuwissen
- Department of Basic Oncology, Health Institute of Ege University, Izmir, Turkey
- Ege University Translational Pulmonary Research Center (EgeSAM), Ege University, Izmir, Turkey
| |
Collapse
|
7
|
Smieja J. Mathematical Modeling Support for Lung Cancer Therapy-A Short Review. Int J Mol Sci 2023; 24:14516. [PMID: 37833963 PMCID: PMC10572824 DOI: 10.3390/ijms241914516] [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: 07/03/2023] [Revised: 09/01/2023] [Accepted: 09/12/2023] [Indexed: 10/15/2023] Open
Abstract
The paper presents a review of models that can be used to describe dynamics of lung cancer growth and its response to treatment at both cell population and intracellular processes levels. To address the latter, models of signaling pathways associated with cellular responses to treatment are overviewed. First, treatment options for lung cancer are discussed, and main signaling pathways and regulatory networks are briefly reviewed. Then, approaches used to model specific therapies are discussed. Following that, models of intracellular processes that are crucial in responses to therapies are presented. The paper is concluded with a discussion of the applicability of the presented approaches in the context of lung cancer.
Collapse
Affiliation(s)
- Jaroslaw Smieja
- Department of Systems Biology and Engineering, Silesian University of Technology, ul. Akademicka 16, 44-100 Gliwice, Poland
| |
Collapse
|
8
|
Shang Z, Li J. Comparison of clinical efficacy between chrono-chemotherapy and conventional chemotherapy in patients with non-small cell lung cancer. Am J Cancer Res 2023; 13:4277-4287. [PMID: 37818045 PMCID: PMC10560957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 08/13/2023] [Indexed: 10/12/2023] Open
Abstract
This work focused on the clinical efficacy of chrono-chemotherapy and conventional chemotherapy on patients with non-small cell lung cancer (NSCLC), providing a theoretical basis for the clinical promotion of chrono-chemotherapy. 60 NSCLS patients in our hospital were randomly enrolled into a chrono-chemotherapy group and a conventional chemotherapy group, with 30 cases in each. Patients were treated with the standardized first-line treatment TP regimen (paclitaxel + cisplatin). After two cycles of chemotherapy, the clinical efficacy and adverse reactions of patients receiving various methods were observed. After the chemotherapy, CD3+, CD4+, and CD28+ increased while NK cells, B cells, and CD28- decreased in the conventional chemotherapy group (P<0.05); CD3+, CD4+, CD4+CD8+, B cells, and CD28+ increased while CD8+, NK cells, and CD28- decreased in chrono-chemotherapy group (P<0.05). The progression-free survival (PFS) of patients in the chrono-chemotherapy group (3.29 ± 0.46 years vs 2.56 ± 0.35 years) was longer (P<0.05). The quality of life (QOL) score in the chrono-chemotherapy group was higher (64.83 ± 1.54 points vs 51.72 ± 1.89 points) (P<0.05). The incidences of leukopenia (63.33%) and nausea and vomiting (53.33%) in the conventional chemotherapy group were higher than those in the chrono-chemotherapy group (30.00% and 30.00, respectively) (P<0.05). The chrono-chemotherapy could improve the cellular immune function of NSCLS patients, prolong their survival period, elevate the QOL, and reduce the side effects.
Collapse
Affiliation(s)
- Ziying Shang
- Department of Respiratory, Shengzhou People’s Hospital (The First Affiliated Hospital of Zhejiang University Shengzhou Branch)Shengzhou 312400, Zhejiang, China
| | - Juan Li
- Department of Radiotherapy, Hangzhou Cancer HospitalHangzhou 310000, Zhejiang, China
| |
Collapse
|
9
|
Wang J, Zhang Y, Zhang G, Xiang L, Pang H, Xiong K, Lu Y, Li J, Dai J, Lin S, Fu S. Radiotherapy-induced enrichment of EGF-modified doxorubicin nanoparticles enhances the therapeutic outcome of lung cancer. Drug Deliv 2022; 29:588-599. [PMID: 35156493 PMCID: PMC8856057 DOI: 10.1080/10717544.2022.2036871] [Citation(s) in RCA: 10] [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: 11/19/2021] [Revised: 01/24/2022] [Accepted: 01/25/2022] [Indexed: 11/30/2022] Open
Abstract
Chemotherapy is the primary treatment for advanced non-small-cell lung cancer (NSCLC). However, related dose-dependent toxicity limits its clinical use. Therefore, it is necessary to explore new strategies for improving the clinical outcomes while reducing the side effects of chemotherapy in the treatment of NSCLC. In this study, we designed and synthesized epidermal growth factor (EGF)-modified doxorubicin nanoparticles (EGF@DOX-NPs) that selectively targets the epidermal growth factor receptor (EGFR) overexpressed in lung tumor cells. When administered in combination with low-dose X-ray radiotherapy (RT), the NPs preferentially accumulated at the tumor site due to radiation-induced outburst of the local intra-tumoral blood vessels. Compared with DOX alone, EGF@DOX-NPs significantly decreased the viability and migration and enhanced the apoptosis rates of tumor cells in vitro. Also, the EGF@DOX-NPs significantly inhibited tumor growth in vivo, increasing the survival of the tumor-bearing mice without apparent systemic toxic effects through RT-induced aggregation. The tumor cell proliferation was greatly inhibited in the RT + EGF@DOX-NPs group. Contrarily, the apoptosis of tumor cells was significantly higher in this group. These results confirm the promising clinical application of radiotherapy in combination with EGF@DOX-NPs for lung cancer treatment.
Collapse
Affiliation(s)
- Jing Wang
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yan Zhang
- Department of Oncology, The Affiliated TCM Hospital of Southwest Medical University, Luzhou, China
| | - GuangPeng Zhang
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Li Xiang
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - HaoWen Pang
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Kang Xiong
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yun Lu
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - JianMei Li
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jie Dai
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Sheng Lin
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - ShaoZhi Fu
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
10
|
Nanomedicine for targeting the lung cancer cells by interpreting the signaling pathways. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
11
|
Zhao J, Wang L, Zhang H, Liao B, Li Y. Progress of Research in In Situ Smart Hydrogels for Local Antitumor Therapy: A Review. Pharmaceutics 2022; 14:pharmaceutics14102028. [PMID: 36297463 PMCID: PMC9611441 DOI: 10.3390/pharmaceutics14102028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/16/2022] [Accepted: 09/21/2022] [Indexed: 11/20/2022] Open
Abstract
Cancer seriously threatens human health. Surgery, radiotherapy and chemotherapy are the three pillars of traditional cancer treatment, with targeted therapy and immunotherapy emerging over recent decades. Standard drug regimens are mostly executed via intravenous injection (IV), especially for chemotherapy agents. However, these treatments pose severe risks, including off-target toxic side effects, low drug accumulation and penetration at the tumor site, repeated administration, etc., leading to inadequate treatment and failure to meet patients’ needs. Arising from these challenges, a local regional anticancer strategy has been proposed to enhance therapeutic efficacy and concomitantly reduce systemic toxicity. With the advances in biomaterials and our understanding of the tumor microenvironment, in situ stimulus-responsive hydrogels, also called smart hydrogels, have been extensively investigated for local anticancer therapy due to their injectability, compatibility and responsiveness to various stimuli (pH, enzyme, heat, light, magnetic fields, electric fields etc.). Herein, we focus on the latest progress regarding various stimuli that cause phase transition and drug release from smart hydrogels in local regional anticancer therapy. Additionally, the challenges and future trends of the reviewed in situ smart hydrogels for local drug delivery are summarized and proposed.
Collapse
|
12
|
Ma S, Cong Z, Wei J, Chen W, Ge D, Yang F, Liao Y. Pulmonary delivery of size-transformable nanoparticles improves tumor accumulation and penetration for chemo-sonodynamic combination therapy. J Control Release 2022; 350:132-145. [PMID: 35940360 DOI: 10.1016/j.jconrel.2022.08.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 07/03/2022] [Accepted: 08/03/2022] [Indexed: 11/26/2022]
Abstract
Very little is currently known about how inhaled nanomedicine for lung cancer treatment overcomes biological barriers hampering the tumor availability of drug and nanoparticles. Here, we developed a size-transformable nanocarrier (~ 119 nm) in which small-size nanoparticles (~ 28 nm) were loaded in the large nanocarrier after the addition of modified hyaluronan and could be released upon size-transformation at tumor tissue. Subsequently, the pulmonary and tumor pharmacokinetics of the two nanocarriers containing 7-ethyl-10-hydroxycamptothecin (SN38) and a covalently linked fluorescent sonosensitizer were comparatively investigated after intratracheal instillation to mice bearing orthotopic Lewis lung carcinoma tumors. The results showed that both instilled nanoparticles seemed to transport drug to tumor by direct access and transcytosis of nanoparticles, and diffusion of the released drug with the latter accounting for a great proportion of the drug tumor bioavailability. Relative to the small-size nanocarrier, the size-transformable counterpart appeared to restrict the mucociliary and absorption clearances from the lung and the clearance from the tumor interstitium to circulation, leading to increases in lung and tumor bioavailability of SN38 by 58.5% and 199%, respectively. In addition, the size-transformable nanoformulation conferred deep tumor penetration and sustained levels of both sonosensitizer and SN38 within tumors and simultaneously exerted sonodynamic- and chemo-therapies. Overall, the pulmonary delivery of size-transformable nanocarrier could co-deliver sonosensitizer and drug to deep tumor sites with enhanced tumor accumulation to realize combination therapy in lung cancer.
Collapse
Affiliation(s)
- Siqi Ma
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicines, Ministry of Education, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151 Malianwa North Road, Haidian District, Beijing 100193, PR China
| | - Zhaoqing Cong
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicines, Ministry of Education, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151 Malianwa North Road, Haidian District, Beijing 100193, PR China
| | - Jiaxing Wei
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicines, Ministry of Education, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151 Malianwa North Road, Haidian District, Beijing 100193, PR China
| | - Weiya Chen
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicines, Ministry of Education, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151 Malianwa North Road, Haidian District, Beijing 100193, PR China
| | - Di Ge
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicines, Ministry of Education, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151 Malianwa North Road, Haidian District, Beijing 100193, PR China
| | - Feifei Yang
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicines, Ministry of Education, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151 Malianwa North Road, Haidian District, Beijing 100193, PR China.
| | - Yonghong Liao
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicines, Ministry of Education, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151 Malianwa North Road, Haidian District, Beijing 100193, PR China.
| |
Collapse
|