1
|
Liu Y, Craig DQM, Parhizkar M. Controlled release of doxorubicin from Poly-(D,L-lactide-co-glycolide) (PLGA) nanoparticles prepared by coaxial electrospraying. Int J Pharm 2024; 666:124724. [PMID: 39312984 DOI: 10.1016/j.ijpharm.2024.124724] [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: 07/15/2024] [Revised: 09/12/2024] [Accepted: 09/13/2024] [Indexed: 09/25/2024]
Abstract
Enhancing the efficacy and reducing the toxicity of chemotherapeutic agents like doxorubicin (DOX) is crucial in cancer treatment. Core-shell nanoparticles (NPs) fabricated by coaxial electrospraying offer controlled release of anticancer agents with the polymer shell protecting drug molecules from rapid degradation, prolonging therapeutic effect. This study developed DOX-loaded poly(lactic-co-glycolic acid) (PLGA) NPs. NPs were fabricated with matrix or core-shell structure via single needle or coaxial electrospraying, respectively. Core-shell NPs exhibited high encapsulation efficiency (>80 %) with controlled DOX distribution. Compared to matrix NPs, core-shell NPs demonstrated slower sustained release (69 % in 144 h) after reduced initial burst (22 % in 8 h). Release kinetics followed a diffusion mechanism when compared to free drug and matrix DOX-loaded NPs. In vitro assays showed core-shell NPs' enhanced cytotoxicity against breast cancer cells MCF-7, with higher uptake observed by fluorescence microscopy and flow cytometry. The IC50 for core-shell NPs displayed a significant drop (0.115 μg/mL) compared to matrix NPs (0.235 μg/mL) and free DOX (1.482 μg/mL) after 72 h. Coaxial electrospraying enables the production of therapeutically advantageous core-shell NPs, offering controlled drug release with high encapsulation efficiency, potentially improving clinical anticancer chemotherapy.
Collapse
Affiliation(s)
- Yinan Liu
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
| | | | - Maryam Parhizkar
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK.
| |
Collapse
|
2
|
Moon Y, Cho H, Kim K. Nano-Delivery of Immunogenic Cell Death Inducers and Immune Checkpoint Blockade Agents: Single-Nanostructure Strategies for Enhancing Immunotherapy. Pharmaceutics 2024; 16:795. [PMID: 38931916 PMCID: PMC11207855 DOI: 10.3390/pharmaceutics16060795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 06/08/2024] [Accepted: 06/09/2024] [Indexed: 06/28/2024] Open
Abstract
Cancer immunotherapy has revolutionized oncology by harnessing the patient's immune system to target and eliminate cancer cells. However, immune checkpoint blockades (ICBs) face limitations such as low response rates, particularly in immunologically 'cold' tumors. Enhancing tumor immunogenicity through immunogenic cell death (ICD) inducers and advanced drug delivery systems represents a promising solution. This review discusses the development and application of various nanocarriers, including polymeric nanoparticles, liposomes, peptide-based nanoparticles, and inorganic nanoparticles, designed to deliver ICD inducers and ICBs effectively. These nanocarriers improve therapeutic outcomes by converting cold tumors into hot tumors, thus enhancing immune responses and reducing systemic toxicity. By focusing on single-nanoparticle systems that co-deliver both ICD inducers and ICBs, this review highlights their potential in achieving higher drug concentrations at tumor sites, improving pharmacokinetics and pharmacodynamics, and facilitating clinical translation. Future research should aim to optimize these nanocarrier systems for better in vivo performance and clinical applications, ultimately advancing cancer immunotherapy.
Collapse
Affiliation(s)
- Yujeong Moon
- Department of Bioengineering, Korea University, Seoul 02841, Republic of Korea;
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Hanhee Cho
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea;
| | - Kwangmeyung Kim
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea;
| |
Collapse
|
3
|
Blaya-Cánovas JL, Griñán-Lisón C, Blancas I, Marchal JA, Ramírez-Tortosa C, López-Tejada A, Benabdellah K, Cortijo-Gutiérrez M, Cano-Cortés MV, Graván P, Navarro-Marchal SA, Gómez-Morales J, Delgado-Almenta V, Calahorra J, Agudo-Lera M, Sagarzazu A, Rodríguez-González CJ, Gallart-Aragón T, Eich C, Sánchez-Martín RM, Granados-Principal S. Autologous patient-derived exhausted nano T-cells exploit tumor immune evasion to engage an effective cancer therapy. Mol Cancer 2024; 23:83. [PMID: 38730475 PMCID: PMC11084007 DOI: 10.1186/s12943-024-01997-x] [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: 01/18/2024] [Accepted: 04/05/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Active targeting by surface-modified nanoplatforms enables a more precise and elevated accumulation of nanoparticles within the tumor, thereby enhancing drug delivery and efficacy for a successful cancer treatment. However, surface functionalization involves complex procedures that increase costs and timelines, presenting challenges for clinical implementation. Biomimetic nanoparticles (BNPs) have emerged as unique drug delivery platforms that overcome the limitations of actively targeted nanoparticles. Nevertheless, BNPs coated with unmodified cells show reduced functionalities such as specific tumor targeting, decreasing the therapeutic efficacy. Those challenges can be overcome by engineering non-patient-derived cells for BNP coating, but these are complex and cost-effective approaches that hinder their wider clinical application. Here we present an immune-driven strategy to improve nanotherapeutic delivery to tumors. Our unique perspective harnesses T-cell exhaustion and tumor immune evasion to develop a groundbreaking new class of BNPs crafted from exhausted T-cells (NExT) of triple-negative breast cancer (TNBC) patients by specific culture methods without sophisticated engineering. METHODS NExT were generated by coating PLGA (poly(lactic-co-glycolic acid)) nanoparticles with TNBC-derived T-cells exhausted in vitro by acute activation. Physicochemical characterization of NExT was made by dynamic light scattering, electrophoretic light scattering and transmission electron microscopy, and preservation and orientation of immune checkpoint receptors by flow cytometry. The efficacy of chemotherapy-loaded NExT was assessed in TNBC cell lines in vitro. In vivo toxicity was made in CD1 mice. Biodistribution and therapeutic activity of NExT were determined in cell-line- and autologous patient-derived xenografts in immunodeficient mice. RESULTS We report a cost-effective approach with a good performance that provides NExT naturally endowed with immune checkpoint receptors (PD1, LAG3, TIM3), augmenting specific tumor targeting by engaging cognate ligands, enhancing the therapeutic efficacy of chemotherapy, and disrupting the PD1/PDL1 axis in an immunotherapy-like way. Autologous patient-derived NExT revealed exceptional intratumor accumulation, heightened chemotherapeutic index and efficiency, and targeted the tumor stroma in a PDL1+ patient-derived xenograft model of triple-negative breast cancer. CONCLUSIONS These advantages underline the potential of autologous patient-derived NExT to revolutionize tailored adoptive cancer nanotherapy and chemoimmunotherapy, which endorses their widespread clinical application of autologous patient-derived NExT.
Collapse
Grants
- PRDJA19001BLAY Fundación Científica Asociación Española Contra el Cáncer
- POSTDOC_21_638 Consejería de Economía, Innovación, Ciencia y Empleo, Junta de Andalucía
- RTI2018.101309B-C22 Ministerio de Ciencia, Innovación y Universidades
- FPU19/04450 Ministerio de Ciencia, Innovación y Universidades
- DOC_01686 Consejería de Transformación Económica, Industria, Conocimiento y Universidades
- PI19/01533 Instituto de Salud Carlos III
- P29/22/02 Consejería de Economía, Conocimiento, Empresas y Universidad, Junta de Andalucía, Spain
Collapse
Affiliation(s)
- José L Blaya-Cánovas
- UGC de Oncología Médica, Hospital Universitario de Jaén, Jaén, 23007, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada- University of Granada, Granada, 18100, Spain
- Centre for Genomics and Oncological Research, GENYO, Pfizer/University of Granada/Andalusian Regional Government, Granada, 18016, Spain
| | - Carmen Griñán-Lisón
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada- University of Granada, Granada, 18100, Spain
- Centre for Genomics and Oncological Research, GENYO, Pfizer/University of Granada/Andalusian Regional Government, Granada, 18016, Spain
- Department of Biochemistry and Molecular Biology 2, Faculty of Pharmacy, University of Granada, Campus de Cartuja s/n, Granada, 18071, Spain
- Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, 18100, Spain
| | - Isabel Blancas
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada- University of Granada, Granada, 18100, Spain
- UGC de Oncología, Hospital Universitario San Cecilio, Granada, 18016, Spain
- Department of Medicine, University of Granada, Granada, 18016, Spain
| | - Juan A Marchal
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada- University of Granada, Granada, 18100, Spain
- Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, 18100, Spain
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, (CIBM), University of Granada, Granada, 18100, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, 18016, Spain
| | - César Ramírez-Tortosa
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada- University of Granada, Granada, 18100, Spain
- UGC de Anatomía Patológica, Hospital San Cecilio, Granada, 18016, Spain
| | - Araceli López-Tejada
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada- University of Granada, Granada, 18100, Spain
- Centre for Genomics and Oncological Research, GENYO, Pfizer/University of Granada/Andalusian Regional Government, Granada, 18016, Spain
- Department of Biochemistry and Molecular Biology 2, Faculty of Pharmacy, University of Granada, Campus de Cartuja s/n, Granada, 18071, Spain
| | - Karim Benabdellah
- Centre for Genomics and Oncological Research, GENYO, Pfizer/University of Granada/Andalusian Regional Government, Granada, 18016, Spain
| | - Marina Cortijo-Gutiérrez
- Centre for Genomics and Oncological Research, GENYO, Pfizer/University of Granada/Andalusian Regional Government, Granada, 18016, Spain
| | - M Victoria Cano-Cortés
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada- University of Granada, Granada, 18100, Spain
- Centre for Genomics and Oncological Research, GENYO, Pfizer/University of Granada/Andalusian Regional Government, Granada, 18016, Spain
- Department of Medicinal & Organic Chemistry and Excellence Research Unit of "Chemistry Applied to Biomedicine and the Environment", Faculty of Pharmacy, University of Granada, Campus de Cartuja s/n, Granada, 18071, Spain
| | - Pablo Graván
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada- University of Granada, Granada, 18100, Spain
- Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, 18100, Spain
- Department of Applied Physics, Faculty of Science, University of Granada, Granada, 18071, Spain
| | - Saúl A Navarro-Marchal
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada- University of Granada, Granada, 18100, Spain
- Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, 18100, Spain
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, (CIBM), University of Granada, Granada, 18100, Spain
- Department of Applied Physics, Faculty of Science, University of Granada, Granada, 18071, Spain
| | - Jaime Gómez-Morales
- Laboratorio de Estudios Cristalográficos IACT-CSIC-UGR, Armilla, 18100, Spain
| | - Violeta Delgado-Almenta
- Centre for Genomics and Oncological Research, GENYO, Pfizer/University of Granada/Andalusian Regional Government, Granada, 18016, Spain
| | - Jesús Calahorra
- UGC de Oncología Médica, Hospital Universitario de Jaén, Jaén, 23007, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada- University of Granada, Granada, 18100, Spain
- Centre for Genomics and Oncological Research, GENYO, Pfizer/University of Granada/Andalusian Regional Government, Granada, 18016, Spain
| | - María Agudo-Lera
- Centre for Genomics and Oncological Research, GENYO, Pfizer/University of Granada/Andalusian Regional Government, Granada, 18016, Spain
| | - Amaia Sagarzazu
- Centre for Genomics and Oncological Research, GENYO, Pfizer/University of Granada/Andalusian Regional Government, Granada, 18016, Spain
| | | | - Tania Gallart-Aragón
- Department of Medicine, University of Granada, Granada, 18016, Spain
- UGC de Cirugía General y del Aparato Digestivo, Hospital Universitario San Cecilio, Granada, 18016, Spain
| | - Christina Eich
- Translational Nanobiomaterials and Imaging, Department of Radiology, Leiden University Medical Center, Leiden, 2333, The Netherlands
| | - Rosario M Sánchez-Martín
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada- University of Granada, Granada, 18100, Spain
- Centre for Genomics and Oncological Research, GENYO, Pfizer/University of Granada/Andalusian Regional Government, Granada, 18016, Spain
- Department of Medicinal & Organic Chemistry and Excellence Research Unit of "Chemistry Applied to Biomedicine and the Environment", Faculty of Pharmacy, University of Granada, Campus de Cartuja s/n, Granada, 18071, Spain
| | - Sergio Granados-Principal
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada- University of Granada, Granada, 18100, Spain.
- Centre for Genomics and Oncological Research, GENYO, Pfizer/University of Granada/Andalusian Regional Government, Granada, 18016, Spain.
- Department of Biochemistry and Molecular Biology 2, Faculty of Pharmacy, University of Granada, Campus de Cartuja s/n, Granada, 18071, Spain.
| |
Collapse
|
4
|
Voznyuk AA, Makarets YA, Advakhova DY, Khafizov KA, Lugovoi ME, Zakharova VA, Senatov FS, Koudan EV. Biodegradable Local Chemotherapy Platform with Prolonged and Controlled Release of Doxorubicin for the Prevention of Local Tumor Recurrence. ACS APPLIED BIO MATERIALS 2024; 7:2472-2487. [PMID: 38480461 DOI: 10.1021/acsabm.4c00078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Local recurrence after surgical and therapeutic treatment remains a significant clinical problem in oncology. Recurrence may be due to imperfections in existing therapies, particularly chemotherapy. To improve antitumor activity and prevent local cancer recurrence while keeping toxicity at acceptable levels, we have developed and demonstrated a biodegradable local chemotherapy platform that provides controlled and prolonged drug release. The platform consists of a polycaprolactone (PCL) substrate, which provides the structural integrity of the platform and the predominant unidirectional drug release, and a thin multilayer coating (∼200 nm) containing doxorubicin (DOX). The coating is an electrostatic complex obtained by the layer-by-layer (LbL) assembly and consists of natural polyelectrolytes [poly-γ-glutamic acid (γ-PGA) and chitosan (CS) or poly-l-lysine (PLL)]. To improve the release stability, an ionic conjugate of DOX and γ-PGA was prepared and incorporated into the multilayer coating. By varying the structure of the coating by adding empty (without DOX) bilayers, we were able to control the kinetics of drug release. The resulting platforms contained equal numbers of empty bilayers and DOX-loaded bilayers (15 + 15 or 30 + 30 bilayers) with a maximum loading of 566 ng/cm2. The platforms demonstrated prolonged and fairly uniform drug release for more than 5 months while retaining antitumor activity in vitro on ovarian cancer cells (SKOV-3). The empty platforms (without DOX) showed good cytocompatibility and no cytotoxicity to human fibroblasts and SKOV-3 cells. This study presents the development of a local chemotherapy platform consisting of a PCL-based substrate which provides structural stability and a biodegradable polyelectrolyte layered coating which combines layers containing a polyanion ionic complex with DOX with empty bilayers to ensure prolonged and controlled drug release. Our results may provide a basis for improving the efficacy of chemotherapy using drug delivery systems.
Collapse
Affiliation(s)
- Amina A Voznyuk
- National University of Science and Technology MISIS, Leninskiy pr. 4, Moscow 119049, Russian Federation
| | - Yulia A Makarets
- National University of Science and Technology MISIS, Leninskiy pr. 4, Moscow 119049, Russian Federation
| | - Darya Yu Advakhova
- National University of Science and Technology MISIS, Leninskiy pr. 4, Moscow 119049, Russian Federation
| | - Krestina A Khafizov
- Haute École de la Province de Namur, Henri Blès st. 192, Namur 5000, Belgium
| | - Maksim E Lugovoi
- National University of Science and Technology MISIS, Leninskiy pr. 4, Moscow 119049, Russian Federation
| | - Vasilina A Zakharova
- National University of Science and Technology MISIS, Leninskiy pr. 4, Moscow 119049, Russian Federation
| | - Fedor S Senatov
- National University of Science and Technology MISIS, Leninskiy pr. 4, Moscow 119049, Russian Federation
| | - Elizaveta V Koudan
- National University of Science and Technology MISIS, Leninskiy pr. 4, Moscow 119049, Russian Federation
| |
Collapse
|
5
|
Hassan AL-Saeedi R, Khalaj-Kondori M, Hosseinpour Feizi MA, Hajavi J. DOX-PLGA Nanoparticles Effectively Suppressed the Expression of Pro-Inflammatory Cytokines TNF-a, IL-6, iNOS, and IL-1β in MCF-7 Breast Cancer Cell Line. Rep Biochem Mol Biol 2024; 12:530-539. [PMID: 39086585 PMCID: PMC11288233 DOI: 10.61186/rbmb.12.4.530] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 06/14/2024] [Indexed: 08/02/2024]
Abstract
Background Inflammation contributes to cancer pathobiology through different mechanisms. Higher levels of pro-inflammatory cytokines can lead to hyperinflammation and promote cancer development and metastasis. For cancer treatment, Doxorubicin (DOX) can be encapsulated into the poly-lactic-glycolic acid (PLGA) nanoparticles. This study aimed to investigate the impact of doxorubicin-loaded PLGA nanoparticles (DOX-PLGA NP) on the expression of pro-inflammatory genes TNF-α, IL-6, iNOS, and IL-1β in the MCF-7 cells. Methods The DOX-PLGA NP was prepared by loading doxorubicin into PLGA and characterized using dynamic light scattering (DLS) and atomic force microscopy (AFM). The cytotoxic effect of the nanoparticles was determined by the MTT assay, and their impacts on the expression of pro-inflammatory genes were assessed by qRT-PCR. Results The encapsulation efficiency and loading capacity were 60±1.5 and 1.13±0.21 percent, respectively. The zeta potential and mean DOX-PLGA nanoparticle size were -18±0.550 mV and 172±55.6 nm, respectively. The 50% inhibitory concentration (IC50) of the DOX-PLGA NP on MCF-7 cell viability was 24.55 µg/mL after 72 hours of treatment. The qRT-PCR results revealed that the 20 µg/mL concentration of the DOX-PLGA NP significantly suppressed the expression of the pro-inflammatory genes TNF-α, IL-6, iNOS, and IL-1β compared to DOX alone (20 µg/mL). Additionally, the suppression effect of DOX-PLGA NP on the expression of these pro-inflammatory genes was dose-dependent. Conclusions These results show that DOX-PLGA NP efficiently suppressed the expression of pro-inflammatory genes. Furthermore, encapsulation of DOX into PLGA nanoparticles significantly improved the effectiveness of DOX in suppressing pro-inflammatory genes in MCF-7 breast cancer cells.
Collapse
Affiliation(s)
- Rawan Hassan AL-Saeedi
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran.
| | - Mohammad Khalaj-Kondori
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran.
| | | | - Jafar Hajavi
- Department of Microbiology, Faculty of Medicine, Infectious Diseases Research Center, Gonabad University of Medical Science, Gonabad, Iran.
- Innovative Medical Research Center, Mashhad Branch, Islamic Azad University, Mashhad, Iran.
| |
Collapse
|
6
|
Yun WS, Kim J, Lim DK, Kim DH, Jeon SI, Kim K. Recent Studies and Progress in the Intratumoral Administration of Nano-Sized Drug Delivery Systems. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:2225. [PMID: 37570543 PMCID: PMC10421122 DOI: 10.3390/nano13152225] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/23/2023] [Accepted: 07/28/2023] [Indexed: 08/13/2023]
Abstract
Over the last 30 years, diverse types of nano-sized drug delivery systems (nanoDDSs) have been intensively explored for cancer therapy, exploiting their passive tumor targetability with an enhanced permeability and retention effect. However, their systemic administration has aroused some unavoidable complications, including insufficient tumor-targeting efficiency, side effects due to their undesirable biodistribution, and carrier-associated toxicity. In this review, the recent studies and advancements in intratumoral nanoDDS administration are generally summarized. After identifying the factors to be considered to enhance the therapeutic efficacy of intratumoral nanoDDS administration, the experimental results on the application of intratumoral nanoDDS administration to various types of cancer therapies are discussed. Subsequently, the reports on clinical studies of intratumoral nanoDDS administration are addressed in short. Intratumoral nanoDDS administration is proven with its versatility to enhance the tumor-specific accumulation and retention of therapeutic agents for various therapeutic modalities. Specifically, it can improve the efficacy of therapeutic agents with poor bioavailability by increasing their intratumoral concentration, while minimizing the side effect of highly toxic agents by restricting their delivery to normal tissues. Intratumoral administration of nanoDDS is considered to expand its application area due to its potent ability to improve therapeutic effects and relieve the systemic toxicities of nanoDDSs.
Collapse
Affiliation(s)
- Wan Su Yun
- Korea Institute of Science and Technology (KU-KIST), Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Jeongrae Kim
- Korea Institute of Science and Technology (KU-KIST), Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Dong-Kwon Lim
- Korea Institute of Science and Technology (KU-KIST), Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Dong-Hwee Kim
- Korea Institute of Science and Technology (KU-KIST), Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Seong Ik Jeon
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Kwangmeyung Kim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| |
Collapse
|
7
|
de Jesús Martín-Camacho U, Rodríguez-Barajas N, Alberto Sánchez-Burgos J, Pérez-Larios A. Weibull β value for the discernment of drug release mechanism of PLGA particles. Int J Pharm 2023; 640:123017. [PMID: 37149112 DOI: 10.1016/j.ijpharm.2023.123017] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 04/27/2023] [Accepted: 04/30/2023] [Indexed: 05/08/2023]
Abstract
Mathematical models are used to characterize and optimize drug release in drug delivery systems (DDS). One of the most widely used DDS is the poly(lactic-co-glycolic acid) (PLGA)-based polymeric matrix owing to its biodegradability, biocompatibility, and easy manipulation of its properties through the manipulation of synthesis processes. Over the years, the Korsmeyer-Peppas model has been the most widely used model for characterizing the release profiles of PLGA DDS. However, owing to the limitations of the Korsmeyer-Peppas model, the Weibull model has emerged as an alternative for the characterization of the release profiles of PLGA polymeric matrices. The purpose of this study was to establish a correlation between the n and β parameters of the Korsmeyer-Peppas and Weibull models and to use the Weibull model to discern the drug release mechanism. A total of 451 datasets describing the overtime drug release of PLGA-based formulations from 173 scientific articles were fitted to both models. The Korsmeyer-Peppas model had a mean Akaike Information Criteria (AIC) value of 54.52 and an n value of 0.42, while the Weibull model had a mean AIC of 51.99 and a β value of 0.55, and by using reduced major axis regression values, a high correlation was found between the n and β values. These results demonstrate the ability of the Weibull model to characterize the release profiles of PLGA-based matrices and the usefulness of the β parameter for determining the drug release mechanism.
Collapse
Affiliation(s)
- Ubaldo de Jesús Martín-Camacho
- Laboratorio de Investigación en Materiales, Agua y Energía, Departamento de Ingeniería, Centro Universitario de los Altos, Universidad de Guadalajara, Tepatitlán de Morelos, Jal., México, 47600
| | - Noé Rodríguez-Barajas
- Laboratorio de Investigación en Materiales, Agua y Energía, Departamento de Ingeniería, Centro Universitario de los Altos, Universidad de Guadalajara, Tepatitlán de Morelos, Jal., México, 47600
| | | | - Alejandro Pérez-Larios
- Laboratorio de Investigación en Materiales, Agua y Energía, Departamento de Ingeniería, Centro Universitario de los Altos, Universidad de Guadalajara, Tepatitlán de Morelos, Jal., México, 47600.
| |
Collapse
|
8
|
Uzhytchak M, Smolková B, Lunova M, Frtús A, Jirsa M, Dejneka A, Lunov O. Lysosomal nanotoxicity: Impact of nanomedicines on lysosomal function. Adv Drug Deliv Rev 2023; 197:114828. [PMID: 37075952 DOI: 10.1016/j.addr.2023.114828] [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: 11/12/2021] [Revised: 03/28/2023] [Accepted: 04/12/2023] [Indexed: 04/21/2023]
Abstract
Although several nanomedicines got clinical approval over the past two decades, the clinical translation rate is relatively small so far. There are many post-surveillance withdrawals of nanomedicines caused by various safety issues. For successful clinical advancement of nanotechnology, it is of unmet need to realize cellular and molecular foundation of nanotoxicity. Current data suggest that lysosomal dysfunction caused by nanoparticles is emerging as the most common intracellular trigger of nanotoxicity. This review analyzes prospect mechanisms of lysosomal dysfunction-mediated toxicity induced by nanoparticles. We summarized and critically assessed adverse drug reactions of current clinically approved nanomedicines. Importantly, we show that physicochemical properties have great impact on nanoparticles interaction with cells, excretion route and kinetics, and subsequently on toxicity. We analyzed literature on adverse reactions of current nanomedicines and hypothesized that adverse reactions might be linked with lysosomal dysfunction caused by nanomedicines. Finally, from our analysis it becomes clear that it is unjustifiable to generalize safety and toxicity of nanoparticles, since different particles possess distinct toxicological properties. We propose that the biological mechanism of the disease progression and treatment should be central in the optimization of nanoparticle design.
Collapse
Affiliation(s)
- Mariia Uzhytchak
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic
| | - Barbora Smolková
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic
| | - Mariia Lunova
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic; Institute for Clinical & Experimental Medicine (IKEM), 14021 Prague, Czech Republic
| | - Adam Frtús
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic
| | - Milan Jirsa
- Institute for Clinical & Experimental Medicine (IKEM), 14021 Prague, Czech Republic
| | - Alexandr Dejneka
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic
| | - Oleg Lunov
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic.
| |
Collapse
|
9
|
Asmawi AA, Salim N, Abdulmalek E, Abdul Rahman MB. Size-Controlled Preparation of Docetaxel- and Curcumin-Loaded Nanoemulsions for Potential Pulmonary Delivery. Pharmaceutics 2023; 15:pharmaceutics15020652. [PMID: 36839974 PMCID: PMC9964819 DOI: 10.3390/pharmaceutics15020652] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 02/09/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
Lung cancer is one of the deadliest pulmonary diseases in the world. Although docetaxel (DTX) has exhibited superior efficacy in lung cancer treatment, it has demonstrated numerous adverse effects and poor bioavailability. The natural product extract, curcumin (CCM), has reportedly reduced toxicity and synergistically improved DTX bioavailability. Nonetheless, the hydrophobic nature of DTX and CCM limits their clinical use. Nanoemulsion pulmonary delivery of DTX and CCM has demonstrated potential as a drug carrier to alleviate these drawbacks. The controlled preparation of inhalable DTX- and CCM-loaded nanoemulsions within the 100 to 200 nm range was explored in this study. A response surface methodology (RSM) based on a central composite design (CCD) was utilized to fabricate the desired size of the nanoemulsion under optimized conditions. Different process parameters were employed to control the size of the nanoemulsions procured through a high-energy emulsification technique. The size of the resultant nanoemulsions decreased with increasing energy input. The actual response according to the targeted sizes for DTX- and CCM-loaded nanoemulsion models exhibited excellent agreement with the predicted value at below 5% residual standard error under optimized conditions. The nanoemulsion of 100 nm particle size demonstrated better membrane permeability than their larger counterparts. Moreover, the formulations documented favorable physicochemical and aerodynamic pulmonary delivery properties and reduced toxicity in human lung fibroblast (MRC-5) cells. Hence, this tunable size of nanoemulsions could be a suitable alternative drug delivery for pulmonary diseases with increased local lung concentration.
Collapse
Affiliation(s)
- Azren Aida Asmawi
- Integrated Chemical BioPhysics Research, Department of Chemistry, Faculty of Science, Universiti Putra Malaysia, Serdang 43400, Malaysia
- UPM-MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Malaysia
- Correspondence: (A.A.A.); (M.B.A.R.); Tel.: +60-397696798 (M.B.A.R.)
| | - Norazlinaliza Salim
- Integrated Chemical BioPhysics Research, Department of Chemistry, Faculty of Science, Universiti Putra Malaysia, Serdang 43400, Malaysia
- Centre of Foundation Studies for Agricultural Science, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Emilia Abdulmalek
- Integrated Chemical BioPhysics Research, Department of Chemistry, Faculty of Science, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Mohd Basyaruddin Abdul Rahman
- Integrated Chemical BioPhysics Research, Department of Chemistry, Faculty of Science, Universiti Putra Malaysia, Serdang 43400, Malaysia
- UPM-MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Malaysia
- Correspondence: (A.A.A.); (M.B.A.R.); Tel.: +60-397696798 (M.B.A.R.)
| |
Collapse
|
10
|
Sokol M, Gulyaev I, Mollaeva M, Kuznetsov S, Zenin V, Klimenko M, Yabbarov N, Chirkina M, Nikolskaya E. Box-Behnken assisted development and validation of high-performance liquid chromatography method for the simultaneous determination of doxorubicin and vorinostat in polymeric nanoparticles. J Sep Sci 2023; 46:e2200731. [PMID: 36427291 DOI: 10.1002/jssc.202200731] [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: 09/09/2022] [Revised: 11/21/2022] [Accepted: 11/23/2022] [Indexed: 11/26/2022]
Abstract
While histone deacetylase inhibitors, such as vorinostat, demonstrate a significant effect against hematological cancers, their application for solid tumor treatment is limited. However, there is strong evidence that combinatorial administration of vorinostat and genotoxic agents (e.g., doxorubicin) enhances the antitumoral action of both drugs against tumors. We developed a high-performance liquid chromatography method for the simultaneous determination of doxorubicin and vorinostat in polymeric nanoparticles designed to provide the parenteral administration of both drugs and increase their safety profile. We performed separation on Nucleodur C-18 Gravity column with a mixture of 10 mM potassium dihydrogen phosphate buffer pH 3.9:ACN (90:10 v/v) as mobile phase at 240 nm. The method was linear within the concentration range of 4.2-52.0 μg/ml for both drugs with limits of detection and quantification of 3.5 and 10.7 μg/ml for doxorubicin and 2.5 and 7.7 μg/ml for vorinostat, respectively. The method was precise and accurate over the concentration range of analysis. Drug loading was 5.4% for doxorubicin and 0.8% for vorinostat. Degradation of doxorubicin after irradiation was less than 5%, while the amount of vorinostat decreased at 88% under the same conditions. Thus, the validated method could be adopted for routine simultaneous analysis of doxorubicin and vorinostat in polymeric nanoparticles.
Collapse
Affiliation(s)
- Maria Sokol
- Laboratory of quantitative oncology, N. M. Emanuel Institute of Biochemical Physics RAS, Moscow, Russia
| | - Ivan Gulyaev
- Laboratory of quantitative oncology, N. M. Emanuel Institute of Biochemical Physics RAS, Moscow, Russia.,Department of Chemistry and Technology of Biomedical Preparations, Mendeleev University of Chemical Technology of Russia, Moscow, Russia
| | - Mariia Mollaeva
- Laboratory of quantitative oncology, N. M. Emanuel Institute of Biochemical Physics RAS, Moscow, Russia
| | - Sergey Kuznetsov
- Department of Nanobiomaterials and Structures, National Research Center "Kurchatov Institute", Moscow, Russia
| | - Vladimir Zenin
- Laboratory of molecular biotechnology, Federal State Institution, Federal Research Centre, Fundamentals of Biotechnology, Russian Academy of Sciences, Moscow, Russia
| | - Maksim Klimenko
- Laboratory of quantitative oncology, N. M. Emanuel Institute of Biochemical Physics RAS, Moscow, Russia.,Department of Chemistry and Technology of Biomedical Preparations, Mendeleev University of Chemical Technology of Russia, Moscow, Russia
| | - Nikita Yabbarov
- Laboratory of quantitative oncology, N. M. Emanuel Institute of Biochemical Physics RAS, Moscow, Russia
| | - Margarita Chirkina
- Laboratory of quantitative oncology, N. M. Emanuel Institute of Biochemical Physics RAS, Moscow, Russia
| | - Elena Nikolskaya
- Laboratory of quantitative oncology, N. M. Emanuel Institute of Biochemical Physics RAS, Moscow, Russia
| |
Collapse
|
11
|
Wang W, Kojima H, Gao M, Yin X, Uchida T, Ni J. Optimization of O/W Emulsion Solvent Evaporation Method for Itraconazole Sustained Release Microspheres. Chem Pharm Bull (Tokyo) 2023; 71:520-527. [PMID: 37394601 DOI: 10.1248/cpb.c22-00747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Itraconazole, a commonly used antifungal drug in the clinic approved by U.S. Food and Drug Administration (FDA), has been gradually found to have anti-tumor, angiogenesis inhibition and other pharmacological activities. However, its poor water solubility and potential toxicity limited its clinical application. In order to improve the water solubility and reduce the side effects caused by the high concentration of itraconazole, a novel preparation method of itraconazole sustained release microspheres was established in this study. Firstly, five kinds of polylactic acid-glycolic acid (PLGA) microspheres loaded with itraconazole were prepared by oil/water (O/W) emulsion solvent evaporation and then characterized by infrared spectroscopy. Then the particle size and morphology of the microspheres were observed by scanning electron microscope (SEM) and transmission electron microscope (TEM). After that, the particle size distribution, drug loading rate, entrapment efficiency, and drug release experiments were evaluated. Our results showed the microspheres prepared in this study had uniform particle size distribution and good integrity. Further study found that the average drug loading of the five kinds of microspheres prepared with PLGA 7505, PLGA 7510, PLGA 7520, PLGA 5020 and PLGA 0020 were 16.88, 17.72, 16.72, 16.57, and 16.64%, respectively, and the encapsulation rate all reached about 100%. More surprisingly, the release experimental results showed that the microspheres prepared with PLGA 7520 did not show sudden release, showing good sustained release performance and high drug release rate. To sum up, this study optimized the preparation method of sustained-release microspheres without sudden release, which provides a new solution for the delivery of itraconazole in the clinic.
Collapse
Affiliation(s)
- Wenping Wang
- Department of Pharmacy, China-Japan Friendship Hospital
| | - Honami Kojima
- Faculty of Pharmaceutical Sciences, Mukogawa Women's University
| | - Ming Gao
- Faculty of Pharmaceutical Sciences, Mukogawa Women's University
| | - Xingbin Yin
- School of Chinese Materia Medica, Beijing University of Chinese Medicine
| | - Takahiro Uchida
- Faculty of Pharmaceutical Sciences, Mukogawa Women's University
| | - Jian Ni
- School of Chinese Materia Medica, Beijing University of Chinese Medicine
| |
Collapse
|
12
|
Harris JC, Sterin EH, Day ES. Membrane-Wrapped Nanoparticles for Enhanced Chemotherapy of Acute Myeloid Leukemia. ACS Biomater Sci Eng 2022; 8:4439-4448. [PMID: 36103274 PMCID: PMC9633094 DOI: 10.1021/acsbiomaterials.2c00832] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
This work reports the development of a biomimetic membrane-wrapped nanoparticle (MWNP) platform for targeted chemotherapy of acute myeloid leukemia (AML). Doxorubicin (DOX), a chemotherapeutic used to treat leukemias, lymphomas, and other cancers, was encapsulated in polymeric NPs that were coated with cytoplasmic membranes derived from human AML cells. The release rate of DOX from the MWNPs was characterized under both storage and physiological conditions, with faster release observed at pH 5.5 than pH 7.4. The system was then introduced to AML cell cultures to test the functionality of the released DOX cargo as compared to DOX delivered freely or via NPs coated with poly(ethylene glycol) (PEG). The MWNPs delivered DOX in an efficient and targeted manner, inducing up to 80% apoptosis in treated cells at a dose of 5 μM, compared to 15% for free DOX and 17% for DOX-loaded PEG-coated NPs at the same drug concentration. The mechanism of cell death was confirmed as DNA double-strand breaks through a γH2A.X assay, indicating that the released DOX retained its expected mechanism of action. These findings designate MWNPs as a robust drug delivery system with great potential for future development in treatments of AML and other blood cancers.
Collapse
Affiliation(s)
- Jenna C Harris
- Department of Materials Science and Engineering, University of Delaware, 201 DuPont Hall, Newark, Delaware 19716, United States
| | - Eric H Sterin
- Department of Biomedical Engineering, University of Delaware, 590 Avenue 1743, 4th Floor, Newark, Delaware 19713, United States
| | - Emily S Day
- Department of Materials Science and Engineering, University of Delaware, 201 DuPont Hall, Newark, Delaware 19716, United States
- Department of Biomedical Engineering, University of Delaware, 590 Avenue 1743, 4th Floor, Newark, Delaware 19713, United States
- Helen F. Graham Cancer Center and Research Institute, 4701 Ogletown-Stanton Road, Newark, Delaware 19713, United States
| |
Collapse
|
13
|
Marshall SK, Saelim B, Taweesap M, Pachana V, Panrak Y, Makchuchit N, Jaroenpakdee P. Anti-EGFR Targeted Multifunctional I-131 Radio-Nanotherapeutic for Treating Osteosarcoma: In Vitro 3D Tumor Spheroid Model. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:3517. [PMID: 36234645 PMCID: PMC9565722 DOI: 10.3390/nano12193517] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/01/2022] [Accepted: 10/04/2022] [Indexed: 06/16/2023]
Abstract
The systemic delivery of doxorubicin (DOX) to treat osteosarcoma requires an adequate drug concentration to be effective, but in doing so, it raises the risk of increasing organ off-target toxicity and developing drug resistance. Herein, this study reveals a multiple therapeutic nanocarrier delivery platform that overcomes off-target toxicity by providing good specificity and imparting enhanced tumor penetration in a three-dimensional (3D) human MG-63 spheroid model. By synthesizing PEG-PLGA nanoparticles by the double emulsion method, encapsulating DOX and Na131I in the inner core, and conjugating with an epidermal growth factor receptor (EGFR) antibody, it is intended to specifically target human MG-63 cells. The nanocarrier is biocompatible with blood and has good stability characteristics. Na131I encapsulation efficiency was >96%, and radiochemical purity was >96% over 96 h. A DOX encapsulation efficacy of ~80% was achieved, with a drug loading efficiency of ~3%, and a sustained DOX release over 5 days. The nanocarrier EGFR antibody achieved a ~80-fold greater targeting efficacy to MG-63 cells (EGFR+) than fibroblast cells (EGFR−). The targeted multiple therapeutic DIE-NPs have a higher penetration and uptake of Na131I to the 3D model and a ~3-fold higher cytotoxicity than the DOX monotherapy (D-NPs). The co-administration of DOX and Na131I (DIE-NPs) disrupts DNA repair and generates free radicals resulting in DNA damage, triggering the activation of apoptosis pathways. This leads to inhibition of MG-63 cell proliferation and promotes cell cycle arrest in the G0/G1 phase. Furthermore, the PEGylated anti-EGFR functionalized DIE-NPs were found to be biocompatible with red blood cells and to have no adverse effects. This anti-EGFR targeted multifunctional I-131 radio-nanotherapeutic signifies a customizable specific targeted treatment for osteosarcoma.
Collapse
Affiliation(s)
- Suphalak Khamruang Marshall
- Department of Radiology, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
- Molecular Imaging and Cyclotron Center, Division of Nuclear Medicine, Department of Radiology, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| | - Boonyisa Saelim
- Department of Radiology, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| | - Maneerat Taweesap
- Department of Radiology, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| | - Verachai Pachana
- Department of Radiology, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| | - Yada Panrak
- Department of Radiology, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| | - Naritsara Makchuchit
- Department of Radiology, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| | - Passara Jaroenpakdee
- Department of Radiology, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| |
Collapse
|
14
|
Kim J, Choi Y, Kim DH, Yoon HY, Kim K. Injectable Hydrogel-Based Combination Cancer Immunotherapy for Overcoming Localized Therapeutic Efficacy. Pharmaceutics 2022; 14:1908. [PMID: 36145656 PMCID: PMC9502377 DOI: 10.3390/pharmaceutics14091908] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/27/2022] [Accepted: 09/05/2022] [Indexed: 02/05/2023] Open
Abstract
Various immunotherapeutic agents that can elicit antitumor immune responses have recently been developed with the potential for improved efficacy in treating cancer. However, insufficient delivery efficiency at the tumor site, along with severe side effects after systemic administration of these anticancer agents, have hindered their therapeutic application in cancer immunotherapy. Hydrogels that can be directly injected into tumor sites have been developed to help modulate or elicit antitumor responses. Based on the biocompatibility, degradability, and controllable mechanochemical properties of these injectable hydrogels, various types of immunotherapeutic agents, such as hydrophobic anticancer drugs, cytokines, antigens, and adjuvants, have been easily and effectively encapsulated, resulting in the successful elicitation of antitumor immune responses and the retention of long-term immunotherapeutic efficacy following administration. This review summarizes recent advances in combination immunotherapy involving injectable hydrogel-based chemoimmunotherapy, photoimmunotherapy, and radioimmunotherapy. Finally, we briefly discuss the current limitations and future perspectives on injectable hydrogels for the effective combination immunotherapy of tumors.
Collapse
Affiliation(s)
- Jeongrae Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seonbuk-gu, Seoul 02841, Korea
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), 14 Gil 5, Hwarang-ro, Seongbuk-gu, Seoul 02792, Korea
| | - Yongwhan Choi
- Noxpharm Co. 924B, 14 Gil 5, Hwarang-ro, Seongbuk-gu, Seoul 02792, Korea
| | - Dong-Hwee Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seonbuk-gu, Seoul 02841, Korea
| | - Hong Yeol Yoon
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), 14 Gil 5, Hwarang-ro, Seongbuk-gu, Seoul 02792, Korea
| | - Kwangmeyung Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seonbuk-gu, Seoul 02841, Korea
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), 14 Gil 5, Hwarang-ro, Seongbuk-gu, Seoul 02792, Korea
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul 03760, Korea
| |
Collapse
|
15
|
Zhang M, Shi M, Yu Y, Sang J, Wang H, Shi J, Duan P, Ge R. The Immune Subtypes and Landscape of Advanced-Stage Ovarian Cancer. Vaccines (Basel) 2022; 10:vaccines10091451. [PMID: 36146529 PMCID: PMC9501495 DOI: 10.3390/vaccines10091451] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/23/2022] [Accepted: 08/29/2022] [Indexed: 12/01/2022] Open
Abstract
Immunotherapy has played a significant role in the treatment of a variety of hematological and solid tumors, but its application in ovarian cancer (OC) remains unclear. This study aimed to identify immune subtypes of OC and delineate an immune landscape for selecting suitable patients for immunotherapy, thereby providing potent therapeutic targets for immunotherapy drug development. Three immune subtypes (IS1–IS3) with distinctive molecular, cellular, and clinical characteristics were identified from the TCGA and GSE32062 cohorts. Compared to IS1, IS3 has a better prognosis and exhibits an immunological “hot”. IS3, in contrast, exhibits an immunological “cold” and has a worse prognosis in OC patients. Moreover, gene mutations, immune modulators, CA125, CA199, and HE4 expression, along with sensitivity either to immunotherapy or chemotherapy, were significantly different among the three immune subtypes. The OC immune landscape was highly heterogeneous between individual patients. Poor prognosis was correlated with low expression of the hub genes CD2, CD3D, and CD3E, which could act not only as biomarkers for predicting prognosis, but also as potential immunotherapy targets. Our study elucidates the immunotyping and molecular characteristics of the immune microenvironment in OC, which could provide an effective immunotherapy stratification method for optimally selecting patients, and also has clinical significance for the development of new immunotherapy as well as rational combination strategies for the treatment of OC patients.
Collapse
Affiliation(s)
- Minjie Zhang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Mengna Shi
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Yang Yu
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Jianmin Sang
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Hong Wang
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Jianhong Shi
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Ping Duan
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Renshan Ge
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Correspondence:
| |
Collapse
|
16
|
Cho H, Jeon SI, Ahn CH, Shim MK, Kim K. Emerging Albumin-Binding Anticancer Drugs for Tumor-Targeted Drug Delivery: Current Understandings and Clinical Translation. Pharmaceutics 2022; 14:728. [PMID: 35456562 PMCID: PMC9028280 DOI: 10.3390/pharmaceutics14040728] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/20/2022] [Accepted: 03/24/2022] [Indexed: 02/01/2023] Open
Abstract
Albumin has shown remarkable promise as a natural drug carrier by improving pharmacokinetic (PK) profiles of anticancer drugs for tumor-targeted delivery. The exogenous or endogenous albumin enhances the circulatory half-lives of anticancer drugs and passively target the tumors by the enhanced permeability and retention (EPR) effect. Thus, the albumin-based drug delivery leads to a potent antitumor efficacy in various preclinical models, and several candidates have been evaluated clinically. The most successful example is Abraxane, an exogenous human serum albumin (HSA)-bound paclitaxel formulation approved by the FDA and used to treat locally advanced or metastatic tumors. However, additional clinical translation of exogenous albumin formulations has not been approved to date because of their unexpectedly low delivery efficiency, which can increase the risk of systemic toxicity. To overcome these limitations, several prodrugs binding endogenous albumin covalently have been investigated owing to distinct advantages for a safe and more effective drug delivery. In this review, we give account of the different albumin-based drug delivery systems, from laboratory investigations to clinical applications, and their potential challenges, and the outlook for clinical translation is discussed. In addition, recent advances and progress of albumin-binding drugs to move more closely to the clinical settings are outlined.
Collapse
Affiliation(s)
- Hanhee Cho
- Department of Materials Science and Engineering, Seoul National University, Seoul 08826, Korea
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| | - Seong Ik Jeon
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| | - Cheol-Hee Ahn
- Department of Materials Science and Engineering, Seoul National University, Seoul 08826, Korea
| | - Man Kyu Shim
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| | - Kwangmeyung Kim
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea
| |
Collapse
|