1
|
Chuang CF, Lin CW, Yeh CK. Ultrasound-triggered drug release and cytotoxicity of microbubbles with diverse drug attributes. ULTRASONICS SONOCHEMISTRY 2024; 112:107182. [PMID: 39631357 DOI: 10.1016/j.ultsonch.2024.107182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 11/28/2024] [Indexed: 12/07/2024]
Abstract
Ultrasound (US)-triggered cavitation of drug-loaded microbubbles (MBs) represents a promising approach for targeted drug delivery, with substantial benefits attainable through precise control over drug release dosage and form. This study investigates Camptothecin-loaded MBs (CPT-MBs) and Doxorubicin-loaded MBs (DOX-MBs), focusing on how properties such as hydrophilicity, hydrophobicity, and charged functional groups affect their interaction with the lipid surfaces of MBs, thereby influencing the fundamental characteristics and acoustic properties of the drug-loaded MBs. In comparison to DOX-MBs, CPT-MBs showed larger MB size (2.2 ± 0.3 and 1.4 ± 0.1 μm, respectively), a 2-fold increase in drug loading, and an 18 % reduction in leakage after 2 h at 37℃. Under 1 MHz US with a 100 ms pulse repetition interval (PRI), 1000 cycles, 5-minute duration, and 550 kPa acoustic pressure, CPT-MBs undergo inertial cavitation, while DOX-MBs undergo stable cavitation. Drug particles released from these MBs under US-induced cavitation were analyzed using dynamic light scattering, NanoSight, cryo-electron microscopy, and density gradient ultracentrifugation. Results showed that CPT-MBs mainly release free CPT, while DOX-MBs release multilayered DOX-lipid aggregates. The cytotoxicity to C6 cells induced by US-triggered cavitation of these two types of MBs also differed. DOX-lipid aggregates delayed initial uptake, leading to less pronounced short-term (2 h) effects compared to the rapid release of free CPT from CPT-MBs. These findings underscore the need to optimize drug delivery strategies by fine-tuning MB composition and US parameters to control drug release kinetics and achieve the best tumoricidal outcomes.
Collapse
Affiliation(s)
- Chi-Fen Chuang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Chia-Wei Lin
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Chih-Kuang Yeh
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
2
|
Lim C, Blocher McTigue WC. Form Equals Function: Influence of Coacervate Architecture on Drug Delivery Applications. ACS Biomater Sci Eng 2024; 10:6766-6789. [PMID: 39423330 PMCID: PMC11558567 DOI: 10.1021/acsbiomaterials.4c01105] [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: 06/17/2024] [Revised: 09/26/2024] [Accepted: 10/03/2024] [Indexed: 10/21/2024]
Abstract
Complex coacervates, formed through electrostatic interactions between oppositely charged polymers, present a versatile platform for drug delivery, providing rapid assembly, selective encapsulation, and responsiveness to environmental stimuli. The architecture and properties of coacervates can be tuned by controlling structural and environmental design factors, which significantly impact the stability and delivery efficiency of the drugs. While environmental design factors such as salt, pH, and temperature play a crucial role in coacervate formation, structural design factors such as polymer concentration, polymer structure, mixing ratio, and chain length serve as the core framework that shapes coacervate architecture. These elements modulate the phase behavior and material properties of coacervates, allowing for a highly tunable system. In this review, we primarily analyze how these structural design factors contribute to the formation of diverse coacervate architecture, ranging from bulk coacervates to polyion complex micelles, vesicles, and cross-linked gels, though environmental design factors are considered. We then examine the effectiveness of these architectures in enhancing the delivery and efficacy of drugs across various administration routes, such as noninvasive (e.g., oral and transdermal) and invasive delivery. This review aims to provide foundational insights into the design of advanced drug delivery systems by examining how the origin and chemical structure of polymers influence coacervate architecture, which in turn defines their material properties. We then explore how the architecture can be tailored to optimize drug delivery for specific administration routes. This approach leverages the intrinsic properties derived from the coacervate architecture to enable targeted, controlled, and efficient drug release, ultimately enhancing therapeutic outcomes in precision medicine.
Collapse
Affiliation(s)
- Chaeyoung Lim
- Department of Chemical and Biomolecular
Engineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Whitney C. Blocher McTigue
- Department of Chemical and Biomolecular
Engineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| |
Collapse
|
3
|
Riet K, Adegoke A, Mashele S, Sekhoacha M. Effective Use of Euphorbia milii DCM Root Extract Encapsulated by Thermosensitive Immunoliposomes for Targeted Drug Delivery in Prostate Cancer Cells. Curr Issues Mol Biol 2024; 46:12037-12060. [PMID: 39590308 PMCID: PMC11593239 DOI: 10.3390/cimb46110714] [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: 07/17/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 11/28/2024] Open
Abstract
The delivery of anticancer drugs using nanotechnology is a promising approach aimed at improving the therapeutic efficacy and reducing the toxicity of chemotherapeutic agents. Liposomes were prepared using HSPC: DSPE-PEG-2000: DSPE-PEG2000-maleimide in the ratio of 4:1:0.2 and conjugated with a PSA antibody. Euphorbia milii extract (EME), doxorubicin (Dox), and docetaxel (Doc) encapsulated in temperature-sensitive immunoliposomes were investigated for their activities against the prostate cancer LNCap and DU145 cell lines. Organic extracts of EME leaves, roots, and stems were screened against both cell lines, inhibiting more than 50% of cell culture at concentrations of 10 μg/mL. The immunoliposomes incorporating the EME and docetaxel were active against the LNCap cells when exposed to heat at 39-40 °C. The liposomes not exposed to heat were inactive against the LNCap cells. The developed heat-sensitive immunoliposomes used for the delivery of both the EME and chemotherapeutic agents was able to successfully release the entrapped contents upon heat exposure above the phase transition temperature of the liposome membrane. The heat-sensitive immunoliposomes conjugated with a PSA antibody encapsulated the extract successfully and showed better cell antiproliferation efficacy against the prostate cancer cell lines in the presence of heat.
Collapse
Affiliation(s)
- Keamogetswe Riet
- Department of Health Sciences, Central University of Technology, Bloemfontein 9300, South Africa; (K.R.); (S.M.)
| | - Ayodeji Adegoke
- Department of Pharmacology, University of the Free State, Bloemfontein 9300, South Africa;
- Cancer Research and Molecular Biology Laboratories, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan 200005, Nigeria
| | - Samson Mashele
- Department of Health Sciences, Central University of Technology, Bloemfontein 9300, South Africa; (K.R.); (S.M.)
| | - Mamello Sekhoacha
- Department of Pharmacology, University of the Free State, Bloemfontein 9300, South Africa;
| |
Collapse
|
4
|
Guo S, Chen M, Yang Y, Zhang Y, Pang X, Shi Y, Zhuang Y, Fan D, Bao J, Ji Z. Magnetic-vortex nanodonuts enhance ferroptosis effect of tumor ablation through an imaging-guided hyperthermia/radiosensitization strategy. iScience 2024; 27:110533. [PMID: 39398248 PMCID: PMC11467572 DOI: 10.1016/j.isci.2024.110533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 06/06/2024] [Accepted: 07/15/2024] [Indexed: 10/15/2024] Open
Abstract
Pursuing more efficient multifunctional treatment is the main challenge of preclinical nanoparticle-mediated theranostics research. Here, nanoscale magnetite vortex donut shape was synthesized as a platform, and then ultrasmall gold nanoparticles were successfully embedded into the nanoring surface, thereby obtaining gold-modified magnetic vortex donut (GMVD). GMVD has a high photothermal conversion efficiency (η = 42.2%), which makes it have excellent photothermal ablation effect on tumors both in vivo and in vitro. Simultaneously, GMVD forms reactive oxygen species (ROS) under the 808 nm laser triggering, inducing ferroptosis. The addition of gold element also makes GMVD have the effect of radiotherapy (RT) sensitization. In conclusion, the synergistic treatment of RT and PTT greatly enhanced tumor ablation, indicating that GMVD has good biocompatibility and antitumor efficacy. This work demonstrates that the proposed GMVD can be a high-performance tumor diagnosis and theranostic treatment agent and may have great potential for clinical application in the future.
Collapse
Affiliation(s)
- S.S. Guo
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450000, China
| | - M.M. Chen
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450000, China
| | - Y.H. Yang
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450000, China
| | - Y.Y. Zhang
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450000, China
| | - X. Pang
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450000, China
| | - Y.P. Shi
- Functional Magnetic Resonance and Molecular Imaging Key Laboratory of Henan Province, Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450000, China
| | - Y.C. Zhuang
- Department of Imaging Sciences, University of Rochester Medical Center, Rochester, NY, USA
| | - D.D. Fan
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450000, China
| | - J.F. Bao
- Functional Magnetic Resonance and Molecular Imaging Key Laboratory of Henan Province, Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450000, China
| | - Z.Y. Ji
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450000, China
| |
Collapse
|
5
|
Monck C, Elani Y, Ceroni F. Genetically programmed synthetic cells for thermo-responsive protein synthesis and cargo release. Nat Chem Biol 2024; 20:1380-1386. [PMID: 38969863 PMCID: PMC11427347 DOI: 10.1038/s41589-024-01673-7] [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] [Accepted: 06/06/2024] [Indexed: 07/07/2024]
Abstract
Synthetic cells containing genetic programs and protein expression machinery are increasingly recognized as powerful counterparts to engineered living cells in the context of biotechnology, therapeutics and cellular modelling. So far, genetic regulation of synthetic cell activity has been largely confined to chemical stimuli; to unlock their potential in applied settings, engineering stimuli-responsive synthetic cells under genetic regulation is imperative. Here we report the development of temperature-sensitive synthetic cells that control protein production by exploiting heat-responsive mRNA elements. This is achieved by combining RNA thermometer technology, cell-free protein expression and vesicle-based synthetic cell design to create cell-sized capsules able to initiate synthesis of both soluble proteins and membrane proteins at defined temperatures. We show that the latter allows for temperature-controlled cargo release phenomena with potential implications for biomedicine. Platforms like the one presented here can pave the way for customizable, genetically programmed synthetic cells under thermal control to be used in biotechnology.
Collapse
Affiliation(s)
- Carolina Monck
- Department of Chemical Engineering, Imperial College London, London, UK
- Imperial College Centre for Synthetic Biology, London, UK
- fabriCELL, Imperial College London, London, UK
| | - Yuval Elani
- Department of Chemical Engineering, Imperial College London, London, UK.
- Imperial College Centre for Synthetic Biology, London, UK.
- fabriCELL, Imperial College London, London, UK.
| | - Francesca Ceroni
- Department of Chemical Engineering, Imperial College London, London, UK.
- Imperial College Centre for Synthetic Biology, London, UK.
| |
Collapse
|
6
|
Xu R, Martinez-Bosch N, Rivera-Hueto F, Mulens-Arias V, Rubio-Moscardo F, Javier Conesa J, Navarro P, Vicente R, Rivera-Gil P. Validation of ZIP4 as a tumour-associated antigen for nanotargeting. J Drug Target 2024:1-13. [PMID: 39283041 DOI: 10.1080/1061186x.2024.2405711] [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: 05/01/2024] [Revised: 09/06/2024] [Accepted: 09/12/2024] [Indexed: 10/02/2024]
Abstract
Pancreatic ductal adenocarcinoma remains a highly aggressive and untreatable cancer. There is a need to develop a new PDAC-associated antigen-targeting drug delivery system to tackle this disease. We validated choosing ZIP4 as a putative target in PDAC theranostics. We developed a nanosystem composed of a fluorescent polystyrene core coated with gold nanoparticles onto which a ZIP4-specific polyclonal antibody is attached. The polystyrene core's fluorescence properties allow the nanosystem tracking by intravital imaging. We also developed two ZIP4-expressing cell lines by stably transfecting HEK293 and RWP1 cells with a ZIP4-coding plasmid that simultaneously provides cells with puromycin resistance. We studied the cell internalisation of the as-synthesised nanoparticles and demonstrated that ZIP4-expressing HEK293 and ZIP4-expressing RWP1 cells tended to take up more ZIP4-targeting nanoparticles. Moreover, we observed that ZIP4-targeting nanoparticles accumulated more in ZIP4-expressing HEK293 and RWP1 tumours when injected intravenously in a subcutaneous xenograft and an orthotopic in vivo model, respectively. Furthermore, the administration of these nanoparticles did not induce any significant systemic toxicity as determined by histological analysis of all organs. Altogether, these results provide the first evidence of the feasibility of using a ZIP4-targeting nanosystem further to design efficient therapeutic and diagnostic tools for PDAC.
Collapse
Affiliation(s)
- Ruixue Xu
- Integrative Biomedical Materials and Nanomedicine Lab, Department of Medicine and Life Sciences (MELIS), Universitat Pompeu Fabra, PRBB, Barcelona, Spain
| | - Neus Martinez-Bosch
- Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM), Unidad Asociada IIBB-CSIC, Barcelona, Spain
| | | | - Vladimir Mulens-Arias
- Integrative Biomedical Materials and Nanomedicine Lab, Department of Medicine and Life Sciences (MELIS), Universitat Pompeu Fabra, PRBB, Barcelona, Spain
| | - Fanny Rubio-Moscardo
- Molecular Physiology Lab, Department of Medicine and Life Sciences (MELIS), Universitat Pompeu Fabra, PRBB, Barcelona, Spain
| | - J Javier Conesa
- Mistral Beamline, Experiment Division, ALBA Synchrotron (ALBA-CELLS), Barcelona, Spain
| | - Pilar Navarro
- Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM), Unidad Asociada IIBB-CSIC, Barcelona, Spain
- Institute of Biomedical Research of Barcelona (IIBB)-CSIC, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Rubén Vicente
- Molecular Physiology Lab, Department of Medicine and Life Sciences (MELIS), Universitat Pompeu Fabra, PRBB, Barcelona, Spain
| | - Pilar Rivera-Gil
- Integrative Biomedical Materials and Nanomedicine Lab, Department of Medicine and Life Sciences (MELIS), Universitat Pompeu Fabra, PRBB, Barcelona, Spain
| |
Collapse
|
7
|
Cheng Z, Fobian SF, Gurrieri E, Amin M, D'Agostino VG, Falahati M, Zalba S, Debets R, Garrido MJ, Saeed M, Seynhaeve ALB, Balcioglu HE, Ten Hagen TLM. Lipid-based nanosystems: the next generation of cancer immune therapy. J Hematol Oncol 2024; 17:53. [PMID: 39030582 PMCID: PMC11265205 DOI: 10.1186/s13045-024-01574-1] [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: 05/23/2024] [Accepted: 07/11/2024] [Indexed: 07/21/2024] Open
Abstract
Immunotherapy has become an important part of the oncotherapy arsenal. Its applicability in various cancer types is impressive, as well as its use of endogenous mechanisms to achieve desired ends. However, off-target or on-target-off-tumor toxicity, limited activity, lack of control in combination treatments and, especially for solid tumors, low local accumulation, have collectively limited clinical use thereof. These limitations are partially alleviated by delivery systems. Lipid-based nanoparticles (NPs) have emerged as revolutionary carriers due to favorable physicochemical characteristics, with specific applications and strengths particularly useful in immunotherapeutic agent delivery. The aim of this review is to highlight the challenges faced by immunotherapy and how lipid-based NPs have been, and may be further utilized to address such challenges. We discuss recent fundamental and clinical applications of NPs in a range of areas and provide a detailed discussion of the main obstacles in immune checkpoint inhibition therapies, adoptive cellular therapies, and cytokine therapies. We highlight how lipid-based nanosystems could address these through either delivery, direct modulation of the immune system, or targeting of the immunosuppressive tumor microenvironment. We explore advanced and emerging liposomal and lipid nanoparticle (LNP) systems for nucleic acid delivery, intrinsic and extrinsic stimulus-responsive formulations, and biomimetic lipid-based nanosystems in immunotherapy. Finally, we discuss the key challenges relating to the clinical use of lipid-based NP immunotherapies, suggesting future research directions for the near term to realize the potential of these innovative lipid-based nanosystems, as they become the crucial steppingstone towards the necessary enhancement of the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Ziyun Cheng
- Precision Medicine in Oncology (PrMiO), Department of Pathology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands
- Nanomedicine Innovation Center Erasmus (NICE), Erasmus Medical Center, Rotterdam, The Netherlands
| | - Seth-Frerich Fobian
- Precision Medicine in Oncology (PrMiO), Department of Pathology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands
- Nanomedicine Innovation Center Erasmus (NICE), Erasmus Medical Center, Rotterdam, The Netherlands
| | - Elena Gurrieri
- Laboratory of Biotechnology and Nanomedicine, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Mohamadreza Amin
- Precision Medicine in Oncology (PrMiO), Department of Pathology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands
- Nanomedicine Innovation Center Erasmus (NICE), Erasmus Medical Center, Rotterdam, The Netherlands
| | - Vito Giuseppe D'Agostino
- Laboratory of Biotechnology and Nanomedicine, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Mojtaba Falahati
- Precision Medicine in Oncology (PrMiO), Department of Pathology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Sara Zalba
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Navarra Institute for Health Research, Pamplona, Spain
| | - Reno Debets
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands
| | - María J Garrido
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Navarra Institute for Health Research, Pamplona, Spain
| | - Mesha Saeed
- Precision Medicine in Oncology (PrMiO), Department of Pathology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ann L B Seynhaeve
- Precision Medicine in Oncology (PrMiO), Department of Pathology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Hayri E Balcioglu
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands.
| | - Timo L M Ten Hagen
- Precision Medicine in Oncology (PrMiO), Department of Pathology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands.
- Nanomedicine Innovation Center Erasmus (NICE), Erasmus Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
8
|
Naser IH, Zaid M, Ali E, Jabar HI, Mustafa AN, Alubiady MHS, Ramadan MF, Muzammil K, Khalaf RM, Jalal SS, Alawadi AH, Alsalamy A. Unveiling innovative therapeutic strategies and future trajectories on stimuli-responsive drug delivery systems for targeted treatment of breast carcinoma. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:3747-3770. [PMID: 38095649 DOI: 10.1007/s00210-023-02885-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 12/02/2023] [Indexed: 05/23/2024]
Abstract
This comprehensive review delineates the latest advancements in stimuli-responsive drug delivery systems engineered for the targeted treatment of breast carcinoma. The manuscript commences by introducing mammary carcinoma and the current therapeutic methodologies, underscoring the urgency for innovative therapeutic strategies. Subsequently, it elucidates the logic behind the employment of stimuli-responsive drug delivery systems, which promise targeted drug administration and the minimization of adverse reactions. The review proffers an in-depth analysis of diverse types of stimuli-responsive systems, including thermoresponsive, pH-responsive, and enzyme-responsive nanocarriers. The paramount importance of material choice, biocompatibility, and drug loading strategies in the design of these systems is accentuated. The review explores characterization methodologies for stimuli-responsive nanocarriers and probes preclinical evaluations of their efficacy, toxicity, pharmacokinetics, and biodistribution in mammary carcinoma models. Clinical applications of stimuli-responsive systems, ongoing clinical trials, the potential of combination therapies, and the utility of multifunctional nanocarriers for the co-delivery of assorted drugs and therapies are also discussed. The manuscript addresses the persistent challenge of drug resistance in mammary carcinoma and the potential of stimuli-responsive systems in surmounting it. Regulatory and safety considerations, including FDA guidelines and biocompatibility assessments, are outlined. The review concludes by spotlighting future trajectories and emergent technologies in stimuli-responsive drug delivery, focusing on pioneering approaches, advancements in nanotechnology, and personalized medicine considerations. This review aims to serve as a valuable compendium for researchers and clinicians interested in the development of efficacious and safe stimuli-responsive drug delivery systems for the treatment of breast carcinoma.
Collapse
Affiliation(s)
- Israa Habeeb Naser
- Medical Laboratories Techniques Department, AL-Mustaqbal University, Hillah, Babil, Iraq
| | - Muhaned Zaid
- Department of Pharmacy, Al-Manara College for Medical Sciences, Maysan, Amarah, Iraq
| | - Eyhab Ali
- Al-Zahraa University for Women, Karbala, Iraq
| | - Hayder Imad Jabar
- Department of Pharmaceutics, College of Pharmacy, University of Al-Ameed, Karbala, Iraq
| | | | | | | | - Khursheed Muzammil
- Department of Public Health, College of Applied Medical Sciences, Khamis Mushait Campus, King Khalid University, Abha, Saudi Arabia
| | | | - Sarah Salah Jalal
- College of Pharmacy, National University of Science and Technology, Dhi Qar, Iraq
| | - Ahmed Hussien Alawadi
- College of Technical Engineering, the Islamic University, Najaf, Iraq
- College of Technical Engineering, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- College of Technical Engineering, the Islamic University of Babylon, Babylon, Iraq
| | - Ali Alsalamy
- College of Technical Engineering, Imam Ja'afar Al-Sadiq University, Al-Muthanna, Iraq.
| |
Collapse
|
9
|
Anup N, Gadeval A, Ramdas Mule S, Gupta T, Kumar Tekade R. Plasmonic laser-responsive BioDissolve 3D-printed graphene@cisplatin-implant for prevention of post-surgical relapse of oral cancer. Int J Pharm 2024; 657:124123. [PMID: 38621618 DOI: 10.1016/j.ijpharm.2024.124123] [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: 12/17/2023] [Revised: 04/11/2024] [Accepted: 04/12/2024] [Indexed: 04/17/2024]
Abstract
The development of chemoresistance is a major obstacle in post-surgical adjuvant therapy of cancer, leading to cancer cell survival, recurrence, and metastasis. This study reports a 3D-printed plasmonic implant developed for the post-surgical adjuvant therapy of cisplatin-resistant cancer cells to prevent relapse. The implant was printed using optimized biomaterial ink containing biodegradable polymers [poly(L-lactide) and hydroxypropyl methylcellulose] blended suitably with laser-responsive graphene and chemo drug (Cisplatin). The irradiation of scar-driven 3D-printed implant with a laser stimulates graphene to generate a series of hyperthermia events leading to photothermolysis of cisplatin-resistant cancer cells under the combined influence of sustained cisplatin release. The developed personalized implant offers pH-responsive sustained drug release for 28 days. The implant exhibited acceptable biophysical properties (Tensile strength: 3.99 ± 0.15 MPa; modulus: 81 ± 9.58 MPa; thickness: 110 μm). The 3D-printed implant effectively reverses the chemoresistance in cisplatin-resistant 3D spheroid tumor models. Cytotoxicity assay performed using cisplatin-resistant (CisR) cell line revealed that the cell viability was reduced to 39.80 ± 0.68 % from 61.37 ± 0.98 % in CisR tumor spheroids on combined chemo-photothermal therapy. The combination therapy reduced the IC50 value from 71.05 μM to 48.73 μM in CisR spheroids. Apoptosis assay revealed an increase in the population of apoptotic cells (35.45 ± 1.56 % →52.53 ± 2.30 %) on combination therapy. A similar trend was observed in gene expression analysis, where the expression of pro-apoptotic genes Caspase 3 (3.73 ± 0.04 fold) and Bcl-2-associated X protein (BAX) (3.35 ± 0.02 fold) was increased on combination therapy. This 3D-printed, biodegradable implant with chemo-combined thermal ablating potential may provide a promising approach for the adjuvant treatment of resistant cancer.
Collapse
Affiliation(s)
- Neelima Anup
- National Institute of Pharmaceutical Education and Research (NIPER) Ahmedabad, An Institute of National Importance, Government of India, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Palaj, Opp. Air Force Station, Gandhinagar 382355, Gujarat, India
| | - Anuradha Gadeval
- National Institute of Pharmaceutical Education and Research (NIPER) Ahmedabad, An Institute of National Importance, Government of India, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Palaj, Opp. Air Force Station, Gandhinagar 382355, Gujarat, India
| | - Shubham Ramdas Mule
- National Institute of Pharmaceutical Education and Research (NIPER) Ahmedabad, An Institute of National Importance, Government of India, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Palaj, Opp. Air Force Station, Gandhinagar 382355, Gujarat, India
| | - Tanisha Gupta
- National Institute of Pharmaceutical Education and Research (NIPER) Ahmedabad, An Institute of National Importance, Government of India, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Palaj, Opp. Air Force Station, Gandhinagar 382355, Gujarat, India
| | - Rakesh Kumar Tekade
- National Institute of Pharmaceutical Education and Research (NIPER) Ahmedabad, An Institute of National Importance, Government of India, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Palaj, Opp. Air Force Station, Gandhinagar 382355, Gujarat, India.
| |
Collapse
|
10
|
Li L, Bo W, Wang G, Juan X, Xue H, Zhang H. Progress and application of lung-on-a-chip for lung cancer. Front Bioeng Biotechnol 2024; 12:1378299. [PMID: 38854856 PMCID: PMC11157020 DOI: 10.3389/fbioe.2024.1378299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 05/08/2024] [Indexed: 06/11/2024] Open
Abstract
Lung cancer is a malignant tumour with the highest incidence and mortality worldwide. Clinically effective therapy strategies are underutilized owing to the lack of efficient models for evaluating drug response. One of the main reasons for failure of anticancer drug therapy is development of drug resistance. Anticancer drugs face severe challenges such as poor biodistribution, restricted solubility, inadequate absorption, and drug accumulation. In recent years, "organ-on-a-chip" platforms, which can directly regulate the microenvironment of biomechanics, biochemistry and pathophysiology, have been developed rapidly and have shown great potential in clinical drug research. Lung-on-a-chip (LOC) is a new 3D model of bionic lungs with physiological functions created by micromachining technology on microfluidic chips. This approach may be able to partially replace animal and 2D cell culture models. To overcome drug resistance, LOC realizes personalized prediction of drug response by simulating the lung-related microenvironment in vitro, significantly enhancing therapeutic effectiveness, bioavailability, and pharmacokinetics while minimizing side effects. In this review, we present an overview of recent advances in the preparation of LOC and contrast it with earlier in vitro models. Finally, we describe recent advances in LOC. The combination of this technology with nanomedicine will provide an accurate and reliable treatment for preclinical evaluation.
Collapse
Affiliation(s)
- Lantao Li
- Department of Anesthesiology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Wentao Bo
- Department of Hepatopancreatobiliary Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Guangyan Wang
- Department of General Internal Medicine, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Xin Juan
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Haiyi Xue
- Department of Intensive Care Unit, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Hongwei Zhang
- Department of Anesthesiology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
11
|
Li ZZ, Zhong NN, Cao LM, Cai ZM, Xiao Y, Wang GR, Liu B, Xu C, Bu LL. Nanoparticles Targeting Lymph Nodes for Cancer Immunotherapy: Strategies and Influencing Factors. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2308731. [PMID: 38327169 DOI: 10.1002/smll.202308731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 01/07/2024] [Indexed: 02/09/2024]
Abstract
Immunotherapy has emerged as a potent strategy in cancer treatment, with many approved drugs and modalities in the development stages. Despite its promise, immunotherapy is not without its limitations, including side effects and suboptimal efficacy. Using nanoparticles (NPs) as delivery vehicles to target immunotherapy to lymph nodes (LNs) can improve the efficacy of immunotherapy drugs and reduce side effects in patients. In this context, this paper reviews the development of LN-targeted immunotherapeutic NP strategies, the mechanisms of NP transport during LN targeting, and their related biosafety risks. NP targeting of LNs involves either passive targeting, influenced by NP physical properties, or active targeting, facilitated by affinity ligands on NP surfaces, while alternative methods, such as intranodal injection and high endothelial venule (HEV) targeting, have uncertain clinical applicability and require further research and validation. LN targeting of NPs for immunotherapy can reduce side effects and increase biocompatibility, but risks such as toxicity, organ accumulation, and oxidative stress remain, although strategies such as biodegradable biomacromolecules, polyethylene glycol (PEG) coating, and impurity addition can mitigate these risks. Additionally, this work concludes with a future-oriented discussion, offering critical insights into the field.
Collapse
Affiliation(s)
- Zi-Zhan Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, #237 Luoyu Road, Wuhan, 430079, China
| | - Nian-Nian Zhong
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, #237 Luoyu Road, Wuhan, 430079, China
| | - Lei-Ming Cao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, #237 Luoyu Road, Wuhan, 430079, China
| | - Ze-Min Cai
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, #237 Luoyu Road, Wuhan, 430079, China
| | - Yao Xiao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, #237 Luoyu Road, Wuhan, 430079, China
| | - Guang-Rui Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, #237 Luoyu Road, Wuhan, 430079, China
| | - Bing Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, Department of Oral & Maxillofacial - Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, #237 Luoyu Road, Wuhan, 430079, China
| | - Chun Xu
- School of Dentistry, The University of Queensland, 288 Herston Road, Brisbane, 4066, Australia
| | - Lin-Lin Bu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, Department of Oral & Maxillofacial - Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, #237 Luoyu Road, Wuhan, 430079, China
| |
Collapse
|
12
|
Opriș O, Mormile C, Lung I, Stegarescu A, Soran ML, Soran A. An Overview of Biopolymers for Drug Delivery Applications. APPLIED SCIENCES 2024; 14:1383. [DOI: 10.3390/app14041383] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Nowadays, drug delivery has an important role in medical therapy. The use of biopolymers in developing drug delivery systems (DDSs) is increasingly attracting attention due to their remarkable and numerous advantages, in contrast to conventional polymers. Biopolymers have many advantages (biodegradability, biocompatibility, renewability, affordability, and availability), which are extremely important for developing materials with applications in the biomedical field. Additionally, biopolymers are appropriate when they improve functioning and have a number of positive effects on human life. Therefore, this review presents the most used biopolymers for biomedical applications, especially in drug delivery. In addition, by combining different biopolymers DDSs with tailored functional properties (e.g., physical properties, biodegradability) can be developed. This review summarizes and provides data on the progress of research on biopolymers (chitosan, alginate, starch, cellulose, albumin, silk fibroin, collagen, and gelatin) used in DDSs, their preparation, and mechanism of action.
Collapse
Affiliation(s)
- Ocsana Opriș
- National Institute for Research and Development of Isotopic and Molecular Technologies, 67-103 Donat, 400293 Cluj-Napoca, Romania
| | - Cristina Mormile
- National Institute for Research and Development of Isotopic and Molecular Technologies, 67-103 Donat, 400293 Cluj-Napoca, Romania
- Faculty of Chemistry, University of Rome La Sapienza, P. le Aldo Moro 5, 00185 Rome, Italy
- INFN—National Laboratories of Frascati, Via Enrico Fermi 54, 00044 Frascati, Italy
| | - Ildiko Lung
- National Institute for Research and Development of Isotopic and Molecular Technologies, 67-103 Donat, 400293 Cluj-Napoca, Romania
| | - Adina Stegarescu
- National Institute for Research and Development of Isotopic and Molecular Technologies, 67-103 Donat, 400293 Cluj-Napoca, Romania
| | - Maria-Loredana Soran
- National Institute for Research and Development of Isotopic and Molecular Technologies, 67-103 Donat, 400293 Cluj-Napoca, Romania
| | - Albert Soran
- Department of Chemistry, Supramolecular Organic and Organometallic Chemistry Centre (SOOMCC), Faculty of Chemistry and Chemical Engineering, Babes-Bolyai University, 11 Arany Janos, 400028 Cluj-Napoca, Romania
| |
Collapse
|
13
|
Ali M, Benfante V, Di Raimondo D, Salvaggio G, Tuttolomondo A, Comelli A. Recent Developments in Nanoparticle Formulations for Resveratrol Encapsulation as an Anticancer Agent. Pharmaceuticals (Basel) 2024; 17:126. [PMID: 38256959 PMCID: PMC10818631 DOI: 10.3390/ph17010126] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/13/2024] [Accepted: 01/15/2024] [Indexed: 01/24/2024] Open
Abstract
Resveratrol is a polyphenolic compound that has gained considerable attention in the past decade due to its multifaceted therapeutic potential, including anti-inflammatory and anticancer properties. However, its anticancer efficacy is impeded by low water solubility, dose-limiting toxicity, low bioavailability, and rapid hepatic metabolism. To overcome these hurdles, various nanoparticles such as organic and inorganic nanoparticles, liposomes, polymeric nanoparticles, dendrimers, solid lipid nanoparticles, gold nanoparticles, zinc oxide nanoparticles, zeolitic imidazolate frameworks, carbon nanotubes, bioactive glass nanoparticles, and mesoporous nanoparticles were employed to deliver resveratrol, enhancing its water solubility, bioavailability, and efficacy against various types of cancer. Resveratrol-loaded nanoparticle or resveratrol-conjugated nanoparticle administration exhibits excellent anticancer potency compared to free resveratrol. This review highlights the latest developments in nanoparticle-based delivery systems for resveratrol, focusing on the potential to overcome limitations associated with the compound's bioavailability and therapeutic effectiveness.
Collapse
Affiliation(s)
- Muhammad Ali
- Ri.MED Foundation, Via Bandiera 11, 90133 Palermo, Italy;
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Molecular and Clinical Medicine, University of Palermo, 90127 Palermo, Italy; (D.D.R.); (A.T.)
| | - Viviana Benfante
- Ri.MED Foundation, Via Bandiera 11, 90133 Palermo, Italy;
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Molecular and Clinical Medicine, University of Palermo, 90127 Palermo, Italy; (D.D.R.); (A.T.)
| | - Domenico Di Raimondo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Molecular and Clinical Medicine, University of Palermo, 90127 Palermo, Italy; (D.D.R.); (A.T.)
| | - Giuseppe Salvaggio
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy;
| | - Antonino Tuttolomondo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Molecular and Clinical Medicine, University of Palermo, 90127 Palermo, Italy; (D.D.R.); (A.T.)
| | - Albert Comelli
- Ri.MED Foundation, Via Bandiera 11, 90133 Palermo, Italy;
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
| |
Collapse
|
14
|
Navaratnarajah A, Daniel C, Bhakta S. Modified HT-SPOTi: An Antimicrobial Susceptibility Testing to Evaluate Formulated Therapeutic Combinations Against Bacterial Growth and Viability. Methods Mol Biol 2024; 2833:35-42. [PMID: 38949698 DOI: 10.1007/978-1-0716-3981-8_4] [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: 07/02/2024]
Abstract
Antimicrobial resistance (AMR) poses a serious threat to global health, potentially causing 10 million deaths per year globally by 2050. To tackle AMR, researchers from all around the world have generated a selection of various formulated (viz. nanoparticulate, liposomal) therapeutic combinations to be evaluated for new antimicrobial drug discovery. To meet the urgent need for accelerating new antibacterial drug development, we need rapid but reliable whole-cell assay methods and models to test formulated therapeutic combinations against several pathogens in different in vitro conditions as models of actual infections.Over the past two decades, high-throughput spot-culture growth inhibition assay (HT-SPOTi) has been demonstrated to be a gold-standard drug susceptibility method for evaluating novel chemotherapeutic entities and existing drugs against various microbes of global concern. Our modified HT-SPOTi method serves the purpose of evaluating drug combinations against Gram-positive/negative microorganisms as well as acid-fast bacilli. The newly developed and modified HT-SPOTi assay builds upon the limitations of our previously published method to incorporate antimicrobial susceptibility testing with formulated therapeutic combinations. The modified HT-SPOTi is compared with a range of other antimicrobial susceptibility testing methods and validated using a library of existing antibiotics as well as formulated therapeutic combinations. The modified HT-SPOTi assay can serve as an efficient and reliable high-throughput drug screening platform to discover new potential antimicrobial molecules, including as part of therapeutic formulations.This chapter describes the generation of drug susceptibility profile for formulated therapeutic combinations using modified HT-SPOTi in a semi-automated system.
Collapse
Affiliation(s)
- Anushandan Navaratnarajah
- Mycobacteria Research Laboratory, Institute of Structural and Molecular Biology, School of Natural Sciences, Birkbeck, University of London, London, UK
| | - Chris Daniel
- Mycobacteria Research Laboratory, Institute of Structural and Molecular Biology, School of Natural Sciences, Birkbeck, University of London, London, UK
| | - Sanjib Bhakta
- Mycobacteria Research Laboratory, Institute of Structural and Molecular Biology, School of Natural Sciences, Birkbeck, University of London, London, UK.
| |
Collapse
|
15
|
Mahajan K, Bhattacharya S. The Advancement and Obstacles in Improving the Stability of Nanocarriers for Precision Drug Delivery in the Field of Nanomedicine. Curr Top Med Chem 2024; 24:686-721. [PMID: 38409730 DOI: 10.2174/0115680266287101240214071718] [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: 10/13/2023] [Revised: 01/19/2024] [Accepted: 01/23/2024] [Indexed: 02/28/2024]
Abstract
Nanocarriers have emerged as a promising class of nanoscale materials in the fields of drug delivery and biomedical applications. Their unique properties, such as high surface area- tovolume ratios and enhanced permeability and retention effects, enable targeted delivery of therapeutic agents to specific tissues or cells. However, the inherent instability of nanocarriers poses significant challenges to their successful application. This review highlights the importance of nanocarrier stability in biomedical applications and its impact on biocompatibility, targeted drug delivery, long shelf life, drug delivery performance, therapeutic efficacy, reduced side effects, prolonged circulation time, and targeted delivery. Enhancing nanocarrier stability requires careful design, engineering, and optimization of physical and chemical parameters. Various strategies and cutting-edge techniques employed to improve nanocarrier stability are explored, with a focus on their applications in drug delivery. By understanding the advances and challenges in nanocarrier stability, this review aims to contribute to the development and implementation of nanocarrier- based therapies in clinical settings, advancing the field of nanomedicine.
Collapse
Affiliation(s)
- Kalpesh Mahajan
- Department of Quality Assurence, Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai, India
| | - Sankha Bhattacharya
- Department of Pharmaceutics, School of Pharmacy and Technology Management, SVKMS NMIMS Maharashtra, Shirpur, 425405, India
| |
Collapse
|
16
|
Yuan J, Ding L, Han L, Pang L, Zhang P, Yang X, Liu H, Zheng M, Zhang Y, Luo W. Thermal/ultrasound-triggered release of liposomes loaded with Ganoderma applanatum polysaccharide from microbubbles for enhanced tumour ablation. J Control Release 2023; 363:84-100. [PMID: 37730090 DOI: 10.1016/j.jconrel.2023.09.030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 09/16/2023] [Accepted: 09/17/2023] [Indexed: 09/22/2023]
Abstract
The effectiveness of thermal ablation for the treatment of liver tumours is limited by the risk of incomplete ablation, which can result in residual tumours. Herein, an enhancement strategy is proposed based on the controlled release of Ganoderma applanatum polysaccharide (GAP) liposome-microbubble complexes (GLMCs) via ultrasound (US)-targeted microbubble destruction (UTMD) and sublethal hyperthermic (SH) field. GLMCs were prepared by conjugating GAP liposomes onto the surface of microbubbles via biotin-avidin linkage. In vitro, UTMD promotes the cellular uptake of liposomes and leads to apoptosis of M2-like macrophages. Secretion of arginase-1 (Arg-1) and transforming growth factor-beta (TGF-β) by M2-like macrophages decreased. In vivo, restriction of tumour volume was observed in rabbit VX2 liver tumours after treatment with GLMCs via UTMD in GLMCs + SH + US group. The expression levels of CD68 and CD163, as markers of tumour-associated macrophages (TAMs) in the GLMCs + SH + US group were reduced in liver tumour tissue. Decreased Arg-1, TGF-β, Ki67, and CD31 factors related to tumour cell proliferation and angiogenesis was evident on histological analysis. In conclusion, thermal/US-triggered drug release from GLMCs suppressed rabbit VX2 liver tumour growth in the SH field by inhibiting TAMs, which represents a potential approach to improve the effectiveness of thermal ablation.
Collapse
Affiliation(s)
- Jiani Yuan
- Department of Ultrasound, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Lei Ding
- Department of Ultrasound, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Lu Han
- Department of Ultrasound, Xi'an Central Hospital, Xi'an, China
| | - Lina Pang
- Department of Ultrasound, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Peidi Zhang
- Department of Ultrasound, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xiao Yang
- Department of Ultrasound, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Haijing Liu
- Department of Ultrasound, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Minjuan Zheng
- Department of Ultrasound, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| | - Yunfei Zhang
- Department of Orthopaedics, Second Affiliated Hospital, Fourth Military Medical University, Xi'an, China.
| | - Wen Luo
- Department of Ultrasound, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
17
|
Sinenko IL, Kuttler F, Simeonov V, Moulin A, Aouad P, Stathopoulos C, Munier FL, Berger A, Dyson PJ. Translational screening platform to evaluate chemotherapy in combination with focal therapy for retinoblastoma. Cancer Sci 2023; 114:3728-3739. [PMID: 37340597 PMCID: PMC10475790 DOI: 10.1111/cas.15878] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 05/15/2023] [Accepted: 05/20/2023] [Indexed: 06/22/2023] Open
Abstract
Retinoblastoma is the most common pediatric eye cancer. It is currently treated with a limited number of drugs, adapted from other pediatric cancer treatments. Drug toxicity and relapse of the disease warrant new therapeutic strategies for these young patients. In this study, we developed a robust tumoroid-based platform to test chemotherapeutic agents in combination with focal therapy (thermotherapy) - a treatment option widely used in clinical practice - in accordance with clinically relevant trial protocols. The model consists of matrix-embedded tumoroids that retain retinoblastoma features and respond to repeated chemotherapeutic drug exposure similarly to advanced clinical cases. Moreover, the screening platform includes a diode laser (810 nm, 0.3 W) to selectively heat the tumoroids, combined with an on-line system to monitor the intratumoral and surrounding temperatures. This allows the reproduction of the clinical settings of thermotherapy and combined chemothermotherapy treatments. When testing the two main drugs currently used in clinics to treat retinoblastoma in our model, we observed results similar to those clinically obtained, validating the utility of the model. This screening platform is the first system to accurately reproduce clinically relevant treatment methods and should lead to the identification of more efficient drugs to treat retinoblastoma.
Collapse
Affiliation(s)
- Irina L. Sinenko
- Institute of Chemical Sciences and EngineeringÉcole Polytechnique Fédérale de Lausanne (EPFL)LausanneSwitzerland
- Ophthalmology DepartmentUniversity of Lausanne, Jules‐Gonin Eye Hospital, Fondation Asile des AveuglesLausanneSwitzerland
| | - Fabien Kuttler
- Biomolecular Screening Facility, School of Life SciencesÉcole Polytechnique Fédérale de Lausanne (EPFL)LausanneSwitzerland
| | - Valentin Simeonov
- Laboratory of Environmental Remote SensingÉcole Polytechnique Fédérale de Lausanne (EPFL)LausanneSwitzerland
| | - Alexandre Moulin
- Ophthalmology DepartmentUniversity of Lausanne, Jules‐Gonin Eye Hospital, Fondation Asile des AveuglesLausanneSwitzerland
| | - Patrick Aouad
- Ophthalmology DepartmentUniversity of Lausanne, Jules‐Gonin Eye Hospital, Fondation Asile des AveuglesLausanneSwitzerland
| | - Christina Stathopoulos
- Ophthalmology DepartmentUniversity of Lausanne, Jules‐Gonin Eye Hospital, Fondation Asile des AveuglesLausanneSwitzerland
| | - Francis L. Munier
- Ophthalmology DepartmentUniversity of Lausanne, Jules‐Gonin Eye Hospital, Fondation Asile des AveuglesLausanneSwitzerland
| | - Adeline Berger
- Ophthalmology DepartmentUniversity of Lausanne, Jules‐Gonin Eye Hospital, Fondation Asile des AveuglesLausanneSwitzerland
| | - Paul J. Dyson
- Institute of Chemical Sciences and EngineeringÉcole Polytechnique Fédérale de Lausanne (EPFL)LausanneSwitzerland
| |
Collapse
|
18
|
Amézaga González MF, Acosta Bezada J, Gómez Flores V, Chapa González C, Farias Mancilla JR, Castillo SJ, Avila Orta C, García-Casillas PE. Effect of Physiological Fluid on the Photothermal Properties of Gold Nanostructured. Int J Mol Sci 2023; 24:ijms24098339. [PMID: 37176046 PMCID: PMC10179537 DOI: 10.3390/ijms24098339] [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: 03/08/2023] [Revised: 04/12/2023] [Accepted: 04/18/2023] [Indexed: 05/15/2023] Open
Abstract
Colloidal gold particles have been extensively studied for their potential in hyperthermia treatment due to their ability to become excited in the presence of an external laser. However, their light-to-heat efficiency is affected by the physiologic environment. In this study, we aimed to evaluate the ability of gold sphere, rod, and star-shaped colloids to elevate the temperature of blood plasma and breast cancer-simulated fluid under laser stimulation. Additionally, the dependence of optical properties and colloid stability of gold nanostructures with physiological medium, particle shape, and coating was determined. The light-to-heat efficiency of the gold particle is shape-dependent. The light-to-heat conversion efficiency of a star-shaped colloid is 36% higher than that of sphere-shaped colloids. However, the raised temperature of the surrounding medium is the lowest in the star-shaped colloid. When gold nanostructures are exited with a laser stimulation in a physiological fluid, the ions/cations attach to the surface of the gold particles, resulting in colloidal instability, which limits electron oscillation and diminishes the energy generated by the plasmonic excitation. Fluorescein (Fl) and polyethylene glycol (PEG) attached to gold spheres enhances their colloidal stability and light-to-heat efficiency; post-treatment, they remand their optical properties.
Collapse
Affiliation(s)
- María Fernanda Amézaga González
- Insituto de Ingenieria y Tecnología, Universidad Autónoma de Ciudad Juárez, Av. del Charro no. 450 Nte. Col. Partido Romero, Ciudad Juárez 32310, CHIH, Mexico
| | - Jazzely Acosta Bezada
- Insituto de Ingenieria y Tecnología, Universidad Autónoma de Ciudad Juárez, Av. del Charro no. 450 Nte. Col. Partido Romero, Ciudad Juárez 32310, CHIH, Mexico
| | - Víctor Gómez Flores
- Insituto de Ingenieria y Tecnología, Universidad Autónoma de Ciudad Juárez, Av. del Charro no. 450 Nte. Col. Partido Romero, Ciudad Juárez 32310, CHIH, Mexico
| | - Christian Chapa González
- Insituto de Ingenieria y Tecnología, Universidad Autónoma de Ciudad Juárez, Av. del Charro no. 450 Nte. Col. Partido Romero, Ciudad Juárez 32310, CHIH, Mexico
| | - Jose Rurik Farias Mancilla
- Insituto de Ingenieria y Tecnología, Universidad Autónoma de Ciudad Juárez, Av. del Charro no. 450 Nte. Col. Partido Romero, Ciudad Juárez 32310, CHIH, Mexico
| | - S J Castillo
- Departamento de Investigación en Física, Universidad de Sonora, Blvd. Luis Encinas y Rosales S/N, Hermosillo 83000, SON, Mexico
| | - Carlos Avila Orta
- Centro de Investigación en Química Aplicada, Blvd. Enrique Reyna Hermosillo No. 140, Saltillo 25294, COAH, Mexico
| | - Perla E García-Casillas
- Centro de Investigación en Química Aplicada, Blvd. Enrique Reyna Hermosillo No. 140, Saltillo 25294, COAH, Mexico
| |
Collapse
|
19
|
Kola P, Nagesh PKB, Roy PK, Deepak K, Reis RL, Kundu SC, Mandal M. Innovative nanotheranostics: Smart nanoparticles based approach to overcome breast cancer stem cells mediated chemo- and radioresistances. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023:e1876. [PMID: 36600447 DOI: 10.1002/wnan.1876] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/29/2022] [Accepted: 12/09/2022] [Indexed: 01/06/2023]
Abstract
The alarming increase in the number of breast cancer patients worldwide and the increasing death rate indicate that the traditional and current medicines are insufficient to fight against it. The onset of chemo- and radioresistances and cancer stem cell-based recurrence make this problem harder, and this hour needs a novel treatment approach. Competent nanoparticle-based accurate drug delivery and cancer nanotheranostics like photothermal therapy, photodynamic therapy, chemodynamic therapy, and sonodynamic therapy can be the key to solving this problem due to their unique characteristics. These innovative formulations can be a better cargo with fewer side effects than the standard chemotherapy and can eliminate the stability problems associated with cancer immunotherapy. The nanotheranostic systems can kill the tumor cells and the resistant breast cancer stem cells by novel mechanisms like local hyperthermia and reactive oxygen species and prevent tumor recurrence. These theranostic systems can also combine with chemotherapy or immunotherapy approaches. These combining approaches can be the future of anticancer therapy, especially to overcome the breast cancer stem cells mediated chemo- and radioresistances. This review paper discusses several novel theranostic systems and smart nanoparticles, their mechanism of action, and their modifications with time. It explains their relevance and market scope in the current era. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Prithwish Kola
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | | | - Pritam Kumar Roy
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - K Deepak
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Rui Luis Reis
- 3Bs Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimaraes, Portugal
| | - Subhas C Kundu
- 3Bs Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimaraes, Portugal
| | - Mahitosh Mandal
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| |
Collapse
|
20
|
Image-guided drug delivery in nanosystem-based cancer therapies. Adv Drug Deliv Rev 2023; 192:114621. [PMID: 36402247 DOI: 10.1016/j.addr.2022.114621] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 10/18/2022] [Accepted: 11/13/2022] [Indexed: 11/18/2022]
Abstract
The past decades have shown significant advancements in the development of solid tumor treatment. For instance, implementation of nanosystems for drug delivery has led to a reduction in side effects and improved delivery to the tumor region. However, clinical translation has faced challenges, as tumor drug levels are still considered to be inadequate. Interdisciplinary research has resulted in the development of more advanced drug delivery systems. These are coined "smart" due to the ability to be followed and actively manipulated in order to have better control over local drug release. Therefore, image-guided drug delivery can be a powerful strategy to improve drug activity at the target site. Being able to visualize the inflow of the administered smart nanosystem within the tumor gives the potential to determine the right moment to apply the facilitator to initiate drug release. Here we provide an overview of available nanosystems, imaging moieties, and imaging techniques. We discuss preclinical application of these smart drug delivery systems, the strength of image-guided drug delivery, and the future of personalized treatment.
Collapse
|
21
|
Toro-Córdova A, Llaguno-Munive M, Jurado R, Garcia-Lopez P. The Therapeutic Potential of Chemo/Thermotherapy with Magnetoliposomes for Cancer Treatment. Pharmaceutics 2022; 14:2443. [PMID: 36432634 PMCID: PMC9697689 DOI: 10.3390/pharmaceutics14112443] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/02/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
Cancer represents a very grave and quickly growing public health problem worldwide. Despite the breakthroughs in treatment and early detection of the disease, an increase is projected in the incidence rate and mortality during the next 30 years. Thus, it is important to develop new treatment strategies and diagnostic tools. One alternative is magnetic hyperthermia, a therapeutic approach that has shown promising results, both as monotherapy and in combination with chemo- and radiotherapy. However, there are still certain limitations and questions with respect to the safety of the systemic administration of magnetic nanoparticles. To deal with these issues, magnetoliposomes were conceived as a new generation of liposomes that incorporate superparamagnetic nanoparticles and oncological pharmaceuticals within their structure. They have the advantage of targeted and selective drug delivery to the diseased organs and tissues. Some of them can avoid the immune response of the host. When exposed to a magnetic field of alternating current, magnetoliposomes produce hyperthermia, which acts synergistically with the released drug. The aim of the present review is to describe the most recent advances in the use of magnetoliposomes and point out what research remains to be done for their application to chemo-thermal therapy in cancer patients.
Collapse
Affiliation(s)
- Alfonso Toro-Córdova
- Laboratorio de Fármaco-Oncología, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Cd, Mexico City 14080, Mexico
- Departamento de Formulación de Vacunas de mRNA, CerTest Biotec S.L., 50840 Zaragoza, Spain
| | - Monserrat Llaguno-Munive
- Laboratorio de Fármaco-Oncología, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Cd, Mexico City 14080, Mexico
- Laboratorio de Física Médica, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Cd, Mexico City 14080, Mexico
| | - Rafael Jurado
- Laboratorio de Fármaco-Oncología, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Cd, Mexico City 14080, Mexico
| | - Patricia Garcia-Lopez
- Laboratorio de Fármaco-Oncología, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Cd, Mexico City 14080, Mexico
| |
Collapse
|
22
|
El Gohary NA, Mahmoud A, Ashraf Nazmy M, Zaabalawi R, El Zahar L, Khalil ISM, Mitwally ME. Magnetic polycaprolactone microspheres: drug encapsulation and control. INT J POLYM MATER PO 2022. [DOI: 10.1080/00914037.2022.2132248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- Nesrine Abdelrehim El Gohary
- Pharmaceutical Chemistry Department, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| | - Abdelrahman Mahmoud
- Materials Engineering Department, Faculty of Engineering and Materials Science, German University in Cairo, Cairo, Egypt
| | | | - Rami Zaabalawi
- Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| | - Loaa El Zahar
- Faculty of Media Engineering and Technology, German University in Cairo, Cairo, Egypt
| | - Islam S. M. Khalil
- Department of Biomechanical Engineering, University of Twente, Enschede, the Netherlands
| | - Mohamed E. Mitwally
- Materials Engineering Department, Faculty of Engineering and Materials Science, German University in Cairo, Cairo, Egypt
| |
Collapse
|
23
|
Aram E, Moeni M, Abedizadeh R, Sabour D, Sadeghi-Abandansari H, Gardy J, Hassanpour A. Smart and Multi-Functional Magnetic Nanoparticles for Cancer Treatment Applications: Clinical Challenges and Future Prospects. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:nano12203567. [PMID: 36296756 PMCID: PMC9611246 DOI: 10.3390/nano12203567] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/16/2022] [Accepted: 09/27/2022] [Indexed: 05/14/2023]
Abstract
Iron oxide nanoparticle (IONPs) have become a subject of interest in various biomedical fields due to their magnetism and biocompatibility. They can be utilized as heat mediators in magnetic hyperthermia (MHT) or as contrast media in magnetic resonance imaging (MRI), and ultrasound (US). In addition, their high drug-loading capacity enabled them to be therapeutic agent transporters for malignancy treatment. Hence, smartening them allows for an intelligent controlled drug release (CDR) and targeted drug delivery (TDD). Smart magnetic nanoparticles (SMNPs) can overcome the impediments faced by classical chemo-treatment strategies, since they can be navigated and release drug via external or internal stimuli. Recently, they have been synchronized with other modalities, e.g., MRI, MHT, US, and for dual/multimodal theranostic applications in a single platform. Herein, we provide an overview of the attributes of MNPs for cancer theranostic application, fabrication procedures, surface coatings, targeting approaches, and recent advancement of SMNPs. Even though MNPs feature numerous privileges over chemotherapy agents, obstacles remain in clinical usage. This review in particular covers the clinical predicaments faced by SMNPs and future research scopes in the field of SMNPs for cancer theranostics.
Collapse
Affiliation(s)
- Elham Aram
- Department of Cancer Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Babol 47138-18981, Iran
- Department of Polymer Engineering, Faculty of Engineering, Golestan University, Gorgan 49188-88369, Iran
| | - Masome Moeni
- School of Chemical and Process Engineering, University of Leeds, Leeds LS2 9JT, UK
| | - Roya Abedizadeh
- Department of Cancer Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Babol 47138-18981, Iran
| | - Davood Sabour
- Department of Cancer Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Babol 47138-18981, Iran
| | - Hamid Sadeghi-Abandansari
- Department of Cancer Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Babol 47138-18981, Iran
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 16635-148, Iran
| | - Jabbar Gardy
- School of Chemical and Process Engineering, University of Leeds, Leeds LS2 9JT, UK
- Correspondence: (J.G.); (A.H.)
| | - Ali Hassanpour
- School of Chemical and Process Engineering, University of Leeds, Leeds LS2 9JT, UK
- Correspondence: (J.G.); (A.H.)
| |
Collapse
|
24
|
Amin M, Lammers T, Ten Hagen TLM. Temperature-sensitive polymers to promote heat-triggered drug release from liposomes: Towards bypassing EPR. Adv Drug Deliv Rev 2022; 189:114503. [PMID: 35998827 DOI: 10.1016/j.addr.2022.114503] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 06/14/2022] [Accepted: 08/17/2022] [Indexed: 01/24/2023]
Abstract
Heat-triggered drug release from temperature-sensitive nanocarriers upon the application of mild hyperthermia is a promising approach to achieve site-specific delivery of drugs. The combination of mild hyperthermia (41-42 °C) and temperature-sensitive liposomes (TSL) that undergo lipid phase-transition and drug release has been studied extensively and has shown promising therapeutic outcome in a variety of animal tumor models as well as initial indications of success in humans. Sensitization of liposomes to mild hyperthermia by means of exploiting the thermal behavior of temperature-sensitive polymers (TSP) provides novel opportunities. Recently, TSP-modified liposomes (TSPL) have shown potential for enhancing tumor-directed drug delivery, either by triggered drug release or by triggered cell interactions in response to heat. In this review, we describe different classes of TSPL, and analyze and discuss the mechanisms and kinetics of content release from TSPL in response to local heating. In addition, the impact of lipid composition, polymer and copolymer characteristics, serum components and PEGylation on the mechanism of content release and TSPL performance is addressed. This is done from the perspective of rationally designing TSPL, with the overall goal of conceiving efficient strategies to increase the efficacy of TSPL plus hyperthermia to improve the outcome of targeted anticancer therapy.
Collapse
Affiliation(s)
- Mohamadreza Amin
- Laboratory of Experimental Oncology (LEO), Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands; Nanomedicine Innovation Center Erasmus (NICE), Erasmus Medical Center, Rotterdam, The Netherlands.
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Center for Biohybrid Medical Systems, Aachen, Germany.
| | - Timo L M Ten Hagen
- Laboratory of Experimental Oncology (LEO), Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands; Nanomedicine Innovation Center Erasmus (NICE), Erasmus Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
25
|
Maleš P, Pem B, Petrov D, Jurašin DD, Bakarić D. Deciphering the origin of the melting profile of unilamellar phosphatidylcholine liposomes by measuring the turbidity of its suspensions. SOFT MATTER 2022; 18:6703-6715. [PMID: 36017811 DOI: 10.1039/d2sm00878e] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The elucidation of the thermal properties of phosphatidylcholine liposomes is often based on the analysis of the thermal capacity profiles of multilamellar liposomes (MLV), which may qualitatively disagree with those of unilamellar liposomes (LUV). Experiments and interpretation of LUV liposomes is further complicated by aggregation and lamellarization of lipid bilayers in a short time period, which makes it almost impossible to distinguish the signatures of the two types of bilayers. To characterize independently MLV and LUV of 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC), the latter were prepared with the addition of small amounts of 1,2-dipalmitoyl-sn-glycero-3-phosphatidylglycerol (DPPG) which, due to the sterical hindrance and negative charge at a given pH value, cause LUV repellence and contribute to their stability. Differential scanning calorimetry curves and temperature-dependent UV/Vis spectra of the prepared MLV and LUV were measured. Multivariate analysis of spectrophotometric data determined the phase transition temperatures (pretransition at Tp and the main phase transition at Tm), and based on the changes in turbidities, the thickness of the lipid bilayer in LUV was determined. The obtained data suggested that the curvature change is a key distinguishing factor in MLV and LUV heat capacity profiles. By combining the experimental results and those obtained by MD simulations, the interfacial water layer was characterized and its contribution to the thermal properties of LUV was discussed.
Collapse
Affiliation(s)
- Petra Maleš
- Division for Organic Chemistry and Biochemistry, Ruđer Bošković Institute, Bijenička 54, 10000 Zagreb, Croatia.
| | - Barbara Pem
- Division for Organic Chemistry and Biochemistry, Ruđer Bošković Institute, Bijenička 54, 10000 Zagreb, Croatia.
| | - Dražen Petrov
- Institute of Molecular Modeling and Simulation, University of Natural Resources and Life Sciences, 1180 Vienna, Austria
| | - Darija Domazet Jurašin
- Division for Physical Chemistry, Ruđer Bošković Institute, Bijenička 54, 10000 Zagreb, Croatia
| | - Danijela Bakarić
- Division for Organic Chemistry and Biochemistry, Ruđer Bošković Institute, Bijenička 54, 10000 Zagreb, Croatia.
| |
Collapse
|
26
|
Dymek M, Sikora E. Liposomes as biocompatible and smart delivery systems – The current state. Adv Colloid Interface Sci 2022; 309:102757. [DOI: 10.1016/j.cis.2022.102757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 08/12/2022] [Accepted: 08/16/2022] [Indexed: 11/01/2022]
|
27
|
Liu P, Ye M, Wu Y, Wu L, Lan K, Wu Z. Hyperthermia combined with immune checkpoint inhibitor therapy: Synergistic sensitization and clinical outcomes. Cancer Med 2022; 12:3201-3221. [PMID: 35908281 PMCID: PMC9939221 DOI: 10.1002/cam4.5085] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 06/24/2022] [Accepted: 06/29/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Within the field of oncotherapy, research interest regarding immunotherapy has risen to the point that it is now seen as a key application. However, inherent disadvantages of immune checkpoint inhibitors (ICIs), such as their low response rates and immune-related adverse events (irAEs), currently restrict their clinical application. Were these disadvantages to be overcome, more patients could derive prolonged benefits from ICIs. At present, many basic experiments and clinical studies using hyperthermia combined with ICI treatment (HIT) have been performed and shown the potential to address the above challenges. Therefore, this review extensively summarizes the knowledge and progress of HIT for analysis and discusses the effect and feasibility. METHODS In this review, we explored the PubMed and clinicaltrials.gov databases, with regard to the searching terms "immune checkpoint inhibitor, immunotherapy, hyperthermia, ablation, photothermal therapy". RESULTS By reviewing the literature, we analyzed how hyperthermia influences tumor immunology and improves the efficacy of ICI. Hyperthermia can trigger a series of multifactorial molecular cascade reactions between tumors and immunization and can significantly induce cytological modifications within the tumor microenvironment (TME). The pharmacological potency of ICIs can be enhanced greatly through the immunomodulatory amelioration of immunosuppression, and the activation of immunostimulation. Emerging clinical trials outcome regarding HIT have verified and enriched the theoretical foundation of synergistic sensitization. CONCLUSION HIT research is now starting to transition from preclinical studies to clinical investigations. Several HIT sensitization mechanisms have been reflected and demonstrated as significant survival benefits for patients through pioneering clinical trials. Further studies into the theoretical basis and practical standards of HIT, combined with larger-scale clinical studies involving more cancer types, will be necessary for the future.
Collapse
Affiliation(s)
- Pengyuan Liu
- Oncology & Radiotherapy DepartmentZhejiang HospitalHangzhouChina,Second Clinical Medical CollegeZhejiang Chinese Medical UniversityHangzhouChina
| | - Mengna Ye
- Second Clinical Medical CollegeZhejiang Chinese Medical UniversityHangzhouChina
| | - Yajun Wu
- Department of TCM PharmacyZhejiang HospitalHangzhouChina
| | - Lichao Wu
- College of Basic Medical SciencesZhejiang Chinese Medical UniversityHangzhouChina
| | - Kaiping Lan
- Oncology Department of Combination of Traditional Chinese and Western MedicineTonglu Hospital of Traditional Chinese MedicineHangzhouChina
| | - Zhibing Wu
- Oncology & Radiotherapy DepartmentZhejiang HospitalHangzhouChina
| |
Collapse
|
28
|
Shabani L, Abbasi M, Amini M, Amani AM, Vaez A. The brilliance of nanoscience over cancer therapy: Novel promising nanotechnology-based methods for eradicating glioblastoma. J Neurol Sci 2022; 440:120316. [DOI: 10.1016/j.jns.2022.120316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 05/28/2022] [Accepted: 05/31/2022] [Indexed: 10/18/2022]
|
29
|
Zhou L, Zou M, Xu Y, Lin P, Lei C, Xia X. Nano Drug Delivery System for Tumor Immunotherapy: Next-Generation Therapeutics. Front Oncol 2022; 12:864301. [PMID: 35664731 PMCID: PMC9160744 DOI: 10.3389/fonc.2022.864301] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 04/07/2022] [Indexed: 12/12/2022] Open
Abstract
Tumor immunotherapy is an artificial stimulation of the immune system to enhance anti-cancer response. It has become a powerful clinical strategy for treating cancer. The number of immunotherapy drug approvals has been increasing in recent years, and many treatments are in clinical and preclinical stages. Despite this progress, the special tumor heterogeneity and immunosuppressive microenvironment of solid tumors made immunotherapy in the majority of cancer cases difficult. Therefore, understanding how to improve the intratumoral enrichment degree and the response rate of various immunotherapy drugs is key to improve efficacy and control adverse reactions. With the development of materials science and nanotechnology, advanced biomaterials such as nanoparticle and drug delivery systems like T-cell delivery therapy can improve effectiveness of immunotherapy while reducing the toxic side effects on non-target cells, which offers innovative ideas for improving immunity therapeutic effectiveness. In this review, we discuss the mechanism of tumor cell immune escape and focus on current immunotherapy (such as cytokine immunotherapy, therapeutic monoclonal antibody immunotherapy, PD-1/PD-L1 therapy, CAR-T therapy, tumor vaccine, oncolytic virus, and other new types of immunity) and its challenges as well as the latest nanotechnology (such as bionic nanoparticles, self-assembled nanoparticles, deformable nanoparticles, photothermal effect nanoparticles, stimuli-responsive nanoparticles, and other types) applications in cancer immunotherapy.
Collapse
Affiliation(s)
- Lili Zhou
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Manshu Zou
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Yilin Xu
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Peng Lin
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Chang Lei
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Xinhua Xia
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
30
|
Salkho NM, Awad NS, Pitt WG, Husseini GA. Photo-Induced Drug Release from Polymeric Micelles and Liposomes: Phototriggering Mechanisms in Drug Delivery Systems. Polymers (Basel) 2022; 14:1286. [PMID: 35406160 PMCID: PMC9003562 DOI: 10.3390/polym14071286] [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: 01/28/2022] [Revised: 03/12/2022] [Accepted: 03/16/2022] [Indexed: 12/13/2022] Open
Abstract
Chemotherapeutic drugs are highly effective in treating cancer. However, the side effects associated with this treatment lower the quality of life of cancer patients. Smart nanocarriers are able to encapsulate these drugs to deliver them to tumors while reducing their contact with the healthy cells and the subsequent side effects. Upon reaching their target, the release of the encapsulated drugs should be carefully controlled to achieve therapeutic levels at the required time. Light is one of the promising triggering mechanisms used as external stimuli to trigger drug release from the light-responsive nanocarriers. Photo-induced drug release can be achieved at a wide range of wavelengths: UV, visible, and NIR depending on many factors. In this review, photo-induced release mechanisms were summarized, focusing on liposomes and micelles. In general, light-triggering mechanisms are based on one of the following: changing the hydrophobicity of a nanocarrier constituent(s) to make it more soluble, introducing local defects within a nanocarrier (by conformational transformation or photo-cleavage of its lipids/polymers chains) to make it more porous or concentrating heat for thermo-sensitive nanocarriers to release their payload. Several research studies were also presented to explore the potentials and limitations of this promising drug release triggering mechanism.
Collapse
Affiliation(s)
- Najla M Salkho
- Department of Chemical Engineering, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
- Materials Science and Engineering Program, College of Arts and Sciences, American University of Sharjah, Sharjah P.O. Box. 26666, United Arab Emirates
| | - Nahid S Awad
- Department of Chemical Engineering, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
| | - William G Pitt
- Chemical Engineering Department, Brigham Young University, Provo, UT 84602, USA
| | - Ghaleb A Husseini
- Department of Chemical Engineering, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
- Materials Science and Engineering Program, College of Arts and Sciences, American University of Sharjah, Sharjah P.O. Box. 26666, United Arab Emirates
| |
Collapse
|
31
|
Zuo Z, Li M, Han T, Zheng X, Yao W, Wang H, Li X, Qu D. A platelet-cloaking tetramethylprazine-loaded microemulsion for improved therapy of myocardial ischemia/reperfusion injury. J Drug Target 2022; 30:646-656. [PMID: 35225125 DOI: 10.1080/1061186x.2022.2048389] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- Zhi Zuo
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | | | - Tao Han
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | | | - Wenming Yao
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Hui Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Xinli Li
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Ding Qu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
32
|
Musolino E, Pagiatakis C, Serio S, Borgese M, Gamberoni F, Gornati R, Bernardini G, Papait R. The Yin and Yang of epigenetics in the field of nanoparticles. NANOSCALE ADVANCES 2022; 4:979-994. [PMID: 36131763 PMCID: PMC9419747 DOI: 10.1039/d1na00682g] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 12/30/2021] [Indexed: 05/02/2023]
Abstract
Nanoparticles (NPs) have become a very exciting research avenue, with multitudinous applications in various fields, including the biomedical one, whereby they have been gaining considerable interest as drug carriers able to increase bioavailability, therapeutic efficiency and specificity of drugs. Epigenetics, a complex network of molecular mechanisms involved in gene expression regulation, play a key role in mediating the effect of environmental factors on organisms and in the etiology of several diseases (e.g., cancers, neurological disorders and cardiovascular diseases). For many of these diseases, epigenetic therapies have been proposed, whose application is however limited by the toxicity of epigenetic drugs. In this review, we will analyze two aspects of epigenetics in the field of NPs: the first is the role that epigenetics play in mediating nanotoxicity, and the second is the possibility of using NPs for delivery of "epi-drugs" to overcome their limitations. We aim to stimulate discussion among specialists, specifically on the potential contribution of epigenetics to the field of NPs, and to inspire newcomers to this exciting technology.
Collapse
Affiliation(s)
- Elettra Musolino
- Department of and Life Sciences, Insubria University Via Dunant 3 21100 Varese Italy
| | - Christina Pagiatakis
- Department of Cardiovascular Medicine, Humanitas Research Hospital Rozzano MI Italy
| | - Simone Serio
- Department of Cardiovascular Medicine, Humanitas Research Hospital Rozzano MI Italy
- Department of Biomedical Sciences, Humanitas University Via Rita Levi Montalcini 4 20090 Pieve Emanuele MI Italy
| | - Marina Borgese
- Department of and Life Sciences, Insubria University Via Dunant 3 21100 Varese Italy
| | - Federica Gamberoni
- Department of and Life Sciences, Insubria University Via Dunant 3 21100 Varese Italy
| | - Rosalba Gornati
- Department of and Life Sciences, Insubria University Via Dunant 3 21100 Varese Italy
| | - Giovanni Bernardini
- Department of and Life Sciences, Insubria University Via Dunant 3 21100 Varese Italy
| | - Roberto Papait
- Department of and Life Sciences, Insubria University Via Dunant 3 21100 Varese Italy
- Department of Cardiovascular Medicine, Humanitas Research Hospital Rozzano MI Italy
| |
Collapse
|
33
|
Ding X, Sun X, Cai H, Wu L, Liu Y, Zhao Y, Zhou D, Yu G, Zhou X. Engineering Macrophages via Nanotechnology and Genetic Manipulation for Cancer Therapy. Front Oncol 2022; 11:786913. [PMID: 35070992 PMCID: PMC8770285 DOI: 10.3389/fonc.2021.786913] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/13/2021] [Indexed: 12/14/2022] Open
Abstract
Macrophages play critical roles in tumor progression. In the tumor microenvironment, macrophages display highly diverse phenotypes and may perform antitumorigenic or protumorigenic functions in a context-dependent manner. Recent studies have shown that macrophages can be engineered to transport drug nanoparticles (NPs) to tumor sites in a targeted manner, thereby exerting significant anticancer effects. In addition, macrophages engineered to express chimeric antigen receptors (CARs) were shown to actively migrate to tumor sites and eliminate tumor cells through phagocytosis. Importantly, after reaching tumor sites, these engineered macrophages can significantly change the otherwise immune-suppressive tumor microenvironment and thereby enhance T cell-mediated anticancer immune responses. In this review, we first introduce the multifaceted activities of macrophages and the principles of nanotechnology in cancer therapy and then elaborate on macrophage engineering via nanotechnology or genetic approaches and discuss the effects, mechanisms, and limitations of such engineered macrophages, with a focus on using live macrophages as carriers to actively deliver NP drugs to tumor sites. Several new directions in macrophage engineering are reviewed, such as transporting NP drugs through macrophage cell membranes or extracellular vesicles, reprogramming tumor-associated macrophages (TAMs) by nanotechnology, and engineering macrophages with CARs. Finally, we discuss the possibility of combining engineered macrophages and other treatments to improve outcomes in cancer therapy.
Collapse
Affiliation(s)
- Xiaoling Ding
- Department of Immunology, Nantong University, School of Medicine, Nantong, China.,Department of Gastroenterology, The Affiliated Hospital of Nantong University, Nantong, China
| | - Xinchen Sun
- Department of Immunology, Nantong University, School of Medicine, Nantong, China.,Department of Clinical Laboratory, Taizhou Peoples' Hospital, Taizhou, China
| | - Huihui Cai
- Department of Immunology, Nantong University, School of Medicine, Nantong, China.,Department of Clinical Laboratory, The Sixth Nantong People's Hospital, Nantong, China
| | - Lei Wu
- Department of Immunology, Nantong University, School of Medicine, Nantong, China
| | - Ying Liu
- Department of Immunology, Nantong University, School of Medicine, Nantong, China
| | - Yu Zhao
- Department of Immunology, Southeast University, School of Medicine, Nanjing, China
| | - Dingjingyu Zhou
- Krieger School of Arts & Sciences, Johns Hopkins University, Baltimore, MD, United States
| | - Guiping Yu
- Department of Cardiothoracic Surgery, The Affiliated Jiangyin Hospital of Nantong University, Jiangyin, China
| | - Xiaorong Zhou
- Department of Immunology, Nantong University, School of Medicine, Nantong, China
| |
Collapse
|
34
|
Liu H, Prachyathipsakul T, Koyasseril-Yehiya TM, Le SP, Thayumanavan S. Molecular bases for temperature sensitivity in supramolecular assemblies and their applications as thermoresponsive soft materials. MATERIALS HORIZONS 2022; 9:164-193. [PMID: 34549764 PMCID: PMC8757657 DOI: 10.1039/d1mh01091c] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Thermoresponsive supramolecular assemblies have been extensively explored in diverse formats, from injectable hydrogels to nanoscale carriers, for a variety of applications including drug delivery, tissue engineering and thermo-controlled catalysis. Understanding the molecular bases behind thermal sensitivity of materials is fundamentally important for the rational design of assemblies with optimal combination of properties and predictable tunability for specific applications. In this review, we summarize the recent advances in this area with a specific focus on the parameters and factors that influence thermoresponsive properties of soft materials. We summarize and analyze the effects of structures and architectures of molecules, hydrophilic and lipophilic balance, concentration, components and external additives upon the thermoresponsiveness of the corresponding molecular assemblies.
Collapse
Affiliation(s)
- Hongxu Liu
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, USA.
| | | | | | - Stephanie P Le
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, USA.
| | - S Thayumanavan
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, USA.
- Department of Biomedical Engineering, University of Massachusetts, Amherst, Massachusetts 01003, USA
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, Massachusetts 01003, USA
- Centre for Bioactive Delivery, Institute for Applied Life Science, University of Massachusetts, Amherst, Massachusetts 01003, USA
| |
Collapse
|
35
|
Ramajayam KK, Newton DA, Haemmerich D. Selecting ideal drugs for encapsulation in thermosensitive liposomes and other triggered nanoparticles. Int J Hyperthermia 2022; 39:998-1009. [PMID: 35876089 PMCID: PMC9774053 DOI: 10.1080/02656736.2022.2086303] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
OBJECTIVE Thermosensitive liposomes (TSL) and other triggered drug delivery systems (DDS) are promising therapeutic strategies for targeted drug delivery. However, successful designs with candidate drugs depend on many variables, including nanoparticle formulation, drug properties, and cancer cell properties. We developed a computational model based on experimental data to predict the potential efficacies of drugs when used with triggered DDS, such as TSL. METHODS A computer model based on the Krogh cylinder was developed to predict uptake and cell survival with four anthracyclines when delivered by intravascular triggered DDS (e.g., TSL): doxorubicin (DOX), idarubicin (IDA), pirarubicin (PIR), and aclarubicin (ACLA). We simulated three tumor types derived from SVR angiosarcoma, LLC lung cancer, or SCC-1 oral carcinoma cells. In vitro cellular drug uptake and cytotoxicity data were obtained experimentally and incorporated into the model. RESULTS For all three cell lines, ACLA and IDA had the fastest cell uptake, with slower uptake for DOX and PIR. Cytotoxicity was highest for IDA and lowest for ACLA. The computer model predicted the highest tumor drug uptake for ACLA and IDA, resulting from their rapid cell uptake. Overall, IDA was most effective and produced the lowest tumor survival fraction, with DOX being the second best. Perivascular drug penetration was reduced for drugs with rapid cell uptake, potentially limiting delivery to cancer cells distant from the vasculature. CONCLUSION Combining simple in vitro experiments with a computer model could provide a powerful screening tool to evaluate the potential efficacy of candidate investigative drugs preceding TSL encapsulation and in vivo studies.
Collapse
Affiliation(s)
- Krishna K. Ramajayam
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC 29425
| | - Danforth A. Newton
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC 29425
| | - Dieter Haemmerich
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC 29425,Corresponding author: (D. Haemmerich)
| |
Collapse
|
36
|
Fobian SF, Cheng Z, ten Hagen TLM. Smart Lipid-Based Nanosystems for Therapeutic Immune Induction against Cancers: Perspectives and Outlooks. Pharmaceutics 2021; 14:26. [PMID: 35056922 PMCID: PMC8779430 DOI: 10.3390/pharmaceutics14010026] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/19/2021] [Accepted: 12/20/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer immunotherapy, a promising and widely applied mode of oncotherapy, makes use of immune stimulants and modulators to overcome the immune dysregulation present in cancer, and leverage the host's immune capacity to eliminate tumors. Although some success has been seen in this field, toxicity and weak immune induction remain challenges. Liposomal nanosystems, previously used as targeting agents, are increasingly functioning as immunotherapeutic vehicles, with potential for delivery of contents, immune induction, and synergistic drug packaging. These systems are tailorable, multifunctional, and smart. Liposomes may deliver various immune reagents including cytokines, specific T-cell receptors, antibody fragments, and immune checkpoint inhibitors, and also present a promising platform upon which personalized medicine approaches can be built, especially with preclinical and clinical potentials of liposomes often being frustrated by inter- and intrapatient variation. In this review, we show the potential of liposomes in cancer immunotherapy, as well as the methods for synthesis and in vivo progression thereof. Both preclinical and clinical studies are included to comprehensively illuminate prospects and challenges for future research and application.
Collapse
Affiliation(s)
| | | | - Timo L. M. ten Hagen
- Laboratory Experimental Oncology (LEO), Department of Pathology, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands; (S.-F.F.); (Z.C.)
| |
Collapse
|
37
|
Feng ZH, Li ZT, Zhang S, Wang JR, Li ZY, Xu MQ, Li H, Zhang SQ, Wang GX, Liao A, Zhang X. A combination strategy based on an Au nanorod/doxorubicin gel via mild photothermal therapy combined with antigen-capturing liposomes and anti-PD-L1 agent promote a positive shift in the cancer-immunity cycle. Acta Biomater 2021; 136:495-507. [PMID: 34619371 DOI: 10.1016/j.actbio.2021.09.052] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 09/14/2021] [Accepted: 09/27/2021] [Indexed: 01/09/2023]
Abstract
The antitumor immune response involves a cascade of cancer-immunity cycles. Developing a combination therapy aimed at the cancer-immunity cycle is of great importance. In this research, we designed and tested a combined therapeutic-Au nanorod (AuNR)/doxorubicin (DOX) gel (AuNR/DOX gel)-in which the sustained release of DOX was controlled by Pluronic gel. DOX served as an immunogenic tumor cell death (ICD) inducer, triggering the production of damage-associated molecular patterns (DAMPs). Mild photothermal therapy (Mild PTT) produced by 880 nm laser-irradiated AuNRs also generated tumor-associated antigens. Maleimide-modified liposomes (L-Mals), as antigen capturing agents, promoted tumor antigen uptake by DCs. Ultimately, more CD8+ T cells and fewer regulatory T cells (Tregs) infiltrated the tumor, eliciting antitumor responses from the PD-L1 antibody. Our results indicate that this combination strategy promotes a positive shift in the cancer-immunity cycle and holds much promise for combination strategy will lead to development of an antitumor drug delivery system. STATEMENT OF SIGNIFICANCE: Developing a combination therapy for cancer-immunity cycle is of great importance due to antitumor immune response involving a cascade of cancer-immunity cycles. Cancer-immunity cycle usually includes tumor antigen release, antigen presentation, immune activation, trafficking, infiltration, specific recognition of tumor cells by T cells, and finally cancer cell killing. In this research, we designed a combination strategy based on Au nanorod/doxorubicin gel via mild photothermal therapy combined with antigen-capturing liposomes and anti-PD-L1 agent promoting a positive shift in the cancer-immunity cycle. Our results indicate that this combination strategy promotes a positive shift in the cancer-immunity cycle and holds much promise for combination strategy will lead to development of an antitumor drug delivery system.
Collapse
Affiliation(s)
- Zhen-Han Feng
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China
| | - Zhan-Tao Li
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China
| | - Shuang Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China
| | - Jing-Ru Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China
| | - Zhuo-Yue Li
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China
| | - Mei-Qi Xu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China
| | - Hui Li
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Shuai-Qiang Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China
| | - Guang-Xue Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China
| | - Ai Liao
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China
| | - Xuan Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China.
| |
Collapse
|
38
|
Raouf I, Gas P, Kim HS. Numerical Investigation of Ferrofluid Preparation during In-Vitro Culture of Cancer Therapy for Magnetic Nanoparticle Hyperthermia. SENSORS (BASEL, SWITZERLAND) 2021; 21:5545. [PMID: 34450987 PMCID: PMC8402254 DOI: 10.3390/s21165545] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/12/2021] [Accepted: 08/12/2021] [Indexed: 12/19/2022]
Abstract
Recently, in-vitro studies of magnetic nanoparticle (MNP) hyperthermia have attracted significant attention because of the severity of this cancer therapy for in-vivo culture. Accurate temperature evaluation is one of the key challenges of MNP hyperthermia. Hence, numerical studies play a crucial role in evaluating the thermal behavior of ferrofluids. As a result, the optimum therapeutic conditions can be achieved. The presented research work aims to develop a comprehensive numerical model that directly correlates the MNP hyperthermia parameters to the thermal response of the in-vitro model using optimization through linear response theory (LRT). For that purpose, the ferrofluid solution is evaluated based on various parameters, and the temperature distribution of the system is estimated in space and time. Consequently, the optimum conditions for the ferrofluid preparation are estimated based on experimental and mathematical findings. The reliability of the presented model is evaluated via the correlation analysis between magnetic and calorimetric methods for the specific loss power (SLP) and intrinsic loss power (ILP) calculations. Besides, the presented numerical model is verified with our experimental setup. In summary, the proposed model offers a novel approach to investigate the thermal diffusion of a non-adiabatic ferrofluid sample intended for MNP hyperthermia in cancer treatment.
Collapse
Affiliation(s)
- Izaz Raouf
- Department of Mechanical, Robotics and Energy Engineering, Dongguk University-Seoul, 30 Pildong-ro 1-gil, Jung-gu, Seoul 100-715, Korea;
| | - Piotr Gas
- Department of Electrical and Power Engineering, AGH University of Science and Technology, Mickiewicza 30 Avenue, 30-059 Krakow, Poland
| | - Heung Soo Kim
- Department of Mechanical, Robotics and Energy Engineering, Dongguk University-Seoul, 30 Pildong-ro 1-gil, Jung-gu, Seoul 100-715, Korea;
| |
Collapse
|
39
|
Emerging Therapeutic Strategies for Brain Tumors. Neuromolecular Med 2021; 24:23-34. [PMID: 34406634 DOI: 10.1007/s12017-021-08681-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 07/10/2021] [Indexed: 12/14/2022]
Abstract
Nearly thirty thousand incidences of primary and 300 thousand incidences of metastatic brain cancer are diagnosed in the USA each year. It has a high mortality rate and is often unresponsive to the standard of care, which includes surgical resection, radiation, and chemotherapy. These treatment strategies are also hindered by their invasiveness and toxic effects on healthy cells and tissues. Furthermore, the blood-brain/tumor barrier severely limits delivery of anti-cancer therapeutics administered intravenously to brain tumors, resulting in poor tumor response to the treatment. There is a critical need to develop new approaches to brain cancer therapy that can overcome these limitations. Focused ultrasound has emerged as a modality that addresses many of these limitations and has the potential to alter the treatment paradigm for brain cancer. Ultrasound transmitted through the skull can be focused on tumors and used for targeted ablation or opening the vascular barriers for drug delivery. This review provides insight on the current status of these unique ultrasound techniques, different strategies of using this technique for brain cancer, experience in preclinical models, and potential for clinical translation. We also debate the safety perspective of these techniques and discuss potential avenues for future work in noninvasive planning, monitoring, and evaluation of the ultrasonic neurointervention.
Collapse
|
40
|
Fernández-Álvarez F, García-García G, Arias JL. A Tri-Stimuli Responsive (Maghemite/PLGA)/Chitosan Nanostructure with Promising Applications in Lung Cancer. Pharmaceutics 2021; 13:1232. [PMID: 34452193 PMCID: PMC8401782 DOI: 10.3390/pharmaceutics13081232] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 07/30/2021] [Accepted: 08/06/2021] [Indexed: 12/13/2022] Open
Abstract
A (core/shell)/shell nanostructure (production performance ≈ 50%, mean diameter ≈ 330 nm) was built using maghemite, PLGA, and chitosan. An extensive characterization proved the complete inclusion of the maghemite nuclei into the PLGA matrix (by nanoprecipitation solvent evaporation) and the disposition of the chitosan shell onto the nanocomposite (by coacervation). Short-term stability and the adequate magnetism of the nanocomposites were demonstrated by size and electrokinetic determinations, and by defining the first magnetization curve and the responsiveness of the colloid to a permanent magnet, respectively. Safety of the nanoparticles was postulated when considering the results from blood compatibility studies, and toxicity assays against human colonic CCD-18 fibroblasts and colon carcinoma T-84 cells. Cisplatin incorporation to the PLGA matrix generated appropriate loading values (≈15%), and a dual pH- and heat (hyperthermia)-responsive drug release behaviour (≈4.7-fold faster release at pH 5.0 and 45 °C compared to pH 7.4 and 37 °C). The half maximal inhibitory concentration of the cisplatin-loaded nanoparticles against human lung adenocarcinoma A-549 cells was ≈1.6-fold less than that of the free chemotherapeutic. Such a biocompatible and tri-stimuli responsive (maghemite/PLGA)/chitosan nanostructure may found a promising use for the effective treatment of lung cancer.
Collapse
Affiliation(s)
- Fátima Fernández-Álvarez
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Granada, 18071 Granada, Spain;
| | - Gracia García-García
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, 28223 Madrid, Spain;
| | - José L. Arias
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Granada, 18071 Granada, Spain;
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain
- Biosanitary Research Institute of Granada (ibs.GRANADA), Andalusian Health Service (SAS), University of Granada, 18071 Granada, Spain
| |
Collapse
|
41
|
Phase Compensation Technique for Effective Heat Focusing in Microwave Hyperthermia Systems. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11135972] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
In this paper, effective electromagnetic (EM) focusing achieved with a phase compensation technique for microwave hyperthermia systems is proposed. To treat tumor cells positioned deep inside a human female breast, EM energy must be properly focused on the target area. A circular antenna array for microwave hyperthermia allows EM energy to concentrate on a specific target inside the breast tumor. Depending on the cancerous cell conditions in the breast, the input phases of each antenna are calculated for single and multiple tumor cell locations. In the case of multifocal breast cancer, sub-array beam focusing via the phase compensation technique is presented to enhance the ability of EM energy to concentrate on multiple targets while minimizing damage to normal cells. To demonstrate the thermal treatment effects on single and multiple tumor locations, the accumulation of the specific absorption rate (SAR) parameter and temperature changes were verified using both simulated and experimental results.
Collapse
|
42
|
Choi SH, Lee DY, Kang S, Lee MK, Lee JH, Lee SH, Lee HL, Lee HY, Jeong YIL. Caffeic Acid Phenethyl Ester-Incorporated Radio-Sensitive Nanoparticles of Phenylboronic Acid Pinacol Ester-Conjugated Hyaluronic Acid for Application in Radioprotection. Int J Mol Sci 2021; 22:6347. [PMID: 34198522 PMCID: PMC8231778 DOI: 10.3390/ijms22126347] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 05/31/2021] [Accepted: 06/08/2021] [Indexed: 12/30/2022] Open
Abstract
We synthesized phenylboronic acid pinacol ester (PBPE)-conjugated hyaluronic acid (HA) via thiobis(ethylamine) (TbEA) linkage (abbreviated as HAsPBPE conjugates) to fabricate the radiosensitive delivery of caffeic acid phenetyl ester (CAPE) and for application in radioprotection. PBPE was primarily conjugated with TbEA and then PBPE-TbEA conjugates were conjugated again with hyaluronic acid using carbodiimide chemistry. CAPE-incorporated nanoparticles of HAsPBPE were fabricated by the nanoprecipitation method and then the organic solvent was removed by dialysis. CAPE-incorporated HAsPBPE nanoparticles have a small particle size of about 80 or 100 nm and they have a spherical shape. When CAPE-incorporated HAsPBPE nanoparticles were irradiated, nanoparticles became swelled or disintegrated and their morphologies were changed. Furthermore, the CAPE release rate from HAsPBPE nanoparticles were increased according to the radiation dose, indicating that CAPE-incorporated HAsPBPE nanoparticles have radio-sensitivity. CAPE and CAPE-incorporated HAsPBPE nanoparticles appropriately prevented radiation-induced cell death and suppressed intracellular accumulation of reactive oxygen species (ROS). CAPE and CAPE-incorporated HAsPBPE nanoparticles efficiently improved survivability of mice from radiation-induced death and reduced apoptotic cell death. We suggest that HAsPBPE nanoparticles are promising candidates for the radio-sensitive delivery of CAPE.
Collapse
Affiliation(s)
- Seon-Hee Choi
- Biomedical R&D Center, Pusan National University Yangsan Hospital, Gyeongnam 50612, Korea; (S.-H.C.); (H.-L.L.)
| | - Dong-Yeon Lee
- Department of Radiation Oncology, Dongnam Institute of Radiological & Medical Sciences, Pusan 46033, Korea;
| | - Sohi Kang
- Biomaterial R&BD Center, Chonnam National University, Gwangju 61186, Korea;
| | - Min-Kyung Lee
- Department of Dental Hygiene, Dong-Eui University, Pusan 47340, Korea;
| | - Jae-Heun Lee
- Department of Radiological Science, Dong-Eui University, Pusan 47340, Korea; (J.-H.L.); (S.-H.L.)
| | - Sang-Heon Lee
- Department of Radiological Science, Dong-Eui University, Pusan 47340, Korea; (J.-H.L.); (S.-H.L.)
| | - Hye-Lim Lee
- Biomedical R&D Center, Pusan National University Yangsan Hospital, Gyeongnam 50612, Korea; (S.-H.C.); (H.-L.L.)
| | - Hyo-Young Lee
- Department of Radiological Science, Dong-Eui University, Pusan 47340, Korea; (J.-H.L.); (S.-H.L.)
| | - Young-IL Jeong
- Biomedical R&D Center, Pusan National University Yangsan Hospital, Gyeongnam 50612, Korea; (S.-H.C.); (H.-L.L.)
| |
Collapse
|
43
|
Lu T, Haemmerich D, Liu H, Seynhaeve AL, van Rhoon GC, Houtsmuller AB, ten Hagen TL. Externally triggered smart drug delivery system encapsulating idarubicin shows superior kinetics and enhances tumoral drug uptake and response. Am J Cancer Res 2021; 11:5700-5712. [PMID: 33897876 PMCID: PMC8058728 DOI: 10.7150/thno.55163] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 02/09/2021] [Indexed: 12/20/2022] Open
Abstract
Rationale: Increasing the bioavailable drug level in a tumor is the key to enhance efficacy of chemotherapy. Thermosensitive smart drug delivery systems (SDDS) in combination with local hyperthermia facilitate high local drug levels, thus improving uptake in the tumor. However, inability to rapidly and efficiently absorb the locally released drug results in reduced efficacy, as well as undesired redistribution of the drug away from the tumor to the system. Methods: Based on this paradigm we propose a novel approach in which we replaced doxorubicin (DXR), one of the classic drugs for nanocarrier-based delivery, with idarubicin (IDA), a hydrophobic anthracycline used solely in the free form for treatment hematologic cancers. We established a series of in vitro and in vivo experiments to in depth study the kinetics of SDDS-based delivery, drug release, intratumor biodistribution and subsequent cell uptake. Results: We demonstrate that IDA is taken up over 10 times more rapidly by cancer cells than DXR in vitro. Similar trend is observed in in vivo online imaging and less drug redistribution is shown for IDA, together resulting in 4-times higher whole tumor drug uptake for IDA vs. DXR. Together his yielded an improved intratumoral drug distribution for IDA-SDDS, translating into superior tumor response compared to DXR-SDDS treatment at the same dose. Thus, IDA - a drug that is not used for treatment of solid cancers - shows superior therapeutic index and better outcome when administered in externally triggered SDDS. Conclusions: We show that a shift in selection of chemotherapeutics is urgently needed, away from the classic drugs towards selection based on properties of a chemotherapeutic in context of the nanoparticle and delivery mode, to maximize the therapeutic efficacy.
Collapse
|