1
|
Gong Z, Zhou D, Wu D, Han Y, Yu H, Shen H, Feng W, Hou L, Chen Y, Xu T. Challenges and material innovations in drug delivery to central nervous system tumors. Biomaterials 2025; 319:123180. [PMID: 39985979 DOI: 10.1016/j.biomaterials.2025.123180] [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: 06/05/2024] [Revised: 01/28/2025] [Accepted: 02/08/2025] [Indexed: 02/24/2025]
Abstract
Central nervous system (CNS) tumors, encompassing a diverse array of neoplasms in the brain and spinal cord, pose significant therapeutic challenges due to their intricate anatomy and the protective presence of the blood-brain barrier (BBB). The primary treatment obstacle is the effective delivery of therapeutics to the tumor site, which is hindered by multiple physiological, biological, and technical barriers, including the BBB. This comprehensive review highlights recent advancements in material science and nanotechnology aimed at surmounting these delivery challenges, with a focus on the development and application of nanomaterials. Nanomaterials emerge as potent tools in designing innovative drug delivery systems that demonstrate the potential to overcome the limitations posed by CNS tumors. The review delves into various strategies, including the use of lipid nanoparticles, polymeric nanoparticles, and inorganic nanoparticles, all of which are engineered to enhance drug stability, BBB penetration, and targeted tumor delivery. Additionally, this review highlights the burgeoning role of theranostic nanoparticles, integrating therapeutic and diagnostic functionalities to optimize treatment efficacy. The exploration extends to biocompatible materials like biodegradable polymers, liposomes, and advanced material-integrated delivery systems such as implantable drug-eluting devices and microfabricated devices. Despite promising preclinical results, the translation of these material-based strategies into clinical practice necessitates further research and optimization.
Collapse
Affiliation(s)
- Zhenyu Gong
- Department of Neurosurgery, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, PR China; Department of Neurosurgery, Klinikum rechts der Isar, Technical University of Munich, Munich, 81675, Germany
| | - Dairan Zhou
- Department of Neurosurgery, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, PR China
| | - Dejun Wu
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, 230601, PR China
| | - Yaguang Han
- Department of Orthopedics, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, PR China
| | - Hao Yu
- National Engineering Research Center of Ophthalmology and Optometry, School of Ophthalmology & Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, PR China
| | - Haotian Shen
- Department of Neurosurgery, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, PR China
| | - Wei Feng
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, PR China
| | - Lijun Hou
- Department of Neurosurgery, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, PR China.
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, PR China.
| | - Tao Xu
- Department of Neurosurgery, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, PR China.
| |
Collapse
|
2
|
Bhattacharjee P, Thaker AH, Patel PK, Ranade VV, Hudson SP. A vortex-based hydrodynamic cavitation manufacturing platform to generate albumin microbubbles for delivery of chemotherapies to cancerous tumours. ULTRASONICS SONOCHEMISTRY 2025; 117:107350. [PMID: 40262476 DOI: 10.1016/j.ultsonch.2025.107350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/10/2025] [Accepted: 04/10/2025] [Indexed: 04/24/2025]
Abstract
A novel approach was developed to create stable protein-based microbubbles using a vortex-driven hydrodynamic cavitation device. Such microbubbles, tiny gas-filled spheres, combined with ultrasound, can enhance drug uptake leading to inhibition of cancerous cell growth, boosting the effectiveness of anti-cancer drug molecules. The optimal conditions for the fabrication of stable bovine serum albumin (BSA) microbubbles were found to be a 15 wt% bovine serum albumin (BSA) solution at 60 °C with a pH of 6 and an ionic strength of 1.0 M. This resulted in stable BSA microbubbles with an approximate diameter of 7 μm. Curcumin-encapsulated BSA microbubbles (CBMs, 63 ± 1 μM curcumin per 101⁰ microbubbles) were created using these optimised fabrication parameters as a model system for delivering chemotherapeutic agents. The maximum percentage of curcumin release from the CBMs into phosphate buffered saline with sonication (85 %) was significantly greater than without sonication (24 %). These microbubbles were then tested to assess their effectiveness in delivering curcumin to triple-negative breast cancer cells (MDAMB-231) using a cell-to-MB ratio of 1:100, an ultrasound intensity of 0.5 W/cm2, and an ultrasound exposure time of 10 s to maximise uptake. Kinetic studies demonstrated a significant enhancement in the uptake of curcumin by MDAMB-231 cells when encapsulated into the microbubbles with ultrasound application. A substantial reduction in cellular proliferation was observed in both 2D cell culture and 3D tumour spheroid models when MDAMB-231 cells were exposed to microbubbles loaded with curcumin and ultrasound was applied. The vortex-based hydrodynamic cavitation device successfully generated curcumin loaded microbubbles with a long shelf life (120 days at 4 °C), mild preparation conditions, and enhanced uptake into cancerous tumour spheroid models. This data demonstrates the potential of this device for the commercial manufacture of drug loaded microbubble-based delivery systems.
Collapse
Affiliation(s)
| | - Abhijeet H Thaker
- Multiphase Reactors and Intensification Group Bernal Institute, University of Limerick, Limerick V94T9PX, Ireland
| | - Pratik Kumar Patel
- Department of Chemical Sciences, Bernal Institute, University of Limerick, Ireland
| | - Vivek V Ranade
- Multiphase Reactors and Intensification Group Bernal Institute, University of Limerick, Limerick V94T9PX, Ireland; SSPC, The Research Ireland Centre for Pharmaceuticals, University of Limerick, Ireland
| | - Sarah P Hudson
- Department of Chemical Sciences, Bernal Institute, University of Limerick, Ireland; SSPC, The Research Ireland Centre for Pharmaceuticals, University of Limerick, Ireland.
| |
Collapse
|
3
|
Jose AD, Chong CHN, Cheah E, Jaiswal J, Wu Z, Thakur SS. Formulation and evaluation of oxygen microbubbles stabilised in a hydrogel to potentiate radiotherapy. Int J Pharm 2025; 674:125443. [PMID: 40064385 DOI: 10.1016/j.ijpharm.2025.125443] [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: 02/02/2025] [Revised: 02/23/2025] [Accepted: 03/07/2025] [Indexed: 03/18/2025]
Abstract
BACKGROUND Tumour hypoxia poses a significant challenge in cancer treatment. There is mounting evidence that reoxygenating tumours increases their sensitivity to conventional cancer therapies. Oxygenated microbubbles (OMB) show promise for this application but suffer from poor stability and rapid clearance. Embedding OMB in a thermosensitive hydrogel (OMBHG) may prolong tumour oxygenation and improve therapeutic outcomes. OBJECTIVES To formulate and evaluate OMB loaded in a temperature sensitive hydrogel on an in vitro model of tumour hypoxia. METHODS OMB generated from a liposomal precursor were dispersed at various concentrations in a poloxamer hydrogel. OMB size, hydrogel rheology, injectability, oxygen loading/release, and impact on efficacy of radiotherapy against HCT116 colon cancer cells under hypoxia/normoxia were evaluated. RESULTS DSPC:DSPE-PEG2000 (94:6 molar ratio) liposomes dispersed in a poloxamer 407: poloxamer 188 (21:6.5 % w/w) hydrogel generated OMB predominantly sized < 1 µm. OMBHG formulations were deemed injectable (force to inject < 38 N) at 20 °C and gelled before 37 °C and demonstrated both greater oxygen loading and prolonged oxygen release than OMB alone. Cancer cells were significantly less sensitive to radiotherapy under hypoxic conditions. Pre-treatment of the cells with OMB or OMBHG enhanced radiotherapy significantly, reducing clonogenic survival rates in HCT116 cells by 78 % in hypoxic conditions and by 68 % in normoxic conditions (p < 0.0001 in both cases). Notably, this treatment restored the radiotherapy sensitivity of hypoxic cells to the levels seen with normoxic cells. CONCLUSION Reoxygenation with a newly developed OMB hydrogel formulation effectively sensitised HCT116 to radiotherapy in vitro. Ongoing studies are exploring the importance of reoxygenation rate and extent for optimal tumour sensitisation.
Collapse
Affiliation(s)
- Ashok David Jose
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Celine Hui-Ning Chong
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Ernest Cheah
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Jagdish Jaiswal
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Zimei Wu
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Sachin Sunil Thakur
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.
| |
Collapse
|
4
|
Lin BZ, Fan AC, Wang Y, Lowerison MR, Dong Z, You Q, Sekaran NVC, Llano D, Borden M, Song P. Combined Nanodrops Imaging and Ultrasound Localization Microscopy for Detecting Intracerebral Hemorrhage. ULTRASOUND IN MEDICINE & BIOLOGY 2025; 51:707-714. [PMID: 39837748 DOI: 10.1016/j.ultrasmedbio.2025.01.002] [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: 08/26/2024] [Revised: 12/25/2024] [Accepted: 01/07/2025] [Indexed: 01/23/2025]
Abstract
OBJECTIVE Advanced imaging methods are crucial for understanding stroke mechanisms and discovering effective treatments to reduce bleeding and enhance recovery. In pre-clinical in vivo stroke imaging, MRI, CT and optical imaging are commonly used to evaluate stroke outcomes in rodent models. However, MRI and CT have limited spatial resolution for rodent brains, and optical imaging is hindered by limited imaging depth of penetration. Here we introduce a novel contrast-enhanced ultrasound imaging method to overcome these challenges and characterize intracerebral hemorrhage with unique insights. METHODS We combined microbubble-based ultrasound localization microscopy (ULM) and nanodrop (ND)-based vessel leakage imaging to achieve simultaneous microvascular imaging and hemorrhage detection. ULM maps brain-wide cerebral vasculature with high spatial resolution and identifies microvascular impairments around hemorrhagic areas. NDs are sub-micron liquid-core particles that can extravasate due to blood-brain barrier breakdown, serving as positive contrast agents to detect hemorrhage sites. RESULTS Our findings demonstrate that NDs could effectively accumulate in the hemorrhagic site and reveal the location of the bleeding areas upon activation by focused ultrasound beams. ULM further reveals the microvascular damage manifested in the form of reduced vascularity and decreased blood flow velocity across areas affected by the hemorrhagic stroke. CONCLUSION The results demonstrate that sequential ULM combined with ND imaging is a useful imaging tool for basic in vivo research in stroke with rodent models where brain-wide detection of active bleeding and microvascular impairment are essential.
Collapse
Affiliation(s)
- Bing-Ze Lin
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL, USA; Department of Electrical and Computer Engineering, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | | | - Yike Wang
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL, USA; Department of Electrical and Computer Engineering, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Matthew R Lowerison
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL, USA; Department of Electrical and Computer Engineering, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Zhijie Dong
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL, USA; Department of Electrical and Computer Engineering, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Qi You
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL, USA; Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Nathiya Vaithiyalingam Chandra Sekaran
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL, USA; Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL, USA; Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Daniel Llano
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL, USA; Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL, USA; Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Mark Borden
- Department of Mechanical Engineering, University of Colorado, Boulder, CO, USA
| | - Pengfei Song
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL, USA; Department of Electrical and Computer Engineering, University of Illinois Urbana-Champaign, Urbana, IL, USA; Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL, USA; Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
5
|
Ran Q, Zhang J, Zhong J, Lin J, Zhang S, Li G, You B. Organ preservation: current limitations and optimization approaches. Front Med (Lausanne) 2025; 12:1566080. [PMID: 40206471 PMCID: PMC11980443 DOI: 10.3389/fmed.2025.1566080] [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: 01/24/2025] [Accepted: 02/28/2025] [Indexed: 04/11/2025] Open
Abstract
Despite the annual rise in patients with end-stage diseases necessitating organ transplantation, the scarcity of high-quality grafts constrains the further development of transplantation. The primary causes of the graft shortage are the scarcity of standard criteria donors, unsatisfactory organ preservation strategies, and mismatching issues. Organ preservation strategies are intimately related to pre-transplant graft viability and the incidence of adverse clinical outcomes. Static cold storage (SCS) is the current standard practice of organ preservation, characterized by its cost-effectiveness, ease of transport, and excellent clinical outcomes. However, cold-induced injury during static cold preservation, toxicity of organ preservation solution components, and post-transplantation reperfusion injury could further exacerbate graft damage. Long-term ex vivo dynamic machine perfusion (MP) preserves grafts in a near-physiological condition, evaluates graft viability, and cures damage to grafts, hence enhancing the usage and survival rates of marginal organs. With the increased use of extended criteria donors (ECD) and advancements in machine perfusion technology, static cold storage is being gradually replaced by machine perfusion. This review encapsulates the latest developments in cryopreservation, subzero non-freezing storage, static cold storage, and machine perfusion. The emphasis is on the injury mechanisms linked to static cold storage and optimization strategies, which may serve as references for the optimization of machine perfusion techniques.
Collapse
Affiliation(s)
- Qiulin Ran
- Department of Cardiovascular Surgery, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Jiayi Zhang
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Jisheng Zhong
- Department of Cardiovascular Surgery, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Ji Lin
- Department of Cardiovascular Surgery, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Shuai Zhang
- Department of Cardiovascular Surgery, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Guang Li
- Department of Cardiovascular Surgery, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Bin You
- Department of Cardiovascular Surgery, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
6
|
Chaudhary S, Kaushik A, Abeid BA, Estrada JB, Fabiilli ML, Aliabouzar M. Optical and Acoustic Characterization of Phase-Shift Droplets with Varying Shell Compositions. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025; 41:6816-6830. [PMID: 40042596 DOI: 10.1021/acs.langmuir.4c05030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Shell-stabilized, phase-shift droplets of perfluorocarbon have shown promising potential in diagnostic and therapeutic ultrasound applications. While the role of shell composition has been well-studied for conventional gas-filled microbubbles, its influence on bubbles generated via acoustic droplet vaporization (ADV) remains understudied. This study investigates the effect of shell composition─lipid, protein, and polymer─on the stability, ADV dynamics, growth behavior, release kinetics, and acoustic response of perfluorohexane phase-shift droplets. Payload-carrying, micron-sized droplets were produced using a microfluidic platform. Both optical, including ultrahigh-speed and timelapse confocal microscopy, and acoustic characterizations of the generated droplets were investigated in fibrin-based, tissue-mimicking hydrogels. Fluorescent markers were used to selectively label fibrin, payload, and droplet shells. Following insonation at 2.5 and 9.6 MHz, lipid-coated droplets had a maximum expansion ratio up to 20% lower than protein- and polymer-coated droplets. Payload release rates were an order of magnitude faster than the bubble growth rate post ADV, with minimal dependence on shell composition. Additionally, lipid shell fluorescence retained up to 40% of its initial intensity 120 s after ADV, while protein shell fluorescence was completely diminished. Acoustic characterization indicated that the ADV and inertial cavitation thresholds were similar across all shell types, suggesting that shell composition has a negligible effect on acoustic characteristics at high driving pressures.
Collapse
Affiliation(s)
- Sugandha Chaudhary
- Department of Radiology, University of Michigan, Ann Arbor, Michigan 48109-5667, United States
| | - Anuj Kaushik
- Department of Radiology, University of Michigan, Ann Arbor, Michigan 48109-5667, United States
| | - Bachir A Abeid
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan 48109-5667, United States
| | - Jonathan B Estrada
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan 48109-5667, United States
| | - Mario L Fabiilli
- Department of Radiology, University of Michigan, Ann Arbor, Michigan 48109-5667, United States
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan 48109-5667, United States
- Applied Physics Program, University of Michigan, Ann Arbor, Michigan 48109-5667, United States
| | - Mitra Aliabouzar
- Department of Radiology, University of Michigan, Ann Arbor, Michigan 48109-5667, United States
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan 48109-5667, United States
| |
Collapse
|
7
|
Sharma D, Czarnota GJ. Using ultrasound and microbubble to enhance the effects of conventional cancer therapies in clinical settings. Cancer Metastasis Rev 2025; 44:39. [PMID: 40088396 PMCID: PMC11910443 DOI: 10.1007/s10555-025-10255-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 02/20/2025] [Indexed: 03/17/2025]
Abstract
It has been demonstrated in preclinical research that the administration of microbubbles with ultrasound can augment the proapoptotic sphingolipid pathway and enhance chemotherapy or radiation therapy-induced vascular endothelial disruption resulting in enhanced tumor cell death. Specifically, ultrasound-stimulated microbubbles (USMB) can increase blood vessel permeability facilitating the release of therapeutic substances in the target area. USMB can also serve as a potential radiation enhancing therapy as USMB exposure increases tumor cell death significantly as observed in preclinical models. Clinical studies have found the combination of USMB and these existing cancer therapies to be safe and also to be associated with greater tumor responses. USMB-based treatment can be applicable in a clinical setting using either ultrasound imaging or magnetic resonance imaging (MRI) guidance for precise treatment. In the latter, the ultrasound device is integrated into the MRI system platform for sonication to facilitate microbubble stimulation. In this review, we concisely present findings related to USMB and existing cancer therapies (chemotherapy and radiation therapy) in clinical trial settings. The possible underlying mechanism involved in USMB-enhanced chemotherapy or radiotherapy enhancement is also discussed. Lastly, the study concludes with some limitations and an examination of the future direction of these combined therapies.
Collapse
Affiliation(s)
- Deepa Sharma
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada.
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada.
- Departments of Medical Biophysics, and Radiation Oncology, University of Toronto, Toronto, Ontario, Canada.
| | - Gregory J Czarnota
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada.
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada.
- Departments of Medical Biophysics, and Radiation Oncology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
8
|
Cho YN, Lim JW, Oh SJ, Han SR, Cho S, Jeong J, Han BH, Jeong JH. O 2-microbubble of iron-porphyrin conjugated polyaspartamide for molecular ultrasound contrast effect. Biotechnol Lett 2025; 47:28. [PMID: 39969614 DOI: 10.1007/s10529-025-03571-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 01/28/2025] [Accepted: 02/10/2025] [Indexed: 02/20/2025]
Abstract
OBJECTIVE This study aimed to prepare oxygen-microbubbles incorporating ferrous porphyrin to emulate the oxygen-capturing ability of hemoglobin porphyrin in red blood cells. RESULTS We synthesized poly(2-hydroxyethyl aspartamide) (PHEA) grafted with ferrous porphyrins (Iron-P-PHEA) and created microbubbles using 1,2-dipalmitoyl-sn-glycero-3-phosphocholine. These microbubbles trapped oxygen and retained it over a 2 h period. The O2-microbubbles demonstrated an enhanced photoacoustic effect as an ultrasound contrast agent, as confirmed by Doppler ultrasound testing. CONCLUSIONS The innovative strategy for O2-microbubble preparation enhances the efficiency of targeted delivery in molecular optical and ultrasound imaging.
Collapse
Affiliation(s)
- Yoon Na Cho
- Department of Chemical Engineering, Soongsil University, 369, Sangdo-Ro, Dongjak-Gu, Seoul, 06978, Republic of Korea
| | - Jun Woo Lim
- Department of Chemical Engineering, Soongsil University, 369, Sangdo-Ro, Dongjak-Gu, Seoul, 06978, Republic of Korea
| | - Seung Joo Oh
- Department of Chemical Engineering, Soongsil University, 369, Sangdo-Ro, Dongjak-Gu, Seoul, 06978, Republic of Korea
| | - Sa Ra Han
- Department of Chemical Engineering, Soongsil University, 369, Sangdo-Ro, Dongjak-Gu, Seoul, 06978, Republic of Korea
| | - Sungwoo Cho
- Department of Chemical Engineering, Soongsil University, 369, Sangdo-Ro, Dongjak-Gu, Seoul, 06978, Republic of Korea
| | - Jimin Jeong
- Department of Chemical Engineering, Soongsil University, 369, Sangdo-Ro, Dongjak-Gu, Seoul, 06978, Republic of Korea
| | - Byoung Hee Han
- Department of Radiology, GangNeung Asan Hospital, Gangneung, Ganwon-do, Republic of Korea.
| | - Jae Hyun Jeong
- Department of Chemical Engineering, Soongsil University, 369, Sangdo-Ro, Dongjak-Gu, Seoul, 06978, Republic of Korea.
| |
Collapse
|
9
|
Mezdrokhin I, Ilovitsh T. Unravelling the dynamics of coated nanobubbles and low frequency ultrasound using the Blake threshold and modified surface tension model. Phys Med Biol 2025; 70:055009. [PMID: 39919379 DOI: 10.1088/1361-6560/adb3e9] [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: 09/05/2024] [Accepted: 02/07/2025] [Indexed: 02/09/2025]
Abstract
Objective.To develop a model that accurately describes the behavior of nanobubbles (NBs) under low-frequency ultrasound (US) insonation (<250 kHz), addressing the limitations of existing numerical models, such as the Marmottant model and Blake's Threshold model, in predicting NB behavior.Approach.A modified surface tension model, derived from empirical data, was introduced to capture the surface tension behavior of NBs as a function of bubble radius. This model was integrated into the Marmottant framework and combined with the Blake threshold to predict cavitation thresholds at low pressures, providing a comprehensive approach to understanding NB dynamics.Main results.Experimentally, inertial cavitation for NBs with a radius of 85 nm was observed at peak negative pressures of 200 kPa at 80 kHz and 1000 kPa at 250 kHz. The Marmottant model significantly overestimated these thresholds (1600 kPa). The modified surface tension model improved predictions at 250 kHz, while combining it with the Blake threshold accurately aligned cavitation thresholds at both frequencies (∼150 kPa at low pressures) with experimental results.Significance.This work bridges a critical gap in understanding the acoustic behavior of NBs at low US frequencies and offers a new theoretical framework for predicting cavitation thresholds of NBs at low US frequencies, advancing their application in biomedical US technologies.
Collapse
Affiliation(s)
- Ilia Mezdrokhin
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Tali Ilovitsh
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
10
|
Kakiuchi K, Borden MA. Effect of Lipid Composition and Stirring Dynamics on Oxygen Microbubble Stability and Oxygen Release. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025; 41:1745-1755. [PMID: 39814569 DOI: 10.1021/acs.langmuir.4c04104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
Lipid-coated oxygen microbubbles (OMBs) are being investigated for biomedical applications to alleviate hypoxia such as systemic oxygenation and image-guided radiosensitization therapy. Additionally, they hold potential for boarder application as oxygen carriers beyond the biomedical filed. Understanding the stability and oxygen release properties of OMBs in dynamic aqueous environments is critical for these applications. In this study, we found that OMBs composed of longer acyl chain phospholipids (DSPC and DBPC) were stable in storage for at least 1 week, unlike the shorter acyl chain phospholipid (DPPC). OMBs were also more stable with a diacyl PEG-PE emulsifier compared with single-chain PEG-40 stearate. Dilution of OMBs did not alter the average diameter. While previous studies have examined the theoretical and experimental aspects of oxygen release from OMBs under static conditions, quantitative evaluations of OMB dispersions under dynamic stirring conditions remain limited. Here, we introduce a novel oxygen measurement method that quantitatively tracks the transition of the dissolved oxygen concentration in an aqueous medium upon mixing with a bolus of OMBs. Our results indicate that a 50 vol % OMB dispersion releases more than 330 mg/L of oxygen, surpassing arterial oxygen levels, and that more than 95% of this oxygen is released within 30 s. The rate of oxygenation of the OMB dispersions was comparable to that of a bolus injection of oxygen-saturated water under sufficient agitation, indicating that convection in the aqueous medium is the limiting transport mechanism. However, the lipid shell had a measurable effect on the oxygen release rate, which correlated with its oxygen permeability. Increasing the stirring speed increased both oxygen release rate and total amount of oxygen released. Overall, this study elucidates the fundamental stability and mass transport properties of the OMB dispersions under practical stirring conditions.
Collapse
Affiliation(s)
- Kenta Kakiuchi
- Biomedical Engineering Program, University of Colorado Boulder, Boulder, Colorado 80309, United States
| | - Mark Andrew Borden
- Biomedical Engineering Program, University of Colorado Boulder, Boulder, Colorado 80309, United States
- Mechanical Engineering Department, University of Colorado Boulder, Boulder, Colorado 80309, United States
| |
Collapse
|
11
|
Rastegar G, Kianpour B, Pathour T, Salman MM, Sirsi SR. Development of hemoglobin microbubbles for acoustic blood oxygen sensing: A study on PEGylation and gas core modification for in vivo applications. Acta Biomater 2025; 193:305-315. [PMID: 39672316 DOI: 10.1016/j.actbio.2024.12.024] [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: 07/12/2024] [Revised: 12/08/2024] [Accepted: 12/10/2024] [Indexed: 12/15/2024]
Abstract
The creation of innovative ultrasound contrast agents (UCAs) with the ability to monitor oxygen levels in real-time holds immense potential for advancing early diagnosis of various medical conditions such as hypoxic/reperfusion injury. In this study, we propose the development of oxygen sensitive UCAs using microbubbles composed of hemoglobin (HbMBs), which can function as sensors for blood oxygen levels. Previously, we performed a study highlighting the initial proof-of-concept efficacy of air-filled HbMBs in detecting oxygenation changes in vitro, offering a promising tool for clinically detecting tissue hypoxia. Nevertheless, a significant drawback of this approach is the potential for immune reactions and toxicity when hemoglobin is outside its natural red blood cell environment. Moreover, in vitro, HbMBs had low stability, with more than 90% decrease in their concentration after 120 minutes. Therefore, careful consideration of the surface properties and the gas core of HbMBs is crucial. Here, we formulated PEGylated HbMBs (PHbMBs), and investigated their stability, immunogenicity, and their acoustic response in oxygenated and deoxygenated media in vitro. We optimized PEGylated HbMBs (PHbMBs), showing a 42% reduction in immunogenicity and significantly improved stability in vitro, while maintaining their oxygen-binding and acoustic response. In vivo, PHbMBs demonstrated similar contrast enhancement to that of non-PEGylated MBs, demonstrating that PEGylation does not decrease HbMBs' acoustic signaling. Finally, changing the gas core from air to PFB increased PHbMBs' mean circulation time more than 11-fold, without diminishing their responsiveness to oxygen. Overall, the proposed oxygen sensitive PHbMBs offer a promising avenue for real-time acoustic detection of blood oxygen levels, paving the way for potential clinical applications in monitoring critically ill patients. STATEMENT OF SIGNIFICANCE: This research explores the emergent field of Acoustic Oxygen Imaging in vivo using hemoglobin-based microbubbles. This innovative contrast agent approach involves imaging using crosslinked biomaterial comprised of the hemoglobin protein, aiming to transform the way we monitor blood oxygen levels with ultrasound. This work fundamentally addresses central concerns of improving bubble stability and circulation life for eventual clinical use, while minimizing toxicity. Importantly, we demonstrate that PEGylation of hemoglobin microbubbles enhances their stability, reduces immunogenicity, and maintains acoustic responsiveness. The incorporation of perfluorobutane into the bubble core increases the longevity of these microbubbles in circulation, while sustaining their oxygen sensitivity. Favorable in vivo results highlight the potential of this technology in real-time acoustic detection of blood oxygen levels.
Collapse
Affiliation(s)
- Ghazal Rastegar
- Department of Bioengineering, Erik Johnson School of Engineering, The University of Texas at Dallas
| | - Bahareh Kianpour
- Department of Bioengineering, Erik Johnson School of Engineering, The University of Texas at Dallas
| | - Teja Pathour
- Department of Bioengineering, Erik Johnson School of Engineering, The University of Texas at Dallas
| | - Mohammad Musa Salman
- Department of Bioengineering, Erik Johnson School of Engineering, The University of Texas at Dallas
| | - Shashank R Sirsi
- Department of Bioengineering, Erik Johnson School of Engineering, The University of Texas at Dallas.
| |
Collapse
|
12
|
Lee MY, Lee D, Choi D, Kim KS, Kang PM. Targeting Reactive Oxygen Species for Diagnosis of Various Diseases. J Funct Biomater 2024; 15:378. [PMID: 39728178 DOI: 10.3390/jfb15120378] [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: 11/04/2024] [Revised: 12/12/2024] [Accepted: 12/12/2024] [Indexed: 12/28/2024] Open
Abstract
Reactive oxygen species (ROS) are generated predominantly during cellular respiration and play a significant role in signaling within the cell and between cells. However, excessive accumulation of ROS can lead to cellular dysfunction, disease progression, and apoptosis that can lead to organ dysfunction. To overcome the short half-life of ROS and the relatively small amount produced, various imaging methods have been developed, using both endogenous and exogenous means to monitor ROS in disease settings. In this review, we discuss the molecular mechanisms underlying ROS production and explore the methods and materials that could be used to detect ROS overproduction, including iron-based materials, ROS-responsive chemical bond containing polymers, and ROS-responsive molecule containing biomaterials. We also discuss various imaging and imaging techniques that could be used to target and detect ROS overproduction. We discuss the ROS imaging potentials of established clinical imaging methods, such as magnetic resonance imaging (MRI), sonographic imaging, and fluorescence imaging. ROS imaging potentials of other imaging methods, such as photoacoustic imaging (PAI) and Raman imaging (RI) that are currently in preclinical stage are also discussed. Finally, this paper focuses on various diseases that are associated with ROS overproduction, and the current and the future clinical applications of ROS-targeted imaging. While the most widely used clinical condition is cardiovascular diseases, its potential extends into non-cardiovascular clinical conditions, such as neurovascular, neurodegenerative, and other ROS-associated conditions, such as cancers, skin aging, acute kidney injury, and inflammatory arthritis.
Collapse
Affiliation(s)
- Moung Young Lee
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
- Centers for Research in ICT based Infectious Diseases, Jeonbuk National University, Jeonju 561-756, Republic of Korea
| | - Donguk Lee
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
- Massachusetts College of Pharmacy and Health Sciences, Boston, MA 02115, USA
| | - Dayun Choi
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
- Massachusetts College of Pharmacy and Health Sciences, Boston, MA 02115, USA
| | - Kye S Kim
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Peter M Kang
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
13
|
Harpster SL, Piñeiro AM, Wong JY. Methods for Rapid Characterization of Tunable Microbubble Formulations. Bioengineering (Basel) 2024; 11:1224. [PMID: 39768042 PMCID: PMC11673760 DOI: 10.3390/bioengineering11121224] [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: 10/28/2024] [Revised: 11/15/2024] [Accepted: 11/25/2024] [Indexed: 01/11/2025] Open
Abstract
To optimize microbubble formulations for clinical applications, the size distribution, concentration, and acoustic intensity must be rapidly measurable to allow for the successful iteration of microbubble design. In this paper, a comprehensive method was developed to compare microbubble formulations with different lipid shell compositions using optical and acoustic methods of measurement to collect the size distribution, concentration, and mean scattering intensity. An open-source ImageJ macro code was modified for the selective counting and sizing of brightfield microbubble images. A high-throughput agarose phantom was designed to collect multiple scattering reflections of microbubble samples to estimate the echogenicity of each microbubble solution. The information contained in the size distribution and concentration, combined with the instantaneous scattering power, can identify modifications needed for prototyping specific microbubble formulations.
Collapse
Affiliation(s)
- Savannah L. Harpster
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; (S.L.H.)
| | - Alexandra M. Piñeiro
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; (S.L.H.)
| | - Joyce Y. Wong
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; (S.L.H.)
- Division of Materials Science & Engineering, Boston University, Boston, MA 02215, USA
| |
Collapse
|
14
|
Gunjkar S, Gupta U, Nair R, Paul P, Aalhate M, Mahajan S, Maji I, Chourasia MK, Guru SK, Singh PK. The Neoteric Paradigm of Biomolecule-Functionalized Albumin-Based Targeted Cancer Therapeutics. AAPS PharmSciTech 2024; 25:265. [PMID: 39500822 DOI: 10.1208/s12249-024-02977-6] [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: 07/31/2024] [Accepted: 10/17/2024] [Indexed: 12/12/2024] Open
Abstract
Albumin is a nature-derived, versatile protein carrier, that has been explored extensively by researchers for anticancer drug delivery due to its role in enhancing drug stability, solubility, circulation time, targeting capabilities, and overall therapeutic efficacy. Albumin nanoparticles possess inherent biocompatibility, biodegradability, and passive tumor-targeting ability due to the enhanced permeability and retention effect. However, non-specific accumulation of cytotoxic agents in healthy tissues remains a challenge. In this paper, the functionalization of albumin nanoparticles using various biomolecules including antibodies, nucleic acids, proteins and peptides, vitamins, chondroitin sulfate, hyaluronic acid, and lactobionic acid have been discussed which enables specific recognition and binding to cancer cells. Furthermore, we highlight the supremacy of such a targeted approach in tumor-specific drug delivery, minimization of off-target effects, potential improvement in therapeutic efficacy, cellular internalization, reduced side effects, and better clinical outcomes. This review centers on how they have revolutionized the field of biomedical research and tuned into an excellent targeted approach. In conclusion, this review highlights in detail the role of albumin as a nanocarrier for tumor-targeted delivery using biomolecules as ligands.
Collapse
Affiliation(s)
- Swati Gunjkar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, Telangana, India
| | - Ujala Gupta
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, Telangana, India
| | - Rahul Nair
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, Telangana, India
| | - Priti Paul
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, Telangana, India
| | - Mayur Aalhate
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, Telangana, India
| | - Srushti Mahajan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, Telangana, India
| | - Indrani Maji
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, Telangana, India
| | - Manish K Chourasia
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, 226031, U.P., India
| | - Santosh Kumar Guru
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, Telangana, India
| | - Pankaj Kumar Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, Telangana, India.
| |
Collapse
|
15
|
Wegierak D, Nittayacharn P, Cooley MB, Berg FM, Kosmides T, Durig D, Kolios MC, Exner AA. Nanobubble Contrast Enhanced Ultrasound Imaging: A Review. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e2007. [PMID: 39511794 PMCID: PMC11567054 DOI: 10.1002/wnan.2007] [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: 02/09/2024] [Revised: 08/07/2024] [Accepted: 09/26/2024] [Indexed: 11/15/2024]
Abstract
Contrast-enhanced ultrasound is currently used worldwide with clinical indications in cardiology and radiology, and it continues to evolve and develop through innovative technological advancements. Clinically utilized contrast agents for ultrasound consist of hydrophobic gas microbubbles stabilized with a biocompatible shell. These agents are used commonly in echocardiography, with emerging applications in cancer diagnosis and therapy. Microbubbles are a blood pool agent with diameters between 1 and 10 μm, which precludes their use in other extravascular applications. To expand the potential use of contrast-enhanced ultrasound beyond intravascular applications, sub-micron agents, often called nanobubbles or ultra-fine bubbles, have recently emerged as a promising tool. Combining the principles of ultrasound imaging with the unique properties of nanobubbles (high concentration and small size), recent work has established their imaging potential. Contrast-enhanced ultrasound imaging using these agents continues to gain traction, with new studies establishing novel imaging applications. We highlight the recent achievements in nonlinear nanobubble contrast imaging, including a discussion on nanobubble formulations and their acoustic characteristics. Ultrasound imaging with nanobubbles is still in its early stages, but it has shown great potential in preclinical research and animal studies. We highlight unexplored areas of research where the capabilities of nanobubbles may offer new advantages. As technology advances, this technique may find applications in various areas of medicine, including cancer detection and treatment, cardiovascular imaging, and drug delivery.
Collapse
Affiliation(s)
- Dana Wegierak
- Department of Biomedical EngineeringCase Western Reserve University (CWRU)ClevelandOhioUSA
| | - Pinunta Nittayacharn
- Department of RadiologyCWRUClevelandOhioUSA
- Department of Biomedical Engineering, Faculty of EngineeringMahidol UniversityPuttamonthonNakorn PathomThailand
| | - Michaela B. Cooley
- Department of Biomedical EngineeringCase Western Reserve University (CWRU)ClevelandOhioUSA
| | - Felipe M. Berg
- Department of RadiologyCWRUClevelandOhioUSA
- Hospital Israelita Albert EinsteinSão PauloSão PauloBrazil
| | - Theresa Kosmides
- Department of Biomedical EngineeringCase Western Reserve University (CWRU)ClevelandOhioUSA
| | - Dorian Durig
- Department of Biomedical EngineeringCase Western Reserve University (CWRU)ClevelandOhioUSA
| | - Michael C. Kolios
- Department of PhysicsToronto Metropolitan UniversityTorontoOntarioCanada
- Institute for Biomedical Engineering, Science and Technology (iBEST), a Partnership Between St. Michael's Hospital, a Site of Unity Health Toronto and Toronto Metropolitan UniversityTorontoOntarioCanada
| | - Agata A. Exner
- Department of Biomedical EngineeringCase Western Reserve University (CWRU)ClevelandOhioUSA
- Department of RadiologyCWRUClevelandOhioUSA
| |
Collapse
|
16
|
Jiang Y, Harberts J, Assadi A, Chen Y, Spatz JP, Duan W, Nisbet DR, Voelcker NH, Elnathan R. The Roles of Micro- and Nanoscale Materials in Cell-Engineering Systems. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2410908. [PMID: 39401098 DOI: 10.1002/adma.202410908] [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: 07/26/2024] [Revised: 09/13/2024] [Indexed: 11/29/2024]
Abstract
Customizable manufacturing of ex vivo cell engineering is driven by the need for innovations in the biomedical field and holds substantial potential for addressing current therapeutic challenges; but it is still only in its infancy. Micro- and nanoscale-engineered materials are increasingly used to control core cell-level functions in cellular engineering. By reprogramming or redirecting targeted cells for extremely precise functions, these advanced materials offer new possibilities. This influences the modularity of cell reprogramming and reengineering, making these materials part of versatile and emerging technologies. Here, the roles of micro- and nanoscale materials in cell engineering are highlighted, demonstrating how they can be adaptively controlled to regulate cellular reprogramming and core cell-level functions, including differentiation, proliferation, adhesion, user-defined gene expression, and epigenetic changes. The current reprogramming routes used to achieve pluripotency from somatic cells and the significant potential of induced pluripotent stem cell technology for translational biomedical research are covered. Recent advances in nonviral intracellular delivery modalities for cell reprogramming and their constraints are evaluated. This paper focuses on emerging physical and combinatorial approaches of intracellular delivery for cell engineering, revealing the capabilities and limitations of these routes. It is showcased how these programmable materials are continually being explored as customizable tools for inducing biophysical stimulation. Harnessing the power of micro- and nanoscale-engineered materials will be a step change in the design of cell engineering, producing a suite of powerful tools for addressing potential future challenges in therapeutic cell engineering.
Collapse
Affiliation(s)
- Yuan Jiang
- Faculty of Health, School of Medicine, Deakin University, Waurn Ponds, Victoria, 3216, Australia
- The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Waurn Ponds, Victoria, 3216, Australia
| | - Jann Harberts
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Victoria, Clayton, 3168, Australia
| | - Artin Assadi
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Victoria, Clayton, 3168, Australia
| | - Yaping Chen
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
- Oujiang Laboratory, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Zhejiang, 325000, China
| | - Joachim P Spatz
- Department of Cellular Biophysics, Max Planck Institute for Medical Research, 69120, Heidelberg, Germany
- Institute for Molecular Systems Engineering (IMSE), Heidelberg University, 69120, Heidelberg, Germany
- Max Planck School Matter to Life, Max Planck Schools, 69120, Heidelberg, Germany
| | - Wei Duan
- Faculty of Health, School of Medicine, Deakin University, Waurn Ponds, Victoria, 3216, Australia
| | - David R Nisbet
- The Graeme Clark Institute, University of Melbourne, Parkville, Victoria, 3010, Australia
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Melbourne, Parkville, Victoria, 3010, Australia
- Medical School, Faculty of Medicine, Dentistry and Health Science, The University of Melbourne, Melbourne, Parkville, VIC, 3010, Australia
| | - Nicolas H Voelcker
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Victoria, Clayton, 3168, Australia
| | - Roey Elnathan
- Faculty of Health, School of Medicine, Deakin University, Waurn Ponds, Victoria, 3216, Australia
- The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Waurn Ponds, Victoria, 3216, Australia
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Victoria, Clayton, 3168, Australia
- Institute for Frontier Materials, Deakin University, Waurn Ponds, Victoria, 3216, Australia
| |
Collapse
|
17
|
Sayed Tabatabaei M, Sayed Tabatabaei FA, Moghimi HR. Drug self-delivery systems: A comprehensive review on small molecule nanodrugs. BIOIMPACTS : BI 2024; 15:30161. [PMID: 40161942 PMCID: PMC11954755 DOI: 10.34172/bi.30161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/11/2023] [Accepted: 12/12/2023] [Indexed: 04/02/2025]
Abstract
Drug self-delivery systems are nanostructures composed of a drug as the main structural unit, having the ability of intracellular trafficking with no additional carrier. In these systems, the drug itself undertakes the functional and structural roles; thereby, the ancillary role of excipients and carrier-related limitations are circumvented and therapeutic effect is achieved at a much lower dose. Such advantages -which are mainly but not exclusively beneficial in cancer treatment- have recently led to an upsurge of research on these systems. Subsequently, various terminologies were utilized to describe them, referring to the same concept with different words. However, not all the systems developed based on the self-delivery approach are introduced using one of these keywords. Using a scoping strategy, this review aims to encompass the systems that have been developed as yet -inspired by the concept of self-delivery- and classify them in a coherent taxonomy. Two main groups are introduced based on the type of building blocks: small molecule-based nanomedicines and self-assembling hybrid prodrugs. Due to the diversity, covering the whole gamut of topics is beyond the scope of a single article, and, inevitably, the latter is just briefly introduced here, whereas the features of the former group are meticulously presented. Depending on whether the drug is merely a carrier for itself or carries a second drug as cargo, two classes of small molecule-based nanomedicines are defined (i.e., pure nanodrugs and carrier-mimicking systems, respectively), each having sub-branches. After introducing each branch and giving some examples, possible strategies for designing each particular system are visually displayed. The resultant mind map can create a macro view of the taken path and its prospects, give a profound insight into opportunities, spark new ideas, and facilitate overcoming obstacles. Taken together, one can foresee a brilliant future for self-delivery systems as a pioneering candidate for the next generation of drug delivery systems.
Collapse
Affiliation(s)
- Mahsa Sayed Tabatabaei
- Department of Pharmaceutics and Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Hamid Reza Moghimi
- Department of Pharmaceutics and Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Protein Technology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
18
|
Patel PB, Latt S, Ravi K, Razavi M. Clinical Applications of Micro/Nanobubble Technology in Neurological Diseases. Biomimetics (Basel) 2024; 9:645. [PMID: 39451851 PMCID: PMC11506587 DOI: 10.3390/biomimetics9100645] [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: 09/04/2024] [Revised: 10/11/2024] [Accepted: 10/18/2024] [Indexed: 10/26/2024] Open
Abstract
Nanomedicine, leveraging the unique properties of nanoparticles, has revolutionized the diagnosis and treatment of neurological diseases. Among various nanotechnological advancements, ultrasound-mediated drug delivery using micro- and nanobubbles offers promising solutions to overcome the blood-brain barrier (BBB), enhancing the precision and efficacy of therapeutic interventions. This review explores the principles, current clinical applications, challenges, and future directions of ultrasound-mediated drug delivery systems in treating stroke, brain tumors, neurodegenerative diseases, and neuroinflammatory disorders. Additionally, ongoing clinical trials and potential advancements in this field are discussed, providing a comprehensive overview of the impact of nanomedicine on neurological diseases.
Collapse
Affiliation(s)
- Parth B. Patel
- University of Central Florida College of Medicine, Orlando, FL 32827, USA; (P.B.P.); (K.R.)
| | - Sun Latt
- Biionix (Bionic Materials, Implants & Interfaces) Cluster, Department of Medicine, University of Central Florida College of Medicine, Orlando, FL 32827, USA;
- Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, Orlando, FL 32827, USA
| | - Karan Ravi
- University of Central Florida College of Medicine, Orlando, FL 32827, USA; (P.B.P.); (K.R.)
| | - Mehdi Razavi
- Biionix (Bionic Materials, Implants & Interfaces) Cluster, Department of Medicine, University of Central Florida College of Medicine, Orlando, FL 32827, USA;
- Department of Material Sciences and Engineering, University of Central Florida, Orlando, FL 32816, USA
- Biomedical Engineering Program, Department of Mechanical and Aerospace Engineering, University of Central Florida, Orlando, FL 32816, USA
| |
Collapse
|
19
|
Singh A, Reynolds JNJ. Therapeutic ultrasound: an innovative approach for targeting neurological disorders affecting the basal ganglia. Front Neuroanat 2024; 18:1469250. [PMID: 39417047 PMCID: PMC11480080 DOI: 10.3389/fnana.2024.1469250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 09/17/2024] [Indexed: 10/19/2024] Open
Abstract
The basal ganglia are involved in motor control and action selection, and their impairment manifests in movement disorders such as Parkinson's disease (PD) and dystonia, among others. The complex neuronal circuitry of the basal ganglia is located deep inside the brain and presents significant treatment challenges. Conventional treatment strategies, such as invasive surgeries and medications, may have limited effectiveness and may result in considerable side effects. Non-invasive ultrasound (US) treatment approaches are becoming increasingly recognized for their therapeutic potential for reversibly permeabilizing the blood-brain barrier (BBB), targeting therapeutic delivery deep into the brain, and neuromodulation. Studies conducted on animals and early clinical trials using ultrasound as a therapeutic modality have demonstrated promising outcomes for controlling symptom severity while preserving neural tissue. These results could improve the quality of life for patients living with basal ganglia impairments. This review article explores the therapeutic frontiers of ultrasound technology, describing the brain mechanisms that are triggered and engaged by ultrasound. We demonstrate that this cutting-edge method could transform the way neurological disorders associated with the basal ganglia are managed, opening the door to less invasive and more effective treatments.
Collapse
Affiliation(s)
| | - John N. J. Reynolds
- Translational Brain Plasticity Laboratory, Department of Anatomy, School of Biomedical Sciences, and the Brain Health Research Center, University of Otago, Dunedin, New Zealand
| |
Collapse
|
20
|
Moosavifar M, Barmin RA, Rama E, Rix A, Gumerov RA, Lisson T, Bastard C, Rütten S, Avraham‐Radermacher N, Koehler J, Pohl M, Kulkarni V, Baier J, Koletnik S, Zhang R, Dasgupta A, Motta A, Weiler M, Potemkin II, Schmitz G, Kiessling F, Lammers T, Pallares RM. Polymeric Microbubble Shell Engineering: Microporosity as a Key Factor to Enhance Ultrasound Imaging and Drug Delivery Performance. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404385. [PMID: 39207095 PMCID: PMC11516050 DOI: 10.1002/advs.202404385] [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: 04/25/2024] [Revised: 07/15/2024] [Indexed: 09/04/2024]
Abstract
Microbubbles (MB) are widely used as contrast agents for ultrasound (US) imaging and US-enhanced drug delivery. Polymeric MB are highly suitable for these applications because of their acoustic responsiveness, high drug loading capability, and ease of surface functionalization. While many studies have focused on using polymeric MB for diagnostic and therapeutic purposes, relatively little attention has thus far been paid to improving their inherent imaging and drug delivery features. This study here shows that manipulating the polymer chemistry of poly(butyl cyanoacrylate) (PBCA) MB via temporarily mixing the monomer with the monomer-mimetic butyl cyanoacetate (BCC) during the polymerization process improves the drug loading capacity of PBCA MB by more than twofold, and the in vitro and in vivo acoustic responses of PBCA MB by more than tenfold. Computer simulations and physisorption experiments show that BCC manipulates the growth of PBCA polymer chains and creates nanocavities in the MB shell, endowing PBCA MB with greater drug entrapment capability and stronger acoustic properties. Notably, because BCC can be readily and completely removed during MB purification, the resulting formulation does not include any residual reagent beyond the ones already present in current PBCA-based MB products, facilitating the potential translation of next-generation PBCA MB.
Collapse
Affiliation(s)
- Mirjavad Moosavifar
- Institute for Experimental Molecular ImagingRWTH Aachen University Hospital52074AachenGermany
| | - Roman A. Barmin
- Institute for Experimental Molecular ImagingRWTH Aachen University Hospital52074AachenGermany
| | - Elena Rama
- Institute for Experimental Molecular ImagingRWTH Aachen University Hospital52074AachenGermany
| | - Anne Rix
- Institute for Experimental Molecular ImagingRWTH Aachen University Hospital52074AachenGermany
| | - Rustam A. Gumerov
- DWI – Leibniz Institute for Interactive MaterialsRWTH Aachen University52074AachenGermany
| | - Thomas Lisson
- Chair for Medical EngineeringRuhr University Bochum44780BochumGermany
| | - Céline Bastard
- DWI – Leibniz Institute for Interactive MaterialsRWTH Aachen University52074AachenGermany
| | - Stephan Rütten
- Electron Microscope FacilityRWTH Aachen University Hospital52074AachenGermany
| | - Noah Avraham‐Radermacher
- Institute of Technical and Macromolecular ChemistryRWTH Aachen University Hospital52074AachenGermany
| | - Jens Koehler
- DWI – Leibniz Institute for Interactive MaterialsRWTH Aachen University52074AachenGermany
| | - Michael Pohl
- DWI – Leibniz Institute for Interactive MaterialsRWTH Aachen University52074AachenGermany
| | - Vedangi Kulkarni
- Institute for Experimental Molecular ImagingRWTH Aachen University Hospital52074AachenGermany
| | - Jasmin Baier
- Institute for Experimental Molecular ImagingRWTH Aachen University Hospital52074AachenGermany
| | - Susanne Koletnik
- Institute for Experimental Molecular ImagingRWTH Aachen University Hospital52074AachenGermany
| | - Rui Zhang
- Institute for Experimental Molecular ImagingRWTH Aachen University Hospital52074AachenGermany
| | - Anshuman Dasgupta
- Institute for Experimental Molecular ImagingRWTH Aachen University Hospital52074AachenGermany
| | - Alessandro Motta
- Institute for Experimental Molecular ImagingRWTH Aachen University Hospital52074AachenGermany
| | - Marek Weiler
- Institute for Experimental Molecular ImagingRWTH Aachen University Hospital52074AachenGermany
| | - Igor I. Potemkin
- DWI – Leibniz Institute for Interactive MaterialsRWTH Aachen University52074AachenGermany
| | - Georg Schmitz
- Chair for Medical EngineeringRuhr University Bochum44780BochumGermany
| | - Fabian Kiessling
- Institute for Experimental Molecular ImagingRWTH Aachen University Hospital52074AachenGermany
| | - Twan Lammers
- Institute for Experimental Molecular ImagingRWTH Aachen University Hospital52074AachenGermany
| | - Roger M. Pallares
- Institute for Experimental Molecular ImagingRWTH Aachen University Hospital52074AachenGermany
| |
Collapse
|
21
|
Kalayeh K, Fowlkes JB, Yeras S, Chen A, Daignault-Newton S, Schultz WW, Sack BS. A Comparative Study of Commercially Available Ultrasound Contrast Agents for Sub-harmonic-Aided Pressure Estimation (SHAPE) in a Bladder Phantom. ULTRASOUND IN MEDICINE & BIOLOGY 2024; 50:1494-1505. [PMID: 39054243 DOI: 10.1016/j.ultrasmedbio.2024.05.019] [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: 04/12/2024] [Revised: 05/14/2024] [Accepted: 05/19/2024] [Indexed: 07/27/2024]
Abstract
OBJECTIVE The goal of this study was to evaluate the performance of different commercial ultrasound contrast microbubbles (MBs) when measuring bladder phantom pressure with sub-harmonic-aided pressure estimation (SHAPE) methodology. We hypothesized that SHAPE performance is dependent on MB formulation. This study aimed to advance the SHAPE application for bladder pressure measurements in humans. METHODS Using a previously designed and built bladder phantom, we tested four different commercial agents: Definity, Lumason, Sonazoid and Optison. A standard clinical cystometrogram (CMG) system was used to infuse a MB-saline mixture into the bladder phantom to measure pressure. Ultrasound imaging was performed using the GE Healthcare LOGIQ E10 scanner. RESULTS All agents showed a predicted inverse linear relationship between change in pressure and SHAPE signal. However, they differ from each other in terms of stability, linear correlation, sensitivity to pressure and error. Generally, Definity and Lumason showed the highest performance during the SHAPE-based bladder phantom pressure assessments. CONCLUSION Our results show that the SHAPE signal decreases as bladder phantom pressures increases, regardless of the agent or CMG phase, suggesting the possibility of using SHAPE for measuring bladder pressure without a catheter. However, the efficacy of SHAPE in measuring pressure varies by MB formulation. These observations support using Lumason and Definity in a human subject feasibility study as we advance toward a catheter-free solution for measuring voiding bladder pressure via SHAPE.
Collapse
Affiliation(s)
- Kourosh Kalayeh
- Department of Urology, University of Michigan, Ann Arbor, MI, USA
| | - J Brian Fowlkes
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Sophia Yeras
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI, USA
| | - Amy Chen
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | | | - William W Schultz
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Bryan S Sack
- Department of Urology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
22
|
Rouhi N, Chakeri Z, Ghorbani Nejad B, Rahimzadegan M, Rafi Khezri M, Kamali H, Nosrati R. A comprehensive review of advanced focused ultrasound (FUS) microbubbles-mediated treatment of Alzheimer's disease. Heliyon 2024; 10:e37533. [PMID: 39309880 PMCID: PMC11416559 DOI: 10.1016/j.heliyon.2024.e37533] [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: 11/18/2023] [Revised: 08/27/2024] [Accepted: 09/04/2024] [Indexed: 09/25/2024] Open
Abstract
Alzheimer's disease (AD) is characterized by progressive neurodegeneration, memory loss, and cognitive impairment leading to dementia and death. The blood-brain barrier (BBB) prevents the delivery of drugs into the brain, which can limit their therapeutic potential in the treatment of AD. Therefore, there is a need to develop new approaches to bypass the BBB for appropriate treatment of AD. Recently, focused ultrasound (FUS) has been shown to disrupt the BBB, allowing therapeutic agents to penetrate the brain. In addition, microbubbles (MBs) as lipophilic carriers can penetrate across the BBB and deliver the active drug into the brain tissue. Therefore, combined with FUS, the drug-encapsulated MBs can pass through the ultrasound-disrupted zone of the BBB and diffuse into the brain tissue. This review provides clear and concise statements on the recent advances of the various FUS-mediated MBs-based carriers developed for delivering AD-related drugs. In addition, the sonogenetics-based FUS/MBs approaches for the treatment of AD are highlighted. The future perspectives and challenges of ultrasound-based MBs drug delivery in AD are then discussed.
Collapse
Affiliation(s)
- Nadiyeh Rouhi
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Zahra Chakeri
- Cardiothoracic Imaging Section, Department of Radiology, University of Washington, Seattle, WA, USA
| | - Behnam Ghorbani Nejad
- Department of Toxicology, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | - Milad Rahimzadegan
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Hossein Kamali
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Rahim Nosrati
- Cellular and Molecular Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
23
|
McCorkell G, Piva T, Highgate D, Nakayama M, Geso M. Ultrasound-stimulated microbubbles to enhance radiotherapy: A scoping review. J Med Imaging Radiat Oncol 2024; 68:740-769. [PMID: 39250692 DOI: 10.1111/1754-9485.13740] [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: 02/24/2024] [Accepted: 07/22/2024] [Indexed: 09/11/2024]
Abstract
INTRODUCTION Primarily used as ultrasound contrast agents, microbubbles have recently emerged as a versatile therapeutic vector that can be 'burst' to deliver payloads in the presence of suitably optimised ultrasound fields. Ultrasound-stimulated microbubbles (USMB) have recently demonstrated improvements in treatment outcomes across a variety of clinical applications. This scoping review investigates whether this potential translates into the context of radiation therapy by evaluating the application of this technology across all three phases of radiation action. METHODS Primary research articles, excluding poster presentations and conference proceedings, were identified through systematic searches of the PubMed NCBI/Medline, Embase/OVID, Web of Science and CINAHL/EBSCOhost databases, with additional articles identified via manual Google Scholar searching. Articles were dual screened for inclusion using the Covidence systematic review platform and classified against all three phases of radiation action. RESULTS Overall, 57 eligible publications from a total of 1389 identified articles were included in the review, with studies dating back to 2012. Study heterogeneity prevented formal statistical analysis; however, most articles reported improved outcomes using USMB in the presence of radiation compared to that of radiation alone. These improvements appear to result from the use of USMB as either a biovascular disruptor causing tumour cell damage via indirect mechanisms, or as a localised treatment vector that directly increases tumour cell uptake of other therapeutic and physical agents designed to enhance radiation action. CONCLUSIONS USMB demonstrate exciting potential to enhance the effects of radiation treatments due to their versatility and capacity to target all three phases of radiation action.
Collapse
Affiliation(s)
- Giulia McCorkell
- RMIT University, Melbourne, Victoria, Australia
- The University of Melbourne, Melbourne, Victoria, Australia
| | | | | | - Masao Nakayama
- RMIT University, Melbourne, Victoria, Australia
- Kobe University, Kobe, Hyogo, Japan
- Kita-Harima Medical Center, Ono, Hyogo, Japan
| | - Moshi Geso
- RMIT University, Melbourne, Victoria, Australia
| |
Collapse
|
24
|
Alkhudhairy F. Canal disinfection using Nd: YAG Laser, synchronized microbubble-photodynamic activation, and carbon quantum dots on microhardness, smear layer removal, and extrusion bond strength of zirconia post to canal dentin. An invitro scanning electron microscopic analysis. Microsc Res Tech 2024; 87:2043-2052. [PMID: 38646819 DOI: 10.1002/jemt.24584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/16/2024] [Accepted: 04/11/2024] [Indexed: 04/23/2024]
Abstract
Evaluation of the impact of the latest root canal disinfectant, that is carbon quantum dots (CQDs), synchronized microbubble-photodynamic activation (SYMPA), and Nd: YAG laser along with ethylenediaminetetraacetic acid (EDTA) as a final irrigant on the Marten hardness (MH), smear layer (SL) removal, and extrusion bond strength (EBS) of zirconia post to the canal dentin. Eighty intact single-rooted premolars were obtained and disinfected using 0.5% chloramine-T solution. Root canal preparation was performed using ProTaper files followed by obturation. The post space was prepared for prefabricated zirconia post and all the teeth were randomly divided into four groups based on the disinfection used (n = 20 each) Group 1: 5.25% NaOCl + 17% EDTA (Control), Group 2: Nd: YAG laser + 17% EDTA, Group 3: SYMPA + 17% EDTA, and Group 4: CQDs + 17% EDTA. MH, SL removal, and EBS of zirconia post-bonded to root dentin were performed using a microhardness tester, scanning electron microscope (SEM), and universal testing machine, respectively. Both intragroup and intergroup comparisons were performed using one-way analysis of variance (ANOVA) and posthoc-Tukey test for significant difference (p < .05). Group 2 samples (Nd: YAG laser + 17% EDTA) (0.24 ± 0.06 GPa) exhibited highest values of MH. Samples in group 3 (SYMPA + 17% EDTA) treated teeth unveiled the lowest MH scores (0.13 ± 0.02 GPa). Moreover, the coronal third of Group 3 specimens (SYMPA and 17% EDTA) (1.54 ± 0.31) eliminated SL from the canal with the greatest efficacy as well as presented the highest EBS (10.13 ± 0.69 MPa). However, the apical third of Group 1 samples (5.25% NaOCl + 17% EDTA) (2.95 ± 0.33) exhibited the least efficient elimination of SL from the radicular dentin as well as the lowest bond strength (5.11 ± 0.19 MPa) of zirconia post to the dentin. The SYMPA technique with 17% EDTA proved highly effective in removing the SL from canal dentin and enhancing the EBS of zirconia posts. The least preferable method for SL removal and MH improvement was found to be 5.25% NaOCl + 17% EDTA. CQDs and Nd: YAG laser demonstrated satisfactory smear layer removal properties from the canal, along with achieving appropriate bond strength of zirconia posts. RESEARCH HIGHLIGHTS: Nd: YAG laser and 17% EDTA as canal disinfectant exhibited the highest values of MH. Specimens irrigated with SYMPA and 17% EDTA eliminated SL from the canal with the greatest efficacy. The coronal third of Group 3 (SYMPA + 17% EDTA) samples unveiled the highest zirconia post-bond integrity score to the canal dentin. Cohesive failure was a dominant failure type among different experimental groups.
Collapse
Affiliation(s)
- Fahad Alkhudhairy
- Restorative Dental Sciences Department, College of Dentistry, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
25
|
Rehman R, Lu W, Shi L, Yang Y, Li P. The effect of pre-treatments on atrazine removal from source water by microbubble ozonation. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024; 31:55145-55157. [PMID: 39222228 DOI: 10.1007/s11356-024-34829-y] [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: 01/08/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
Ozone-based advanced oxidation processes (AOPs) have emerged a promising avenue for water treatment, offering effective removal of micropollutants. Recent research underscores the potential of ozone microbubbles to enhance ozone mass transfer during water treatment, particularly when combined with pre-treatment steps. This study aimed to evaluate the efficacy of three different combined processes (chlorine/KMnO4/PAC pre-treatment followed by ozonation) in removing atrazine, a common micropollutant from natural source water. Results revealed that all combined processes achieved higher atrazine removal rates compared to individual pre-treatment or ozonation methods. Notably, the highest atrazine removal rates were observed under alkaline pH conditions, with treatment outcomes influenced by oxidant dose and pH levels. Among the combined processes, chlorine pre-treatment followed by ozonation emerged as the most effective approach, achieving a removal rate of 59.7% that exceeded the sum of individual treatments. However, this treatment efficacy was affected by water quality parameters, particularly the presence of organic matter and elevated ammonia nitrogen concentration (> 0.5 mg/L). This study highlights the potential for utilizing ozone micro/nanobubbles to enhance ozone mass transfer and offers valuable insights for optimizing the combined application of pre-treatment and ozonation strategies for efficient atrazine removal from natural water sources.
Collapse
Affiliation(s)
- Ratul Rehman
- School of Environmental Science and Engineering, Tongji University, 1239 Siping Road, Shanghai, P.R. China
| | - Wanmeng Lu
- School of Civil Engineering, Lanzhou University of Technology, 287 Langongping Road, Lanzhou City, Gansu, P.R. China
| | - Lifang Shi
- School of Environmental Science and Engineering, Tongji University, 1239 Siping Road, Shanghai, P.R. China
| | - Yahong Yang
- School of Civil Engineering, Lanzhou University of Technology, 287 Langongping Road, Lanzhou City, Gansu, P.R. China
| | - Pan Li
- School of Environmental Science and Engineering, Tongji University, 1239 Siping Road, Shanghai, P.R. China.
- UNEP-Institute of Environment and Sustainable Development (IESD), Tongji University, Shanghai, China.
| |
Collapse
|
26
|
Duan X, Wang P, He L, He Z, Wang S, Yang F, Gao C, Ren W, Lin J, Chen T, Xu C, Li J, Wu A. Peptide-Functionalized Inorganic Oxide Nanomaterials for Solid Cancer Imaging and Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2311548. [PMID: 38333964 DOI: 10.1002/adma.202311548] [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: 11/01/2023] [Revised: 01/15/2024] [Indexed: 02/10/2024]
Abstract
The diagnosis and treatment of solid tumors have undergone significant advancements marked by a trend toward increased specificity and integration of imaging and therapeutic functions. The multifaceted nature of inorganic oxide nanomaterials (IONs), which boast optical, magnetic, ultrasonic, and biochemical modulatory properties, makes them ideal building blocks for developing multifunctional nanoplatforms. A promising class of materials that have emerged in this context are peptide-functionalized inorganic oxide nanomaterials (PFIONs), which have demonstrated excellent performance in multifunctional imaging and therapy, making them potential candidates for advancing solid tumor diagnosis and treatment. Owing to the functionalities of peptides in tumor targeting, penetration, responsiveness, and therapy, well-designed PFIONs can specifically accumulate and release therapeutic or imaging agents at the solid tumor sites, enabling precise imaging and effective treatment. This review provides an overview of the recent advances in the use of PFIONs for the imaging and treatment of solid tumors, highlighting the superiority of imaging and therapeutic integration as well as synergistic treatment. Moreover, the review discusses the challenges and prospects of PFIONs in depth, aiming to promote the intersection of the interdisciplinary to facilitate their clinical translation and the development of personalized diagnostic and therapeutic systems by optimizing the material systems.
Collapse
Affiliation(s)
- Xiaolin Duan
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Cixi Institute of Biomedical Engineering, Cixi, 315300, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Pin Wang
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Cixi Institute of Biomedical Engineering, Cixi, 315300, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lulu He
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Cixi Institute of Biomedical Engineering, Cixi, 315300, China
| | - Zhen He
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Cixi Institute of Biomedical Engineering, Cixi, 315300, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shiwei Wang
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Cixi Institute of Biomedical Engineering, Cixi, 315300, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Fang Yang
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Cixi Institute of Biomedical Engineering, Cixi, 315300, China
| | - Changyong Gao
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Cixi Institute of Biomedical Engineering, Cixi, 315300, China
| | - Wenzhi Ren
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Cixi Institute of Biomedical Engineering, Cixi, 315300, China
| | - Jie Lin
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Cixi Institute of Biomedical Engineering, Cixi, 315300, China
| | - Tianxiang Chen
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Cixi Institute of Biomedical Engineering, Cixi, 315300, China
| | - Chen Xu
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Cixi Institute of Biomedical Engineering, Cixi, 315300, China
| | - Juan Li
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Cixi Institute of Biomedical Engineering, Cixi, 315300, China
| | - Aiguo Wu
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Cixi Institute of Biomedical Engineering, Cixi, 315300, China
| |
Collapse
|
27
|
Snipstad S, Sulheim E, Åslund AKO, Hyldbakk A, Wågbø AM, Klinkenberg G, Mørch Y. Nanoparticle-loaded microbubbles for treatment of lung cancer. Eur J Pharm Sci 2024; 199:106804. [PMID: 38763448 DOI: 10.1016/j.ejps.2024.106804] [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: 10/05/2023] [Revised: 03/20/2024] [Accepted: 05/16/2024] [Indexed: 05/21/2024]
Abstract
Lung cancer is one of the most common cancers and a leading cause of death, with poor prognosis and high unmet clinical need. Chemotherapy is a common part of the treatment, either alone or in combination with other treatment modalities, but with limited efficacy and severe side effects. Encapsulation of drugs into nanoparticles can enable a more targeted delivery with reduced off-target toxicity. Delivery to the lungs is however often insufficient due to various biological barriers in the body and in the tumor microenvironment. Here we demonstrate that by incorporating drug-loaded nanoparticles into air-filled microbubbles, a more effective targeting to the lungs can be achieved. Fluorescence imaging and mass spectrometry revealed that the microbubbles could significantly improve accumulation of drug in the lungs of mice, compared to injecting either the free drug by itself or only the drug-loaded nanoparticles. Therapeutic efficacy was verified in a preclinical mouse model with non-small cell lung cancer, monitoring tumor growth by luminescence.
Collapse
Affiliation(s)
- Sofie Snipstad
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway; Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway; Cancer Clinic, St. Olavs Hospital, Trondheim, Norway.
| | - Einar Sulheim
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway; Institute for Research in Biomedicine, Bellinzona, Switzerland
| | - Andreas K O Åslund
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | - Astrid Hyldbakk
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway; Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Ane Marit Wågbø
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | - Geir Klinkenberg
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | - Yrr Mørch
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway; NaDeNo Nanoscience AS, Trondheim, Norway
| |
Collapse
|
28
|
Ya J, Zhang H, Qin G, Huang C, Zhao C, Ren J, Qu X. A Biocompatible Hydrogen-Bonded Organic Framework (HOF) as Sonosensitizer and Artificial Enzyme for In-Depth Treatment of Alzheimer's Disease. Adv Healthc Mater 2024:e2402342. [PMID: 39031538 DOI: 10.1002/adhm.202402342] [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: 06/26/2024] [Revised: 07/09/2024] [Indexed: 07/22/2024]
Abstract
Current phototherapeutic approaches for Alzheimer's disease (AD) exhibit restricted clinical outcomes due to the limited physical penetration and comprised brain microenvironment of noninvasive nanomedicine. Herein, a hydrogen-bonded organic framework (HOF) based sonosensitizer is designed and synthesized. Mn-TCPP, a planar molecule where Mn2+ ion is chelated in the core with a large p-conjugated system and 4 carboxylate acid groups, has been successfully used as building blocks to construct an ultrasound-sensitive HOF (USI-MHOF), which can go deep in the brain of AD animal models. The both in vitro and in vivo studies indicate that USI-MHOF can generate singlet oxygen (1O2) and oxidize β-amyloid (Aβ) to inhibit aggregation, consequently attenuating Aβ neurotoxicity. More intriguingly, USI-MHOF exhibits catalase (CAT)- and superoxide dismutase (SOD)-like activities, mitigating neuron oxidative stress and reprograming the brain microenvironment. For better crossing the blood-brain barrier (BBB), the peptide KLVFFAED (KD8) has been covalently grafted to USI-MHOF for improving BBB permeability and Aβ selectivity. Further, in vivo experiments demonstrate a significant reduction of the craniocerebral Aβ plaques and improvement of the cognition deficits in triple-transgenic AD (3×Tg-AD) mice models following deep-penetration ultrasound treatment. The work provides the first example of an ultrasound-responsive biocompatible HOF as non-invasive nanomedicine for in-depth treatment of AD.
Collapse
Affiliation(s)
- Junlin Ya
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230029, P. R. China
| | - Haochen Zhang
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230029, P. R. China
| | - Geng Qin
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230029, P. R. China
| | - Congcong Huang
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230029, P. R. China
| | - Chuanqi Zhao
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230029, P. R. China
| | - Jinsong Ren
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230029, P. R. China
| | - Xiaogang Qu
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230029, P. R. China
| |
Collapse
|
29
|
Huang H, Zheng Y, Chang M, Song J, Xia L, Wu C, Jia W, Ren H, Feng W, Chen Y. Ultrasound-Based Micro-/Nanosystems for Biomedical Applications. Chem Rev 2024; 124:8307-8472. [PMID: 38924776 DOI: 10.1021/acs.chemrev.4c00009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2024]
Abstract
Due to the intrinsic non-invasive nature, cost-effectiveness, high safety, and real-time capabilities, besides diagnostic imaging, ultrasound as a typical mechanical wave has been extensively developed as a physical tool for versatile biomedical applications. Especially, the prosperity of nanotechnology and nanomedicine invigorates the landscape of ultrasound-based medicine. The unprecedented surge in research enthusiasm and dedicated efforts have led to a mass of multifunctional micro-/nanosystems being applied in ultrasound biomedicine, facilitating precise diagnosis, effective treatment, and personalized theranostics. The effective deployment of versatile ultrasound-based micro-/nanosystems in biomedical applications is rooted in a profound understanding of the relationship among composition, structure, property, bioactivity, application, and performance. In this comprehensive review, we elaborate on the general principles regarding the design, synthesis, functionalization, and optimization of ultrasound-based micro-/nanosystems for abundant biomedical applications. In particular, recent advancements in ultrasound-based micro-/nanosystems for diagnostic imaging are meticulously summarized. Furthermore, we systematically elucidate state-of-the-art studies concerning recent progress in ultrasound-based micro-/nanosystems for therapeutic applications targeting various pathological abnormalities including cancer, bacterial infection, brain diseases, cardiovascular diseases, and metabolic diseases. Finally, we conclude and provide an outlook on this research field with an in-depth discussion of the challenges faced and future developments for further extensive clinical translation and application.
Collapse
Affiliation(s)
- Hui Huang
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, P. R. China
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Yi Zheng
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P. R. China
| | - Meiqi Chang
- Laboratory Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P. R. China
| | - Jun Song
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Lili Xia
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Chenyao Wu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Wencong Jia
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Hongze Ren
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Wei Feng
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, P. R. China
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Yu Chen
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, P. R. China
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| |
Collapse
|
30
|
Shen Q, Li Z, Wang Y, Meyer MD, De Guzman MT, Lim JC, Xiao H, Bouchard RR, Lu GJ. 50-nm Gas-Filled Protein Nanostructures to Enable the Access of Lymphatic Cells by Ultrasound Technologies. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2307123. [PMID: 38533973 PMCID: PMC11550859 DOI: 10.1002/adma.202307123] [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: 07/18/2023] [Revised: 03/14/2024] [Indexed: 03/28/2024]
Abstract
Ultrasound imaging and ultrasound-mediated gene and drug delivery are rapidly advancing diagnostic and therapeutic methods; however, their use is often limited by the need for microbubbles, which cannot transverse many biological barriers due to their large size. Here, the authors introduce 50-nm gas-filled protein nanostructures derived from genetically engineered gas vesicles(GVs) that are referred to as 50 nmGVs. These diamond-shaped nanostructures have hydrodynamic diameters smaller than commercially available 50-nm gold nanoparticles and are, to the authors' knowledge, the smallest stable, free-floating bubbles made to date. 50 nmGVs can be produced in bacteria, purified through centrifugation, and remain stable for months. Interstitially injected 50 nmGVs can extravasate into lymphatic tissues and gain access to critical immune cell populations, and electron microscopy images of lymph node tissues reveal their subcellular location in antigen-presenting cells adjacent to lymphocytes. The authors anticipate that 50 nmGVs can substantially broaden the range of cells accessible to current ultrasound technologies and may generate applications beyond biomedicine as ultrasmall stable gas-filled nanomaterials.
Collapse
Affiliation(s)
- Qionghua Shen
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA
| | - Zongru Li
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA
| | - Yixian Wang
- Department of Chemistry, Rice University, Houston, TX, 77005, USA
| | - Matthew D Meyer
- Shared Equipment Authority, Rice University, Houston, TX, 77005, USA
| | - Marc T De Guzman
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA
| | - Janie C Lim
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA
| | - Han Xiao
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA
- Department of Chemistry, Rice University, Houston, TX, 77005, USA
- SynthX Center, Rice University, Houston, TX, 77005, USA
| | - Richard R Bouchard
- Department of Imaging Physics, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - George J Lu
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA
- Department of BioSciences, Rice University, Houston, TX, 77005, USA
- Rice Synthetic Biology Institute, Rice University, Houston, TX, 77005, USA
| |
Collapse
|
31
|
Jayasankar G, Koilpillai J, Narayanasamy D. A Systematic Study on Long-acting Nanobubbles: Current Advancement and Prospects on Theranostic Properties. Adv Pharm Bull 2024; 14:278-301. [PMID: 39206408 PMCID: PMC11347731 DOI: 10.34172/apb.2024.042] [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: 08/23/2023] [Revised: 03/16/2024] [Accepted: 03/17/2024] [Indexed: 09/04/2024] Open
Abstract
Delivery of diagnostic drugs via nanobubbles (NBs) has shown to be an emerging field of study. Due to their small size, NBs may more easily travel through constricted blood vessels and precisely target certain bodily parts. NB is considered the major treatment for cancer treatment and other diseases which are difficult to diagnose. The field of NBs is dynamic and continues to grow as researchers discover new properties and seek practical applications in various fields. The predominant usage of NBs in novel drug delivery is to enhance the bioavailability, and controlled drug release along with imaging properties NBs are important because they may change interfacial characteristics including surface force, lubrication, and absorption. The quick diffusion of gas into the water was caused by a hypothetical film that was stimulated and punctured by a strong acting force at the gas/water contact of the bubble. In this article, various prominent aspects of NBs have been discussed, along with the long-acting nature, and the theranostical aspect which elucidates the potential marketed drugs along with clinical trial products. The article also covers quality by design aspects, different production techniques that enable method-specific therapeutic applications, increasing the floating time of the bubble, and refining its properties to enhance the prepared NB's quality. NB containing both analysis and curing properties makes it special from other nano-carriers. This work includes all the possible methods of preparing NB, its application, all marketed drugs, and products in clinical trials.
Collapse
Affiliation(s)
| | | | - Damodharan Narayanasamy
- Department of Pharmaceutics, SRM College of Pharmacy, SRM Institution of Science and Technology, Kattankulathur, Chengalpattu, India
| |
Collapse
|
32
|
Prasher P, Sharma M, Agarwal V, Singh SK, Gupta G, Dureja H, Dua K. Cationic cycloamylose based nucleic acid nanocarriers. Chem Biol Interact 2024; 395:111000. [PMID: 38614318 DOI: 10.1016/j.cbi.2024.111000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 04/02/2024] [Accepted: 04/07/2024] [Indexed: 04/15/2024]
Abstract
Nucleic acid delivery by viral and non-viral methods has been a cornerstone for the contemporary gene therapy aimed at correcting the defective genes, replacing of the missing genes, or downregulating the expression of anomalous genes is highly desirable for the management of various diseases. Ostensibly, it becomes paramount for the delivery vectors to intersect the biological barriers for accessing their destined site within the cellular environment. However, the lipophilic nature of biological membranes and their potential to limit the entry of large sized, charged, hydrophilic molecules thus presenting a sizeable challenge for the cellular integration of negatively charged nucleic acids. Furthermore, the susceptibility of nucleic acids towards the degrading enzymes (nucleases) in the lysosomes present in cytoplasm is another matter of concern for their cellular and nuclear delivery. Hence, there is a pressing need for the identification and development of cationic delivery systems which encapsulate the cargo nucleic acids where the charge facilitates their cellular entry by evading the membrane barriers, and the encapsulation shields them from the enzymatic attack in cytoplasm. Cycloamylose bearing a closed loop conformation presents a robust candidature in this regard owing to its remarkable encapsulating tendency towards nucleic acids including siRNA, CpG DNA, and siRNA. The presence of numerous hydroxyl groups on the cycloamylose periphery provides sites for its chemical modification for the introduction of cationic groups, including spermine, (3-Chloro-2 hydroxypropyl) trimethylammonium chloride (Q188), and diethyl aminoethane (DEAE). The resulting cationic cycloamylose possesses a remarkable transfection efficiency and provides stability to cargo oligonucleotides against endonucleases, in addition to modulating the undesirable side effects such as unwanted immune stimulation. Cycloamylose is known to interact with the cell membranes where they release certain membrane components such as phospholipids and cholesterol thereby resulting in membrane destabilization and permeabilization. Furthermore, cycloamylose derivatives also serve as formulation excipients for improving the efficiency of other gene delivery systems. This review delves into the various vector and non-vector-based gene delivery systems, their advantages, and limitations, eventually leading to the identification of cycloamylose as an ideal candidate for nucleic acid delivery. The synthesis of cationic cycloamylose is briefly discussed in each section followed by its application for specific delivery/transfection of a particular nucleic acid.
Collapse
Affiliation(s)
- Parteek Prasher
- Department of Chemistry, University of Petroleum & Energy Studies, Energy Acres, Dehradun, 248007, India.
| | - Mousmee Sharma
- Department of Chemistry, Uttaranchal University, Dehradun, 248007, India
| | - Vipul Agarwal
- Cluster for Advanced Macromolecular Design (CAMD), School of Chemical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India; Faculty of Health, Australian Research Center in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia; School of Medical and Life Sciences, Sunway University, 47500 Sunway City, Malaysia
| | - Gaurav Gupta
- School of Pharmacy, Graphic Era Hill University, Dehradun, 248007, India; Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman 346, United Arab Emirates
| | - Harish Dureja
- Department of Pharmaceutical Sciences, Maharishi Dayanand University, Rohtak, 124001, India
| | - Kamal Dua
- Faculty of Health, Australian Research Center in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia.
| |
Collapse
|
33
|
Pan X, Huang W, Nie G, Wang C, Wang H. Ultrasound-Sensitive Intelligent Nanosystems: A Promising Strategy for the Treatment of Neurological Diseases. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2303180. [PMID: 37871967 DOI: 10.1002/adma.202303180] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 09/26/2023] [Indexed: 10/25/2023]
Abstract
Neurological diseases are a major global health challenge, affecting hundreds of millions of people worldwide. Ultrasound therapy plays an irreplaceable role in the treatment of neurological diseases due to its noninvasive, highly focused, and strong tissue penetration capabilities. However, the complexity of brain and nervous system and the safety risks associated with prolonged exposure to ultrasound therapy severely limit the applicability of ultrasound therapy. Ultrasound-sensitive intelligent nanosystems (USINs) are a novel therapeutic strategy for neurological diseases that bring greater spatiotemporal controllability and improve safety to overcome these challenges. This review provides a detailed overview of therapeutic strategies and clinical advances of ultrasound in neurological diseases, focusing on the potential of USINs-based ultrasound in the treatment of neurological diseases. Based on the physical and chemical effects induced by ultrasound, rational design of USINs is a prerequisite for improving the efficacy of ultrasound therapy. Recent developments of ultrasound-sensitive nanocarriers and nanoagents are systemically reviewed. Finally, the challenges and developing prospects of USINs are discussed in depth, with a view to providing useful insights and guidance for efficient ultrasound treatment of neurological diseases.
Collapse
Affiliation(s)
- Xueting Pan
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Wenping Huang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Changyong Wang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Beijing, 100850, China
| | - Hai Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
34
|
Gusliakova OI, Kurochkin MA, Barmin RA, Prikhozhdenko ES, Estifeeva TM, Rudakovskaya PG, Sindeeva OA, Galushka VV, Vavaev ES, Komlev AS, Lyubin EV, Fedyanin AA, Dey KK, Gorin DA. Magnetically navigated microbubbles coated with albumin/polyarginine and superparamagnetic iron oxide nanoparticles. BIOMATERIALS ADVANCES 2024; 158:213759. [PMID: 38227987 DOI: 10.1016/j.bioadv.2024.213759] [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: 09/22/2023] [Revised: 12/31/2023] [Accepted: 01/01/2024] [Indexed: 01/18/2024]
Abstract
While microbubbles (MB) are routinely used for ultrasound (US) imaging, magnetic MB are increasingly explored as they can be guided to specific sites of interest by applied magnetic field gradient. This requires the MB shell composition tuning to prolong MB stability and provide functionalization capabilities with magnetic nanoparticles. Hence, we developed air-filled MB stabilized by a protein-polymer complex of bovine serum albumin (BSA) and poly-L-arginine (pArg) of different molecular weights, showing that pArg of moderate molecular weight distribution (15-70 kDa) enabled MB with greater stability and acoustic response while preserving MB narrow diameters and the relative viability of THP-1 cells after 48 h of incubation. After MB functionalization with superparamagnetic iron oxide nanoparticles (SPION), magnetic moment values provided by single MB confirmed the sufficient SPION deposition onto BSA + pArg MB shells. During MB magnetic navigation in a blood vessel mimicking phantom with magnetic tweezers and in a Petri dish with adherent mouse renal carcinoma cell line, we demonstrated the effectiveness of magnetic MB localization in the desired area by magnetic field gradient. Magnetic MB co-localization with cells was further exploited for effective doxorubicin delivery with drug-loaded MB. Taken together, these findings open new avenues in control over albumin MB properties and magnetic navigation of SPION-loaded MB, which can envisage their applications in diagnostic and therapeutic needs.
Collapse
Affiliation(s)
- Olga I Gusliakova
- Science Medical Center, Saratov State University, Saratov 410012, Russia; Vladimir Zelman Center for Neurobiology and Brain Rehabilitation, Skolkovo Institute of Science and Technology, Moscow 121205, Russia.
| | - Maxim A Kurochkin
- Center for Photonic Science and Engineering, Skolkovo Institute of Science and Technology, Moscow 121205, Russia
| | - Roman A Barmin
- Center for Photonic Science and Engineering, Skolkovo Institute of Science and Technology, Moscow 121205, Russia
| | | | - Tatyana M Estifeeva
- Center for Photonic Science and Engineering, Skolkovo Institute of Science and Technology, Moscow 121205, Russia
| | - Polina G Rudakovskaya
- Center for Photonic Science and Engineering, Skolkovo Institute of Science and Technology, Moscow 121205, Russia
| | - Olga A Sindeeva
- Vladimir Zelman Center for Neurobiology and Brain Rehabilitation, Skolkovo Institute of Science and Technology, Moscow 121205, Russia
| | - Victor V Galushka
- Education and Research Institute of Nanostructures and Biosystems, Saratov State University, Saratov 410012, Russia
| | - Evgeny S Vavaev
- Faculty of Physics, Lomonosov Moscow State University, Moscow 119991, Russia
| | - Aleksei S Komlev
- Faculty of Physics, Lomonosov Moscow State University, Moscow 119991, Russia
| | - Evgeny V Lyubin
- Faculty of Physics, Lomonosov Moscow State University, Moscow 119991, Russia
| | - Andrey A Fedyanin
- Faculty of Physics, Lomonosov Moscow State University, Moscow 119991, Russia
| | - Krishna Kanti Dey
- Department of Physics, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382055, India
| | - Dmitry A Gorin
- Center for Photonic Science and Engineering, Skolkovo Institute of Science and Technology, Moscow 121205, Russia.
| |
Collapse
|
35
|
Liu J, Wang C, Qiu S, Sun W, Yang G, Yuan L. Toward Ultrasound Molecular Imaging of Endothelial Dysfunction in Diabetes: Targets, Strategies, and Challenges. ACS APPLIED BIO MATERIALS 2024; 7:1416-1428. [PMID: 38391247 DOI: 10.1021/acsabm.4c00053] [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: 02/24/2024]
Abstract
Diabetes vasculopathy is a significant complication of diabetes mellitus (DM), and early identification and timely intervention can effectively slow the progression. Accumulating studies have shown that diabetes causes vascular complications directly or indirectly through a variety of mechanisms. Direct imaging of the endothelial molecular changes not only identifies the early stage of diabetes vasculopathy but also sheds light on the precise treatment. Targeted ultrasound contrast agent (UCA)-based ultrasound molecular imaging (UMI) can noninvasively detect the expression status of molecular biomarkers overexpressed in the vasculature, thereby being a potential strategy for the diagnosis and treatment response evaluation of DM. Amounts of efforts have been focused on identification of the molecular targets expressed in the vasculature, manufacturing strategies of the targeted UCA, and the clinical translation for the diagnosis and evaluation of therapeutic efficacy in both micro- and macrovasculopathy in DM. This review summarizes the latest research progress on endothelium-targeted UCA and discusses their promising future and challenges in diabetes vasculopathy theranostics.
Collapse
Affiliation(s)
- Jiahan Liu
- Department of Ultrasound Medicine, Tangdu Hospital, Fourth Military Medical University, Shaanxi 710038, China
| | - Chen Wang
- Department of Ultrasound Medicine, Tangdu Hospital, Fourth Military Medical University, Shaanxi 710038, China
| | - Shuo Qiu
- Department of Ultrasound Medicine, Tangdu Hospital, Fourth Military Medical University, Shaanxi 710038, China
| | - Wenqi Sun
- Department of Ultrasound Medicine, Tangdu Hospital, Fourth Military Medical University, Shaanxi 710038, China
| | - Guodong Yang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular Biology, Fourth Military Medical University Xi'an, Shaanxi 710032, China
| | - Lijun Yuan
- Department of Ultrasound Medicine, Tangdu Hospital, Fourth Military Medical University, Shaanxi 710038, China
| |
Collapse
|
36
|
Dhara P, Shah N, Sundaram V, Srivastava A, Solovev AA, Mei Y, Gorin DA, Dey KK. Influence of protein nativity on the stability of bovine serum albumin coated microbubbles. iScience 2024; 27:109286. [PMID: 38482489 PMCID: PMC10933459 DOI: 10.1016/j.isci.2024.109286] [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: 10/03/2023] [Revised: 01/31/2024] [Accepted: 02/16/2024] [Indexed: 11/02/2024] Open
Abstract
Protein-coated microbubbles have become one of the emerging platforms in biomedical research as theranostic agents. In recent years, microbubbles have been extensively used as ultrasound contrast agents and carriers of molecular cargoes, pertaining to which several studies have focused on tuning the properties of these bubbles to achieve a higher degree of biocompatibility and extended stability. Synthesis of microbubbles has so far been traditionally carried out with pre-heated proteins like bovine serum albumin (BSA) as shell coatings, owing to the ease in making BSA crosslinked structures through disulfide bridge formation. We, however, have performed experiments to demonstrate that air core microbubbles formed with native BSA are more stable compared with those formed using denatured BSA. The experimental observations have been supported with analytical modeling and computational studies, which offer insights into the effect of BSA conformation in stabilizing the microbubbles shells and prolonging their lifetimes.
Collapse
Affiliation(s)
- Palash Dhara
- Laboratory of Soft and Living Materials, Department of Physics, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar, Gujarat 382055, India
| | - Niyati Shah
- Laboratory of Soft and Living Materials, Department of Physics, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar, Gujarat 382055, India
| | - Vidya Sundaram
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar, Gujarat 382055, India
| | - Ashutosh Srivastava
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar, Gujarat 382055, India
| | | | - Yongfeng Mei
- Department of Materials Science, Fudan University, Shanghai 200433 P.R. China
| | - Dmitry A. Gorin
- Center for Photonics Sciences and Engineering, Skolkovo Institute of Science and Technology, 3 Nobelya Str., 121205 Moscow, Russia
| | - Krishna Kanti Dey
- Laboratory of Soft and Living Materials, Department of Physics, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar, Gujarat 382055, India
| |
Collapse
|
37
|
Qin B, Chen X, Zhu J, Kopechek J, Helfield B, Yu F, Cyriac J, Lavery L, Grandis JR, Villanueva FS. Ultrasound enhanced siRNA delivery using cationic liposome-microbubble complexes for the treatment of squamous cell carcinoma. Nanotheranostics 2024; 8:285-297. [PMID: 38577322 PMCID: PMC10988211 DOI: 10.7150/ntno.90516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 02/20/2024] [Indexed: 04/06/2024] Open
Abstract
Rationale: Microbubble (MB) contrast agents combined with ultrasound targeted microbubble cavitation (UTMC) are a promising platform for site-specific therapeutic oligonucleotide delivery. We investigated UTMC-mediated delivery of siRNA directed against epidermal growth factor receptor (EGFR), to squamous cell carcinoma (SCC) via a novel MB-liposome complex (LPX). Methods: LPXs were constructed by conjugation of cationic liposomes to the surface of C4F10 gas-filled lipid MBs using biotin/avidin chemistry, then loaded with siRNA via electrostatic interaction. Luciferase-expressing SCC-VII cells (SCC-VII-Luc) were cultured in Petri dishes. The Petri dishes were filled with media in which LPXs loaded with siRNA against firefly luciferase (Luc siRNA) were suspended. Ultrasound (US) (1 MHz, 100-µs pulse, 10% duty cycle) was delivered to the dishes for 10 sec at varying acoustic pressures and luciferase assay was performed 24 hr later. In vivo siRNA delivery was studied in SCC-VII tumor-bearing mice intravenously infused with a 0.5 mL saline suspension of EGFR siRNA LPX (7×108 LPX, ~30 µg siRNA) for 20 min during concurrent US (1 MHz, 0.5 MPa spatial peak temporal peak negative pressure, five 100-µs pulses every 1 ms; each pulse train repeated every 2 sec to allow reperfusion of LPX into the tumor). Mice were sacrificed 2 days post treatment and tumor EGFR expression was measured (Western blot). Other mice (n=23) received either EGFR siRNA-loaded LPX + UTMC or negative control (NC) siRNA-loaded LPX + UTMC on days 0 and 3, or no treatment ("sham"). Tumor volume was serially measured by high-resolution 3D US imaging. Results: Luc siRNA LPX + UTMC caused significant luciferase knockdown vs. no treatment control, p<0.05) in SCC-VII-Luc cells at acoustic pressures 0.25 MPa to 0.9 MPa, while no significant silencing effect was seen at lower pressure (0.125 MPa). In vivo, EGFR siRNA LPX + UTMC reduced tumor EGFR expression by ~30% and significantly inhibited tumor growth by day 9 (~40% decrease in tumor volume vs. NC siRNA LPX + UTMC, p<0.05). Conclusions: Luc siRNA LPXs + UTMC achieved functional delivery of Luc siRNA to SCC-VII-Luc cells in vitro. EGFR siRNA LPX + UTMC inhibited tumor growth and suppressed EGFR expression in vivo, suggesting that this platform holds promise for non-invasive, image-guided targeted delivery of therapeutic siRNA for cancer treatment.
Collapse
Affiliation(s)
- Bin Qin
- Center for Ultrasound for Molecular Imaging and Therapeutics, University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xucai Chen
- Center for Ultrasound for Molecular Imaging and Therapeutics, University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jianhui Zhu
- Center for Ultrasound for Molecular Imaging and Therapeutics, University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jonathan Kopechek
- Center for Ultrasound for Molecular Imaging and Therapeutics, University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Brandon Helfield
- Center for Ultrasound for Molecular Imaging and Therapeutics, University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Francois Yu
- Center for Ultrasound for Molecular Imaging and Therapeutics, University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jissy Cyriac
- Center for Ultrasound for Molecular Imaging and Therapeutics, University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Linda Lavery
- Center for Ultrasound for Molecular Imaging and Therapeutics, University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jennifer R. Grandis
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco, CA, USA
| | - Flordeliza S. Villanueva
- Center for Ultrasound for Molecular Imaging and Therapeutics, University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
38
|
Nittayacharn P, Abenojar E, Cooley MB, Berg FM, Counil C, Sojahrood AJ, Khan MS, Yang C, Berndl E, Golczak M, Kolios MC, Exner AA. Efficient ultrasound-mediated drug delivery to orthotopic liver tumors - Direct comparison of doxorubicin-loaded nanobubbles and microbubbles. J Control Release 2024; 367:135-147. [PMID: 38237687 PMCID: PMC11700473 DOI: 10.1016/j.jconrel.2024.01.028] [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: 09/14/2023] [Revised: 01/11/2024] [Accepted: 01/15/2024] [Indexed: 01/28/2024]
Abstract
Liver metastasis is a major obstacle in treating aggressive cancers, and current therapeutic options often prove insufficient. To overcome these challenges, there has been growing interest in ultrasound-mediated drug delivery using lipid-shelled microbubbles (MBs) and nanobubbles (NBs) as promising strategies for enhancing drug delivery to tumors. Our previous work demonstrated the potential of Doxorubicin-loaded C3F8 NBs (hDox-NB, 280 ± 123 nm) in improving cancer treatment in vitro using low-frequency unfocused therapeutic ultrasound (TUS). In this study, we investigated the pharmacokinetics and biodistribution of sonicated hDox-NBs in orthotopic rat liver tumors. We compared their delivery and therapeutic efficiency with size-isolated MBs (hDox-MB, 1104 ± 373 nm) made from identical shell material and core gas. Results showed a similar accumulation of hDox in tumors treated with hDox-MBs and unfocused therapeutic ultrasound (hDox-MB + TUS) and hDox-NB + TUS. However, significantly increased apoptotic cell death in the tumor and fewer off-target apoptotic cells in the normal liver were found upon the treatment with hDox-NB + TUS. The tumor-to-liver apoptotic ratio was elevated 9.4-fold following treatment with hDox-NB + TUS compared to hDox-MB + TUS, suggesting that the therapeutic efficacy and specificity are significantly increased when using hDox-NB + TUS. These findings highlight the potential of this approach as a viable treatment modality for liver tumors. By elucidating the behavior of drug-loaded bubbles in vivo, we aim to contribute to developing more effective liver cancer treatments that could ultimately improve patient outcomes and decrease off-target side effects.
Collapse
Affiliation(s)
- Pinunta Nittayacharn
- Department of Radiology, Case Western Reserve University, Cleveland, OH, USA; Department of Biomedical Engineering, Faculty of Engineering, Mahidol University, Puttamonthon, Nakorn Pathom, Thailand
| | - Eric Abenojar
- Department of Radiology, Case Western Reserve University, Cleveland, OH, USA
| | - Michaela B Cooley
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Felipe M Berg
- Department of Radiology, Case Western Reserve University, Cleveland, OH, USA; Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | - Claire Counil
- Department of Radiology, Case Western Reserve University, Cleveland, OH, USA
| | - Amin Jafari Sojahrood
- Department of Physics, Toronto Metropolitan University, Toronto, Canada; Institute for Biomedical Engineering, Science and Technology (iBEST), a partnership between St. Michael's Hospital, a site of Unity Health Toronto and Toronto Metropolitan University, Toronto, Canada
| | - Muhammad Saad Khan
- Department of Physics, Toronto Metropolitan University, Toronto, Canada; Institute for Biomedical Engineering, Science and Technology (iBEST), a partnership between St. Michael's Hospital, a site of Unity Health Toronto and Toronto Metropolitan University, Toronto, Canada
| | - Celina Yang
- Department of Physics, Toronto Metropolitan University, Toronto, Canada; Institute for Biomedical Engineering, Science and Technology (iBEST), a partnership between St. Michael's Hospital, a site of Unity Health Toronto and Toronto Metropolitan University, Toronto, Canada
| | - Elizabeth Berndl
- Department of Physics, Toronto Metropolitan University, Toronto, Canada; Institute for Biomedical Engineering, Science and Technology (iBEST), a partnership between St. Michael's Hospital, a site of Unity Health Toronto and Toronto Metropolitan University, Toronto, Canada
| | - Marcin Golczak
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, USA
| | - Michael C Kolios
- Department of Physics, Toronto Metropolitan University, Toronto, Canada; Institute for Biomedical Engineering, Science and Technology (iBEST), a partnership between St. Michael's Hospital, a site of Unity Health Toronto and Toronto Metropolitan University, Toronto, Canada
| | - Agata A Exner
- Department of Radiology, Case Western Reserve University, Cleveland, OH, USA; Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
39
|
Bouakaz A, Michel Escoffre J. From concept to early clinical trials: 30 years of microbubble-based ultrasound-mediated drug delivery research. Adv Drug Deliv Rev 2024; 206:115199. [PMID: 38325561 DOI: 10.1016/j.addr.2024.115199] [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: 10/06/2023] [Revised: 01/03/2024] [Accepted: 02/02/2024] [Indexed: 02/09/2024]
Abstract
Ultrasound mediated drug delivery, a promising therapeutic modality, has evolved remarkably over the past three decades. Initially designed to enhance contrast in ultrasound imaging, microbubbles have emerged as a main vector for drug delivery, offering targeted therapy with minimized side effects. This review addresses the historical progression of this technology, emphasizing the pivotal role microbubbles play in augmenting drug extravasation and targeted delivery. We explore the complex mechanisms behind this technology, from stable and inertial cavitation to diverse acoustic phenomena, and their applications in medical fields. While the potential of ultrasound mediated drug delivery is undeniable, there are still challenges to overcome. Balancing therapeutic efficacy and safety and establishing standardized procedures are essential areas requiring attention. A multidisciplinary approach, gathering collaborations between researchers, engineers, and clinicians, is important for exploiting the full potential of this technology. In summary, this review highlights the potential of using ultrasound mediated drug delivery in improving patient care across various medical conditions.
Collapse
Affiliation(s)
- Ayache Bouakaz
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France.
| | | |
Collapse
|
40
|
Tran NLH, Lam TQ, Duong PVQ, Doan LH, Vu MP, Nguyen KHP, Nguyen KT. Review on the Significant Interactions between Ultrafine Gas Bubbles and Biological Systems. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:984-996. [PMID: 38153335 DOI: 10.1021/acs.langmuir.3c03223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
Having sizes comparable with living cells and high abundance, ultrafine bubbles (UBs) are prone to inevitable interactions with different types of cells and facilitate alterations in physiological properties. The interactions of four typical cell types (e.g., bacterial, fungal, plant, and mammalian cells) with UBs have been studied over recent years. For bacterial cells, UBs have been utilized in creating the capillary force to tear down biofilms. The release of high amounts of heat, pressure, and free radicals during bubble rupture is also found to affect bacterial cell growth. Similarly, the bubble gas core identity plays an important role in the development of fungal cells. By the proposed mechanism of attachment of UBs on hydrophobin proteins in the fungal cell wall, oxygen and ozone gas-filled ultrafine bubbles can either promote or hinder the cell growth rate. On the other hand, reactive oxygen species (ROS) formation and mass transfer facilitation are two means of indirect interactions between UBs and plant cells. Likewise, the use of different gas cores in generating bubbles can produce different physical effects on these cells, for example, hydrogen gas for antioxidation against infections and oxygen for oxidation of toxic metal ions. For mammalian cells, the importance of investigating their interactions with UBs lies in the bubbles' action on cell viability as membrane poration for drug delivery can greatly affect cells' survival. UBs have been utilized and tested in forming the pores by different methods, ranging from bubble oscillation and microstream generation through acoustic cavitation to bubble implosion.
Collapse
Affiliation(s)
- Nguyen Le Hanh Tran
- School of Biotechnology, International University, Vietnam National University, Ho Chi Minh City 700000, Vietnam
| | - Thien Quang Lam
- School of Biotechnology, International University, Vietnam National University, Ho Chi Minh City 700000, Vietnam
| | - Phuong Vu Quynh Duong
- School of Biotechnology, International University, Vietnam National University, Ho Chi Minh City 700000, Vietnam
| | - Linh Hai Doan
- School of Biotechnology, International University, Vietnam National University, Ho Chi Minh City 700000, Vietnam
| | - Mai Phuong Vu
- School of Biotechnology, International University, Vietnam National University, Ho Chi Minh City 700000, Vietnam
| | - Khang Huy Phuc Nguyen
- School of Biotechnology, International University, Vietnam National University, Ho Chi Minh City 700000, Vietnam
| | - Khoi Tan Nguyen
- School of Biotechnology, International University, Vietnam National University, Ho Chi Minh City 700000, Vietnam
| |
Collapse
|
41
|
Liu M, Dasgupta A, Qu N, Rama E, Kiessling F, Lammers T. Strategies to Maximize Anthracycline Drug Loading in Albumin Microbubbles. ACS Biomater Sci Eng 2024; 10:82-88. [PMID: 34931809 DOI: 10.1021/acsbiomaterials.1c01203] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Human serum albumin (HSA) microbubbles (MBs) are attracting increasing attention as image-guided and stimuli-responsive drug delivery systems. To better understand and maximize drug encapsulation in HSA MBs, we investigated the impact of the loading strategy and the drugs' physicochemical properties on their entrapment in the MB shell. Regarding loading strategy, we explored preloading, i.e., incubating drugs with HSA prior to MB formation, as well as postloading, i.e., incubating drugs with preformed MB. Both strategies were utilized to encapsulate six anthracyclines with different physicochemical properties. We demonstrate that drug loading in the HSA MB shell profits from preloading as well as from employing drugs with high intrinsic HSA binding affinity. These findings exemplify the potential of exploiting the natural bioconjugation interactions between drugs and HSA to formulate optimally loaded MBs, and they promote the development of HSA MBs for ultrasound-triggered drug delivery.
Collapse
Affiliation(s)
- Mengjiao Liu
- Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Forckenbeckstrasse 55, Aachen 52074, Germany
| | - Anshuman Dasgupta
- Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Forckenbeckstrasse 55, Aachen 52074, Germany
| | - Na Qu
- Liaoning University, No. 66 Chongshan Middle Road, Huanggu District, Shenyang 110036, China
| | - Elena Rama
- Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Forckenbeckstrasse 55, Aachen 52074, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Forckenbeckstrasse 55, Aachen 52074, Germany
| | - Twan Lammers
- Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Forckenbeckstrasse 55, Aachen 52074, Germany
| |
Collapse
|
42
|
Barmin RA, Dasgupta A, Rix A, Weiler M, Appold L, Rütten S, Padilla F, Kuehne AJC, Pich A, De Laporte L, Kiessling F, Pallares RM, Lammers T. Enhanced Stable Cavitation and Nonlinear Acoustic Properties of Poly(butyl cyanoacrylate) Polymeric Microbubbles after Bioconjugation. ACS Biomater Sci Eng 2024; 10:75-81. [PMID: 36315422 DOI: 10.1021/acsbiomaterials.2c01021] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Microbubbles (MB) are used as ultrasound (US) contrast agents in clinical settings because of their ability to oscillate upon exposure to acoustic pulses and generate nonlinear responses with a stable cavitation profile. Polymeric MB have recently attracted increasing attention as molecular imaging probes and drug delivery agents based on their tailorable acoustic responses, high drug loading capacity, and surface functionalization capabilities. While many of these applications require MB to be functionalized with biological ligands, the impact of bioconjugation on polymeric MB cavitation and acoustic properties remains poorly understood. Hence, we here evaluated the effects of MB shell hydrolysis and subsequent streptavidin conjugation on the acoustic behavior of poly(butyl cyanoacrylate) (PBCA) MB. We show that upon biofunctionalization, MB display higher acoustic stability, stronger stable cavitation, and enhanced second harmonic generation. Furthermore, functionalized MB preserve the binding capabilities of streptavidin conjugated on their surface. These findings provide insights into the effects of bioconjugation chemistry on polymeric MB acoustic properties, and they contribute to improving the performance of polymer-based US imaging and theranostic agents.
Collapse
Affiliation(s)
- Roman A Barmin
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Anshuman Dasgupta
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Anne Rix
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Marek Weiler
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Lia Appold
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Stephan Rütten
- Electron Microscope Facility, RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Frederic Padilla
- Focused Ultrasound Foundation, Charlottesville, Virginia 22903, United States
- LabTAU, INSERM, Centre Léon Bérard, Université Lyon 1, Univ-Lyon, Lyon F-69003, France
- Department of Radiology, University of Virginia, Charlottesville, Virginia 22903, United States
| | - Alexander J C Kuehne
- DWI - Leibniz Institute for Interactive Materials, RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Andrij Pich
- DWI - Leibniz Institute for Interactive Materials, RWTH Aachen University Hospital, Aachen 52074, Germany
- Institute for Technical and Macromolecular Chemistry, RWTH Aachen University, Aachen 52074, Germany
- Aachen Maastricht Institute for Biobased Materials (AMIBM), Maastricht University, Brightlands Chemelot Campus, 6167 RD Geleen, The Netherlands
| | - Laura De Laporte
- DWI - Leibniz Institute for Interactive Materials, RWTH Aachen University Hospital, Aachen 52074, Germany
- Institute for Technical and Macromolecular Chemistry, RWTH Aachen University, Aachen 52074, Germany
- Institute of Applied Medical Engineering, Department of Advanced Materials for Biomedicine, RWTH Aachen University, Aachen 52074, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Roger M Pallares
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Twan Lammers
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen 52074, Germany
| |
Collapse
|
43
|
Li P, Du P, Peng J, Zhao Z, Li H, Yu W, Wang S, Liu L. Pharmacokinetics and pharmacodynamics of perfluoropropane after intra-venous bolus injection of perflutren lipid microsphere injection (DEFINITY®) in healthy Chinese volunteers. BMC Pharmacol Toxicol 2024; 25:6. [PMID: 38167238 PMCID: PMC10763448 DOI: 10.1186/s40360-023-00729-z] [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: 08/23/2023] [Accepted: 12/27/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND AND OBJECTIVE Definity is an ultrasound contrast agent consisting of phospholipids-encapsulated perfluoropropane (PFP), also known as perflutren, microspheres, which is initially designed to enhance echocardiographic ultrasound images. With no pharmacologic action, Definity can increase the backscatter of ultrasound resulting enhanced ultrasound signals. The objective of this study was to determine the pharmacokinetics (PKs), Pharmacodynamics (PDs) and safety of Definity in healthy male and female Chinese volunteers. METHODS A simple GC-MS method was developed and applied to simultaneously quantify PFP both in human whole blood and in expired air using Perfluorobutane (PFB) as internal standard. Meanwhile, the blood microbubble Doppler intensities were continuously monitored as companion PDs by a Doppler ultrasonography system using a non-imaging method. RESULTS After intravenous infusion of 10 µL/kg of PFP within 30 seconds, the mean AUClast of the pharmacokinetic analysis set was 0.000653 (uL/mL)*min, the average AUC∞ was 0.001051 (uL/mL)*min. The main coefficient of variation of parameters were within 30%. In this trial, the blood drug concentration of female subjects was lower than that of males. Female Cmax, AUClast and AUC∞ were lower than males', Tmax and t1/2 was close to males', Vss and CL were slightly higher than males'. The concentration of PFP in the expired air of the subject reached the maximum value 1-2 min after administration and the PFP accumulation curve in the expired air began to become flat at 9.5-11 min after administration. The PFP in the expired air at the last sampling point of most subjects was still measurable. The results of the analysis showed that female subjects had slightly more and faster PFP excretion via the lungs than males. The change of blood drug concentration in this trial was related to the change process of Doppler signal intensity. The trend of the two was close, but the peak time of blood drug concentration was slightly delayed compared with the peak time of the Doppler signal intensity. The results showed that female tmax-pd, t10 was earlier than male, and women have lower AUCpd than men. CONCLUSION The pharmacokinetics and pharmacodynamics of Definity in blood and expired air were systematically evaluated for the first time in this study. The PK/PD analysis results of this trial showed that the change of blood concentration was related to the change process of Doppler signal intensity, the trend of the two was close and expired air are the main excretion pathways of Definity. Definity was well tolerated by all subjects in the trial. TRIAL REGISTRATION This study was registered on 08 December 2017 at the Chinese Clinical Trial Registry (CTR20171087).
Collapse
Affiliation(s)
- Pengfei Li
- Department of Pharmacy, Beijing An Ding Hospital, Capital Medical University, No.5 Ankang Hutong, Xicheng District, Beijing, 100088, China.
- Phase I Clinical Trial Unit, Beijing Chao-Yang Hospital, Capital Medical University, No.8 Gongti South Road, Chaoyang District, Beijing, 100020, China.
| | - Ping Du
- Phase I Clinical Trial Unit, Beijing Chao-Yang Hospital, Capital Medical University, No.8 Gongti South Road, Chaoyang District, Beijing, 100020, China
| | - Jun Peng
- Guoke Excellence (Beijing) Medicine Technology Research Co., Ltd, No.18 Zhonghe street, Daxing District, Beijing, 100176, China
| | - Zhixia Zhao
- Department of Pharmacy, China-Japan Friendship Hospital, No.2 Yinghuayuan East Street, Chaoyang District, Beijing, 100029, China
| | - Huiling Li
- Phase I Clinical Trial Unit, Beijing Chao-Yang Hospital, Capital Medical University, No.8 Gongti South Road, Chaoyang District, Beijing, 100020, China
| | - Weiyue Yu
- Phase I Clinical Trial Unit, Beijing Chao-Yang Hospital, Capital Medical University, No.8 Gongti South Road, Chaoyang District, Beijing, 100020, China
| | - Shumin Wang
- Phase I Clinical Trial Unit, Beijing Chao-Yang Hospital, Capital Medical University, No.8 Gongti South Road, Chaoyang District, Beijing, 100020, China.
| | - Lihong Liu
- Department of Pharmacy, China-Japan Friendship Hospital, No.2 Yinghuayuan East Street, Chaoyang District, Beijing, 100029, China.
| |
Collapse
|
44
|
Rajora MA, Dhaliwal A, Zheng M, Choi V, Overchuk M, Lou JWH, Pellow C, Goertz D, Chen J, Zheng G. Quantitative Pharmacokinetics Reveal Impact of Lipid Composition on Microbubble and Nanoprogeny Shell Fate. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304453. [PMID: 38032129 PMCID: PMC10811482 DOI: 10.1002/advs.202304453] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 10/19/2023] [Indexed: 12/01/2023]
Abstract
Microbubble-enabled focused ultrasound (MB-FUS) has revolutionized nano and molecular drug delivery capabilities. Yet, the absence of longitudinal, systematic, quantitative studies of microbubble shell pharmacokinetics hinders progress within the MB-FUS field. Microbubble radiolabeling challenges contribute to this void. This barrier is overcome by developing a one-pot, purification-free copper chelation protocol able to stably radiolabel diverse porphyrin-lipid-containing Definity® analogues (pDefs) with >95% efficiency while maintaining microbubble physicochemical properties. Five tri-modal (ultrasound-, positron emission tomography (PET)-, and fluorescent-active) [64 Cu]Cu-pDefs are created with varying lipid acyl chain length and charge, representing the most prevalently studied microbubble compositions. In vitro, C16 chain length microbubbles yield 2-3x smaller nanoprogeny than C18 microbubbles post FUS. In vivo, [64 Cu]Cu-pDefs are tracked in healthy and 4T1 tumor-bearing mice ± FUS over 48 h qualitatively through fluorescence imaging (to characterize particle disruption) and quantitatively through PET and γ-counting. These studies reveal the impact of microbubble composition and FUS on microbubble dissolution rates, shell circulation, off-target tissue retention (predominantly the liver and spleen), and FUS enhancement of tumor delivery. These findings yield pharmacokinetic microbubble structure-activity relationships that disrupt conventional knowledge, the implications of which on MB-FUS platform design, safety, and nanomedicine delivery are discussed.
Collapse
Affiliation(s)
- Maneesha A. Rajora
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
- Institute of Biomedical EngineeringUniversity of TorontoTorontoOntarioM5G 1L7Canada
| | - Alexander Dhaliwal
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
- Department of Medical BiophysicsUniversity of TorontoTorontoOntarioM5G 1L7Canada
| | - Mark Zheng
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
| | - Victor Choi
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
| | - Marta Overchuk
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
- Institute of Biomedical EngineeringUniversity of TorontoTorontoOntarioM5G 1L7Canada
- Joint Department of Biomedical EngineeringUniversity of North Carolina at Chapel Hill and North Carolina State UniversityChapel HillNC27599USA
| | - Jenny W. H. Lou
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
- Department of Medical BiophysicsUniversity of TorontoTorontoOntarioM5G 1L7Canada
| | - Carly Pellow
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
- Department of Medical BiophysicsUniversity of TorontoTorontoOntarioM5G 1L7Canada
- Sunnybrook Research InstituteTorontoOntarioM4N 3M5Canada
| | - David Goertz
- Department of Medical BiophysicsUniversity of TorontoTorontoOntarioM5G 1L7Canada
- Sunnybrook Research InstituteTorontoOntarioM4N 3M5Canada
| | - Juan Chen
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
| | - Gang Zheng
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
- Institute of Biomedical EngineeringUniversity of TorontoTorontoOntarioM5G 1L7Canada
- Department of Medical BiophysicsUniversity of TorontoTorontoOntarioM5G 1L7Canada
| |
Collapse
|
45
|
Rivera J, Digklia A, Christou AS, Anibal J, Vallis KA, Wood BJ, Stride E. A Review of Ultrasound-Mediated Checkpoint Inhibitor Immunotherapy. ULTRASOUND IN MEDICINE & BIOLOGY 2024; 50:1-7. [PMID: 37798210 DOI: 10.1016/j.ultrasmedbio.2023.08.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 08/11/2023] [Accepted: 08/26/2023] [Indexed: 10/07/2023]
Abstract
Over the past decade, immunotherapy has emerged as a major modality in cancer medicine. However, despite its unprecedented success, immunotherapy currently benefits only a subgroup of patients, may induce responses of limited duration and is associated with potentially treatment-limiting side effects. In addition, responses to immunotherapeutics are sometimes diminished by the emergence of a complex array of resistance mechanisms. The efficacy of immunotherapy depends on dynamic interactions between tumour cells and the immune landscape in the tumour microenvironment. Ultrasound, especially in conjunction with cavitation-promoting agents such as microbubbles, can assist in the uptake and/or local release of immunotherapeutic agents at specific target sites, thereby increasing treatment efficacy and reducing systemic toxicity. There is also increasing evidence that ultrasound and/or cavitation may themselves directly stimulate a beneficial immune response. In this review, we summarize the latest developments in the use of ultrasound and cavitation agents to promote checkpoint inhibitor immunotherapy.
Collapse
Affiliation(s)
- Jocelyne Rivera
- Center for Interventional Oncology, Interventional Radiology, National Institutes of Health Clinical Center, National Cancer Institute, Bethesda, MD, USA; Botnar Research Centre, Institute of Biomedical Engineering, University of Oxford, Oxford, UK
| | - Antonia Digklia
- Department of Oncology, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Anna S Christou
- Center for Interventional Oncology, Interventional Radiology, National Institutes of Health Clinical Center, National Cancer Institute, Bethesda, MD, USA
| | - James Anibal
- Center for Interventional Oncology, Interventional Radiology, National Institutes of Health Clinical Center, National Cancer Institute, Bethesda, MD, USA; Computational Health Informatics Lab, Institute of Biomedical Engineering, University of Oxford, Oxford, UK
| | | | - Bradford J Wood
- Center for Interventional Oncology, Interventional Radiology, National Institutes of Health Clinical Center, National Cancer Institute, Bethesda, MD, USA
| | - Eleanor Stride
- Botnar Research Centre, Institute of Biomedical Engineering, University of Oxford, Oxford, UK.
| |
Collapse
|
46
|
Sharma D, Czarnota GJ. Ultrasound-Based Radiation Enhancement: Concepts, Mechanisms and Therapeutic Applications. Technol Cancer Res Treat 2024; 23:15330338241298864. [PMID: 39540206 PMCID: PMC11561977 DOI: 10.1177/15330338241298864] [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: 09/06/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024] Open
Abstract
Microbubbles have emerged as versatile carriers used both for cancer diagnosis and therapy. Microbubbles in the presence of ultrasound waves undergo cavitation, generating bioeffects near the cell's vicinity. Studies have shown ultrasound-stimulated microbubbles (USMB) to cause mechanical perturbation of endothelial cells, resulting in acid sphingomyelinase (ASMase)-induced ceramide production. Disruption of endothelial cells further causes vascular deterioration, leading to secondary tumor cell death. These effects are known to be synergistically higher when USMB is combined with radiation. This paper provides insight into the use of USMB as a potential radioenhancer. The possible underlying mechanism and therapeutic effects of combining USMB and radiation therapy are also presented.
Collapse
Affiliation(s)
- Deepa Sharma
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
- Departments of Medical Biophysics, and Radiation Oncology, University of Toronto, Toronto, Ontario, Canada
- Physical Sciences, Sunnybrook Research, Toronto, Ontario, Canada
| | - Gregory J. Czarnota
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
- Departments of Medical Biophysics, and Radiation Oncology, University of Toronto, Toronto, Ontario, Canada
- Physical Sciences, Sunnybrook Research, Toronto, Ontario, Canada
| |
Collapse
|
47
|
Jang Y, Park J, Kim P, Park EJ, Sun H, Baek Y, Jung J, Song TK, Doh J, Kim H. Development of exosome membrane materials-fused microbubbles for enhanced stability and efficient drug delivery of ultrasound contrast agent. Acta Pharm Sin B 2023; 13:4983-4998. [PMID: 38045059 PMCID: PMC10692476 DOI: 10.1016/j.apsb.2023.08.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 07/21/2023] [Accepted: 07/27/2023] [Indexed: 12/05/2023] Open
Abstract
Lipid-coated microbubbles are widely used as an ultrasound contrast agent, as well as drug delivery carriers. However, the two main limitations in ultrasound diagnosis and drug delivery using microbubbles are the short half-life in the blood system, and the difficulty of surface modification of microbubbles for active targeting. The exosome, a type of extracellular vesicle, has a preferentially targeting ability for its original cell. In this study, exosome-fused microbubbles (Exo-MBs) were developed by embedding the exosome membrane proteins into microbubbles. As a result, the stability of Exo-MBs is improved over the conventional microbubbles. On the same principle that under the exposure of ultrasound, microbubbles are cavitated and self-assembled into nano-sized particles, and Exo-MBs are self-assembled into exosome membrane proteins-embedded nanoparticles (Exo-NPs). The Exo-NPs showed favorable targeting properties to their original cells. A photosensitizer, chlorin e6, was loaded into Exo-MBs to evaluate therapeutic efficacy as a drug carrier. Much higher therapeutic efficacy of photodynamic therapy was confirmed, followed by cancer immunotherapy from immunogenic cell death. We have therefore developed a novel ultrasound image-guided drug delivery platform that overcomes the shortcomings of the conventional ultrasound contrast agent and is capable of simultaneous photodynamic therapy and cancer immunotherapy.
Collapse
Affiliation(s)
- Yongho Jang
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul 04107, Republic of Korea
| | - Jeehun Park
- Research Institute of Advanced Materials (RIAM), Institute of Engineering Research, Seoul National University, Seoul 08826, Republic of Korea
| | - Pilsu Kim
- Department of Electronic Engineering, Sogang University, Seoul 04107, Republic of Korea
| | - Eun-Joo Park
- Biomedical Research Institute & Department of Radiology, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Hyungjin Sun
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Yujin Baek
- Department of Materials Science and Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Jaehun Jung
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul 04107, Republic of Korea
| | - Tai-kyong Song
- Department of Electronic Engineering, Sogang University, Seoul 04107, Republic of Korea
| | - Junsang Doh
- Research Institute of Advanced Materials (RIAM), Institute of Engineering Research, Seoul National University, Seoul 08826, Republic of Korea
- Department of Materials Science and Engineering, Seoul National University, Seoul 08826, Republic of Korea
- Institute of Engineering Research, Bio-MAX Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyuncheol Kim
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul 04107, Republic of Korea
- Department of Biomedical Engineering, Sogang University, Seoul 04107, Republic of Korea
| |
Collapse
|
48
|
Xing Z, Guo J, Wu Z, He C, Wang L, Bai M, Liu X, Zhu B, Guan Q, Cheng C. Nanomaterials-Enabled Physicochemical Antibacterial Therapeutics: Toward the Antibiotic-Free Disinfections. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2303594. [PMID: 37626465 DOI: 10.1002/smll.202303594] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 07/17/2023] [Indexed: 08/27/2023]
Abstract
Bacterial infection continues to be an increasing global health problem with the most widely accepted treatment paradigms restricted to antibiotics. However, the overuse and misuse of antibiotics have triggered multidrug resistance of bacteria, frustrating therapeutic outcomes, and leading to higher mortality rates. Even worse, the tendency of bacteria to form biofilms on living and nonliving surfaces further increases the difficulty in confronting bacteria because the extracellular matrix can act as a robust barrier to prevent the penetration of antibiotics and resist environmental damage. As a result, the inability to eliminate bacteria and biofilms often leads to persistent infection, implant failure, and device damage. Therefore, it is of paramount importance to develop alternative antimicrobial agents while avoiding the generation of bacterial resistance to prevent the large-scale growth of bacterial resistance. In recent years, nano-antibacterial materials have played a vital role in the antibacterial field because of their excellent physical and chemical properties. This review focuses on new physicochemical antibacterial strategies and versatile antibacterial nanomaterials, especially the mechanism and types of 2D antibacterial nanomaterials. In addition, this advanced review provides guidance on the development direction of antibiotic-free disinfections in the antibacterial field in the future.
Collapse
Affiliation(s)
- Zhenyu Xing
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Jiusi Guo
- Department of Orthodontics, Department of Endodontics, State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Zihe Wu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Chao He
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Liyun Wang
- Department of Medical Ultrasound, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Mingru Bai
- Department of Orthodontics, Department of Endodontics, State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Xikui Liu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Bihui Zhu
- Department of Medical Ultrasound, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qiuyue Guan
- Department of Geriatrics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Chong Cheng
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| |
Collapse
|
49
|
Lyons B, Balkaran JPR, Dunn-Lawless D, Lucian V, Keller SB, O’Reilly CS, Hu L, Rubasingham J, Nair M, Carlisle R, Stride E, Gray M, Coussios C. Sonosensitive Cavitation Nuclei-A Customisable Platform Technology for Enhanced Therapeutic Delivery. Molecules 2023; 28:7733. [PMID: 38067464 PMCID: PMC10708135 DOI: 10.3390/molecules28237733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/14/2023] [Accepted: 11/16/2023] [Indexed: 12/18/2023] Open
Abstract
Ultrasound-mediated cavitation shows great promise for improving targeted drug delivery across a range of clinical applications. Cavitation nuclei-sound-sensitive constructs that enhance cavitation activity at lower pressures-have become a powerful adjuvant to ultrasound-based treatments, and more recently emerged as a drug delivery vehicle in their own right. The unique combination of physical, biological, and chemical effects that occur around these structures, as well as their varied compositions and morphologies, make cavitation nuclei an attractive platform for creating delivery systems tuned to particular therapeutics. In this review, we describe the structure and function of cavitation nuclei, approaches to their functionalization and customization, various clinical applications, progress toward real-world translation, and future directions for the field.
Collapse
Affiliation(s)
- Brian Lyons
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Joel P. R. Balkaran
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Darcy Dunn-Lawless
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Veronica Lucian
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Sara B. Keller
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Colm S. O’Reilly
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford OX1 3PJ, UK;
| | - Luna Hu
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Jeffrey Rubasingham
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Malavika Nair
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Robert Carlisle
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Eleanor Stride
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Michael Gray
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Constantin Coussios
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| |
Collapse
|
50
|
Hani U, Gowda BHJ, Haider N, Ramesh K, Paul K, Ashique S, Ahmed MG, Narayana S, Mohanto S, Kesharwani P. Nanoparticle-Based Approaches for Treatment of Hematological Malignancies: a Comprehensive Review. AAPS PharmSciTech 2023; 24:233. [PMID: 37973643 DOI: 10.1208/s12249-023-02670-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 10/03/2023] [Indexed: 11/19/2023] Open
Abstract
Blood cancer, also known as hematological malignancy, is one of the devastating types of cancer that has significantly paved its mortality mark globally. It persists as an extremely deadly cancer type and needs utmost attention owing to its negligible overall survival rate. Major challenges in the treatment of blood cancer include difficulties in early diagnosis, as well as severe side effects resulting from chemotherapy. In addition, immunotherapies and targeted therapies can be prohibitively expensive. Over the past two decades, scientists have devised a few nanoparticle-based drug delivery systems aimed at overcoming this challenge. These therapeutic strategies are engineered to augment the cellular uptake, pharmacokinetics, and effectiveness of anticancer drugs. However, there are still numerous types of nanoparticles that could potentially improve the efficacy of blood cancer treatment, while also reducing treatment costs and mitigating drug-related side effects. To the best of our knowledge, there has been limited reviews published on the use of nano-based drug delivery systems for the treatment of hematological malignancies. Therefore, we have made a concerted effort to provide a comprehensive review that draws upon recent literature and patents, with a focus on the most promising results regarding the use of nanoparticle-based approaches for the treatment of hematological malignancies. All these crucial points covered under a common title would significantly help researchers and scientists working in the area.
Collapse
Affiliation(s)
- Umme Hani
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, 61421, Abha, Saudi Arabia.
| | - B H Jaswanth Gowda
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore, 575018, Karnataka, India.
- School of Pharmacy, Medical Biology Centre, Queen's University Belfast, Belfast, BT9 7BL, UK.
| | - Nazima Haider
- Department of Pathology, College of Medicine, King Khalid University, 61421, Abha, Saudi Arabia
| | - Kvrns Ramesh
- Department of Pharmaceutics, RAK College of Pharmaceutical Sciences, RAK Medical and Health Sciences University, 11172, Ras Al Khaimah, United Arab Emirates
| | - Karthika Paul
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, 570015, Karnataka, India
| | - Sumel Ashique
- Department of Pharmaceutics, Pandaveswar School of Pharmacy, Pandaveswar, West Bengal, 713378, India
| | - Mohammed Gulzar Ahmed
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore, 575018, Karnataka, India
| | - Soumya Narayana
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore, 575018, Karnataka, India
| | - Sourav Mohanto
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore, 575018, Karnataka, India
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India.
- Center for Global health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Tamil Nadu, India.
| |
Collapse
|