1
|
Patrick PS, Stuckey DJ, Zhu H, Kalber TL, Iftikhar H, Southern P, Bear JC, Lythgoe MF, Hattersley SR, Pankhurst QA. Improved tumour delivery of iron oxide nanoparticles for magnetic hyperthermia therapy of melanoma via ultrasound guidance and 111In SPECT quantification. NANOSCALE 2024; 16:19715-19729. [PMID: 39044561 PMCID: PMC11488578 DOI: 10.1039/d4nr00240g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 07/12/2024] [Indexed: 07/25/2024]
Abstract
Magnetic field hyperthermia relies on the intra-tumoural delivery of magnetic nanoparticles by interstitial injection, followed by their heating on exposure to a remotely-applied alternating magnetic field (AMF). This offers a potential sole or adjuvant route to treating drug-resistant tumours for which no alternatives are currently available. However, two challenges in nanoparticle delivery currently hinder the effective clinical translation of this technology: obtaining enough magnetic material within the tumour to enable sufficient heating; and doing this accurately to limit or avoid damage to surrounding healthy tissue. A further complication is the lack of established methods to non-invasively quantify nanoparticle biodistribution, which is necessary to evaluate the performance of improved delivery strategies. Here we employ 111In radiolabelling and single-photon emission computed tomography (SPECT) to non-invasively quantify distribution of a clinical grade iron-oxide-based nanoparticle in a mouse model of melanoma. We show that compared to manual injection, ultrasound guided delivery together with syringe-pump-controlled infusion improves both the nanoparticle concentration within the tumour, and the accuracy of delivery - reducing off-target peri-tumoural delivery. Following AMF heating, injected melanomas shrank significantly compared to non-injected controls, validating therapeutic efficacy. Systemic off-target delivery was quantified and extrapolated to predict off-target energy absorbance within safe limits for the main sites of background accumulation. With many nanoparticle-based therapies currently in development for cancer, this image-guided delivery strategy has wide potential impact beyond the field of magnetic hyperthermia. Future use in representative patient cohorts would also be enabled by the high clinical availability of both SPECT and ultrasound imaging.
Collapse
Affiliation(s)
- P Stephen Patrick
- Centre for Advanced Biomedical Imaging (CABI), Department of Medicine, University College London, London WC1E 6DD, UK.
| | - Daniel J Stuckey
- Centre for Advanced Biomedical Imaging (CABI), Department of Medicine, University College London, London WC1E 6DD, UK.
| | - Huachen Zhu
- Centre for Advanced Biomedical Imaging (CABI), Department of Medicine, University College London, London WC1E 6DD, UK.
| | - Tammy L Kalber
- Centre for Advanced Biomedical Imaging (CABI), Department of Medicine, University College London, London WC1E 6DD, UK.
| | - Haadi Iftikhar
- Healthcare Biomagnetics Laboratory, University College London, 21 Albemarle Street, London, W1S 4BS, UK
| | - Paul Southern
- Healthcare Biomagnetics Laboratory, University College London, 21 Albemarle Street, London, W1S 4BS, UK
- Resonant Circuits Limited, 21 Albemarle Street, London, W1S 4BS, UK
| | - Joseph C Bear
- School of Life Science, Pharmacy & Chemistry, Kingston University, Penrhyn Road, Kingston upon Thames, KT1 2EE, UK
| | - Mark F Lythgoe
- Centre for Advanced Biomedical Imaging (CABI), Department of Medicine, University College London, London WC1E 6DD, UK.
| | | | - Quentin A Pankhurst
- Healthcare Biomagnetics Laboratory, University College London, 21 Albemarle Street, London, W1S 4BS, UK
- Resonant Circuits Limited, 21 Albemarle Street, London, W1S 4BS, UK
| |
Collapse
|
2
|
Zhu W, Pan S, Zhang J, Xu J, Zhang R, Zhang Y, Fu Z, Wang Y, Hu C, Xu Z. The role of hyperthermia in the treatment of tumor. Crit Rev Oncol Hematol 2024:104541. [PMID: 39461607 DOI: 10.1016/j.critrevonc.2024.104541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 09/19/2024] [Accepted: 10/17/2024] [Indexed: 10/29/2024] Open
Abstract
Despite recent advancements in the diagnosis and treatment options for cancer, it remains one of the most serious threats to health. Hyperthermia (HT) has emerged as a highly promising area of research due to its safety and cost-effectiveness. Currently, based on temperature, HT can be categorized into thermal ablation and mild hyperthermia. Thermal ablation involves raising the temperature within the tumor to over 60°C, resulting in direct necrosis in the central region of the tumor. In contrast, mild hyperthermia operates at relatively lower temperatures, typically in the range of 41-45°C, to induce damage to tumor cells. Furthermore, HT also serves as an immune adjuvant strategy in radiotherapy, chemotherapy, and immunotherapy, enhancing the effectiveness of radiotherapy, increasing the uptake of chemotherapy drugs, and reprogramming the tumor microenvironment through the induction of immunogenic cell death, thereby promoting the recruitment of endogenous immune cells. This article reviews the current status and development of hyperthermia, outlines potential mechanisms by which hyperthermia inhibits tumors, describes clinical trial attempts combining hyperthermia with radiotherapy, chemotherapy, and immunotherapy, and discusses the relationship between nanoparticles and hyperthermia.
Collapse
Affiliation(s)
- Weiwei Zhu
- Second clinical medical College,Zhejiang Chinese Medical University, Hangzhou 310053, China; Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Siwei Pan
- Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China; Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Jiaqing Zhang
- Second clinical medical College,Zhejiang Chinese Medical University, Hangzhou 310053, China; Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Jingli Xu
- Second clinical medical College,Zhejiang Chinese Medical University, Hangzhou 310053, China; Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Ruolan Zhang
- Second clinical medical College,Zhejiang Chinese Medical University, Hangzhou 310053, China; Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Yanqiang Zhang
- Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China; Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Zhenjie Fu
- Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China; Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Yuqi Wang
- Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China; Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Can Hu
- Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China; Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou 310022, China.
| | - Zhiyuan Xu
- Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China; Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou 310022, China.
| |
Collapse
|
3
|
Rousseau C, Vuong QL, Gossuin Y, Maes B, Rosolen G. Concurrent photothermal therapy and nuclear magnetic resonance imaging with plasmonic-magnetic nanoparticles: A numerical study. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2024; 257:108453. [PMID: 39426140 DOI: 10.1016/j.cmpb.2024.108453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 09/18/2024] [Accepted: 09/30/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND AND OBJECTIVE Theranostics is the combination of the diagnostic and therapeutic phases. Here we focus on simultaneous use of photothermal therapy and magnetic resonance imaging, employing a contrast-photothermal agent that converts incident light into heat and affects the transverse relaxation time, a key magnetic resonance imaging parameter. Our work considers a gold-magnetite nanoshell platform to gauge the feasibility of magnetic resonance imaging monitoring of the heating associated with phototherapy, by studying the modification of the transverse relaxation rate induced by laser illumination of a solution containing these hybrid nanoparticles. METHODS We simulate a system composed of an aqueous solution with hybrid nanoshells under continuous laser irradiation, enabling the evaluation of spatial variations of the transverse relaxation rate within the sample. We work with the hybrid nanoshell platform comprising a metal/gold shell for thermoplasmonic effects and a magnetite core for magnetic resonance imaging contrast enhancement. The optical properties of the nanoshells are first obtained through simulations using the finite element method. Next, the heating generated by the laser illumination is calculated by numerical integration. Finally, the transverse relaxation rate is obtained through the application of an analytical model. Additionally, we conduct an optimization of the nanoshell geometry to fulfill requirements of both magnetic resonance imaging and phototherapy techniques. RESULTS Our findings demonstrate a narrow range of nanoshell sizes exhibiting both a plasmonic absorption peak in the human biological window and a high response to laser illumination of the transverse relaxation rate. Furthermore, the illumination can induce up to a 30% modification in transverse relaxation rate compared to the non-illuminated scenario in this range of nanoshell sizes. CONCLUSIONS In this work we establish the numerical understanding of the interplay between phototherapy and nuclear magnetic resonance imaging when employed concurrently. This allows magnetic resonance imaging monitoring of the heating associated with phototherapy.
Collapse
Affiliation(s)
- C Rousseau
- Micro- and Nanophotonic Materials Group, Research Institute for Materials Science and Engineering, University of Mons, 20 Place du Parc, B-7000 Mons, Belgium.
| | - Q L Vuong
- Biomedical Physics Unit, University of Mons, 20 Place du Parc, B-7000 Mons, Belgium
| | - Y Gossuin
- Biomedical Physics Unit, University of Mons, 20 Place du Parc, B-7000 Mons, Belgium
| | - B Maes
- Micro- and Nanophotonic Materials Group, Research Institute for Materials Science and Engineering, University of Mons, 20 Place du Parc, B-7000 Mons, Belgium
| | - G Rosolen
- Micro- and Nanophotonic Materials Group, Research Institute for Materials Science and Engineering, University of Mons, 20 Place du Parc, B-7000 Mons, Belgium
| |
Collapse
|
4
|
Yue Y, Ai J, Chi W, Zhao X, Huo F, Yin C. Biomedical-Optical-Window Tailored Cyanines for Steerable Inflammatory Bowel Disease Theranostic. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024:e2408450. [PMID: 39240024 DOI: 10.1002/adma.202408450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/23/2024] [Indexed: 09/07/2024]
Abstract
Tailored photophysical properties and chemical activity is the ultimate pursuit of functional dyes for in vivo biomedical theranostics. In this work, the independent regulation of the absorption and fluorescence emission wavelengths of heptamethine cyanines is reported. These dyes retain near-infrared fluorescence emission (except a nitro-modified dye) while feature variable absorption wavelengths ranging from 590 to 860 nm. This enables to obtain customized functional dyes that meet the excitation and fluorescence wavelength requirements defined by the optical properties of tissues for in vivo biomedical applications. Typically, a nitro-modified photothermal active derivative Cy-Mu-7-9 is used, which features strong absorption at 810 nm in PBS, a wavelength that balanced the tissue penetration depth and non-specific photothermal effect, to realize non-destructive inflammatory bowel disease (IBD) therapy via photothermal induced up-regulation of heat shock protein 70 in the intestinal epithelial cells. The corresponding amino-modified dye Cy-Mu-7-9-NH2, which can be formed in health enteric cavity by Cy-Mu-7-9 after oral administration, is a fluorescence compound with the emission of 800 nm in PBS. Based on the IBD sensitive transformation of Cy-Mu-7-9 and Cy-Mu-7-9-NH2, in vivo IBD theranostic and therapeutic effect evaluation is realized via the synergy of fluorescence imaging and photothermal therapy for the first time.
Collapse
Affiliation(s)
- Yongkang Yue
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institute of Molecular Science, Shanxi University, Taiyuan, 030006, P. R. China
| | - Jiahong Ai
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institute of Molecular Science, Shanxi University, Taiyuan, 030006, P. R. China
| | - Weijie Chi
- School of Chemistry and Chemical Engineering, Hainan University, Haikou, 570228, China
| | - Xiaoni Zhao
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institute of Molecular Science, Shanxi University, Taiyuan, 030006, P. R. China
| | - Fangjun Huo
- Research Institute of Applied Chemistry, Shanxi University, Taiyuan, 030006, P. R. China
| | - Caixia Yin
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institute of Molecular Science, Shanxi University, Taiyuan, 030006, P. R. China
| |
Collapse
|
5
|
Kadian P, Kesari A, Singh IR, Choudhury S, Singh A, Panda JJ, Randhawa JK. Temporal Optimization in the Synthesis of Multifunctional Nano Agents for Enhanced MRI Contrast, Hyperthermia Therapy, and ROS Generation. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:17337-17347. [PMID: 39113429 DOI: 10.1021/acs.langmuir.4c01466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/21/2024]
Abstract
Advanced methodologies, such as hyperthermia and modulation of reactive oxygen species (ROS), exhibit considerable promise in the therapeutic landscape of cancer. These strategies offer a targeted paradigm for combating malignant cells while mitigating damage to healthy tissue. Noteworthy among these approaches is the utilization of superparamagnetic iron oxide nanoparticles, which are renowned for their ability to enhance both hyperthermia and ROS generation specifically within tumor microenvironments. The objective of this investigation is to scrutinize the relationship between the reaction duration and the characteristics of carbon-doped silica core-shell iron oxide nanoparticles (CSIONPs). Specifically, we focus on CSIONP-12, CSIONP-24, and CSIONP-36, synthesized by using varying reaction periods. Through a comprehensive analysis, we primarily evaluate the impact of these formulations on T1 and T2 magnetic resonance imaging (MRI), aiming to elucidate their mechanisms and therapeutic potential in promoting hyperthermia and ROS-mediated cancer therapy. CSIONP-24 emerges as a compelling candidate due to its dual influence on magnetic hyperthermia and ROS generation, suggesting its promise in enhancing cancer treatment outcomes. Furthermore, the findings underscore the exceptional T1-T2 MRI capabilities of this technology, underscoring its versatility and efficacy in the nuanced realm of cancer theranostic.
Collapse
Affiliation(s)
- Pallavi Kadian
- School of Chemical Sciences, Indian Institute of Technology, Mandi 175005, India
| | - Anshika Kesari
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | | | | | - Anup Singh
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Jiban Jyoti Panda
- Institute of Nano Science and Technology, Mohali, Punjab 160062, India
| | - Jaspreet Kaur Randhawa
- School of Mechanical and Materials Engineering, Indian Institute of Technology, Mandi 175005, India
| |
Collapse
|
6
|
Kejík Z, Hajduch J, Abramenko N, Vellieux F, Veselá K, Fialová JL, Petrláková K, Kučnirová K, Kaplánek R, Tatar A, Skaličková M, Masařík M, Babula P, Dytrych P, Hoskovec D, Martásek P, Jakubek M. Cyanine dyes in the mitochondria-targeting photodynamic and photothermal therapy. Commun Chem 2024; 7:180. [PMID: 39138299 PMCID: PMC11322665 DOI: 10.1038/s42004-024-01256-6] [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: 02/08/2024] [Accepted: 07/26/2024] [Indexed: 08/15/2024] Open
Abstract
Mitochondrial dysregulation plays a significant role in the carcinogenesis. On the other hand, its destabilization strongly represses the viability and metastatic potential of cancer cells. Photodynamic and photothermal therapies (PDT and PTT) target mitochondria effectively, providing innovative and non-invasive anticancer therapeutic modalities. Cyanine dyes, with strong mitochondrial selectivity, show significant potential in enhancing PDT and PTT. The potential and limitations of cyanine dyes for mitochondrial PDT and PTT are discussed, along with their applications in combination therapies, theranostic techniques, and optimal delivery systems. Additionally, novel approaches for sonodynamic therapy using photoactive cyanine dyes are presented, highlighting advances in cancer treatment.
Collapse
Affiliation(s)
- Zdeněk Kejík
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Prague, Czech Republic.
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455, 120 00, Prague, Czech Republic.
| | - Jan Hajduch
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Prague, Czech Republic
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455, 120 00, Prague, Czech Republic
| | - Nikita Abramenko
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Prague, Czech Republic
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455, 120 00, Prague, Czech Republic
| | - Frédéric Vellieux
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Prague, Czech Republic
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455, 120 00, Prague, Czech Republic
| | - Kateřina Veselá
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Prague, Czech Republic
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455, 120 00, Prague, Czech Republic
| | | | - Kateřina Petrláková
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, CZ-625 00, Brno, Czech Republic
| | - Kateřina Kučnirová
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Prague, Czech Republic
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455, 120 00, Prague, Czech Republic
| | - Robert Kaplánek
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Prague, Czech Republic
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455, 120 00, Prague, Czech Republic
| | - Ameneh Tatar
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Prague, Czech Republic
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455, 120 00, Prague, Czech Republic
| | - Markéta Skaličková
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Prague, Czech Republic
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455, 120 00, Prague, Czech Republic
| | - Michal Masařík
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Prague, Czech Republic
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455, 120 00, Prague, Czech Republic
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, CZ-625 00, Brno, Czech Republic
- Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - Petr Babula
- Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - Petr Dytrych
- 1st Department of Surgery-Department of Abdominal, Thoracic Surgery and Traumatology, First Faculty of Medicine, Charles University and General University Hospital in Prague, U Nemocnice 2, 121 08, Prague, Czech Republic
| | - David Hoskovec
- 1st Department of Surgery-Department of Abdominal, Thoracic Surgery and Traumatology, First Faculty of Medicine, Charles University and General University Hospital in Prague, U Nemocnice 2, 121 08, Prague, Czech Republic
| | - Pavel Martásek
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455, 120 00, Prague, Czech Republic.
| | - Milan Jakubek
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Prague, Czech Republic.
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455, 120 00, Prague, Czech Republic.
| |
Collapse
|
7
|
Righini MF, Durham A, Tsoutsou PG. Hyperthermia and radiotherapy: physiological basis for a synergistic effect. Front Oncol 2024; 14:1428065. [PMID: 39165690 PMCID: PMC11333208 DOI: 10.3389/fonc.2024.1428065] [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: 05/05/2024] [Accepted: 07/17/2024] [Indexed: 08/22/2024] Open
Abstract
In cancer treatment, mild hyperthermia (HT) represents an old, but recently revived opportunity to increase the efficacy of radiotherapy (RT) without increasing side effects, thereby widening the therapeutic window. HT disrupts cellular homeostasis by acting on multiple targets, and its combination with RT produces synergistic antitumoral effects on specific pathophysiological mechanisms, associated to DNA damage and repair, hypoxia, stemness and immunostimulation. HT is furthermore associated to direct tumor cell kill, particularly in higher temperature levels. A phenomenon of temporary resistance to heat, known as thermotolerance, follows each HT session. Cancer treatment requires innovative concepts and combinations to be tested but, for a meaningful development of clinical trials, the understanding of the underlying mechanisms of the tested modalities is essential. In this mini-review, we aimed to describe the synergistic effects of the combination of HT with RT as well as the phenomena of thermal shock and thermotolerance, in order to stimulate clinicians in new, clinically relevant concepts and combinations, which become particularly relevant in the era of technological advents in both modalities but also cancer immunotherapy.
Collapse
Affiliation(s)
| | - André Durham
- Faculty of Medicine, University of Geneva (UNIGE), Geneva, Switzerland
- Department of Radiation Oncology, Geneva University Hospitals (HUG), Geneva, Switzerland
| | - Pelagia G. Tsoutsou
- Faculty of Medicine, University of Geneva (UNIGE), Geneva, Switzerland
- Department of Radiation Oncology, Geneva University Hospitals (HUG), Geneva, Switzerland
| |
Collapse
|
8
|
Guo Y, Li B, Xie H, Wu C, Wang G, Yao K, Li L. The therapeutic efficacy of different configuration nano-polydopamine drug carrier systems with photothermal synergy against head and neck squamous cell carcinoma. Regen Biomater 2024; 11:rbae073. [PMID: 39027362 PMCID: PMC11256922 DOI: 10.1093/rb/rbae073] [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: 03/07/2024] [Revised: 06/02/2024] [Accepted: 06/05/2024] [Indexed: 07/20/2024] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is the sixth most common malignant tumor worldwide. Considering its special anatomical site and the progressive resistance to chemotherapy drugs, the development of more effective, minimally invasive and precise treatment methods is urgently needed. Nanomaterials, given their special properties, can be used as drug carrier systems to improve the therapeutic effect and reduce the adverse effects. The drug carrier systems with photothermal effect can promote the killing of cancer cells and help overcome drug resistance through heat stress. We selected dopamine, a simple raw material, and designed and synthesized three different configurations of nano-polydopamine (nPDA) nanomaterials, including nPDA balls, nPDA plates and porous nPDA balls. In addition to the self-polymerization and self-assembly, nPDA has high photothermal conversion efficiency and can be easily modified. Moreover, we loaded cisplatin into three different configurations of nPDA, creating nPDA-cis (the nano-drug carrier system with cisplatin), and comparatively studied the properties and antitumor effects of all the nPDA and nPDA-cis materials in vitro and nPDA-cis in vivo. We found that the photothermal effect of the nPDA-cis balls drug carrier system had synergistic effect with cisplatin, resulting in excellent antitumor effect and good clinical application prospects. The comparison of the three different configurations of drug carrier systems suggested the importance of optimizing the spatial configuration design and examining the physical and chemical properties in the future development of nano-drug carrier systems. In this study, we also noted the duality and complexity of the influences of heat stress on tumors in vitro and in vivo. The specific mechanisms and the synergy with chemotherapy and immunotherapy will be an important research direction in the future.
Collapse
Affiliation(s)
- Yuhao Guo
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041,China
- Department of Stomatology, Xinqiao Hospital of Army Medical University, Chongqing 400037,China
| | - Bo Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041,China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041,China
| | - Huixu Xie
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041,China
- Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041,China
| | - Chenzhou Wu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041,China
- Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041,China
| | - Guixue Wang
- State and Local Joint Engineering Laboratory, Bioengineering College of Chongqing University, Chongqing 400044,China
| | - Kexin Yao
- Multi-Scale Porous Materials Center, Institute of Advanced Interdisciplinary Studies, School of Chemistry and Chemical Engineering, Chongqing University, Chongqing 400044,China
| | - Longjiang Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041,China
- Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041,China
| |
Collapse
|
9
|
Sun C, Bai J, Sun J, Sun Y, Zhang F, Li H, Liu Y, Meng L, Wang X. OTU deubiquitinase 7B facilitates the hyperthermia-induced inhibition of lung cancer progression through enhancing Smac-mediated mitochondrial dysfunction. ENVIRONMENTAL TOXICOLOGY 2024; 39:1989-2005. [PMID: 38088504 DOI: 10.1002/tox.24080] [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: 08/25/2023] [Revised: 11/18/2023] [Accepted: 11/28/2023] [Indexed: 03/09/2024]
Abstract
Hyperthermia, as an adjuvant therapy, has shown promising anti-tumor effects. Ovarian tumor domain-containing 7B (OTUD7B) is a deubiquitinating enzyme that is frequently found in a variety of cancers. The aim of this study is to investigate the role of OTUD7B in lung cancer hyperthermia and the underlying mechanism. A549 and CALU-3 cells were respectively exposed to 42 or 44°C for the indicated times (0, 1, 3, or 6 h) followed by incubation at 37°C for 24 h. We found a temperature- and time-dependent decrease in cell viability and an increase in apoptosis levels. Compared with 0 h, heat treatment for 3 h inhibited the proliferation and invasion of A549 cells, reduced the expression levels of mitochondrial membrane potential, IAP family members (cIAP-1 and XIAP) proteins and ubiquitination of Smac, and increased Smac protein expression. Treatment with 10 μM Smac mimic BV6 further enhanced the anti-tumor effect of hyperthermia. Next, co-IP validation showed that OTUD7B interacted with Smac and stabilized Smac through deubiquitination. OTUD7B overexpression induced damage in A549 and CALU-3 cells, while silencing OTUD7B caused opposite effects. Overexpressing OTUD7B enhanced the anti-cancer effect of hyperthermia, while si-OTUD7B reversed the anti-cancer effect of hyperthermia, which was verified in the xenograft tumor model in nude mice. Taken together, OTUD7B may serve as a potential anticancer factor with potential clinical efficacy in the thermotherapeutic treatment of lung cancer.
Collapse
Affiliation(s)
- Chao Sun
- Department of Medical Oncology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Jun Bai
- Department of Medical Oncology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Jingying Sun
- Shaanxi Provincial Key Laboratory of Infectious and Immunological Diseases, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Yang Sun
- Data Center, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Fan Zhang
- Department of Medical Oncology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - He Li
- Department of Medical Oncology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Ying Liu
- Department of Medical Oncology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Lian Meng
- Department of Pathology, The First Affiliated Hospital of Shihezi University, Shihezi, China
| | - Xifang Wang
- Department of Medical Oncology, Shaanxi Provincial People's Hospital, Xi'an, China
| |
Collapse
|
10
|
Lin TY, Jia JS, Luo WR, Lin XL, Xiao SJ, Yang J, Xia JW, Zhou C, Zhou ZH, Lin SJ, Li QW, Yang ZZ, Lei Y, Yang WQ, Shen HF, Huang SH, Wang SC, Chen LB, Yang YL, Xue SW, Li YL, Dai GQ, Zhou Y, Li YC, Wei F, Rong XX, Luo XJ, Zhao BX, Huang WH, Xiao D, Sun Y. ThermomiR-377-3p-induced suppression of Cirbp expression is required for effective elimination of cancer cells and cancer stem-like cells by hyperthermia. J Exp Clin Cancer Res 2024; 43:62. [PMID: 38419081 PMCID: PMC10903011 DOI: 10.1186/s13046-024-02983-3] [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: 02/09/2024] [Accepted: 02/12/2024] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND In recent years, the development of adjunctive therapeutic hyperthermia for cancer therapy has received considerable attention. However, the mechanisms underlying hyperthermia resistance are still poorly understood. In this study, we investigated the roles of cold‑inducible RNA binding protein (Cirbp) in regulating hyperthermia resistance and underlying mechanisms in nasopharyngeal carcinoma (NPC). METHODS CCK-8 assay, colony formation assay, tumor sphere formation assay, qRT-PCR, Western blot were employed to examine the effects of hyperthermia (HT), HT + oridonin(Ori) or HT + radiotherapy (RT) on the proliferation and stemness of NPC cells. RNA sequencing was applied to gain differentially expressed genes upon hyperthermia. Gain-of-function and loss-of-function experiments were used to evaluate the effects of RNAi-mediated Cirbp silencing or Cirbp overexpression on the sensitivity or resistance of NPC cells and cancer stem-like cells to hyperthermia by CCK-8 assay, colony formation assay, tumorsphere formation assay and apoptosis assay, and in subcutaneous xenograft animal model. miRNA transient transfection and luciferase reporter assay were used to demonstrate that Cirbp is a direct target of miR-377-3p. The phosphorylation levels of key members in ATM-Chk2 and ATR-Chk1 pathways were detected by Western blot. RESULTS Our results firstly revealed that hyperthermia significantly attenuated the stemness of NPC cells, while combination treatment of hyperthermia and oridonin dramatically increased the killing effect on NPC cells and cancer stem cell (CSC)‑like population. Moreover, hyperthermia substantially improved the sensitivity of radiation‑resistant NPC cells and CSC‑like cells to radiotherapy. Hyperthermia noticeably suppressed Cirbp expression in NPC cells and xenograft tumor tissues. Furthermore, Cirbp inhibition remarkably boosted anti‑tumor‑killing activity of hyperthermia against NPC cells and CSC‑like cells, whereas ectopic expression of Cirbp compromised tumor‑killing effect of hyperthermia on these cells, indicating that Cirbp overexpression induces hyperthermia resistance. ThermomiR-377-3p improved the sensitivity of NPC cells and CSC‑like cells to hyperthermia in vitro by directly suppressing Cirbp expression. More importantly, our results displayed the significantly boosted sensitization of tumor xenografts to hyperthermia by Cirbp silencing in vivo, but ectopic expression of Cirbp almost completely counteracted hyperthermia-mediated tumor cell-killing effect against tumor xenografts in vivo. Mechanistically, Cirbp silencing-induced inhibition of DNA damage repair by inactivating ATM-Chk2 and ATR-Chk1 pathways, decrease in stemness and increase in cell death contributed to hyperthermic sensitization; conversely, Cirbp overexpression-induced promotion of DNA damage repair, increase in stemness and decrease in cell apoptosis contributed to hyperthermia resistance. CONCLUSION Taken together, these findings reveal a previously unrecognized role for Cirbp in positively regulating hyperthermia resistance and suggest that thermomiR-377-3p and its target gene Cirbp represent promising targets for therapeutic hyperthermia.
Collapse
Affiliation(s)
- Tao-Yan Lin
- Laboratory Animal Management Center, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jun-Shuang Jia
- Laboratory Animal Management Center, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Wei-Ren Luo
- Cancer Research Institute, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen Third People's Hospital, Shenzhen, 518112, China
| | - Xiao-Lin Lin
- Laboratory Animal Management Center, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315, China
| | - Sheng-Jun Xiao
- Department of Pathology, The Second Affiliated Hospital of Guilin Medical University, Guilin, 541199, China
| | - Jie Yang
- Laboratory Animal Management Center, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
- Department of Imaging, Central Hospital of Shaoyang, Shaoyang, 422000, China
| | - Jia-Wei Xia
- The Third People's Hospital of Kunming (The Sixth Affiliated Hospital of Dali University), Kunming, 650041, China
| | - Chen Zhou
- Department of Pathology, The Second Affiliated Hospital of Guilin Medical University, Guilin, 541199, China
| | - Zhi-Hao Zhou
- Laboratory Animal Management Center, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Shu-Jun Lin
- Laboratory Animal Management Center, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Qi-Wen Li
- Laboratory Animal Management Center, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Zhi-Zhi Yang
- Laboratory Animal Management Center, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Ye Lei
- Laboratory Animal Management Center, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Wen-Qing Yang
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, China
| | - Hong-Fen Shen
- Laboratory Animal Management Center, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Shi-Hao Huang
- Laboratory Animal Management Center, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Sheng-Chun Wang
- Laboratory Animal Management Center, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
- Department of Pathology, School of Basic Medicine, Guangdong Medical University, Dongguan, 523808, China
| | - Lin-Bei Chen
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, China
| | - Yu-Lin Yang
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, China
| | - Shu-Wen Xue
- Laboratory Animal Management Center, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Yong-Long Li
- Laboratory Animal Management Center, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Guan-Qi Dai
- Laboratory Animal Management Center, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Ying Zhou
- Laboratory Animal Management Center, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Ying-Chun Li
- Laboratory Animal Management Center, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Fang Wei
- Laboratory Animal Management Center, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xiao-Xiang Rong
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guang‑zhou, 510515, China
| | - Xiao-Jun Luo
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315, China
| | - Bing-Xia Zhao
- Laboratory Animal Management Center, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
| | - Wen-Hua Huang
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Medical Biomechanics, National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
- Guangdong Medical Innovation Platform for Translation of 3D Printing Application, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, 510000, China.
- Orthopaedic Center, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, 524001, China.
| | - Dong Xiao
- Laboratory Animal Management Center, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
- Guangzhou Southern Medical Laboratory Animal Sci.&Tech. Co.,Ltd, Guangzhou, 510515, China.
- National Demonstration Center for Experimental Education of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
- Department of Stomatology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
| | - Yan Sun
- Laboratory Animal Management Center, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
| |
Collapse
|
11
|
Tang X, Zhang L, Huang M, Wang F, Xie G, Huo R, Gao R. Selective enhanced cytotoxicity of amino acid deprivation for cancer therapy using thermozyme functionalized nanocatalyst. J Nanobiotechnology 2024; 22:53. [PMID: 38326899 PMCID: PMC10848425 DOI: 10.1186/s12951-024-02326-6] [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: 04/13/2023] [Accepted: 01/30/2024] [Indexed: 02/09/2024] Open
Abstract
BACKGROUND Enzyme therapy based on differential metabolism of cancer cells has demonstrated promising potential as a treatment strategy. Nevertheless, the therapeutic benefit of reported enzyme drugs is compromised by their uncontrollable activity and weak stability. Additionally, thermozymes with high thermal-stability suffer from low catalytic activity at body temperature, preventing them from functioning independently. RESULTS Herein, we have developed a novel thermo-enzymatic regulation strategy for near-infrared (NIR)-triggered precise-catalyzed photothermal treatment of breast cancer. Our strategy enables efficient loading and delivery of thermozymes (newly screened therapeutic enzymes from thermophilic bacteria) via hyaluronic acid (HA)-coupled gold nanorods (GNRs). These nanocatalysts exhibit enhanced cellular endocytosis and rapid enzyme activity enhancement, while also providing biosafety with minimized toxic effects on untargeted sites due to temperature-isolated thermozyme activity. Locally-focused NIR lasers ensure effective activation of thermozymes to promote on-demand amino acid deprivation and photothermal therapy (PTT) of superficial tumors, triggering apoptosis, G1 phase cell cycle arrest, inhibiting migration and invasion, and potentiating photothermal sensitivity of malignancies. CONCLUSIONS This work establishes a precise, remotely controlled, non-invasive, efficient, and biosafe nanoplatform for accurate enzyme therapy, providing a rationale for promising personalized therapeutic strategies and offering new prospects for high-precision development of enzyme drugs.
Collapse
Affiliation(s)
- Xiuhui Tang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Lijuan Zhang
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China
| | - Mingwang Huang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Fang Wang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Guiqiu Xie
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China
| | - Rui Huo
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China
| | - Renjun Gao
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130012, China.
| |
Collapse
|
12
|
Lukácsi S, Munkácsy G, Győrffy B. Harnessing Hyperthermia: Molecular, Cellular, and Immunological Insights for Enhanced Anticancer Therapies. Integr Cancer Ther 2024; 23:15347354241242094. [PMID: 38818970 PMCID: PMC11143831 DOI: 10.1177/15347354241242094] [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: 12/28/2023] [Revised: 02/25/2024] [Accepted: 03/11/2024] [Indexed: 06/01/2024] Open
Abstract
Hyperthermia, the raising of tumor temperature (≥39°C), holds great promise as an adjuvant treatment for cancer therapy. This review focuses on 2 key aspects of hyperthermia: its molecular and cellular effects and its impact on the immune system. Hyperthermia has profound effects on critical biological processes. Increased temperatures inhibit DNA repair enzymes, making cancer cells more sensitive to chemotherapy and radiation. Elevated temperatures also induce cell cycle arrest and trigger apoptotic pathways. Furthermore, hyperthermia modifies the expression of heat shock proteins, which play vital roles in cancer therapy, including enhancing immune responses. Hyperthermic treatments also have a significant impact on the body's immune response against tumors, potentially improving the efficacy of immune checkpoint inhibitors. Mild systemic hyperthermia (39°C-41°C) mimics fever, activating immune cells and raising metabolic rates. Intense heat above 50°C can release tumor antigens, enhancing immune reactions. Using photothermal nanoparticles for targeted heating and drug delivery can also modulate the immune response. Hyperthermia emerges as a cost-effective and well-tolerated adjuvant therapy when integrated with immunotherapy. This comprehensive review serves as a valuable resource for the selection of patient-specific treatments and the guidance of future experimental studies.
Collapse
Affiliation(s)
- Szilvia Lukácsi
- HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- Semmelweis University, Budapest, Hungary
| | - Gyöngyi Munkácsy
- HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- Semmelweis University, Budapest, Hungary
| | - Balázs Győrffy
- HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- Semmelweis University, Budapest, Hungary
- University of Pécs, Pécs, Hungary
- National Laboratory for Drug Research and Development, Budapest, Hungary
| |
Collapse
|
13
|
Malekzadeh R, Mortezazadeh T, Abdulsahib WK, Babaye Abdollahi B, Hamblin MR, Mansoori B, Alsaikhan F, Zeng B. Nanoarchitecture-based photothermal ablation of cancer: A systematic review. ENVIRONMENTAL RESEARCH 2023; 236:116526. [PMID: 37487920 DOI: 10.1016/j.envres.2023.116526] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/17/2023] [Accepted: 06/28/2023] [Indexed: 07/26/2023]
Abstract
Photothermal therapy (PTT) is an emerging non-invasive method used in cancer treatment. In PTT, near-infrared laser light is absorbed by a chromophore and converted into heat within the tumor tissue. PTT for cancer usually combines a variety of interactive plasmonic nanomaterials with laser irradiation. PTT enjoys PT agents with high conversion efficiency to convert light into heat to destroy malignant tissue. In this review, published studies concerned with the use of nanoparticles (NPs) in PTT were collected by a systematic and comprehensive search of PubMed, Cochrane, Embase, and Scopus databases. Gold, silver and iron NPs were the most frequent choice in PTT. The use of surface modified NPs allowed selective delivery and led to a precise controlled increase in the local temperature. The presence of NPs during PTT can increase the reactive generation of oxygen species, damage the DNA and mitochondria, leading to cancer cell death mainly via apoptosis. Many studies recently used core-shell metal NPs, and the effects of the polymer coating or ligands targeted to specific cellular receptors in order to increase PTT efficiency were often reported. The effective parameters (NP type, size, concentration, coated polymers or attached ligands, exposure conditions, cell line or type, and cell death mechanisms) were investigated individually. With the advances in chemical synthesis technology, NPs with different shapes, sizes, and coatings can be prepared with desirable properties, to achieve multimodal cancer treatment with precision and specificity.
Collapse
Affiliation(s)
- Reza Malekzadeh
- Department of Medical Physics, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Medical Radiation Science Research Team, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Tohid Mortezazadeh
- Department of Medical Physics, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Waleed K Abdulsahib
- Department of Pharmacology and Toxicology, College of Pharmacy, Al Farahidi University, Baghdad, Iraq
| | - Behnaz Babaye Abdollahi
- Department of Medical Physics, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, 2028, South Africa
| | - Behzad Mansoori
- The Wistar Institute, Cellular and Molecular Oncogenesis Program, Philadelphia, PA, USA.
| | - Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| | - Bo Zeng
- Department of Thoracic Surgery, The First Affiliated Hospital, Sun Yat-sen University, 510080, Guangzhou, China.
| |
Collapse
|
14
|
Song S, Wang Q, Xie J, Dai J, Ouyang D, Huang G, Guo Y, Chen C, Wu M, Huang T, Ruan J, Cheng X, Lin X, He Y, Rozhkova EA, Chen Z, Yang H. Dual-Responsive Turn-On T 1 Imaging-Guided Mild Photothermia for Precise Apoptotic Cancer Therapy. Adv Healthc Mater 2023; 12:e2301437. [PMID: 37379009 DOI: 10.1002/adhm.202301437] [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: 05/05/2023] [Revised: 06/23/2023] [Accepted: 06/26/2023] [Indexed: 06/29/2023]
Abstract
Apoptosis has gained increasing attention in cancer therapy as an intrinsic signaling pathway, which leads to minimal leakage of waste products from a dying cell to neighboring normal cells. Among various stimuli to trigger apoptosis, mild hyperthermia is attractive but confronts limitations of non-specific heating and acquired resistance from elevated expression of heat shock proteins. Here, a dual-stimulation activated turn-on T1 imaging-based nanoparticulate system (DAS) is developed for mild photothermia (≈43 °C)-mediated precise apoptotic cancer therapy. In the DAS, a superparamagnetic quencher (ferroferric oxide nanoparticles, Fe3 O4 NPs) and a paramagnetic enhancer (Gd-DOTA complexes) are connected via the N6-methyladenine (m6 A)-caged, Zn2+ -dependent DNAzyme molecular device. The substrate strand of the DNAzyme contains one segment of Gd-DOTA complex-labeled sequence and another one of HSP70 antisense oligonucleotide. When the DAS is taken up by cancer cells, overexpressed fat mass and obesity-associated protein (FTO) specifically demethylates the m6 A group, thereby activating DNAzymes to cleave the substrate strand and simultaneously releasing Gd-DOTA complex-labeled oligonucleotides. The restored T1 signal from the liberated Gd-DOTA complexes lights up the tumor to guide the location and time of deploying 808 nm laser irradiation. Afterward, locally generated mild photothermia works in concert with HSP70 antisense oligonucleotides to promote apoptosis of tumor cells. This highly integrated design provides an alternative strategy for mild hyperthermia-mediated precise apoptotic cancer therapy.
Collapse
Affiliation(s)
- Sijie Song
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, and State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350108, P. R. China
| | - Qi Wang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, and State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350108, P. R. China
| | - Jiangao Xie
- Fujian Medical University Union Hospital, Fuzhou, 350108, P. R. China
| | - Junduan Dai
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, and State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350108, P. R. China
| | - Dilan Ouyang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, and State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350108, P. R. China
| | - Guoming Huang
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, P. R. China
| | - Yuheng Guo
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, and State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350108, P. R. China
| | - Chen Chen
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, and State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350108, P. R. China
| | - Mengnan Wu
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, and State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350108, P. R. China
| | - Tingjing Huang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, and State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350108, P. R. China
| | - Jingwen Ruan
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, and State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350108, P. R. China
| | - Xiaofeng Cheng
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, and State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350108, P. R. China
| | - Xucong Lin
- Engineering Technology Research Center on Reagent and Instrument for Rapid Detection of Product Quality and Food Safety in Fujian Province, College of Chemistry, Fuzhou University, Fuzhou, 350108, P. R. China
| | - Yu He
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, and State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350108, P. R. China
| | - Elena A Rozhkova
- Center for Nanoscale Materials, Argonne National Laboratory, Argonne, IL, 60439, USA
| | - Zhaowei Chen
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, and State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350108, P. R. China
| | - Huanghao Yang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, and State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350108, P. R. China
| |
Collapse
|
15
|
Yang Y, Huangfu L, Li H, Yang D. Research progress of hyperthermia in tumor therapy by influencing metabolic reprogramming of tumor cells. Int J Hyperthermia 2023; 40:2270654. [PMID: 37871910 DOI: 10.1080/02656736.2023.2270654] [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: 04/25/2023] [Accepted: 10/09/2023] [Indexed: 10/25/2023] Open
Abstract
Cellular metabolic reprogramming is an important feature of malignant tumors. Metabolic reprogramming causes changes in the levels or types of specific metabolites inside and outside the cell, which affects tumorigenesis and progression by influencing gene expression, the cellular state, and the tumor microenvironment. During tumorigenesis, a series of changes in the glucose metabolism, fatty acid metabolism, amino acid metabolism, and cholesterol metabolism of tumor cells occur, which are involved in the process of cellular carcinogenesis and constitute part of the underlying mechanisms of tumor formation. Hyperthermia, as one of the main therapeutic tools for malignant tumors, has obvious effects on tumor cell metabolism. In this paper, we will combine the latest research progress in the field of cellular metabolic reprogramming and focus on the current experimental research and clinical treatment of hyperthermia in cellular metabolic reprogramming to discuss the feasibility of cellular metabolic reprogramming-related mechanisms guiding hyperthermia in malignant tumor treatment, so as to provide more ideas for hyperthermia to treat malignant tumors through the direction of cellular metabolic reprogramming.
Collapse
Affiliation(s)
- Yuchuan Yang
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| | - Linkuan Huangfu
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| | - Huizhen Li
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| | - Daoke Yang
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| |
Collapse
|
16
|
Ahn CR, Baek SH. Enhancing Gastric Cancer Therapeutic Efficacy through Synergistic Cotreatment of Linderae Radix and Hyperthermia in AGS Cells. Biomedicines 2023; 11:2710. [PMID: 37893084 PMCID: PMC10604735 DOI: 10.3390/biomedicines11102710] [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: 06/26/2023] [Revised: 09/21/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023] Open
Abstract
Gastric cancer remains a global health threat, particularly in Asian countries. Current treatment methods include surgery, chemotherapy, and radiation therapy. However, they all have limitations, such as adverse side effects, tumor resistance, and patient tolerance. Hyperthermia therapy uses heat to selectively target and destroy cancer cells, but it has limited efficacy when used alone. Linderae Radix (LR), a natural compound with thermogenic effects, has the potential to enhance the therapeutic efficacy of hyperthermia treatment. In this study, we investigated the synergistic anticancer effects of cotreatment with LR and 43 °C hyperthermia in AGS gastric cancer cells. The cotreatment inhibited AGS cell proliferation, induced apoptosis, caused cell cycle arrest, suppressed heat-induced heat shock responses, increased reactive oxygen species (ROS) generation, and promoted mitogen-activated protein kinase phosphorylation. N-acetylcysteine pretreatment abolished the apoptotic effect of LR and hyperthermia cotreatment, indicating the crucial role of ROS in mediating the observed anticancer effects. These findings highlight the potential of LR as an adjuvant to hyperthermia therapy for gastric cancer. Further research is needed to validate these findings in vivo, explore the underlying molecular pathways, and optimize treatment protocols for the development of novel and effective therapeutic strategies for patients with gastric cancer.
Collapse
Affiliation(s)
- Chae-Ryeong Ahn
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Seung-Ho Baek
- College of Korean Medicine, Dongguk University, 32 Dongguk-ro, Goyang-si 10326, Republic of Korea
| |
Collapse
|
17
|
Sarogni P, Zamborlin A, Mapanao AK, Logghe T, Brancato L, van Zwol E, Menicagli M, Giannini N, Gonnelli A, Linsalata S, Colenbier R, Van den Bossche J, Paiar F, Bogers J, Voliani V. Hyperthermia Reduces Irradiation-Induced Tumor Repopulation in an In Vivo Pancreatic Carcinoma Model. Adv Biol (Weinh) 2023; 7:e2200229. [PMID: 36861331 DOI: 10.1002/adbi.202200229] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 12/19/2022] [Indexed: 03/03/2023]
Abstract
Pancreatic cancer has a poor prognosis due to its aggressive nature and ability to metastasize at an early stage. Currently, its management is still a challenge because this neoplasm is resistant to conventional treatment approaches, among which is chemo-radiotherapy (CRT), due to the abundant stromal compartment involved in the mechanism of hypoxia. Hyperthermia, among other effects, counteracts hypoxia by promoting blood perfusion and thereby can enhance the therapeutic effect of radiotherapy (RT). Therefore, the establishment of integrated treatments would be a promising strategy for the management of pancreatic carcinoma. Here, the effects of joint radiotherapy/hyperthermia (RT/HT) on optimized chick embryo chorioallantoic membrane (CAM) pancreatic tumor models are investigated. This model enables a thorough assessment of the tumor-arresting effect of the combined approach as well as the quantitative evaluation of hypoxia and cell cycle-associated mechanisms by both gene expression analysis and histology. The analysis of the lower CAM allows to investigate the variation of the metastatic behaviors of the cancer cells associated with the treatments. Overall, this study provides a potentially effective combined strategy for the non-invasive management of pancreatic carcinoma.
Collapse
Affiliation(s)
- Patrizia Sarogni
- Center for Nanotechnology Innovation@NEST, Istituto Italiano di Tecnologia, Piazza San Silvestro 12, Pisa, 56127, Italy
| | - Agata Zamborlin
- Center for Nanotechnology Innovation@NEST, Istituto Italiano di Tecnologia, Piazza San Silvestro 12, Pisa, 56127, Italy
- NEST-Scuola Normale Superiore, Piazza San Silvestro 12, Pisa, 56127, Italy
| | - Ana Katrina Mapanao
- Center for Nanotechnology Innovation@NEST, Istituto Italiano di Tecnologia, Piazza San Silvestro 12, Pisa, 56127, Italy
- Center for Radiopharmaceutical Sciences, Paul Scherrer Institute, 5232 Villigen-PSI, Forschungsstrasse, Switzerland
| | - Tine Logghe
- ElmediX NV, Dellingstraat 34-1, Mechelen, 2800, Belgium
| | | | - Eke van Zwol
- ElmediX NV, Dellingstraat 34-1, Mechelen, 2800, Belgium
| | - Michele Menicagli
- Fondazione Pisana per la Scienza ONLUS, via Ferruccio Giovannini 13, S. Giuliano Terme, Pisa, 56017, Italy
| | - Noemi Giannini
- Center for Nanotechnology Innovation@NEST, Istituto Italiano di Tecnologia, Piazza San Silvestro 12, Pisa, 56127, Italy
- Radiation Oncology Unit, Pisa University Hospital "Azienda Ospedaliero-Universitaria Pisana", Via Roma 67, 56126, Pisa, Italy
| | - Alessandra Gonnelli
- Center for Nanotechnology Innovation@NEST, Istituto Italiano di Tecnologia, Piazza San Silvestro 12, Pisa, 56127, Italy
- Radiation Oncology Unit, Pisa University Hospital "Azienda Ospedaliero-Universitaria Pisana", Via Roma 67, 56126, Pisa, Italy
| | - Stefania Linsalata
- Unit of Medical Physics, Pisa University Hospital "Azienda Ospedaliero-Universitaria Pisana", Pisa, 56126, Italy
| | - Robin Colenbier
- University of Antwerp, Laboratory of Cell Biology and Histology, University of Antwerp, Antwerpen, 2610, Belgium
| | | | - Fabiola Paiar
- Radiation Oncology Unit, Pisa University Hospital "Azienda Ospedaliero-Universitaria Pisana", Via Roma 67, 56126, Pisa, Italy
| | - Johannes Bogers
- ElmediX NV, Dellingstraat 34-1, Mechelen, 2800, Belgium
- University of Antwerp, Laboratory of Cell Biology and Histology, University of Antwerp, Antwerpen, 2610, Belgium
| | - Valerio Voliani
- Center for Nanotechnology Innovation@NEST, Istituto Italiano di Tecnologia, Piazza San Silvestro 12, Pisa, 56127, Italy
- Department of Pharmacy, University of Genoa, Viale Cembrano, 4, Genoa, 16148, Italy
| |
Collapse
|
18
|
Gelissen JH, Adjei NN, McNamara B, Mutlu L, Harold JA, Clark M, Altwerger G, Dottino PR, Huang GS, Santin AD, Azodi M, Ratner E, Schwartz PE, Andikyan V. Hyperthermic Intraperitoneal Chemotherapy in Ovarian Cancer. Ann Surg Oncol 2023; 30:5597-5609. [PMID: 37358686 DOI: 10.1245/s10434-023-13757-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 06/04/2023] [Indexed: 06/27/2023]
Abstract
Hyperthermic intraperitoneal chemotherapy (HIPEC) is a treatment modality that aims to target the main site of tumor dissemination in ovarian cancer, the peritoneum, by combining the benefits of intraperitoneal chemotherapy with the synergistic effects of hyperthermia all during a single administration at the time of cytoreductive surgery. High-quality evidence currently only supports the use of HIPEC with cisplatin at the time of interval cytoreduction after neoadjuvant chemotherapy for stage III epithelial ovarian cancer. Many questions remain, including HIPEC's role at other timepoints in ovarian cancer treatment, who are optimal candidates, and specifics of HIPEC protocols. This article reviews the history of normothermic and hyperthermic intraperitoneal chemotherapy in ovarian cancer and evidence regarding HIPEC implementation and patient outcomes. Additionally, this review explores details of HIPEC technique and perioperative care, cost considerations, complication and quality of life data, disparities in HIPEC use, and unresolved issues.
Collapse
Affiliation(s)
- Julia H Gelissen
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA.
| | - Naomi N Adjei
- Gynecologic Oncology and Reproductive Medicine Department, MD Anderson Cancer Center, Houston, TX, USA
| | - Blair McNamara
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Levent Mutlu
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Justin A Harold
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Mitchell Clark
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Gary Altwerger
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Peter R Dottino
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Gloria S Huang
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Alessandro D Santin
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Masoud Azodi
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Elena Ratner
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Peter E Schwartz
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Vaagn Andikyan
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
19
|
Chiang CF, Wang ZZ, Hsu YH, Miaw SC, Lin WL. Exercise improves the outcome of anticancer treatment with ultrasound-hyperthermia-enhanced nanochemotherapy and autophagy inhibitor. PLoS One 2023; 18:e0288380. [PMID: 37437011 DOI: 10.1371/journal.pone.0288380] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 06/21/2023] [Indexed: 07/14/2023] Open
Abstract
It has been shown that exercise has a direct impact on tumor growth along with functional improvement. Previous studies have shown that exercise decreases the risk of cancer recurrence across various types of cancer. It was indicated that exercise stimulates the immune system to fight cancer. Previous study demonstrated that pulsed-wave ultrasound hyperthermia (pUH) combined with PEGylated liposomal doxorubicin (PLD) and chloroquine (CQ) inhibits 4T1 tumors growth and delays their recurrence. In this study, we investigated if the combinatorial treatment with high-intensity interval training (HIIT) combined with pUH-enhanced PLD delivery and CQ improved the outcome. The mouse experiment composed of three groups, HIIT+PLD+pUH+CQ group, PLD+pUH+CQ group, and the control group. HIIT+PLD+pUH+CQ group received 6 weeks of HIIT (15 min per day, 5 days per week) before 4T1 tumor implantation. Seven days later, they received therapy with PLD (10 mg/kg) + pUH (3 MHz, 50% duty cycle, 0.65 W/cm2, 15min) + CQ (50 mg/kg daily). Results showed that HIIT+PLD+pUH+CQ significantly reduced the tumor volumes and brought about longer survival of tumor-bearing mice than PLD+pUH+CQ did. Blood cell components were analyzed and showed that neutrophil and reticulocytes decreased while lymphocytes increased after exercise.
Collapse
Affiliation(s)
- Chi-Feng Chiang
- Department of Biomedical Engineering, National Taiwan University, Taipei, Taiwan
| | - Zi-Zong Wang
- Department of Biomedical Engineering, National Taiwan University, Taipei, Taiwan
| | - Yu-Hone Hsu
- Division of Neurosurgery, Department of Surgery, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Shi-Chuen Miaw
- Graduate Institute of Immunology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Win-Li Lin
- Department of Biomedical Engineering, National Taiwan University, Taipei, Taiwan
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Zhunan Town, Miaoli, Taiwan
| |
Collapse
|
20
|
Wang T, Wu C, Hu Y, Zhang Y, Ma J. Stimuli-responsive nanocarrier delivery systems for Pt-based antitumor complexes: a review. RSC Adv 2023; 13:16488-16511. [PMID: 37274408 PMCID: PMC10233443 DOI: 10.1039/d3ra00866e] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 03/30/2023] [Indexed: 06/06/2023] Open
Abstract
Platinum-based anticancer drugs play a crucial role in the clinical treatment of various cancers. However, the application of platinum-based drugs is heavily restricted by their severe toxicity and drug resistance/cross resistance. Various drug delivery systems have been developed to overcome these limitations of platinum-based chemotherapy. Stimuli-responsive nanocarrier drug delivery systems as one of the most promising strategies attract more attention. And huge progress in stimuli-responsive nanocarrier delivery systems of platinum-based drugs has been made. In these systems, a variety of triggers including endogenous and extracorporeal stimuli have been employed. Endogenous stimuli mainly include pH-, thermo-, enzyme- and redox-responsive nanocarriers. Extracorporeal stimuli include light-, magnetic field- and ultrasound responsive nanocarriers. In this review, we present the recent advances in stimuli-responsive drug delivery systems with different nanocarriers for improving the efficacy and reducing the side effects of platinum-based anticancer drugs.
Collapse
Affiliation(s)
- Tianshuai Wang
- Hubei Key Lab of Wudang Local Chinese Medicine Research, Hubei University of Medicine Shiyan 442000 Hubei China
- College of Pharmaceutical Sciences, Hubei University of Medicine Shiyan 442000 Hubei China
| | - Chen Wu
- College of Pharmaceutical Sciences, Hubei University of Medicine Shiyan 442000 Hubei China
| | - Yanggen Hu
- Hubei Key Lab of Wudang Local Chinese Medicine Research, Hubei University of Medicine Shiyan 442000 Hubei China
- College of Pharmaceutical Sciences, Hubei University of Medicine Shiyan 442000 Hubei China
| | - Yan Zhang
- College of Pharmaceutical Sciences, Hubei University of Medicine Shiyan 442000 Hubei China
| | - Junkai Ma
- Hubei Key Lab of Wudang Local Chinese Medicine Research, Hubei University of Medicine Shiyan 442000 Hubei China
- College of Pharmaceutical Sciences, Hubei University of Medicine Shiyan 442000 Hubei China
| |
Collapse
|
21
|
Nguyen QA, Chou WH, Hsieh MC, Chang CM, Luo WT, Tai YT, Chang WC. Genetic alterations in peritoneal metastatic tumors predicted the outcomes for hyperthermic intraperitoneal chemotherapy. Front Oncol 2023; 13:1054406. [PMID: 37182141 PMCID: PMC10170308 DOI: 10.3389/fonc.2023.1054406] [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: 09/26/2022] [Accepted: 03/27/2023] [Indexed: 05/16/2023] Open
Abstract
Introduction Cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) are considered for patients with peritoneal metastasis (PM). However, patients selection that relies on conventional prognostic factors is not yet optimal. In this study, we performed whole exome sequencing (WES) to establish tumor molecular characteristics and expect to identify prognosis profiles for PM management. Methods In this study, blood and tumor samples were collected from patients with PM before HIPEC. Tumor molecular signatures were determined using WES. Patient cohort was divided into responders and non-responders according to 12-month progression-free survival (PFS). Genomic characteristics between the two cohorts were compared to study potential targets. Results In total, 15 patients with PM were enrolled in this study. Driver genes and enriched pathways were identified from WES results. AGAP5 mutation was found in all responders. This mutation was significantly associated with better OS (p = 0.00652). Conclusions We identified prognostic markers that might be useful to facilitate decision-making before CRS/HIPEC.
Collapse
Affiliation(s)
- Quynh-Anh Nguyen
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Wan-Hsuan Chou
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Mao-Chih Hsieh
- Department of General Surgery, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Che-Mai Chang
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Wei-Tzu Luo
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Yu-Ting Tai
- Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Anesthesiology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Wei-Chiao Chang
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taipei, Taiwan
- Master Program in Clinical Genomics and Proteomics, School of Pharmacy, Taipei Medical University, Taipei, Taiwan
- Department of Pharmacy, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Integrative Research Center for Critical Care, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
22
|
Montazersaheb P, Pishgahzadeh E, Jahani VB, Farahzadi R, Montazersaheb S. Magnetic nanoparticle-based hyperthermia: A prospect in cancer stem cell tracking and therapy. Life Sci 2023; 323:121714. [PMID: 37088411 DOI: 10.1016/j.lfs.2023.121714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 04/10/2023] [Accepted: 04/18/2023] [Indexed: 04/25/2023]
Abstract
Tumor heterogeneity is a major problem in cancer treatment. Cancer stem cells (CSCs) are a subpopulation of tumor masses that produce proliferating and quiescent cells. Under stress-related conditions, quiescent cells are capable of repopulating tumor masses. Consequently, many attempts have been made to identify, isolate, and eradicate CSCs from various tumors. Research has found that quiescent CSCs are less susceptible to conventional therapy than bulk cancer cells. This could be due to reduced cell cycling and increased DNA repair capacity of these cells. Indeed, disease progression is temporarily suppressed by eliminating fast-proliferating tumor cells and sparing quiescent CSCs lead to cancer relapse. Among all the available therapeutic modalities for cancer treatment, hyperthermia uses moderate heat to kill tumor cells. Nanoparticle-based platforms have the potential to deposit heat locally and selectively with the simultaneous activation of nanoparticles as heat transducers. Over the past few decades, magnetic nanoparticles (MNPs) have been widely investigated in the biomedical field. Magnetic hyperthermia therapy (MHT) is a promising therapeutic approach in which MNPs are delivered directly through targeting (systemic) or by direct injection into a tumor under exposure to an alternating magnetic field (AMF). Heat is generated by the MNPs subjected to AMF at a frequency of 100 kHz. Despite the widespread use of MHT alone or in combination therapies, its effectiveness in targeting CSCs remains unclear. This review discusses various types of MHT and their related mechanisms in cancer therapy, particularly concerning the eradication of CSCs.
Collapse
Affiliation(s)
- Parsa Montazersaheb
- Department of Materials Engineering, Institute of Mechanical Engineering, University of Tabriz, Tabriz, Iran
| | - Elahe Pishgahzadeh
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Vahid Bayrami Jahani
- School of Metallurgy and Materials Engineering, Iran University of Science and Technology (IUST), Narmak, Tehran, Iran
| | - Raheleh Farahzadi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Soheila Montazersaheb
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
23
|
Mohammadi A, Eivazzadeh-Keihan R, Aliabadi HAM, Kashtiaray A, Cohan RA, Bani MS, Komijani S, Etminan A, salehpour N, Maleki A, Mahdavi M. Magnetic carboxymethyl cellulose-silk fibroin hydrogel: a ternary nanobiocomposite exhibiting excellent biological activity and in vitro hyperthermia of cancer therapy. J Biotechnol 2023; 367:71-80. [PMID: 37028560 DOI: 10.1016/j.jbiotec.2023.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 03/30/2023] [Accepted: 04/03/2023] [Indexed: 04/08/2023]
Abstract
In this work, a magnetic nanobiocomposite scaffold based on carboxymethylcellulose (CMC) hydrogel, silk fibroin (SF), and magnetite nanoparticles was fabricated. The structural properties of this new magnetic nanobiocomposite were characterized by various analyses such as FT-IR, XRD, EDX, FE-SEM, TGA and VSM. According to the particle size histogram, most of the particles were between 55-77nm and the value of saturation magnetization of this nanobiocomposite was reported 41.65emu.g- 1. Hemolysis and MTT tests showed that the designed magnetic nanobiocomposite was compatible with the blood. In addition, the viability percentage of HEK293T normal cells did not change significantly, and the proliferation rate of BT549 cancer cells decreased in its vicinity. EC50 values for HEK293T normal cells after 48h and 72h were 3958 and 2566, respectively. Also, these values for BT549 cancer cells after 48h and 72h were 0.4545 and 0.9967, respectively. The efficiency of fabricated magnetic nanobiocomposite was appraised in a magnetic fluid hyperthermia manner. The specific absorption rate (SAR) of 69W/g (for the 1mg/mL sample at 200kHz) was measured under the alternating magnetic field (AMF).
Collapse
|
24
|
Peng C, Li X, Ao F, Li T, Guo J, Liu J, Zhang X, Gu J, Mao J, Zhou B. Mitochondrial ROS driven by NOX4 upregulation promotes hepatocellular carcinoma cell survival after incomplete radiofrequency ablation by inducing of mitophagy via Nrf2/PINK1. J Transl Med 2023; 21:218. [PMID: 36964576 PMCID: PMC10039571 DOI: 10.1186/s12967-023-04067-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 03/17/2023] [Indexed: 03/26/2023] Open
Abstract
BACKGROUND The recurrence of hepatocellular carcinoma (HCC) after radiofrequency ablation (RFA) remains a major clinical problem. Cells that survive the sublethal heat stress that is induced by incomplete RFA are the main source of HCC relapse. Heat stress has long been reported to increase intracellular reactive oxygen species (ROS) generation. Although ROS can induce apoptosis, a pro-survival effect of ROS has also been demonstrated. However, the role of ROS in HCC cells exposed to sublethal heat stress remains unclear. METHODS HepG2 and HuH7 cells were used for this experiment. Insufficient RFA was performed in cells and in a xenograft model. ROS and antioxidant levels were measured. Apoptosis was analyed by Annexin-V/PI staining and flow cytometry. Protein expression was measured using western blotting. Colocalization of lysosomes and mitochondria was analyzed to assess mitophagy. Corresponding activators or inhibitors were applied to verify the function of specific objectives. RESULTS Here,we showed that sublethal heat stress induced a ROS burst, which caused acute oxidative stress. This ROS burst was generated by mitochondria, and it was initiated by upregulated NOX4 expression in the mitochondria. N-acetylcysteine (NAC) decreased HCC cell survival under sublethal heat stress conditions in vivo and in vitro. NOX4 triggers the production of mitochondrial ROS (mtROS), and NOX4 inhibitors or siNOX4 also decreased HCC cell survival under sublethal heat stress conditions in vitro. Increased mtROS trigger PINK1-dependent mitophagy to eliminate the mitochondria that are damaged by sublethal heat stress and to protect cells from apoptosis. Nrf2 expression was elevated in response to this ROS burst and mediated the ROS burst-induced increase in PINK1 expression after sublethal heat stress. CONCLUSION These data confirmed that the ROS burst that occurs after iRFA exerted a pro-survival effect. NOX4 increased the generation of ROS by mitochondria. This short-term ROS burst induced PINK1-dependent mitophagy to eliminate damaged mitochondria by increasing Nrf2 expression.
Collapse
Affiliation(s)
- Chao Peng
- Department of Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, Guangdong, China
| | - Xi Li
- Department of Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, Guangdong, China
| | - Feng Ao
- Department of Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, Guangdong, China
| | - Ting Li
- Department of Anesthesiology, Gansu Provincial People's Hospital, Lanzhou, 730000, Gansu, China
| | - Jingpei Guo
- Department of Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, Guangdong, China
| | - Junfeng Liu
- Department of Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, Guangdong, China
| | - Xiaoting Zhang
- Department of Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, Guangdong, China
| | - Jinyan Gu
- Library Department, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, Guangdong, China.
| | - Junjie Mao
- Department of Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, Guangdong, China.
| | - Bin Zhou
- Department of Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, Guangdong, China.
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, Guangdong, China.
| |
Collapse
|
25
|
Anup N, Gadeval A, Tekade RK. A 3D-Printed Graphene BioFuse Implant for Postsurgical Adjuvant Therapy of Cancer: Proof of Concept in 2D- and 3D-Spheroid Tumor Models. ACS APPLIED BIO MATERIALS 2023; 6:1195-1212. [PMID: 36893437 DOI: 10.1021/acsabm.2c01031] [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: 03/11/2023]
Abstract
Three-dimensional printing is an emerging technology that is finding its niche applications in diverse fields owing to its flexibility concerning personalization and design. Surgery followed by adjuvant therapy is the standard treatment plan in most cancers from stage I to stage III. Most of the available adjuvant therapies, like chemotherapy, radiation therapy, immunotherapy, hormonal therapy, etc., are associated with severe side effects that considerably reduce the quality of life of patients. In addition, there is always the chance of tumor recurrence or metastasis development followed by surgery. This investigation reports the development of a 3D-printed, biodegradable, laser-responsive implant with a chemo-combined thermal ablating potential for adjuvant therapy of cancer. The 3D-printable ink was developed using poly(l-lactide) and hydroxypropyl methylcellulose as the base polymer, doxorubicin as the chemotherapeutic agent, and reduced graphene oxide as the photothermal ablating agent. The personalized implant released the drug pH-dependently (p value < 0.0001) for an extended period (93.55 ± 1.80% → 28 days). The 3D-printed implant exhibited acceptable biophysical properties (tensile strength: 3.85 ± 0.15 MPa; modulus: 92.37 ± 11.50 MPa; thickness: 110 μm) with laser-responsive hyperthermia (ΔT: 37 ± 0.9 °C → 48.5 ± 1.07 °C; 5 min; 1.5 W/cm2) and inherent biodegradable property (SEM analysis). The 3D-printed implant was evaluated for its therapeutic potential in 2D- and 3D-spheroid tumor models (MDA-MB 231 and SCC 084 2D cells) employing MTT cytotoxicity assay, apoptosis assay, cell cycle analysis, and gene expression analysis. The biomolecular aspects and biomechanics of the 3D-printed BioFuse implant were also evaluated by determining the effect of treatment on the expression levels of HSP1A, Hsp70, BAX, and PTEN. It is advocated that the knowledge developed in this project will significantly assist and advance the science aiming to develop a clinically translatable postsurgical adjuvant therapy for cancer.
Collapse
Affiliation(s)
- Neelima Anup
- National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, An Institute of National Importance, Government of India, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Palaj, Opp. Air force station, Gandhinagar 382355, Gujarat, India
| | - Anuradha Gadeval
- National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, An Institute of National Importance, Government of India, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Palaj, Opp. Air force station, Gandhinagar 382355, Gujarat, India
| | - Rakesh Kumar Tekade
- National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, An Institute of National Importance, Government of India, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Palaj, Opp. Air force station, Gandhinagar 382355, Gujarat, India
| |
Collapse
|
26
|
Ruvalcaba-Ontiveros RI, Murillo-Ramírez JG, Medina-Vázquez JA, Carrasco-Hernández AR, Duarte-Möller JA, Esparza-Ponce HE. Synthesis of gold decorated silica nanoparticles and their photothermal properties. Micron 2023; 166:103415. [PMID: 36657307 DOI: 10.1016/j.micron.2023.103415] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 11/16/2022] [Accepted: 01/13/2023] [Indexed: 01/18/2023]
Abstract
Silica-Gold Nanostructures (SGNs), composed of a silica core decorated by gold nanoparticles, have the photothermal capacity to transform near-infrared (NIR) wavelengths into heat. This work presents a simple, efficient, and replicable method of synthesis of SGNs and their characterization by: (1) transmission electron microscopy to obtain micrographs of the particles and their corresponding diameter distribution; (2) diffraction patterns showing the amorphous atomic arraignment of the silica and the crystalline atomic arrangement of the gold nanoparticles; (3) zeta potential confirming the stability of the SGNs in a colloidal solution; and (4) thermal images displaying the capacity of SGNs to convert NIR irradiation into heat and their respective increment in temperature. SGNs were synthesized over silica cores with diameters of 63, 83, and 132 nm and decorated with a partial gold shell. They were heated with a coherent light intensity of 340 mW/cm2 with a wavelength of 852 nm. This wavelength is within the range of the optical window of the human body; therefore, SGNs may be used for the photothermal ablation of tumors with no damage to the tissue. The heating of different dimensions of SGNs took 6-8 min of NIR radiation, and their cooling, once the laser was turned off, was in the order of 2-3 min. It was found that SGNs, with a core diameter of 132 nm, have a notable photothermal capacity. That enables them to increase the temperature of their surroundings by 4.4 ºC. This increment in temperature is sufficient to induce cellular necrosis, which makes SGNs a good option for photothermal treatments.
Collapse
Affiliation(s)
| | - José G Murillo-Ramírez
- Centro de Investigación en Materiales Avanzados, Complejo Industrial Chihuahua, 31136 Chihuahua, Mexico
| | - José A Medina-Vázquez
- Centro de Investigación en Materiales Avanzados, Complejo Industrial Chihuahua, 31136 Chihuahua, Mexico
| | - Anel R Carrasco-Hernández
- Centro de Investigación en Materiales Avanzados, Complejo Industrial Chihuahua, 31136 Chihuahua, Mexico
| | - José A Duarte-Möller
- División de Ciencias e Ingeniería. Universidad de Sonora, Unidad Regional Sur Lázaro Cárdenas del Río 100, Colonia Francisco Villa, 85880 Navojoa, Sonora, Mexico
| | - Hilda E Esparza-Ponce
- Centro de Investigación en Materiales Avanzados, Complejo Industrial Chihuahua, 31136 Chihuahua, Mexico.
| |
Collapse
|
27
|
Ponciri Fructus Immatarus Sensitizes the Apoptotic Effect of Hyperthermia Treatment in AGS Gastric Cancer Cells through ROS-Dependent HSP Suppression. Biomedicines 2023; 11:biomedicines11020405. [PMID: 36830941 PMCID: PMC9953356 DOI: 10.3390/biomedicines11020405] [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: 12/20/2022] [Revised: 01/21/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
Gastric cancer has been associated with a high incidence and mortality, accompanied by a poor prognosis. Given the limited therapeutic options to treat gastric cancer, alternative treatments need to be urgently developed. Hyperthermia therapy is a potentially effective and safe treatment option for cancer; however, certain limitations need to be addressed. We applied 43 °C hyperthermia to AGS gastric cancer cells combined with Ponciri Fructus Immaturus (PF) to establish their synergistic effects. Co-treatment with PF and hyperthermia synergistically suppressed AGS cell proliferation by inducing extrinsic and intrinsic apoptotic pathways. Additionally, PF and hyperthermia suppressed factors related to metastasis. Cell cycle arrest was determined by flow cytometry, revealing that co-treatment induced arrest at the G2/M phase. As reactive oxygen species (ROS) are critical in hyperthermia therapy, we next examined changes in ROS generation. Co-treatment with PF and hyperthermia increased ROS levels, and apoptotic induction mediated by this combination was partially dependent on ROS generation. Furthermore, heat shock factor 1 and heat shock proteins (HSPs) were notably suppressed following co-treatment with PF and hyperthermia. The HSP-regulating effect was also dependent on ROS generation. Overall, these findings suggest that co-treatment with PF and hyperthermia could afford a promising anticancer therapy for gastric cancer.
Collapse
|
28
|
Ran J, Liu T, Song C, Wei Z, Tang C, Cao Z, Zou H, Zhang X, Cai Y, Han W. Rhythm Mild-Temperature Photothermal Therapy Enhancing Immunogenic Cell Death Response in Oral Squamous Cell Carcinoma. Adv Healthc Mater 2023; 12:e2202360. [PMID: 36401600 DOI: 10.1002/adhm.202202360] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/09/2022] [Indexed: 11/20/2022]
Abstract
The low antitumor efficiency and unexpected thermo-tolerance activation of mild-temperature photothermal therapy (mPTT) severely impede the therapeutic efficacy, thereby the implementation of reasonable mPTT procedure to improve antitumor efficiency is of great significance for clinical transformation. Herein, a rhythm mPTT with organic photothermal nanoparticles (PBDB-T NPs) is demonstrated, synergistically increasing tumor elimination and intense immunogenic cancer cell death (ICD) to elicit tumor-specific immune responses for tumor treatment. Specifically, PBDB-T NPs are characterized by favorable biocompatibility, excellent and controllable photothermal properties, exhibit the properties of noninvasive diagnostic imaging, and effective mPTT against oral squamous cell carcinoma (OSCC). Encouragingly, a temperature-dependent release of damage-associated molecular patterns (DAMPs) is discovered during the mPTT-induced ICD. Meanwhile, orchestrated rhythm mPTT referring to radiotherapy procedure amplifies and balances antitumor efficiency and abundant DAMPs generation to gain optimal immune activation within clinical-recommended hyperthermia temperature compared with conventional PTT. The in vitro and in vivo results show that the rhythm mPTT unites the killing effect and ICD induction, generating strong mPTT efficacy and active tumor-specific adaptive immune responses. The study offers a promising strategy and a new opportunity for the clinical application of mPTT.
Collapse
Affiliation(s)
- Jianchuan Ran
- Nanjing Stomatological Hospital, Medical School of Nanjing University, 30 Zhongyang Road, Nanjing, 210008, China
| | - Tao Liu
- Department of Oral Medicine, Nanjing Stomatological Hospital, Medical School of Nanjing University, 30 Zhongyang Road, Nanjing, 210008, China
| | - Chuanhui Song
- Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Zheng Wei
- Nanjing Stomatological Hospital, Medical School of Nanjing University, 30 Zhongyang Road, Nanjing, 210008, China
| | - Chuanchao Tang
- Nanjing Stomatological Hospital, Medical School of Nanjing University, 30 Zhongyang Road, Nanjing, 210008, China
| | - Zichen Cao
- Department of Oral Medicine, Nanjing Stomatological Hospital, Medical School of Nanjing University, 30 Zhongyang Road, Nanjing, 210008, China
| | - Huihui Zou
- Nanjing Stomatological Hospital, Medical School of Nanjing University, 30 Zhongyang Road, Nanjing, 210008, China
| | - Xinyu Zhang
- Department of Oral Medicine, Nanjing Stomatological Hospital, Medical School of Nanjing University, 30 Zhongyang Road, Nanjing, 210008, China
| | - Yu Cai
- Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China
| | - Wei Han
- Nanjing Stomatological Hospital, Medical School of Nanjing University, 30 Zhongyang Road, Nanjing, 210008, China
| |
Collapse
|
29
|
Shields NJ, Peyroux EM, Campbell K, Mehta S, Woolley AG, Counoupas C, Neumann S, Young SL. Calpains Released from Necrotic Tumor Cells Enhance Antigen Cross-Presentation to Activate CD8 +T Cells In Vitro. THE JOURNAL OF IMMUNOLOGY 2022; 209:1635-1651. [DOI: 10.4049/jimmunol.2100500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 08/19/2022] [Indexed: 11/07/2022]
|
30
|
Madani F, Esnaashari SS, Webster TJ, Khosravani M, Adabi M. Polymeric nanoparticles for drug delivery in glioblastoma: State of the art and future perspectives. J Control Release 2022; 349:649-661. [PMID: 35878729 DOI: 10.1016/j.jconrel.2022.07.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 07/16/2022] [Accepted: 07/19/2022] [Indexed: 11/16/2022]
Abstract
Glioblastoma (GBM) is an aggressive, fatal and malignant primary brain tumor. Despite the current standard treatment for glioblastoma patients including neurosurgical resection, followed by concomitant radiation and chemotherapy, the median survival rate is only about 15 months. An unresolved challenge for current therapies is related to getting drugs through the blood-brain barrier (BBB), which hinders many chemotherapeutic agents from reaching tumors cells. Although a large amount of research has been done to circumvent the BBB and deliver drugs to the brain, with nanoparticles (NPs) taking the lead, the challenge is still high. In this regard, the BBB and how to transfer drug pathways through the BBB, especially using NPs, are introduced here. Afterwards, the latest advances in drug delivery, co-drug delivery, and combination modalities are described specifically for GBM treatments using natural and synthetic polymeric NPs and adjuvant therapies including hyperthermia, photodynamic therapy and also ketogenic regimens. In addition, receptor-mediated endocytosis agents that exist in endothelial capillary cells of the brain are explained. Lastly, future directions to finally deliver drugs through the BBB for GBM treatment are emphasized. It is the hope that this review can provide a number of practical pathways for the future development of BBB permeable nanochemotherapeutics against GBM.
Collapse
Affiliation(s)
- Fatemeh Madani
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyedeh Sara Esnaashari
- Department of Medical Nanotechnology, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Thomas J Webster
- School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin, China
| | - Masood Khosravani
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mahdi Adabi
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran; Food Microbiology Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
31
|
Shi L, Cao H, Fu S, Jia Z, Lu X, Cui Z, Yu D. Cordycepin enhances hyperthermia-induced apoptosis and cell cycle arrest by modulating the MAPK pathway in human lymphoma U937 cells. Mol Biol Rep 2022; 49:8673-8683. [PMID: 35763180 DOI: 10.1007/s11033-022-07705-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 06/14/2022] [Indexed: 12/22/2022]
Abstract
BACKGROUND Hyperthermia induces cancer cell death. However, the cytotoxic effect of hyperthermia is not sufficient. Cordycepin can also induce apoptosis in cancer cells and enhance the antitumoral activity of irradiation. To examine cordycepin-mediated enhancement of hyperthermia-induced apoptosis, this study investigated the combined effects and apoptotic mechanisms of hyperthermia and cordycepin on human leukemia U937 cells. METHODS Cell viability and apoptosis were measured using MTT assays, Hoechst 33342 staining and Annexin V/PI double staining. The distribution of the cell cycle and sub-G1 phase, reactive oxygen species (ROS) and mitochondrial membrane potential (MMP) were examined by flow cytometry. The expression of related proteins was analyzed by western blotting. RESULTS Combined treatment with hyperthermia and cordycepin markedly augmented apoptosis by upregulating Bax and suppressing Bcl-2, Bid and activated caspase 3 and 8 expression, and apoptosis was decreased by Z-VAD-fmk (a pan caspase inhibitor). We also found that the MMP was significantly decreased and excessive ROS generation occurred. The combination treatment also induced arrest in the G2/M phase by downregulating cyclin dependent kinase 1 (CDK1) and cyclin B1 protein expression. Furthermore, it was observed that mitogen-activated protein kinase (MAPK) pathway including ERK, JNK and p38 signals was involved in the induction of apoptosis. The phosphorylated p38 and JNK were increased and ERK phosphorylation was decreased by the combined treatment. In addition, N-acetyl-L-cysteine (NAC) significantly protected the cells by restoring ROS levels and the activity of caspase-3, inactivating the MAPK pathway. CONCLUSION Cordycepin significantly enhanced hyperthermia-induced apoptosis and G2/M phase arrest in U937 cells. The combined treatment enhanced apoptosis through the MAPK pathway and mitochondrial dysfunction, and these effects could be rescued by NAC. We report for the first time that cordycepin can be used as a hyperthermia sensitizer to treat leukemia.
Collapse
Affiliation(s)
- Liying Shi
- The School of Life Science and Biotechnology, Dalian University, Dalian, 116622, People's Republic of China
| | - He Cao
- The School of Life Science and Biotechnology, Dalian University, Dalian, 116622, People's Republic of China
| | - Siyu Fu
- The School of Life Science and Biotechnology, Dalian University, Dalian, 116622, People's Republic of China
| | - Zixian Jia
- The School of Life Science and Biotechnology, Dalian University, Dalian, 116622, People's Republic of China
| | - Xuan Lu
- The School of Life Science and Biotechnology, Dalian University, Dalian, 116622, People's Republic of China
| | - Zhengguo Cui
- Department of Environmental Health, University of Fukui School of Medical Science, University of Fukui, Eiheiji, 910-1193, Japan.
| | - Dayong Yu
- The School of Life Science and Biotechnology, Dalian University, Dalian, 116622, People's Republic of China.
| |
Collapse
|
32
|
Wu Q, Wei M, Yao L, Cheng X, Lu W, Xie X, Li X. Hyperthermia synergistically enhances antitumor efficacy of PARP inhibitor through impacting homologous recombination repair and oxidative stress in vitro. Biochem Biophys Res Commun 2022; 619:49-55. [PMID: 35738064 DOI: 10.1016/j.bbrc.2022.06.049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/07/2022] [Accepted: 06/15/2022] [Indexed: 11/28/2022]
Abstract
Tumors with homologous recombination (HR) deficiency are particularly responsive to PARP inhibitors, however strategies to improve the sensitivity of epithelial ovarian carcinoma (EOC) with sufficient HR abilities still need to be deeply explored. In the present study, we firstly validated that hyperthermia (HT) changed diverse genes and signal pathways related to HR and oxidative stress in HR proficient EOC cells. HT impaired HR efficiency through inhibiting Olaparib (Olap) induced RAD51 foci formation in EOC cells, which was independent of the expression level of RAD51. Combination therapy of HT and Olap synergistically induced oxidative stress and oxidative DNA damage of EOC cells. Furthermore, we revealed that HT and Olap synergistically aggravated double-strand breaks of DNA in EOC cells. Conclusively, our findings confirmed that HT could synergistically enhance HR proficient EOC cells' sensitivity to PARP inhibitor through impairing HR efficiency and increasing oxidative stress.
Collapse
Affiliation(s)
- Qianqian Wu
- Department of Gynecologic Oncology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310006, China; Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310006, China
| | - Mingjing Wei
- Department of Gynecologic Oncology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310006, China; Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310006, China
| | - Lifang Yao
- Department of Gynecologic Oncology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310006, China; Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310006, China; Department of Obstetrics and Gynecology, Shaoxing Maternal and Child Health Hospital, Shaoxing, Zhejiang, 312000, China
| | - Xiaodong Cheng
- Department of Gynecologic Oncology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310006, China; Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310006, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Weiguo Lu
- Department of Gynecologic Oncology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310006, China; Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310006, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, China; Center for Uterine Cancer Diagnosis & Therapy Research of Zhejiang Province, Hangzhou, 310006, China
| | - Xing Xie
- Department of Gynecologic Oncology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310006, China; Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310006, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Xiao Li
- Department of Gynecologic Oncology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310006, China; Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310006, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, China.
| |
Collapse
|
33
|
Mohanty A, Parida A, Raut RK, Behera RK. Ferritin: A Promising Nanoreactor and Nanocarrier for Bionanotechnology. ACS BIO & MED CHEM AU 2022; 2:258-281. [PMID: 37101573 PMCID: PMC10114856 DOI: 10.1021/acsbiomedchemau.2c00003] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
The essence of bionanotechnology lies in the application of nanotechnology/nanomaterials to solve the biological problems. Quantum dots and nanoparticles hold potential biomedical applications, but their inherent problems such as low solubility and associated toxicity due to their interactions at nonspecific target sites is a major concern. The self-assembled, thermostable, ferritin protein nanocages possessing natural iron scavenging ability have emerged as a potential solution to all the above-mentioned problems by acting as nanoreactor and nanocarrier. Ferritins, the cellular iron repositories, are hollow, spherical, symmetric multimeric protein nanocages, which sequester the excess of free Fe(II) and synthesize iron biominerals (Fe2O3·H2O) inside their ∼5-8 nm central cavity. The electrostatics and dynamics of the pore residues not only drives the natural substrate Fe2+ inside ferritin nanocages but also uptakes a set of other metals ions/counterions during in vitro synthesis of nanomaterial. The current review aims to report the recent developments/understanding on ferritin structure (self-assembly, surface/pores electrostatics, metal ion binding sites) and chemistry occurring inside these supramolecular protein cages (protein mediated metal ion uptake and mineralization/nanoparticle formation) along with its surface modification to exploit them for various nanobiotechnological applications. Furthermore, a better understanding of ferritin self-assembly would be highly useful for optimizing the incorporation of nanomaterials via the disassembly/reassembly approach. Several studies have reported the successful engineering of these ferritin protein nanocages in order to utilize them as potential nanoreactor for synthesizing/incorporating nanoparticles and as nanocarrier for delivering imaging agents/drugs at cell specific target sites. Therefore, the combination of nanoscience (nanomaterials) and bioscience (ferritin protein) projects several benefits for various applications ranging from electronics to medicine.
Collapse
|
34
|
NIR-Mediated drug release and tumor theranostics using melanin-loaded liposomes. Biomater Res 2022; 26:22. [PMID: 35659113 PMCID: PMC9164422 DOI: 10.1186/s40824-022-00270-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 05/23/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Heat generation in a drug delivery carrier by exposure to near-infrared (NIR) light with excellent tissue transmittance is an effective strategy for drug release and tumor therapy. Because liposomes have amphiphilic properties, they are useful as drug carriers. Liposomes are also very suitable for drug delivery strategies using heat generation by NIR laser because lipid bilayers are easily broken by heat. Thermally generated bubbles from liposomes not only induce drug release, but also enable ultrasound imaging. METHODS Melanin, perfluorohexane (PFH), and 5-fluorouracil (5-FU)-loaded liposomes (melanin@PFH@5-FU-liposomes) that can generate heat and bubble by NIR laser irradiation were prepared by a thin film method. Conversion of light to heat and bubble generation of melanin@PFH@5-FU-liposomes were evaluated using an infrared (IR) thermal imaging camera and an ultrasound imaging system both in vitro and in vivo. To investigate tumor therapeutic effect, NIR laser of 808 nm was used to irradiate tumor site for 10 min after injecting melanin@PFH@5-FU-liposome into tail veins of CT26-bearing mice. RESULTS Melanin@PFH@5-FU-liposomes showed a spherical shape with a size of 209.6 ± 4.3 nm. Upon NIR laser irradiation, melanin@PFH@5-FU-liposomes exhibited effective temperature increase both in vitro and in vivo. In this regard, temperature increase caused a phase transition of PFH to induce bubble generation dramatically, resulting in effective drug release behavior and ultrasound imaging. The temperature of the tumor site was increased to 52 t and contrast was greatly enhanced during ultrasound imaging due to the generation of bubble. More importantly, tumor growth was effectively inhibited by injection of melanin@PFH@5-FU-liposomes with laser irradiation. CONCLUSIONS Based on intrinsic photothermal properties of melanin and phase transition properties of PFH, melanin@PFH@5-FU-liposomes exhibited effective heat and bubble generation upon NIR laser irradiation. The elevated temperature induced bubble generation, resulting in contrast enhancement of ultrasound imaging. Melanin@PFH@5-FU-liposomes under NIR laser irradiation induced the death of cancer cells, thereby effectively inhibiting tumor growth. These results suggest that melanin@PFH@5-FU-liposomes can be utilized as a promising agent for photothermal tumor therapy and ultrasound imaging.
Collapse
|
35
|
Dellinger TH, Han ES, Raoof M, Lee B, Wu X, Cho H, He TF, Lee P, Razavi M, Liang WS, Schmolze D, Priceman SJ, Lee S, Lin WC, Lin JF, Kebria M, Hakim A, Ruel N, Stewart DB, Wang EW, Paz BI, Wakabayashi MT, Cristea MC, Rodriguez-Rodriguez L. Hyperthermic Intraperitoneal Chemotherapy-Induced Molecular Changes in Humans Validate Preclinical Data in Ovarian Cancer. JCO Precis Oncol 2022; 6:e2100239. [PMID: 35357903 PMCID: PMC8984280 DOI: 10.1200/po.21.00239] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Hyperthermic intraperitoneal chemotherapy (HIPEC) confers a survival benefit in epithelial ovarian cancer (EOC) and in preclinical models. However, the molecular changes induced by HIPEC have not been corroborated in humans.
Collapse
Affiliation(s)
- Thanh H Dellinger
- Division of Gynecologic Oncology, Department of Surgery, City of Hope National Medical Center, Duarte, CA
| | - Ernest S Han
- Division of Gynecologic Oncology, Department of Surgery, City of Hope National Medical Center, Duarte, CA
| | - Mustafa Raoof
- Division of Surgical Oncology, Department of Surgery, City of Hope National Medical Center, Duarte, CA
| | - Byrne Lee
- Department of Surgery, Stanford University, Stanford, CA
| | - Xiwei Wu
- Integrative Genomics Core, City of Hope National Medical Center Beckman Research Institute, Duarte, CA
| | - Hyejin Cho
- Integrative Genomics Core, City of Hope National Medical Center Beckman Research Institute, Duarte, CA
| | - Ting-Fang He
- Immuno-oncology Core, City of Hope National Medical Center Beckman Research Institute, Duarte, CA
| | - Peter Lee
- Immuno-oncology Core, City of Hope National Medical Center Beckman Research Institute, Duarte, CA
| | - Marianne Razavi
- Women's Cancer Center, City of Hope National Medical Center, Duarte, CA
| | | | - Daniel Schmolze
- Department of Pathology, City of Hope National Medical Center, Duarte, CA
| | - Saul J Priceman
- Hematology & Hematopoietic Cell Transplantation and Immuno-Oncology, City of Hope National Medical Center Beckman Research Institute, Duarte, CA
| | - Stephen Lee
- Division of Gynecologic Oncology, Department of Surgery, City of Hope National Medical Center, Duarte, CA
| | - Wei-Chien Lin
- Division of Gynecologic Oncology, Department of Surgery, City of Hope National Medical Center, Duarte, CA
| | - Jeff F Lin
- Division of Gynecologic Oncology, Department of Surgery, City of Hope National Medical Center, Duarte, CA
| | - Mehdi Kebria
- Division of Gynecologic Oncology, Department of Surgery, City of Hope National Medical Center, Duarte, CA
| | - Amy Hakim
- Division of Gynecologic Oncology, Department of Surgery, City of Hope National Medical Center, Duarte, CA
| | - Nora Ruel
- Biostatistics Core, City of Hope National Medical Center Beckman Research Institute, Duarte, CA
| | - Daphne B Stewart
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA
| | - Edward W Wang
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA
| | - Benjamin I Paz
- Department of Surgery, Stanford University, Stanford, CA
| | - Mark T Wakabayashi
- Division of Gynecologic Oncology, Department of Surgery, City of Hope National Medical Center, Duarte, CA
| | - Mihaela C Cristea
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA
| | - Lorna Rodriguez-Rodriguez
- Division of Gynecologic Oncology, Department of Surgery, City of Hope National Medical Center, Duarte, CA
| |
Collapse
|
36
|
Dewhirst MW, Oleson JR, Kirkpatrick J, Secomb TW. Accurate Three-Dimensional Thermal Dosimetry and Assessment of Physiologic Response Are Essential for Optimizing Thermoradiotherapy. Cancers (Basel) 2022; 14:1701. [PMID: 35406473 PMCID: PMC8997141 DOI: 10.3390/cancers14071701] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 02/04/2023] Open
Abstract
Numerous randomized trials have revealed that hyperthermia (HT) + radiotherapy or chemotherapy improves local tumor control, progression free and overall survival vs. radiotherapy or chemotherapy alone. Despite these successes, however, some individuals fail combination therapy; not every patient will obtain maximal benefit from HT. There are many potential reasons for failure. In this paper, we focus on how HT influences tumor hypoxia, since hypoxia negatively influences radiotherapy and chemotherapy response as well as immune surveillance. Pre-clinically, it is well established that reoxygenation of tumors in response to HT is related to the time and temperature of exposure. In most pre-clinical studies, reoxygenation occurs only during or shortly after a HT treatment. If this were the case clinically, then it would be challenging to take advantage of HT induced reoxygenation. An important question, therefore, is whether HT induced reoxygenation occurs in the clinic that is of radiobiological significance. In this review, we will discuss the influence of thermal history on reoxygenation in both human and canine cancers treated with thermoradiotherapy. Results of several clinical series show that reoxygenation is observed and persists for 24-48 h after HT. Further, reoxygenation is associated with treatment outcome in thermoradiotherapy trials as assessed by: (1) a doubling of pathologic complete response (pCR) in human soft tissue sarcomas, (2) a 14 mmHg increase in pO2 of locally advanced breast cancers achieving a clinical response vs. a 9 mmHg decrease in pO2 of locally advanced breast cancers that did not respond and (3) a significant correlation between extent of reoxygenation (as assessed by pO2 probes and hypoxia marker drug immunohistochemistry) and duration of local tumor control in canine soft tissue sarcomas. The persistence of reoxygenation out to 24-48 h post HT is distinctly different from most reported rodent studies. In these clinical series, comparison of thermal data with physiologic response shows that within the same tumor, temperatures at the higher end of the temperature distribution likely kill cells, resulting in reduced oxygen consumption rate, while lower temperatures in the same tumor improve perfusion. However, reoxygenation does not occur in all subjects, leading to significant uncertainty about the thermal-physiologic relationship. This uncertainty stems from limited knowledge about the spatiotemporal characteristics of temperature and physiologic response. We conclude with recommendations for future research with emphasis on retrieving co-registered thermal and physiologic data before and after HT in order to begin to unravel complex thermophysiologic interactions that appear to occur with thermoradiotherapy.
Collapse
Affiliation(s)
- Mark W Dewhirst
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA
| | - James R Oleson
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA
| | - John Kirkpatrick
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Timothy W Secomb
- Department of Physiology, University of Arizona, Tucson, AZ 85724, USA
| |
Collapse
|
37
|
Hyperthermia Treatment as a Promising Anti-Cancer Strategy: Therapeutic Targets, Perspective Mechanisms and Synergistic Combinations in Experimental Approaches. Antioxidants (Basel) 2022; 11:antiox11040625. [PMID: 35453310 PMCID: PMC9030926 DOI: 10.3390/antiox11040625] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/18/2022] [Accepted: 03/19/2022] [Indexed: 02/04/2023] Open
Abstract
Despite recent developments in diagnosis and treatment options, cancer remains one of the most critical threats to health. Several anti-cancer therapies have been identified, but further research is needed to provide more treatment options that are safe and effective for cancer. Hyperthermia (HT) is a promising treatment strategy for cancer because of its safety and cost-effectiveness. This review summarizes studies on the anti-cancer effects of HT and the detailed mechanisms. In addition, combination therapies with anti-cancer drugs or natural products that can effectively overcome the limitations of HT are reviewed because HT may trigger protective events, such as an increase of heat shock proteins (HSPs). In the 115 reports included, the mechanisms related to apoptosis, cell cycle, reactive oxygen species, mitochondrial membrane potential, DNA damage, transcription factors and HSPs were considered important. This review shows that HT is an effective inducer of apoptosis. Moreover, the limitations of HT may be overcome using combined therapy with anti-cancer drugs or natural products. Therefore, appropriate combinations of such agents with HT will exert maximal effects to treat cancer.
Collapse
|
38
|
Wang Y, Xu Z, Li W, Wei W, Qin M, Li Q, Liu X, Zhang X, Wang X. A graphene-Ag based near-infrared defined accurate anti-scarring strategy for ocular glaucoma surgery. Biomater Sci 2022; 10:1281-1291. [PMID: 35083991 DOI: 10.1039/d1bm01614h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Excessive fibrosis is the major factor in the failure of glaucoma filtration surgery. So far, the dominant approach for inhibiting fibrosis is the use of an antimetabolite drug, but the complications it causes, such as filtering bleb leakage, bacterial endophthalmitis and ocular hypotony, are also inevitable. Herein, a multifunctional anti-scarring platform (PVA@rGO-Ag/5-Fu) integrated with outstanding photothermal, antibacterial and drug delivery abilities is developed. PVA@rGO-Ag shows favorable biocompatibility as well as an accurate regional photothermal killing ability on both conjunctival fibroblasts and bacteria under 808 nm near-infrared (NIR) irradiation. Furthermore, PVA@rGO-Ag/5-Fu improves bleb survival rates and results in the satisfactory reduction of intraocular pressure (IOP) by decreasing the fibrous reaction in vivo. In summary, PVA@rGO-Ag/5-Fu has promising potential as an efficacious and safe anti-scarring agent for filtering surgery.
Collapse
Affiliation(s)
- Yanan Wang
- Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Research Institute of Ophthalmology and Visual Science, Affiliated Eye Hospital of Nanchang University, Nanchang, Jiangxi, 330006, P.R. China.
| | - Zikang Xu
- National Engineering Research Centre for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330088, P.R. China.
| | - Wenchi Li
- National Engineering Research Centre for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330088, P.R. China.
| | - Wei Wei
- Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Research Institute of Ophthalmology and Visual Science, Affiliated Eye Hospital of Nanchang University, Nanchang, Jiangxi, 330006, P.R. China.
| | - Mengqi Qin
- Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Research Institute of Ophthalmology and Visual Science, Affiliated Eye Hospital of Nanchang University, Nanchang, Jiangxi, 330006, P.R. China.
| | - Qun Li
- National Engineering Research Centre for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330088, P.R. China.
| | - Xuexia Liu
- National Engineering Research Centre for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330088, P.R. China. .,College of Chemistry, Nanchang University, Nanchang, Jiangxi, 330088, P.R. China
| | - Xu Zhang
- Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Research Institute of Ophthalmology and Visual Science, Affiliated Eye Hospital of Nanchang University, Nanchang, Jiangxi, 330006, P.R. China.
| | - Xiaolei Wang
- National Engineering Research Centre for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330088, P.R. China. .,College of Chemistry, Nanchang University, Nanchang, Jiangxi, 330088, P.R. China
| |
Collapse
|
39
|
Smart Magnetic Nanocarriers for Multi-Stimuli On-Demand Drug Delivery. NANOMATERIALS 2022; 12:nano12030303. [PMID: 35159647 PMCID: PMC8840331 DOI: 10.3390/nano12030303] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/10/2021] [Accepted: 01/12/2022] [Indexed: 02/06/2023]
Abstract
In this study, we report the realization of drug-loaded smart magnetic nanocarriers constituted by superparamagnetic iron oxide nanoparticles encapsulated in a dual pH- and temperature-responsive poly (N-vinylcaprolactam-co-acrylic acid) copolymer to achieve highly controlled drug release and localized magnetic hyperthermia. The magnetic core was constituted by flower-like magnetite nanoparticles with a size of 16.4 nm prepared by the polyol approach, with good saturation magnetization and a high specific absorption rate. The core was encapsulated in poly (N-vinylcaprolactam-co-acrylic acid) obtaining magnetic nanocarriers that revealed reversible hydration/dehydration transition at the acidic condition and/or at temperatures above physiological body temperature, which can be triggered by magnetic hyperthermia. The efficacy of the system was proved by loading doxorubicin with very high encapsulation efficiency (>96.0%) at neutral pH. The double pH- and temperature-responsive nature of the magnetic nanocarriers facilitated a burst, almost complete release of the drug at acidic pH under hyperthermia conditions, while a negligible amount of doxorubicin was released at physiological body temperature at neutral pH, confirming that in addition to pH variation, drug release can be improved by hyperthermia treatment. These results suggest this multi-stimuli-sensitive nanoplatform is a promising candidate for remote-controlled drug release in combination with magnetic hyperthermia for cancer treatment.
Collapse
|
40
|
Gao Y, Tong H, Li J, Li J, Huang D, Shi J, Xia B. Mitochondria-Targeted Nanomedicine for Enhanced Efficacy of Cancer Therapy. Front Bioeng Biotechnol 2021; 9:720508. [PMID: 34490227 PMCID: PMC8418302 DOI: 10.3389/fbioe.2021.720508] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 07/14/2021] [Indexed: 12/27/2022] Open
Abstract
Nanomedicines have been designed and developed to deliver anticancer drugs or exert anticancer therapy more selectively to tumor sites. Recent investigations have gone beyond delivering drugs to tumor tissues or cells, but to intracellular compartments for amplifying therapy efficacy. Mitochondria are attractive targets for cancer treatment due to their important functions for cells and close relationships to tumor occurrence and metastasis. Accordingly, multifunctional nanoplatforms have been constructed for cancer therapy with the modification of a variety of mitochondriotropic ligands, to trigger the mitochondria-mediated apoptosis of tumor cells. On this basis, various cancer therapeutic modalities based on mitochondria-targeted nanomedicines are developed by strategies of damaging mitochondria DNA (mtDNA), increasing reactive oxygen species (ROS), disturbing respiratory chain and redox balance. Herein, in this review, we highlight mitochondria-targeted cancer therapies enabled by nanoplatforms including chemotherapy, photothermal therapy (PTT), photodynamic therapy (PDT), chemodynamic therapy (CDT), sonodynamic therapy (SDT), radiodynamic therapy (RDT) and combined immunotherapy, and discussed the ongoing challenges.
Collapse
Affiliation(s)
- Yan Gao
- College of Science, Key Laboratory of Forest Genetics and Biotechnology (Ministry of Education of China), Nanjing Forestry University, Nanjing, China
| | - Haibei Tong
- College of Science, Key Laboratory of Forest Genetics and Biotechnology (Ministry of Education of China), Nanjing Forestry University, Nanjing, China
| | - Jialiang Li
- College of Science, Key Laboratory of Forest Genetics and Biotechnology (Ministry of Education of China), Nanjing Forestry University, Nanjing, China
| | - Jiachen Li
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, University of Helsinki, Helsinki, Finland
| | - Di Huang
- College of Science, Key Laboratory of Forest Genetics and Biotechnology (Ministry of Education of China), Nanjing Forestry University, Nanjing, China
| | - Jisen Shi
- College of Science, Key Laboratory of Forest Genetics and Biotechnology (Ministry of Education of China), Nanjing Forestry University, Nanjing, China
| | - Bing Xia
- College of Science, Key Laboratory of Forest Genetics and Biotechnology (Ministry of Education of China), Nanjing Forestry University, Nanjing, China
| |
Collapse
|
41
|
The anti-tumor effects of the combination of microwave hyperthermia and lobaplatin against breast cancer cells in vitro and in vivo. Biosci Rep 2021; 42:229268. [PMID: 34282830 PMCID: PMC8829017 DOI: 10.1042/bsr20190878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 06/10/2021] [Accepted: 06/29/2021] [Indexed: 11/17/2022] Open
Abstract
Background: Breast cancer is the main lethal disease among females. The combination of lobaplatin and microwave hyperthermia plays a crucial role in several kinds of cancer in the clinic, but its possible mechanism in breast cancer has remained indistinct. Methods: Mouse models were used to detect breast cancer progression. Cell growth was explored with MTS (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulphonyl)-2H-tetrazolium) and colony formation assays. Cell migration and invasion were investigated with a transwell assay. Cell apoptosis was probed with flow cytometry. The expression of apoptosis-associated proteins was examined with Western blots. Result: Combination treatment decreased breast cancer cell viability, colony formation, cell invasion and metastasis. In addition, the treatment-induced breast cancer cell apoptosis and autophagy, activated the c-Jun N-terminal kinase (JNK) signaling pathway, suppressed the protein kinase B (AKT)/mammalian target of rapamycin (mTOR) signaling pathway, and down-regulated IAP and Bcl-2 family protein expression. Conclusion: These results indicate that lobaplatin is an effective breast cancer anti-tumor agent. Microwave hyperthermia was a useful adjunctive treatment. Combination treatment was more efficient than any single therapy. The possible mechanism for this effect was mainly associated with activation of the JNK signaling pathway, inactivation of the AKT/mTOR signaling pathway and down-regulation of the Bcl-2 and IAP families.
Collapse
|
42
|
Danyuo Y, Obayemi JD, Salifu AA, Oyewole OK, Azeko ST, Ani CJ, Dozie-Nwachukwu S, Yirijor J, Abade-Abugre M, Odusanya OS, McBagonluri F, Soboyejo WO. Cell-surface interactions on gold-coated polydimethylsiloxane nanocomposite structures: Localized laser heating on cell viability. J Biomed Mater Res A 2021; 109:2611-2624. [PMID: 34180577 DOI: 10.1002/jbm.a.37254] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 06/11/2021] [Accepted: 06/18/2021] [Indexed: 12/26/2022]
Abstract
This article presents the results of cell-surface interactions on polydimethylsiloxane (PDMS)-based substrates coated with nanoscale gold (Au) thin films. The surfaces of PDMS and PDMS-magnetite (MNP)-based substrates were treated with UV-ozone, prior to thermal vapor deposition (sputter-coated) of thin films of titanium (Ti) onto the substrates to improve the adhesion of Au coatings. The thin layer of Ti was thermally evaporated to improve interfacial adhesion, which was enhanced by a 40-nm thick film microwrinkled/buckled wavy layer of Au, that was coated to enhance cell-surface interactions and protein absorption. Cell-surface interactions were studied on the hybrid surfaces using a combination of optical and fluorescence microscopy. Consequently, cell proliferation and surface cytotoxicity (of the sputter-coated PDMS surfaces) were elucidated by characterizing the metabolic activity in the presence of breast cancer and normal breast cells. The photothermal conversion efficiency associated with laser-materials interactions with the PDMS/PDMS-magnetite-based composites was shown to have an optimum efficiency of ~31.8%. The implications of the results are discussed for potential applications of PDMS nanocomposites in implantable biomedical devices.
Collapse
Affiliation(s)
- Yiporo Danyuo
- Department of Mechanical Engineering, Ashesi University, 1 University Avenue, Berekuso, Ghana.,Department of Materials Science and Engineering, African University of Science and Technology, FCT, Abuja, Nigeria
| | - John David Obayemi
- Department of Mechanical Engineering, Higgins Labs, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
| | - Ali Azeko Salifu
- Department of Mechanical Engineering, Higgins Labs, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
| | - Oluwaseun Kehinde Oyewole
- Department of Mechanical Engineering, Higgins Labs, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
| | - Salifu Tahiru Azeko
- Department of Mechanical Engineering, Tamale Technical University, Tamale, Northern Region, Ghana
| | - Chukwuemeka Joseph Ani
- Department of Theoretical and Applied Physics, African University of Science and Technology (AUST), FCT, Abuja, Nigeria.,Department of Electrical and Electronics Engineering, Nile University of Nigeria, FCT, Abuja, Nigeria
| | - Stella Dozie-Nwachukwu
- Department of Materials Science and Engineering, African University of Science and Technology, FCT, Abuja, Nigeria.,Biotechnology Advance Research Centre, Sheda Science and Technology Complex (SHESTCO), FCT, Abuja, Nigeria
| | - John Yirijor
- Department of Mechanical Engineering, Academic City University College, Accra, Ghana
| | - Miriam Abade-Abugre
- Department of Mechanical Engineering, Ashesi University, 1 University Avenue, Berekuso, Ghana
| | - Olushola Segun Odusanya
- Department of Materials Science and Engineering, African University of Science and Technology, FCT, Abuja, Nigeria.,Biotechnology Advance Research Centre, Sheda Science and Technology Complex (SHESTCO), FCT, Abuja, Nigeria
| | - Fred McBagonluri
- Department of Mechanical Engineering, Academic City University College, Accra, Ghana
| | - Winston Oluwole Soboyejo
- Department of Mechanical Engineering, Higgins Labs, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
| |
Collapse
|
43
|
Quintana M, Saavedra E, del Rosario H, González I, Hernández I, Estévez F, Quintana J. Ethanol Enhances Hyperthermia-Induced Cell Death in Human Leukemia Cells. Int J Mol Sci 2021; 22:ijms22094948. [PMID: 34066632 PMCID: PMC8125413 DOI: 10.3390/ijms22094948] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/04/2021] [Accepted: 05/04/2021] [Indexed: 12/23/2022] Open
Abstract
Ethanol has been shown to exhibit therapeutic properties as an ablative agent alone and in combination with thermal ablation. Ethanol may also increase sensitivity of cancer cells to certain physical and chemical antitumoral agents. The aim of our study was to assess the potential influence of nontoxic concentrations of ethanol on hyperthermia therapy, an antitumoral modality that is continuously growing and that can be combined with classical chemotherapy and radiotherapy to improve their efficiency. Human leukemia cells were included as a model in the study. The results indicated that ethanol augments the cytotoxicity of hyperthermia against U937 and HL60 cells. The therapeutic benefit of the hyperthermia/ethanol combination was associated with an increase in the percentage of apoptotic cells and activation of caspases-3, -8 and -9. Apoptosis triggered either by hyperthermia or hyperthermia/ethanol was almost completely abolished by a caspase-8 specific inhibitor, indicating that this caspase plays a main role in both conditions. The role of caspase-9 in hyperthermia treated cells acquired significance whether ethanol was present during hyperthermia since the alcohol enhanced Bid cleavage, translocation of Bax from cytosol to mitochondria, release of mitochondrial apoptogenic factors, and decreased of the levels of the anti-apoptotic factor myeloid cell leukemia-1 (Mcl-1). The enhancement effect of ethanol on hyperthermia-activated cell death was associated with a reduction in the expression of HSP70, a protein known to interfere in the activation of apoptosis at different stages. Collectively, our findings suggest that ethanol could be useful as an adjuvant in hyperthermia therapy for cancer.
Collapse
|
44
|
Emamzadeh M, Pasparakis G. Polymer coated gold nanoshells for combinational photochemotherapy of pancreatic cancer with gemcitabine. Sci Rep 2021; 11:9404. [PMID: 33931720 PMCID: PMC8087785 DOI: 10.1038/s41598-021-88909-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 04/14/2021] [Indexed: 12/29/2022] Open
Abstract
Pancreatic cancer is one of the most lethal malignancies with limited therapeutic options and dismal prognosis. Gemcitabine is the front-line drug against pancreatic cancer however with limited improvement of therapeutic outcomes. In this study we envisaged the integration of GEM with gold nanoshells which constitute an interesting class of nanomaterials with excellent photothermal conversion properties. Nanoshells were coated with thiol-capped poly(ethylene glycol) methacrylate polymers of different molecular weight via Au-S attachment. It was found that the molecular weight of the polymers affects the in vitro performance of the formulations; more importantly we demonstrate that the EC50 of nanoshell loaded GEM can be suppressed but fully restored and even improved upon laser irradiation. Our proposed nanoformulations outperformed the cytotoxicity of the parent drug and showed confined synergism under the tested in vitro conditions.
Collapse
Affiliation(s)
- Mina Emamzadeh
- School of Pharmacy, University College London, London, WC1N 1AX, UK
| | - George Pasparakis
- School of Pharmacy, University College London, London, WC1N 1AX, UK.
- Department of Chemical Engineering, University of Patras, Patras, Greece.
| |
Collapse
|
45
|
Synergistic Photothermal-Chemotherapy Based on the Use of Biomimetic Magnetic Nanoparticles. Pharmaceutics 2021; 13:pharmaceutics13050625. [PMID: 33924828 PMCID: PMC8144968 DOI: 10.3390/pharmaceutics13050625] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 04/22/2021] [Accepted: 04/25/2021] [Indexed: 12/18/2022] Open
Abstract
MamC-mediated biomimetic magnetic nanoparticles (BMNPs) have emerged as one of the most promising nanomaterials due to their magnetic features (superparamagnetic character and large magnetic moment per particle), their novel surface properties determined by MamC, their biocompatibility and their ability as magnetic hyperthermia agents. However, the current clinical application of magnetic hyperthermia is limited due to the fact that, in order to be able to reach an effective temperature at the target site, relatively high nanoparticle concentration, as well as high magnetic field strength and/or AC frequency are needed. In the present study, the potential of BMNPs to increase the temperature upon irradiation of a laser beam in the near infrared, at a wavelength at which tissues become partially transparent, is explored. Moreover, our results also demonstrate the synergy between photothermia and chemotherapy in terms of drug release and cytotoxicity, by using BMNPs functionalized with doxorubicin, and the effectiveness of this combination therapy against tumor cells in in vitro experiments. Therefore, the findings of the present study open the possibility of a novel, alternative approach to fight localized tumors.
Collapse
|
46
|
Zhang X, Bobeica M, Unger M, Bednarz A, Gerold B, Patties I, Melzer A, Landgraf L. Focused ultrasound radiosensitizes human cancer cells by enhancement of DNA damage. Strahlenther Onkol 2021; 197:730-743. [PMID: 33885910 PMCID: PMC8292237 DOI: 10.1007/s00066-021-01774-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 03/23/2021] [Indexed: 12/19/2022]
Abstract
Purpose High-intensity focused ultrasound (HIFU/FUS) has expanded as a noninvasive quantifiable option for hyperthermia (HT). HT in a temperature range of 40–47 °C (thermal dose CEM43 ≥ 25) could work as a sensitizer to radiation therapy (RT). Here, we attempted to understand the tumor radiosensitization effect at the cellular level after a combination treatment of FUS+RT. Methods An in vitro FUS system was developed to induce HT at frequencies of 1.147 and 1.467 MHz. Human head and neck cancer (FaDU), glioblastoma (T98G), and prostate cancer (PC-3) cells were exposed to FUS in ultrasound-penetrable 96-well plates followed by single-dose X‑ray irradiation (10 Gy). Radiosensitizing effects of FUS were investigated by cell metabolic activity (WST‑1 assay), apoptosis (annexin V assay, sub-G1 assay), cell cycle phases (propidium iodide staining), and DNA double-strand breaks (γH2A.X assay). Results The FUS intensities of 213 (1.147 MHz) and 225 W/cm2 (1.467 MHz) induced HT for 30 min at mean temperatures of 45.20 ± 2.29 °C (CEM43 = 436 ± 88) and 45.59 ± 1.65 °C (CEM43 = 447 ± 79), respectively. FUS improves the effect of RT significantly by reducing metabolic activity in T98G cells 48 h (RT: 96.47 ± 8.29%; FUS+RT: 79.38 ± 14.93%; p = 0.012) and in PC-3 cells 72 h (54.20 ± 10.85%; 41.01 ± 11.17%; p = 0.016) after therapy, but not in FaDu cells. Mechanistically, FUS+RT leads to increased apoptosis and enhancement of DNA double-strand breaks compared to RT alone in T98G and PC-3 cells. Conclusion Our in vitro findings demonstrate that FUS has good potential to sensitize glioblastoma and prostate cancer cells to RT by mainly enhancing DNA damage. Supplementary Information The online version of this article (10.1007/s00066-021-01774-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xinrui Zhang
- Innovation Center Computer Assisted Surgery (ICCAS), University of Leipzig, Semmelweisstr. 14, Haus 14, Leipzig, 04103, Germany.
| | - Mariana Bobeica
- Institute for Medical Science and Technology (IMSaT), University of Dundee, Wilson House, 1 Wurzburg Loan, Dundee MediPark, Dundee, DD2 1FD, UK.,Extreme Light Infrastructure - Nuclear Physics ELI-NP, "Horia Hulubei" National Institute for Physics and Nuclear Engineering, 30 Reactorului Street, Bucharest-Magurele, 077125, Romania
| | - Michael Unger
- Innovation Center Computer Assisted Surgery (ICCAS), University of Leipzig, Semmelweisstr. 14, Haus 14, Leipzig, 04103, Germany
| | - Anastasia Bednarz
- Innovation Center Computer Assisted Surgery (ICCAS), University of Leipzig, Semmelweisstr. 14, Haus 14, Leipzig, 04103, Germany
| | - Bjoern Gerold
- Institute for Medical Science and Technology (IMSaT), University of Dundee, Wilson House, 1 Wurzburg Loan, Dundee MediPark, Dundee, DD2 1FD, UK.,Theraclion, 102 Rue Etienne Dolet, Malakoff, 92240, France
| | - Ina Patties
- Innovation Center Computer Assisted Surgery (ICCAS), University of Leipzig, Semmelweisstr. 14, Haus 14, Leipzig, 04103, Germany.,Department of Radiation Oncology, University of Leipzig, Stephanstr. 9a, Leipzig, 04103, Germany
| | - Andreas Melzer
- Innovation Center Computer Assisted Surgery (ICCAS), University of Leipzig, Semmelweisstr. 14, Haus 14, Leipzig, 04103, Germany. .,Institute for Medical Science and Technology (IMSaT), University of Dundee, Wilson House, 1 Wurzburg Loan, Dundee MediPark, Dundee, DD2 1FD, UK.
| | - Lisa Landgraf
- Innovation Center Computer Assisted Surgery (ICCAS), University of Leipzig, Semmelweisstr. 14, Haus 14, Leipzig, 04103, Germany
| |
Collapse
|
47
|
Chiancone F, Fabiano M, Carrino M, Fedelini M, Meccariello C, Fedelini P. Impact of systemic inflammatory markers on the response to Hyperthermic IntraVEsical Chemotherapy (HIVEC) in patients with non-muscle-invasive bladder cancer after bacillus Calmette-Guérin failure. Arab J Urol 2021; 19:86-91. [PMID: 33763253 PMCID: PMC7954479 DOI: 10.1080/2090598x.2021.1874627] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Objectives: To evaluate the impact of pre- and post-treatment systemic inflammatory markers on the response to Hyperthermic IntraVEsical Chemotherapy (HIVEC) treatment in a cohort of patients with high-grade non-muscle-invasive bladder cancer with bacillus Calmette–Guérin (BCG) failure or intolerance who were unsuitable or unwilling to undergo early radical cystectomy. As a secondary endpoint, we assessed the influence of some demographic, clinical and pathological factors on the response to chemo-hyperthermia. Patients and methods: Between March 2017 and December 2019, 72 consecutive patients were retrospectively analysed. Patients with diseases or conditions that could interfere with systemic inflammatory status or full blood count were excluded. The HIVEC protocol consisted of six weekly intravesical treatments with 40 mg Mitomycin-C diluted in 50 mL distilled water. The drug was heated to a temperature of 43°C. Association of categorical variables with response to HIVEC was evaluated using Yates’ chi-squared test and differences in continuous variable were analysed using the Mann–Whitney test. Logistic regression analysis was performed to define independent predictors of response to HIVEC. Results: Patients who failed HIVEC were more likely to have multiple tumours (P = 0.039) at transurethral resection of bladder and a recurrence rate of >1/year (P = 0.046). Lower post-HIVEC inflammatory indices [C-reactive protein (P = 0.021), erythrocyte sedimentation rate (P = 0.027)] and lower pre- (P = 0.014) and post-treatment (P = 0.004) neutrophil-to-lymphocyte ratio (NLR) values were significantly associated with the response to the HIVEC regimen (no bladder cancer recurrence or progression). In the multivariate analysis, patients with a recurrence rate of >1/year were eight-times more likely to experience failure of HIVEC (P = 0.007). Higher pre- (P = 0.023) and post-treatment NLR values (P = 0.046) were associated with a worse response to the HIVEC regimen. Conclusions: The recurrence rate and systemic inflammatory response markers could be useful tools to predict the likelihood of obtaining a response with the HIVEC regimen. These markers might help to guide patients about the behaviour of the tumour after BCG failure, predicting failure or success of a conservative treatment. Abbreviations: CHT: chemo-hyperthermia; CIS: carcinoma in situ; CRP: C-reactive protein; EAU: European Association of Urology; ESR: erythrocyte sedimentation rate; HG: high grade; HIVEC: Hyperthermic IntraVEsical Chemotherapy; ICD: immunogenic cell death; IL: interleukin; MMC: Mitomycin-C; NK: natural killer; NLR: neutrophil-to-lymphocyte ratio; NMIBC: non-muscle-invasive bladder cancer; PLR: platelet-to-lymphocyte ratio; RC: radical cystectomy; SIR: systemic inflammatory response; TURB: transurethral resection of bladder
Collapse
Affiliation(s)
- Francesco Chiancone
- Department of Urology, Azienda Ospedaliera di Rilievo Nazionale (A.O.R.N.) Antonio Cardarelli, Naples, Italy
| | - Marco Fabiano
- Department of Urology, Azienda Ospedaliera di Rilievo Nazionale (A.O.R.N.) Antonio Cardarelli, Naples, Italy
| | - Maurizio Carrino
- Department of Urology, Azienda Ospedaliera di Rilievo Nazionale (A.O.R.N.) Antonio Cardarelli, Naples, Italy
| | - Maurizio Fedelini
- Department of Urology, Azienda Ospedaliera di Rilievo Nazionale (A.O.R.N.) Antonio Cardarelli, Naples, Italy
| | - Clemente Meccariello
- Department of Urology, Azienda Ospedaliera di Rilievo Nazionale (A.O.R.N.) Antonio Cardarelli, Naples, Italy
| | - Paolo Fedelini
- Department of Urology, Azienda Ospedaliera di Rilievo Nazionale (A.O.R.N.) Antonio Cardarelli, Naples, Italy
| |
Collapse
|
48
|
Dellinger TH, Han ES. State of the Science: The role of HIPEC in the treatment of ovarian cancer. Gynecol Oncol 2021; 160:364-368. [PMID: 33419611 DOI: 10.1016/j.ygyno.2020.12.029] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Thanh H Dellinger
- Department of Surgery, Division of Gynecologic Oncology, City of Hope Comprehensive Cancer Center, 1500 E. Duarte Road, Duarte, CA 91010, United States of America
| | - Ernest S Han
- Department of Surgery, Division of Gynecologic Oncology, City of Hope Comprehensive Cancer Center, 1500 E. Duarte Road, Duarte, CA 91010, United States of America.
| |
Collapse
|
49
|
Nodooshan SJ, Amini P, Ashrafizadeh M, Tavakoli S, Aryafar T, Khalafi L, Musa AE, Mahdavi SR, Najafi M, Ahmadi A, Farhood B. Suberosin Attenuates the Proliferation of MCF-7 Breast Cancer Cells in Combination with Radiotherapy or Hyperthermia. Curr Drug Res Rev 2021; 13:148-153. [PMID: 33371865 DOI: 10.2174/2589977512666201228104528] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 08/08/2020] [Accepted: 10/05/2020] [Indexed: 06/12/2023]
Abstract
AIM The aim of this study was to determine the proliferation of MCF-7 following irradiation or hyperthermia as alone or pre-treatment with suberosin. BACKGROUND Radiotherapy is a major therapeutic modality for the control of breast cancer. However, hyperthermia can be prescribed for relief of pain or enhancing cancer cell death. Some studies have attempted its use as an adjuvant to improve therapeutic efficiency. Suberosin is a cumarin- derived natural agent that has shown anti-inflammatory properties. OBJECTIVE In this in vitro study, possible sensitization effect of suberosin in combination with radiation or hyperthermia was evaluated. METHODS MCF-7 breast cancer cells were irradiated or received hyperthermia with or without treatment with suberosin. The incidence of apoptosis as well as viability of MCF-7 cells were observed. Furthermore, the expressions of pro-apoptotic genes such as Bax, Bcl-2, and some caspases were evaluated using real-time PCR. RESULTS Both radiotherapy or hyperthermia reduced the proliferation of MCF-7 cells. Suberosin amplified the effects of radiotherapy or hyperthermia for induction of pro-apoptotic genes and reducing cell viability. CONCLUSION Suberosin has a potent anti-cancer effect when combined with radiotherapy or hyperthermia. It could be a potential candidate for killing breast cancer cells as well as increasing the therapeutic efficiency of radiotherapy or hyperthermia.
Collapse
Affiliation(s)
- Saeedeh Jafari Nodooshan
- School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Peyman Amini
- Department of Radiology, Faculty of Paramedical Science, Tehran University of Medical Sciences, Tehran, Iran
| | - Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla 34956, Istanbul, Turkey
| | - Saeed Tavakoli
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran
| | - Tayebeh Aryafar
- Department of Medical Physics and Biomedical Engineering, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Leila Khalafi
- Omid Tehran Radiation Oncology Center, Physics Section, Tehran, Iran
| | - Ahmed Eleojo Musa
- Department of Medical Physics and Biomedical Engineering, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Rabie Mahdavi
- Medical Physics Department, Iran University of Medical Sciences, Tehran, Iran
| | - Masoud Najafi
- Medical Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Akbar Ahmadi
- School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Farhood
- Departments of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
50
|
Ebrahimi A, Nowzari-Dalini A, Jalili M, Masoudi-Nejad A. Appropriate time to apply control input to complex dynamical systems. Sci Rep 2020; 10:22035. [PMID: 33328499 PMCID: PMC7744535 DOI: 10.1038/s41598-020-78909-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 11/12/2020] [Indexed: 12/13/2022] Open
Abstract
Controlling a network structure has many potential applications many fields. In order to have an effective network control, not only finding good driver nodes is important, but also finding the optimal time to apply the external control signals to network nodes has a critical role. If applied in an appropriate time, one might be to control a network with a smaller control signals, and thus less energy. In this manuscript, we show that there is a relationship between the strength of the internal fluxes and the effectiveness of the external control signal. To be more effective, external control signals should be applied when the strength of the internal states is the smallest. We validate this claim on synthetic networks as well as a number of real networks. Our results may have important implications in systems medicine, in order to find the most appropriate time to inject drugs as a signal to control diseases.
Collapse
Affiliation(s)
- Ali Ebrahimi
- Laboratory of Systems Biology and Bioinformatics (LBB), Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | | | - Mahdi Jalili
- School of Engineering, RMIT University, Melbourne, Australia
| | - Ali Masoudi-Nejad
- Laboratory of Systems Biology and Bioinformatics (LBB), Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran.
| |
Collapse
|