1
|
López-Méndez R, Dubrova A, Reguera J, Magro R, Esteban-Betegón F, Parente A, Ángel García M, Camarero J, Fonda E, Wilhelm C, Muñoz-Noval Á, Espinosa A. Multiscale Thermal Analysis of Gold Nanostars in 3D Tumor Spheroids: Integrating Cellular-Level Photothermal Effects and Nanothermometry via X-Ray Spectroscopy. Adv Healthc Mater 2024:e2403799. [PMID: 39663736 DOI: 10.1002/adhm.202403799] [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: 10/03/2024] [Revised: 11/08/2024] [Indexed: 12/13/2024]
Abstract
In the pursuit of enhancing cancer treatment efficacy while minimizing side effects, near-infrared (NIR) photothermal therapy (PTT) has emerged as a promising approach. By using photothermally active nanomaterials, PTT enables localized hyperthermia, effectively eliminating cancer cells with minimal invasiveness and toxicity. Among these nanomaterials, gold nanostars (AuNS) stand out due to their tunable plasmon resonance and efficient light absorption. This study addresses the challenge of measuring nanoscale temperatures during AuNS-mediated PTT by employing X-ray absorption spectroscopy (XAS) within 3D tumor spheroids. It also aims to investigate the heat generated at the nanoscale and the resultant biological damage observed at a larger scale, utilizing confocal microscopy to establish connections between AuNS heat generation, tissue damage, and their impacts on cellular structure. These nanoscale and microscale thermal effects have been compared with macroscopic values obtained from infrared thermography, as part of a multiscale thermal analysis. The findings underscore the efficacy of AuNS in enhancing PTT and provide insights into the spatial distribution of thermal effects within tumor tissues. This research advances the understanding of localized hyperthermia in cancer therapy and underscores the potential of AuNS-based PTT for clinical applications.
Collapse
Affiliation(s)
| | - Anastasiia Dubrova
- Laboratoire Physico Chimie Curie, PCC, CNRS UMR168, Institut Curie, Sorbonne University, PSL University, Paris, 75005, France
| | - Javier Reguera
- BCMaterials, Basque Center Centre for Materials, Applications and Nanostructures, UPV/EHU Science Park, Leioa, 48940, Spain
- Departamento de Física de la Materia Condensada - GIR Bioforge, Universidad de Valladolid, edificio LUCIA, paseo de Belén 19, Valladolid, 47011, Spain
| | - Rául Magro
- Instituto de Ciencia de Materiales de Madrid, ICMM-CSIC, c/ Sor Juana Inés de la Cruz, 3, Madrid, 28049, Spain
| | - Fátima Esteban-Betegón
- Instituto de Ciencia de Materiales de Madrid, ICMM-CSIC, c/ Sor Juana Inés de la Cruz, 3, Madrid, 28049, Spain
| | - Ana Parente
- Dpto. Física Materiales, Facultad CC. Físicas, Universidad Complutense de Madrid, Madrid, 28040, Spain
| | - Miguel Ángel García
- Departamento de Electrocerámica, Instituto de Cerámica y Vidrio, ICV-CSIC, Kelsen 5, Madrid, 28049, Spain
| | - Julio Camarero
- IMDEA Nanociencia, c/Faraday, 9, Madrid, 28049, Spain
- Departamento de Física de la Materia Condensada and Instituto 'Nicolás Cabrera', Universidad Autónoma de Madrid, Madrid, 28049, Spain
| | - Emiliano Fonda
- Synchrotron SOLEIL, L'Orme des Merisiers - St. Aubin-BP 48, Gif s/Yvette, 91192, France
| | - Claire Wilhelm
- Laboratoire Physico Chimie Curie, PCC, CNRS UMR168, Institut Curie, Sorbonne University, PSL University, Paris, 75005, France
| | - Álvaro Muñoz-Noval
- IMDEA Nanociencia, c/Faraday, 9, Madrid, 28049, Spain
- Dpto. Física Materiales, Facultad CC. Físicas, Universidad Complutense de Madrid, Madrid, 28040, Spain
| | - Ana Espinosa
- Instituto de Ciencia de Materiales de Madrid, ICMM-CSIC, c/ Sor Juana Inés de la Cruz, 3, Madrid, 28049, Spain
- Unidad de Nanomateriales Avanzados, IMDEA Nanociencia, Unidad Asociada al CSIC por el ICMM, c/ Faraday 9, Madrid, 28049, Spain
| |
Collapse
|
2
|
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
|
3
|
Smadja DM. Hyperthermia for Targeting Cancer and Cancer Stem Cells: Insights from Novel Cellular and Clinical Approaches. Stem Cell Rev Rep 2024; 20:1532-1539. [PMID: 38795304 DOI: 10.1007/s12015-024-10736-0] [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] [Accepted: 05/14/2024] [Indexed: 05/27/2024]
Abstract
The Cellular Heat Shock Response and in particular heat shock protein activation are vital stress reactions observed in both healthy and cancer cells. Hyperthermia (HT) has been proposed for several years as an advancing non-invasive cancer therapy. It selectively targets cancer cells through mechanisms influenced by temperature and temperature variations. This article delves into the impact of HT on cancer cells, especially cancer stem cells (CSCs), essential contributors to cancer recurrence and metastasis. HT has shown promise in eliminating CSCs, sensitizing them to conventional treatments and modulating the tumor microenvironment. The exploration extends to mesenchymal stem cells (MSCs), which exhibit both pro-tumorigenic and anti-tumorigenic effects. HT's potential in recruiting therapeutic MSCs for targeted delivery of antitumoral agents is also discussed. Furthermore, the article introduces Brain Thermodynamics-guided Hyperthermia (BTGH) technology, a breakthrough in temperature control and modulation of heat transfer under different conditions. This non-invasive method leverages the brain-eyelid thermal tunnel (BTT) to monitor and regulate internal brain temperature. BTGH technology, with its precision and noninvasive continuous monitoring capabilities, is under clinical investigation for applications in neurological disorders and cancer. The innovative three-phase approach involves whole-body HT, targeted brain HT, and organ-specific HT. In conclusion, the exploration of localized or whole-body HT offers promising avenues for cancer, psychiatric and neurological diseases. The ongoing clinical investigations and potential applications underscore the significance of understanding and harnessing heat's responses to enhance human health.
Collapse
Affiliation(s)
- David M Smadja
- Paris Cité University, INSERM, Innovative Therapies in Hemostasis, Paris, F-75006, France.
- Hematology Department, AP-HP, Georges Pompidou European Hospital, 20 rue Leblanc, Paris, F-75015, France.
| |
Collapse
|
4
|
Soeiro JF, Sousa FL, Monteiro MV, Gaspar VM, Silva NJO, Mano JF. Advances in screening hyperthermic nanomedicines in 3D tumor models. NANOSCALE HORIZONS 2024; 9:334-364. [PMID: 38204336 PMCID: PMC10896258 DOI: 10.1039/d3nh00305a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 01/03/2024] [Indexed: 01/12/2024]
Abstract
Hyperthermic nanomedicines are particularly relevant for tackling human cancer, providing a valuable alternative to conventional therapeutics. The early-stage preclinical performance evaluation of such anti-cancer treatments is conventionally performed in flat 2D cell cultures that do not mimic the volumetric heat transfer occurring in human tumors. Recently, improvements in bioengineered 3D in vitro models have unlocked the opportunity to recapitulate major tumor microenvironment hallmarks and generate highly informative readouts that can contribute to accelerating the discovery and validation of efficient hyperthermic treatments. Leveraging on this, herein we aim to showcase the potential of engineered physiomimetic 3D tumor models for evaluating the preclinical efficacy of hyperthermic nanomedicines, featuring the main advantages and design considerations under diverse testing scenarios. The most recent applications of 3D tumor models for screening photo- and/or magnetic nanomedicines will be discussed, either as standalone systems or in combinatorial approaches with other anti-cancer therapeutics. We envision that breakthroughs toward developing multi-functional 3D platforms for hyperthermia onset and follow-up will contribute to a more expedited discovery of top-performing hyperthermic therapies in a preclinical setting before their in vivo screening.
Collapse
Affiliation(s)
- Joana F Soeiro
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal.
- Department of Physics, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Filipa L Sousa
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal.
| | - Maria V Monteiro
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal.
| | - Vítor M Gaspar
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal.
| | - Nuno J O Silva
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal.
- Department of Physics, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - João F Mano
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal.
| |
Collapse
|
5
|
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
|
6
|
Zhao Y, Sheldon M, Sun Y, Ma L. New Insights into YAP/TAZ-TEAD-Mediated Gene Regulation and Biological Processes in Cancer. Cancers (Basel) 2023; 15:5497. [PMID: 38067201 PMCID: PMC10705714 DOI: 10.3390/cancers15235497] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/07/2023] [Accepted: 11/17/2023] [Indexed: 02/12/2024] Open
Abstract
The Hippo pathway is conserved across species. Key mammalian Hippo pathway kinases, including MST1/2 and LATS1/2, inhibit cellular growth by inactivating the TEAD coactivators, YAP, and TAZ. Extensive research has illuminated the roles of Hippo signaling in cancer, development, and regeneration. Notably, dysregulation of Hippo pathway components not only contributes to tumor growth and metastasis, but also renders tumors resistant to therapies. This review delves into recent research on YAP/TAZ-TEAD-mediated gene regulation and biological processes in cancer. We focus on several key areas: newly identified molecular patterns of YAP/TAZ activation, emerging mechanisms that contribute to metastasis and cancer therapy resistance, unexpected roles in tumor suppression, and advances in therapeutic strategies targeting this pathway. Moreover, we provide an updated view of YAP/TAZ's biological functions, discuss ongoing controversies, and offer perspectives on specific debated topics in this rapidly evolving field.
Collapse
Affiliation(s)
- Yang Zhao
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (Y.Z.); (M.S.)
| | - Marisela Sheldon
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (Y.Z.); (M.S.)
| | - Yutong Sun
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Li Ma
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (Y.Z.); (M.S.)
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| |
Collapse
|
7
|
Lad Y, Jangam A, Carlton H, Abu-Ayyad M, Hadjipanayis C, Ivkov R, Zacharia BE, Attaluri A. Development of a Treatment Planning Framework for Laser Interstitial Thermal Therapy (LITT). Cancers (Basel) 2023; 15:4554. [PMID: 37760524 PMCID: PMC10526178 DOI: 10.3390/cancers15184554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/29/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
PURPOSE Develop a treatment planning framework for neurosurgeons treating high-grade gliomas with LITT to minimize the learning curve and improve tumor thermal dose coverage. METHODS Deidentified patient images were segmented using the image segmentation software Materialize MIMICS©. Segmented images were imported into the commercial finite element analysis (FEA) software COMSOL Multiphysics© to perform bioheat transfer simulations. The laser probe was modeled as a cylindrical object with radius 0.7 mm and length 100 mm, with a constant beam diameter. A modeled laser probe was placed in the tumor in accordance with patient specific patient magnetic resonance temperature imaging (MRTi) data. The laser energy was modeled as a deposited beam heat source in the FEA software. Penne's bioheat equation was used to model heat transfer in brain tissue. The cerebrospinal fluid (CSF) was modeled as a solid with convectively enhanced conductivity to capture heat sink effects. In this study, thermal damage-dependent blood perfusion was assessed. Pulsed laser heating was modeled based on patient treatment logs. The stationary heat source and pullback heat source techniques were modeled to compare the calculated tissue damage. The developed bioheat transfer model was compared to MRTi data obtained from a laser log during LITT procedures. The application builder module in COMSOL Multiphysics© was utilized to create a Graphical User Interface (GUI) for the treatment planning framework. RESULTS Simulations predicted increased thermal damage (10-15%) in the tumor for the pullback heat source approach compared with the stationary heat source. The model-predicted temperature profiles followed trends similar to those of the MRTi data. Simulations predicted partial tissue ablation in tumors proximal to the CSF ventricle. CONCLUSION A mobile platform-based GUI for bioheat transfer simulation was developed to aid neurosurgeons in conveniently varying the simulation parameters according to a patient-specific treatment plan. The convective effects of the CSF should be modeled with heat sink effects for accurate LITT treatment planning.
Collapse
Affiliation(s)
- Yash Lad
- Department of Mechanical Engineering, School of Science, Engineering, and Technology, The Pennsylvania State University Harrisburg, Harrisburg, PA 17057, USA
| | - Avesh Jangam
- Department of Mechanical Engineering, School of Science, Engineering, and Technology, The Pennsylvania State University Harrisburg, Harrisburg, PA 17057, USA
| | - Hayden Carlton
- Department of Radiation Oncology and Molecular Radiation Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Ma’Moun Abu-Ayyad
- Department of Mechanical Engineering, School of Science, Engineering, and Technology, The Pennsylvania State University Harrisburg, Harrisburg, PA 17057, USA
| | - Constantinos Hadjipanayis
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Robert Ivkov
- Department of Radiation Oncology and Molecular Radiation Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Mechanical Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Brad E. Zacharia
- Department of Neurosurgery, Pennsylvania State Health, Hershey, PA 17033, USA
| | - Anilchandra Attaluri
- Department of Mechanical Engineering, School of Science, Engineering, and Technology, The Pennsylvania State University Harrisburg, Harrisburg, PA 17057, USA
| |
Collapse
|
8
|
Chen L, Zhao D, Ren X, Ren J, Meng X, Fu C, Li X. Shikonin-Loaded Hollow Fe-MOF Nanoparticles for Enhanced Microwave Thermal Therapy. ACS Biomater Sci Eng 2023; 9:5405-5417. [PMID: 37638660 PMCID: PMC10498989 DOI: 10.1021/acsbiomaterials.3c00644] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 08/15/2023] [Indexed: 08/29/2023]
Abstract
Microwave (MW) thermal therapy has been widely used for the treatment of cancer in clinics, but it still shows limited efficacy and a high recurrence rate owing to non-selective heat delivery and thermo-resistance. Regulating glycolysis shows great promise to improve MW thermal therapy since glycolysis plays an important role in thermo-resistance, progression, metabolism, and recurrence. Herein, we developed a delivery nanosystem of shikonin (SK)-loaded and hyaluronic acid (HA)-modified hollow Fe-MOF (HFM), HFM@SK@HA, as an efficient glycolysis-meditated agent to improve the efficacy of MW thermal therapy. The HFM@SK@HA nanosystem shows a high SK loading capacity of 31.7 wt %. The loaded SK can be effectively released from the HFM@SK@HA under the stimulation of an acidic tumor microenvironment and MW irradiation, overcoming the intrinsically low solubility and severe toxicity of SK. We also find that the HFM@SK@HA can not only greatly improve the heating effect of MW in the tumor site but also mediate MW-enhancing dynamic therapy efficiency by catalyzing the endogenous H2O2 to generate reactive oxygen species (ROS). As such, the MW irradiation treatment in the presence of HFM@SK@HA in vitro enables a highly improved anti-tumor efficacy due to the combined effect of released SK and generated ROS on inhibiting glycolysis in cancer cells. Our in vivo experiments show that the tumor inhibition rate is up to 94.75% ± 3.63% with no obvious recurrence during the 2 weeks after treatment. This work provides a new strategy for improving the efficacy of MW thermal therapy.
Collapse
Affiliation(s)
- Lufeng Chen
- Department
of Radiation Oncology, First Clinical Medical
School and First Hospital of Shanxi Medical University, No.85 Jiefang Road, Taiyuan City 030001, PR China
| | - Dongming Zhao
- Department
of Radiation Oncology, First Clinical Medical
School and First Hospital of Shanxi Medical University, No.85 Jiefang Road, Taiyuan City 030001, PR China
- Department
of Pathology, Basic Medical School, Shanxi
Medical University, No.56 Xinjian Road, Taiyuan City 030001, PR China
| | - Xiangling Ren
- Laboratory
of Controllable Preparation and Application of Nanomaterials, Technical
Institute of Physics and Chemistry, Chinese
Academy of Sciences, No.29 East Road Zhongguancun, Beijing 100190, PR China
- CAS
Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Jun Ren
- Laboratory
of Controllable Preparation and Application of Nanomaterials, Technical
Institute of Physics and Chemistry, Chinese
Academy of Sciences, No.29 East Road Zhongguancun, Beijing 100190, PR China
- CAS
Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Xianwei Meng
- Laboratory
of Controllable Preparation and Application of Nanomaterials, Technical
Institute of Physics and Chemistry, Chinese
Academy of Sciences, No.29 East Road Zhongguancun, Beijing 100190, PR China
- CAS
Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Changhui Fu
- Laboratory
of Controllable Preparation and Application of Nanomaterials, Technical
Institute of Physics and Chemistry, Chinese
Academy of Sciences, No.29 East Road Zhongguancun, Beijing 100190, PR China
- CAS
Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Xianfeng Li
- Department
of Radiation Oncology, First Clinical Medical
School and First Hospital of Shanxi Medical University, No.85 Jiefang Road, Taiyuan City 030001, PR China
- Department
of Pathology, Basic Medical School, Shanxi
Medical University, No.56 Xinjian Road, Taiyuan City 030001, PR China
| |
Collapse
|
9
|
Chia DKA, Demuytere J, Ernst S, Salavati H, Ceelen W. Effects of Hyperthermia and Hyperthermic Intraperitoneal Chemoperfusion on the Peritoneal and Tumor Immune Contexture. Cancers (Basel) 2023; 15:4314. [PMID: 37686590 PMCID: PMC10486595 DOI: 10.3390/cancers15174314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 08/12/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
Hyperthermia combined with intraperitoneal (IP) drug delivery is increasingly used in the treatment of peritoneal metastases (PM). Hyperthermia enhances tumor perfusion and increases drug penetration after IP delivery. The peritoneum is increasingly recognized as an immune-privileged organ with its own distinct immune microenvironment. Here, we review the immune landscape of the healthy peritoneal cavity and immune contexture of peritoneal metastases. Next, we review the potential benefits and unwanted tumor-promoting effects of hyperthermia and the associated heat shock response on the tumor immune microenvironment. We highlight the potential modulating effect of hyperthermia on the biomechanical properties of tumor tissue and the consequences for immune cell infiltration. Data from translational and clinical studies are reviewed. We conclude that (mild) hyperthermia and HIPEC have the potential to enhance antitumor immunity, but detailed further studies are required to distinguish beneficial from tumor-promoting effects.
Collapse
Affiliation(s)
- Daryl K. A. Chia
- Department of Surgery, National University Hospital, National University Health System, Singapore 119074, Singapore
| | - Jesse Demuytere
- Department of Human Structure and Repair, Experimental Surgery Lab, Ghent University, 9052 Ghent, Belgium; (J.D.); (S.E.); (H.S.)
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
| | - Sam Ernst
- Department of Human Structure and Repair, Experimental Surgery Lab, Ghent University, 9052 Ghent, Belgium; (J.D.); (S.E.); (H.S.)
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
| | - Hooman Salavati
- Department of Human Structure and Repair, Experimental Surgery Lab, Ghent University, 9052 Ghent, Belgium; (J.D.); (S.E.); (H.S.)
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
| | - Wim Ceelen
- Department of Human Structure and Repair, Experimental Surgery Lab, Ghent University, 9052 Ghent, Belgium; (J.D.); (S.E.); (H.S.)
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
- Department of GI Surgery, Ghent University Hospital, 9000 Ghent, Belgium
| |
Collapse
|
10
|
Scutigliani EM, Lobo-Cerna F, Mingo Barba S, Scheidegger S, Krawczyk PM. The Effects of Heat Stress on the Transcriptome of Human Cancer Cells: A Meta-Analysis. Cancers (Basel) 2022; 15:cancers15010113. [PMID: 36612111 PMCID: PMC9817844 DOI: 10.3390/cancers15010113] [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: 11/17/2022] [Revised: 12/21/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
Hyperthermia is clinically applied cancer treatment in conjunction with radio- and/or chemotherapy, in which the tumor volume is exposed to supraphysiological temperatures. Since cells can effectively counteract the effects of hyperthermia by protective measures that are commonly known as the heat stress response, the identification of cellular processes that are essential for surviving hyperthermia could lead to novel treatment strategies that improve its therapeutic effects. Here, we apply a meta-analytic approach to 18 datasets that capture hyperthermia-induced transcriptome alterations in nine different human cancer cell lines. We find, in line with previous reports, that hyperthermia affects multiple processes, including protein folding, cell cycle, mitosis, and cell death, and additionally uncover expression changes of genes involved in KRAS signaling, inflammatory responses, TNF-a signaling and epithelial-to-mesenchymal transition (EMT). Interestingly, however, we also find a considerable inter-study variability, and an apparent absence of a 'universal' heat stress response signature, which is likely caused by the differences in experimental conditions. Our results suggest that gene expression alterations after heat stress are driven, to a large extent, by the experimental context, and call for a more extensive, controlled study that examines the effects of key experimental parameters on global gene expression patterns.
Collapse
Affiliation(s)
- Enzo M. Scutigliani
- Department of Medical Biology, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Treatment and Quality of Life, 1081HV Amsterdam, The Netherlands
- Correspondence: (E.M.S.); (P.M.K.)
| | - Fernando Lobo-Cerna
- Department of Medical Biology, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Treatment and Quality of Life, 1081HV Amsterdam, The Netherlands
| | - Sergio Mingo Barba
- ZHAW School of Engineering, University of Applied Sciences, CH 8401 Winterthur, Switzerland
- Chemistry Department, University of Fribourg, 1700 Fribourg, Switzerland
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland
| | - Stephan Scheidegger
- ZHAW School of Engineering, University of Applied Sciences, CH 8401 Winterthur, Switzerland
| | - Przemek M. Krawczyk
- Department of Medical Biology, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Treatment and Quality of Life, 1081HV Amsterdam, The Netherlands
- Correspondence: (E.M.S.); (P.M.K.)
| |
Collapse
|
11
|
Evaluation of the Heat Shock Protein 90 Inhibitor Ganetespib as a Sensitizer to Hyperthermia-Based Cancer Treatments. Cancers (Basel) 2022; 14:cancers14215250. [PMID: 36358669 PMCID: PMC9654690 DOI: 10.3390/cancers14215250] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/17/2022] [Accepted: 10/17/2022] [Indexed: 11/24/2022] Open
Abstract
Simple Summary Hyperthermia boosts the effects of radio- and chemotherapy regimens, but its clinical potential is hindered by the ability of (cancer) cells to activate a protective mechanism known as the heat stress response. Strategies that inhibit its activation or functions have the potential, therefore, to improve the overall efficacy of hyperthermia-based treatments. In this study, we evaluated the efficacy of the HSP90 inhibitor ganetespib in promoting the effects of radiotherapy or cisplatin combined with hyperthermia in vitro and in a cervix cancer mouse model. Abstract Hyperthermia is being used as a radio- and chemotherapy sensitizer for a growing range of tumor subtypes in the clinic. Its potential is limited, however, by the ability of cancer cells to activate a protective mechanism known as the heat stress response (HSR). The HSR is marked by the rapid overexpression of molecular chaperones, and recent advances in drug development make their inhibition an attractive option to improve the efficacy of hyperthermia-based therapies. Our previous in vitro work showed that a single, short co-treatment with a HSR (HSP90) inhibitor ganetespib prolongs and potentiates the effects of hyperthermia on DNA repair, enhances hyperthermic sensitization to radio- and chemotherapeutic agents, and reduces thermotolerance. In the current study, we first validated these results using an extended panel of cell lines and more robust methodology. Next, we examined the effects of hyperthermia and ganetespib on global proteome changes. Finally, we evaluated the potential of ganetespib to boost the efficacy of thermo-chemotherapy and thermo-radiotherapy in a xenograft murine model of cervix cancer. Our results revealed new insights into the effects of HSR inhibition on cellular responses to heat and show that ganetespib could be employed to increase the efficacy of hyperthermia when combined with radiation.
Collapse
|
12
|
Javid H, Hashemian P, Yazdani S, Sharbaf Mashhad A, Karimi-Shahri M. The role of heat shock proteins in metastatic colorectal cancer: A review. J Cell Biochem 2022; 123:1704-1735. [PMID: 36063530 DOI: 10.1002/jcb.30326] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 08/20/2022] [Accepted: 08/24/2022] [Indexed: 01/18/2023]
Abstract
Heat shock proteins (HSPs) are a large molecular chaperone family classified by their molecular weights, including HSP27, HSP40, HSP60, HSP70, HSP90, and HSP110. HSPs are likely to have antiapoptotic properties and participate actively in various processes such as tumor cell proliferation, invasion, metastases, and death. In this review, we discuss comprehensively the functions of HSPs associated with the progression of colorectal cancer (CRC) and metastasis and resistance to cancer therapy. Taken together, HSPs have numerous clinical applications as biomarkers for cancer diagnosis and prognosis and potential therapeutic targets for CRC and its related metastases.
Collapse
Affiliation(s)
- Hossein Javid
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Laboratory Sciences, Varastegan Institute for Medical Sciences, Mashhad, Iran
| | - Pedram Hashemian
- Jahad Daneshgahi Research Committee, Jahad Daneshgahi Institute, Mashhad, Iran
| | - Shaghayegh Yazdani
- Department of Medical Laboratory Sciences, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Laboratory Sciences, Ilam University of Medical Sciences, Ilam, Iran
| | - Alireza Sharbaf Mashhad
- Department of Medical Laboratory Sciences, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehdi Karimi-Shahri
- Department of Pathology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pathology, School of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
| |
Collapse
|
13
|
Liu P, Ye M, Wu Y, Wu L, Lan K, Wu Z. Hyperthermia combined with immune checkpoint inhibitor therapy: Synergistic sensitization and clinical outcomes. Cancer Med 2022; 12:3201-3221. [PMID: 35908281 PMCID: PMC9939221 DOI: 10.1002/cam4.5085] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 06/24/2022] [Accepted: 06/29/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Within the field of oncotherapy, research interest regarding immunotherapy has risen to the point that it is now seen as a key application. However, inherent disadvantages of immune checkpoint inhibitors (ICIs), such as their low response rates and immune-related adverse events (irAEs), currently restrict their clinical application. Were these disadvantages to be overcome, more patients could derive prolonged benefits from ICIs. At present, many basic experiments and clinical studies using hyperthermia combined with ICI treatment (HIT) have been performed and shown the potential to address the above challenges. Therefore, this review extensively summarizes the knowledge and progress of HIT for analysis and discusses the effect and feasibility. METHODS In this review, we explored the PubMed and clinicaltrials.gov databases, with regard to the searching terms "immune checkpoint inhibitor, immunotherapy, hyperthermia, ablation, photothermal therapy". RESULTS By reviewing the literature, we analyzed how hyperthermia influences tumor immunology and improves the efficacy of ICI. Hyperthermia can trigger a series of multifactorial molecular cascade reactions between tumors and immunization and can significantly induce cytological modifications within the tumor microenvironment (TME). The pharmacological potency of ICIs can be enhanced greatly through the immunomodulatory amelioration of immunosuppression, and the activation of immunostimulation. Emerging clinical trials outcome regarding HIT have verified and enriched the theoretical foundation of synergistic sensitization. CONCLUSION HIT research is now starting to transition from preclinical studies to clinical investigations. Several HIT sensitization mechanisms have been reflected and demonstrated as significant survival benefits for patients through pioneering clinical trials. Further studies into the theoretical basis and practical standards of HIT, combined with larger-scale clinical studies involving more cancer types, will be necessary for the future.
Collapse
Affiliation(s)
- Pengyuan Liu
- Oncology & Radiotherapy DepartmentZhejiang HospitalHangzhouChina,Second Clinical Medical CollegeZhejiang Chinese Medical UniversityHangzhouChina
| | - Mengna Ye
- Second Clinical Medical CollegeZhejiang Chinese Medical UniversityHangzhouChina
| | - Yajun Wu
- Department of TCM PharmacyZhejiang HospitalHangzhouChina
| | - Lichao Wu
- College of Basic Medical SciencesZhejiang Chinese Medical UniversityHangzhouChina
| | - Kaiping Lan
- Oncology Department of Combination of Traditional Chinese and Western MedicineTonglu Hospital of Traditional Chinese MedicineHangzhouChina
| | - Zhibing Wu
- Oncology & Radiotherapy DepartmentZhejiang HospitalHangzhouChina
| |
Collapse
|
14
|
Van Hattum JW, Scutigliani EM, Helderman RFCPA, Zweije R, Rodermond HM, Oei AL, Crezee J, Oddens JR, De Reijke TM, Krawczyk PM. A scalable hyperthermic intravesical chemotherapy (HIVEC) setup for rat models of bladder cancer. Sci Rep 2022; 12:7017. [PMID: 35488115 PMCID: PMC9054747 DOI: 10.1038/s41598-022-11016-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 03/24/2022] [Indexed: 12/24/2022] Open
Abstract
Hyperthermic intravesical chemotherapy (HIVEC)—whereby the bladder is heated to ± 43 °C during a chemotherapy instillation—can improve outcomes of non-muscle invasive bladder cancer (NMIBC) treatments. Experiments in animal models are required to explore new hyperthermia based treatments. Existing HIVEC devices are not suitable for rodents or large-scale animal trials. We present a HIVEC setup compatible with orthotopic rat models. An externally heated chemotherapeutic solution is circulated in the bladder through a double-lumen catheter with flow rates controlled using a peristaltic pump. Temperature sensors in the inflow channel, bladder and outflow channel allow temperature monitoring and adjustments in real-time. At a constant flow rate of 2.5 mL/min the system rapidly reaches the desired bladder temperature of 42–43 °C with minimal variability throughout a one-hour treatment in a rat bladder phantom, as well as in euthanised and live rats. Mean intraluminal bladder temperatures were 42.92 °C (SD = 0.15 °C), 42.45 °C (SD = 0.37 °C) and 42.52 °C (SD = 0.09 °C) in the bladder phantom, euthanised, and live rats respectively. Thermal camera measurements showed homogenous heat distributions over the bladder wall. The setup provides well-controlled thermal dose and the upscaling needed for performing large scale HIVEC experiments in rats.
Collapse
Affiliation(s)
- J W Van Hattum
- Department of Urology, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 15, 1105 AZ, Amsterdam, The Netherlands
| | - E M Scutigliani
- Department of Medical Biology, Amsterdam University Medical Centers, Cancer Center Amsterdam, University of Amsterdam, Meibergdreef 15, 1105 AZ, Amsterdam, The Netherlands
| | - R F C P A Helderman
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers, Cancer Center Amsterdam, University of Amsterdam, P.O. Box 22700, 1100 DE, Amsterdam, The Netherlands.,Department of Radiation Oncology, Amsterdam University Medical Centers, Cancer Center Amsterdam, University of Amsterdam, P.O. Box 22700, 1100 DE, Amsterdam, The Netherlands
| | - R Zweije
- Department of Radiation Oncology, Amsterdam University Medical Centers, Cancer Center Amsterdam, University of Amsterdam, P.O. Box 22700, 1100 DE, Amsterdam, The Netherlands
| | - H M Rodermond
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers, Cancer Center Amsterdam, University of Amsterdam, P.O. Box 22700, 1100 DE, Amsterdam, The Netherlands
| | - A L Oei
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers, Cancer Center Amsterdam, University of Amsterdam, P.O. Box 22700, 1100 DE, Amsterdam, The Netherlands.,Department of Radiation Oncology, Amsterdam University Medical Centers, Cancer Center Amsterdam, University of Amsterdam, P.O. Box 22700, 1100 DE, Amsterdam, The Netherlands
| | - J Crezee
- Department of Radiation Oncology, Amsterdam University Medical Centers, Cancer Center Amsterdam, University of Amsterdam, P.O. Box 22700, 1100 DE, Amsterdam, The Netherlands
| | - J R Oddens
- Department of Urology, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 15, 1105 AZ, Amsterdam, The Netherlands
| | - T M De Reijke
- Department of Urology, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 15, 1105 AZ, Amsterdam, The Netherlands
| | - P M Krawczyk
- Department of Medical Biology, Amsterdam University Medical Centers, Cancer Center Amsterdam, University of Amsterdam, Meibergdreef 15, 1105 AZ, Amsterdam, The Netherlands.
| |
Collapse
|
15
|
Ganguly S, Margel S. Design of Magnetic Hydrogels for Hyperthermia and Drug Delivery. Polymers (Basel) 2021; 13:4259. [PMID: 34883761 PMCID: PMC8659876 DOI: 10.3390/polym13234259] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 11/30/2021] [Accepted: 12/02/2021] [Indexed: 12/28/2022] Open
Abstract
Hydrogels are spatially organized hydrophilic polymeric systems that exhibit unique features in hydrated conditions. Among the hydrogel family, composite hydrogels are a special class that are defined as filler-containing systems with some tailor-made properties. The composite hydrogel family includes magnetic-nanoparticle-integrated hydrogels. Magnetic hydrogels (MHGs) show magneto-responsiveness, which is observed when they are placed in a magnetic field (static or oscillating). Because of their tunable porosity and internal morphology they can be used in several biomedical applications, especially diffusion-related smart devices. External stimuli may influence physical and chemical changes in these hydrogels, particularly in terms of volume and shape morphing. One of the most significant external stimuli for hydrogels is a magnetic field. This review embraces a brief overview of the fabrication of MHGs and two of their usages in the biomedical area: drug delivery and hyperthermia-based anti-cancer activity. As for the saturation magnetization imposed on composite MHGs, they are easily heated in the presence of an alternating magnetic field and the temperature increment is dependent on the magnetic nanoparticle concentration and exposure time. Herein, we also discuss the mode of different therapies based on non-contact hyperthermia heating.
Collapse
Affiliation(s)
- Sayan Ganguly
- Bar-Ilan Institute for Nanotechnology and Advanced Materials, Department of Chemistry, Bar-Ilan University, Ramat-Gan 52900, Israel
| | - Shlomo Margel
- Bar-Ilan Institute for Nanotechnology and Advanced Materials, Department of Chemistry, Bar-Ilan University, Ramat-Gan 52900, Israel
| |
Collapse
|
16
|
Bai L, Pfeifer T, Gross W, De La Torre C, Zhao S, Liu L, Schaefer M, Herr I. Establishment of Tumor Treating Fields Combined With Mild Hyperthermia as Novel Supporting Therapy for Pancreatic Cancer. Front Oncol 2021; 11:738801. [PMID: 34804927 PMCID: PMC8597267 DOI: 10.3389/fonc.2021.738801] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 10/18/2021] [Indexed: 12/22/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly malignant tumor with poor prognosis and limited therapeutic options. Alternating electrical fields with low intensity called "Tumor Treating Fields" (TTFields) are a new, non-invasive approach with almost no side effects and phase 3 trials are ongoing in advanced PDAC. We evaluated TTFields in combination with mild hyperthermia. Three established human PDAC cell lines and an immortalized pancreatic duct cell line were treated with TTFields and hyperthermia at 38.5°C, followed by microscopy, assays for MTT, migration, colony and sphere formation, RT-qPCR, FACS, Western blot, microarray and bioinformatics, and in silico analysis using the online databases GSEA, KEGG, Cytoscape-String, and Kaplan-Meier Plotter. Whereas TTFields and hyperthermia alone had weak effects, their combination strongly inhibited the viability of malignant, but not those of nonmalignant cells. Progression features and the cell cycle were impaired, and autophagy was induced. The identified target genes were key players in autophagy, the cell cycle and DNA repair. The expression profiles of part of these target genes were significantly involved in the survival of PDAC patients. In conclusion, the combination of TTFields with mild hyperthermia results in greater efficacy without increased toxicity and could be easily clinically approved as supporting therapy.
Collapse
Affiliation(s)
- Liping Bai
- Molecular OncoSurgery, Section Surgical Research, Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Tobias Pfeifer
- Molecular OncoSurgery, Section Surgical Research, Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Wolfgang Gross
- Molecular OncoSurgery, Section Surgical Research, Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Carolina De La Torre
- Medical Research Center, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Shuyang Zhao
- Department of Hematology, Oncology and Rheumatology, Internal Medicine V, University Hospital of Heidelberg, Heidelberg, Germany
| | - Li Liu
- Molecular OncoSurgery, Section Surgical Research, Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Michael Schaefer
- Molecular OncoSurgery, Section Surgical Research, Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Ingrid Herr
- Molecular OncoSurgery, Section Surgical Research, Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
17
|
Preclinical Studies in Small Animals for Advanced Drug Delivery Using Hyperthermia and Intravital Microscopy. Cancers (Basel) 2021; 13:cancers13205146. [PMID: 34680296 PMCID: PMC8534089 DOI: 10.3390/cancers13205146] [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: 08/26/2021] [Revised: 10/05/2021] [Accepted: 10/05/2021] [Indexed: 01/15/2023] Open
Abstract
This paper presents three devices suitable for the preclinical application of hyperthermia via the simultaneous high-resolution imaging of intratumoral events. (Pre)clinical studies have confirmed that the tumor micro-environment is sensitive to the application of local mild hyperthermia. Therefore, heating is a promising adjuvant to aid the efficacy of radiotherapy or chemotherapy. More so, the application of mild hyperthermia is a useful stimulus for triggered drug release from heat-sensitive nanocarriers. The response of thermosensitive nanoparticles to hyperthermia and ensuing intratumoral kinetics are considerably complex in both space and time. To obtain better insight into intratumoral processes, longitudinal imaging (preferable in high spatial and temporal resolution) is highly informative. Our devices are based on (i) an external electric heating adaptor for the dorsal skinfold model, (ii) targeted radiofrequency application, and (iii) a microwave antenna for heating of internal tumors. These models, while of some technical complexity, significantly add to the understanding of effects of mild hyperthermia warranting implementation in research on hyperthermia.
Collapse
|
18
|
Priester MI, Curto S, van Rhoon GC, ten Hagen TLM. External Basic Hyperthermia Devices for Preclinical Studies in Small Animals. Cancers (Basel) 2021; 13:cancers13184628. [PMID: 34572855 PMCID: PMC8470307 DOI: 10.3390/cancers13184628] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/09/2021] [Accepted: 09/10/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary The application of mild hyperthermia can be beneficial for solid tumor treatment by induction of sublethal effects on a tissue- and cellular level. When designing a hyperthermia experiment, several factors should be taken into consideration. In this review, multiple elementary hyperthermia devices are described in detail to aid standardization of treatment design. Abstract Preclinical studies have shown that application of mild hyperthermia (40–43 °C) is a promising adjuvant to solid tumor treatment. To improve preclinical testing, enhance reproducibility, and allow comparison of the obtained results, it is crucial to have standardization of the available methods. Reproducibility of methods in and between research groups on the same techniques is crucial to have a better prediction of the clinical outcome and to improve new treatment strategies (for instance with heat-sensitive nanoparticles). Here we provide a preclinically oriented review on the use and applicability of basic hyperthermia systems available for solid tumor thermal treatment in small animals. The complexity of these techniques ranges from a simple, low-cost water bath approach, irradiation with light or lasers, to advanced ultrasound and capacitive heating devices.
Collapse
Affiliation(s)
- Marjolein I. Priester
- Laboratory of Experimental Oncology, Department of Pathology, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands;
- Department of Radiotherapy, Erasmus MC Cancer Institute, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (S.C.); (G.C.v.R.)
| | - Sergio Curto
- Department of Radiotherapy, Erasmus MC Cancer Institute, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (S.C.); (G.C.v.R.)
| | - Gerard C. van Rhoon
- Department of Radiotherapy, Erasmus MC Cancer Institute, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (S.C.); (G.C.v.R.)
| | - Timo L. M. ten Hagen
- Laboratory of Experimental Oncology, Department of Pathology, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands;
- Correspondence:
| |
Collapse
|
19
|
Understanding MNPs Behaviour in Response to AMF in Biological Milieus and the Effects at the Cellular Level: Implications for a Rational Design That Drives Magnetic Hyperthermia Therapy toward Clinical Implementation. Cancers (Basel) 2021; 13:cancers13184583. [PMID: 34572810 PMCID: PMC8465027 DOI: 10.3390/cancers13184583] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/03/2021] [Accepted: 09/07/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Magnetic hyperthermia therapy is an alternative treatment for cancer that complements traditional therapies and that has shown great promise in recent years. In this review, we assess the current applications of this therapy in order to understand why its translation from the laboratory to the clinic has been less smooth than was anticipated, identifying the possible bottlenecks and proposing solutions to the problems encountered. Abstract Hyperthermia has emerged as a promising alternative to conventional cancer therapies and in fact, traditional hyperthermia is now commonly used in combination with chemotherapy or surgery during cancer treatment. Nevertheless, non-specific application of hyperthermia generates various undesirable side-effects, such that nano-magnetic hyperthermia has arisen a possible solution to this problem. This technique to induce hyperthermia is based on the intrinsic capacity of magnetic nanoparticles to accumulate in a given target area and to respond to alternating magnetic fields (AMFs) by releasing heat, based on different principles of physics. Unfortunately, the clinical implementation of nano-magnetic hyperthermia has not been fluid and few clinical trials have been carried out. In this review, we want to demonstrate the need for more systematic and basic research in this area, as many of the sub-cellular and molecular mechanisms associated with this approach remain unclear. As such, we shall consider here the biological effects that occur and why this theoretically well-designed nano-system fails in physiological conditions. Moreover, we will offer some guidelines that may help establish successful strategies through the rational design of magnetic nanoparticles for magnetic hyperthermia.
Collapse
|
20
|
Gupta R, Tomar R, Chakraverty S, Sharma D. Effect of manganese doping on the hyperthermic profile of ferrite nanoparticles using response surface methodology. RSC Adv 2021; 11:16942-16954. [PMID: 35479670 PMCID: PMC9032483 DOI: 10.1039/d1ra02376d] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 04/30/2021] [Indexed: 12/25/2022] Open
Abstract
Magnetic hyperthermia-based cancer therapy mediated by magnetic nanomaterials is a promising antitumoral nanotherapy, owning to its power to generate heat under the application of an alternating magnetic field. However, although the ultimate targets of these treatments, the heating potential and its relation with the magnetic behavior of the employed magnetic nanomaterials are rarely studied. Here we provide a bridge between the heating potential and magnetic properties such as anisotropy energy constant and saturation magnetization of the nano-magnets by simultaneous investigation of both hyperthermia and magnetism under a controlled set of variables given by response surface methodology. In the study, we have simultaneously investigated the effect of various synthesis parameters like cation ratio, reaction temperature and time on the magnetic response and heat generation of manganese-doped ferrite nanomaterials synthesized by a simple hydrothermal route. The optimum generation of heat and magnetization is obtained at a cationic ratio of approximately 42 at a temperature of 100 °C for a time period of 4 h. The optimized nanomaterial was then evaluated for in vitro magnetic hyperthermia application for cancer therapy against glioblastoma in terms of cell viability, effect on cellular cytoskeleton and morphological alterations. Furthermore, the correlation between the magnetic properties of the synthesized nanomaterial with its hyperthermia output was also established using K.V.M s variable where K, V and M s are the anisotropy energy constant, volume, and saturation magnetization of the nanomaterial respectively. It was found that the intensity of heat generation decreases with an increase in K.V.M s value, beyond 950 J emu g-1.
Collapse
Affiliation(s)
- Ruby Gupta
- Institute of Nano Science and Technology Knowledge City, Phase 81 Mohali-140306 Punjab India
| | - Ruchi Tomar
- Institute of Nano Science and Technology Knowledge City, Phase 81 Mohali-140306 Punjab India
| | - Suvankar Chakraverty
- Institute of Nano Science and Technology Knowledge City, Phase 81 Mohali-140306 Punjab India
| | - Deepika Sharma
- Institute of Nano Science and Technology Knowledge City, Phase 81 Mohali-140306 Punjab India
| |
Collapse
|