1
|
Berclaz LM, Burkhard-Meier A, Lechner A, Völkl M, Güler SE, Abdel-Rahman S, Mansoorian S, Kunz WG, Knösel T, Canis M, von Bergwelt-Baildon M, Issels RD, Di Gioia D, Lindner LH. Durable response to nivolumab in combination with regional hyperthermia in a patient with PD-L1-negative metastatic head and neck squamous cell carcinoma. Cancer Immunol Immunother 2025; 74:174. [PMID: 40244424 PMCID: PMC12006646 DOI: 10.1007/s00262-025-04029-9] [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: 12/16/2024] [Accepted: 03/19/2025] [Indexed: 04/18/2025]
Abstract
We report a long-lasting response to the immune checkpoint inhibitor nivolumab in combination with regional hyperthermia (RHT) in a patient with recurrent metastatic Head and Neck Squamous Cell Carcinoma (HNSCC) and negative programmed death ligand 1 (PD-L1) expression. Treatment was well tolerated with no local side effects. Tumor-related symptoms in the orbital and masticator area gradually decreased under treatment with nivolumab and RHT. Over the course of treatment, magnetic resonance imaging (MRI) showed a local tumor control in the heated tumor areas, while metastatic lesions developed in areas outside of the RHT field. This is the first case report demonstrating the feasibility and clinical potential of the addition of RHT in this patient collective with poor outcomes and low response rates to immune checkpoint inhibitors. RHT might be an additional tool to activate an immunogenic milieu responsive to immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Luc M Berclaz
- Department of Internal Medicine III, University Hospital, LMU Munich, Munich, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany.
| | - Anton Burkhard-Meier
- Department of Internal Medicine III, University Hospital, LMU Munich, Munich, Germany
| | - Axel Lechner
- Deparment of Otorhinolaryngology, University Hospital, LMU Munich, Munich, Germany
| | - Michael Völkl
- Department of Internal Medicine III, University Hospital, LMU Munich, Munich, Germany
| | - Sinan E Güler
- Department of Internal Medicine III, University Hospital, LMU Munich, Munich, Germany
| | - Sultan Abdel-Rahman
- Department of Internal Medicine III, University Hospital, LMU Munich, Munich, Germany
| | - Sina Mansoorian
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Wolfgang G Kunz
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | | | - Martin Canis
- Deparment of Otorhinolaryngology, University Hospital, LMU Munich, Munich, Germany
| | - Michael von Bergwelt-Baildon
- Department of Internal Medicine III, University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Rolf D Issels
- Department of Internal Medicine III, University Hospital, LMU Munich, Munich, Germany
| | - Dorit Di Gioia
- Department of Internal Medicine III, University Hospital, LMU Munich, Munich, Germany
| | - Lars H Lindner
- Department of Internal Medicine III, University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
2
|
De Lazzari M, Carrapiço-Seabra C, Marder D, van Rhoon GC, Curto S, Dobšíček Trefná H. Toward enhanced quality assurance guidelines for deep hyperthermia devices: a multi-institution study. Int J Hyperthermia 2024; 41:2436005. [PMID: 39658024 DOI: 10.1080/02656736.2024.2436005] [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: 08/13/2024] [Revised: 10/11/2024] [Accepted: 11/25/2024] [Indexed: 12/12/2024] Open
Abstract
INTRODUCTION Hyperthermia efficacy depends on the temperatures achieved in the target area. Therefore, hyperthermia systems must deliver both controlled and conformal heating. This study presents a comprehensive multi-institutional quality assurance (QA) evaluation of deep hyperthermia devices. METHODS Six European institutions equipped with BSD- Sigma 60 and Sigma Eye deep hyperthermia applicators participated in the study. Up to six measurements per applicator were performed in each institution. The thermal distribution in cylindrical homogeneous phantoms after 10 minutes of heating with a total power delivered of 1000 watts was assessed using the applicator's integrated mapping thermometry system. Evaluated quality parameters included temperature increase, focus location, and focus symmetry. RESULTS A total of 54 measurements were conducted, with 43 included in the analysis. All applicators, except one, achieved a temperature increase of 6 °C in 10 minutes. Central heating capabilities were demonstrated, with mean deviations from the intended location of -1.4 ± 1.6 cm for Sigma 60 and 1.5 ± 1.4 cm for Sigma Eye. Symmetry evaluations showed differences in radial temperature profiles of 6.2 ± 4.5 % for the Sigma 60 and 5.9 ± 4.4 % for the Sigma Eye. We propose minimum acceptable values for each quality parameter based on these results. CONCLUSION The measurements were reproducible with acceptable values for the various quality parameters. Potential deviations might be attributed to inaccuracies in the mapping thermometry system rather than the heating system. The presented protocol and practical recommendations should be applied for future QA measurements in deep hyperthermia.
Collapse
Affiliation(s)
- Mattia De Lazzari
- Department of Electrical Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Carolina Carrapiço-Seabra
- Department of Radiotherapy, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Dietmar Marder
- Center for Radiation Oncology KSA-KSB, Cantonal Hospital Aarau, Aarau, Switzerland
| | - Gerard C van Rhoon
- Department of Radiotherapy, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Radiation Science and Technology, Delft University of Technology, Faculty of Applied Sciences, Delft, The Netherlands
| | - Sergio Curto
- Department of Radiotherapy, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Hana Dobšíček Trefná
- Department of Electrical Engineering, Chalmers University of Technology, Gothenburg, Sweden
| |
Collapse
|
3
|
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
|
4
|
Van Dieren L, Quisenaerts T, Licata M, Beddok A, Lellouch AG, Ysebaert D, Saldien V, Peeters M, Gorbaslieva I. Combined Radiotherapy and Hyperthermia: A Systematic Review of Immunological Synergies for Amplifying Radiation-Induced Abscopal Effects. Cancers (Basel) 2024; 16:3656. [PMID: 39518094 PMCID: PMC11545184 DOI: 10.3390/cancers16213656] [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: 09/11/2024] [Revised: 10/22/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
INTRODUCTION The abscopal effect is a systemic immune response characterized by metastases regression at sites distant from the irradiated lesion. This systematic review aims to explore the immunological mechanisms of action underlying the abscopal effect and to investigate how hyperthermia (HT) can increase the chances of radiotherapy (RT) triggering systemic anti-tumor immune responses. METHODS This review is created in accordance with the PRISMA guidelines. RESULTS AND CONCLUSION HT and RT have both complementary and synergistic immunological effects. Both methods trigger danger signal release, promoting cytokine and chemokine secretion, which increases T-cell infiltration and facilitates cell death. Both treatments upregulate extracellular tumor HSP70, which could amplify DAMP recognition by macrophages and DCs, leading to stronger tumor antigen presentation and CTL-mediated immune responses. Additionally, the combined increase in cell adhesion molecules (VCAM-1, ICAM-1, E-selectin, L-selectin) could enhance leukocyte adhesion to tumors, improving lymphocyte trafficking and boosting systemic anti-tumor effects. Lastly, HT causes vasodilation and improves blood flow, which might exacerbate those distant effects. We suggest the combination of local radiotherapy with fever-range whole-body hyperthermia to optimally enhance the chances of triggering the abscopal effect mediated by the immune system.
Collapse
Affiliation(s)
- Loïc Van Dieren
- Vascularized Composite Allotransplantation Laboratory, Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Wilrijk, Belgium
| | - Tom Quisenaerts
- Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Wilrijk, Belgium
| | | | - Arnaud Beddok
- Institut Godinot, Radiation Oncology Department, 85054 Reims, France
- GCMI, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Alexandre G. Lellouch
- Vascularized Composite Allotransplantation Laboratory, Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Dirk Ysebaert
- Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Wilrijk, Belgium
- Department of Hepatobiliary, Transplantation and Endocrine Surgery, University Hospital of Antwerp, 2650 Edegem, Belgium
| | - Vera Saldien
- Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Wilrijk, Belgium
- Department of Hepatobiliary, Transplantation and Endocrine Surgery, University Hospital of Antwerp, 2650 Edegem, Belgium
| | - Marc Peeters
- Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Wilrijk, Belgium
- Department of Hepatobiliary, Transplantation and Endocrine Surgery, University Hospital of Antwerp, 2650 Edegem, Belgium
| | - Ivana Gorbaslieva
- Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Wilrijk, Belgium
- Department of Hepatobiliary, Transplantation and Endocrine Surgery, University Hospital of Antwerp, 2650 Edegem, Belgium
| |
Collapse
|
5
|
Stigliano RV, Danelyan I, Gabriadze G, Shoshiashvili L, Baker I, Hoopes PJ, Jobava R, Shubitidze F. Alternating magnetic field guiding system for MNP hyperthermia treatment of deep-seated cancers. Int J Hyperthermia 2024; 41:2391008. [PMID: 39205623 DOI: 10.1080/02656736.2024.2391008] [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: 03/08/2024] [Revised: 06/19/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024] Open
Abstract
OBJECTIVES Demonstrate the potential application of a novel, endoscope-like device to guide and focus an alternating magnetic field (AMF) for treating deep-seated cancers via magnetic nanoparticle hyperthermia (MNPH). METHODS AMF delivery, MNP activation, and eddy current distribution characteristics are investigated through experimental studies in phantoms and computational simulations using a full 3-dimensional human model. The 3D simulations compare the novel device to traditional AMF designs, including a MagForce-like, two-coil system (used clinically) and a single surface-coil system. RESULTS The results demonstrate that this approach can deliver the same magnetic field strength at the prostate's centroid as traditional AMF designs, while reducing eddy current heating by 2 to 6 times. At the same level of normal tissue heating, this method provides 5.0 times, 1.5 times, and 0.92 times the magnetic field strength to the nearest, centroid, and farthest regions of the prostate, respectively. CONCLUSIONS These results demonstrate proof-of-concept for an endoscopic magnetic field guiding and focusing system capable of delivering clinically relevant AMF from a distance. This innovative approach offers a promising alternative to conventional field delivery methods by directing AMF through the body, concentrating it in the tumor region, reducing eddy currents in surrounding healthy tissue, and avoiding exposure of nearby metallic implants.
Collapse
Affiliation(s)
| | | | | | - Levan Shoshiashvili
- Department of Electrical and Electronics Engineering, Faculty of Exact and Natural Sciences, Ivane Javakhishvili Tbilisi State University, Tbilisi, Georgia
| | - Ian Baker
- Thayer School of Engineering at Dartmouth College, Hanover, NH, USA
| | - P Jack Hoopes
- Thayer School of Engineering at Dartmouth College, Hanover, NH, USA
- Geisel School of Medicine at Dartmouth College, Hanover, NH, USA
| | | | | |
Collapse
|
6
|
Ashar H, Singh A, Kishore D, Neel T, More S, Liu C, Dugat D, Ranjan A. Enabling Chemo-Immunotherapy with HIFU in Canine Cancer Patients. Ann Biomed Eng 2024; 52:1859-1872. [PMID: 37162696 DOI: 10.1007/s10439-023-03194-1] [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: 01/31/2023] [Accepted: 03/23/2023] [Indexed: 05/11/2023]
Abstract
High intensity focused ultrasound (HIFU) is a promising non-invasive technique for treating solid tumors using thermal and histotripsy-based mechanical ablation. However, its clinical significance in different tumor types is not fully understood. To assess its therapeutic efficacy and immunomodulatory properties, we compared HIFU thermal ablation and histotripsy ablation in dogs with spontaneous tumors. We also evaluated the ability of non-ablative HIFU-based mild hyperthermia (40-45 ºC) to improve Doxorubicin delivery and immunomodulation. Our results showed that HIFU thermal ablation induced tumor remission in the majority of treated patients over 60 days, while histotripsy achieved partial response to stable disease persistence. The adverse effects of thermal ablation were minor to moderate, while histotripsy exposures were relatively well-tolerated. Furthermore, we observed a correlation between HIFU-therapeutic response and serum anti-tumor cytokine profiles and the presence of functionally active cytotoxic immune cells in patients. Similarly, Doxorubicin-treated patients showed improved drug delivery, efficacy, and anti-tumor immune responses with HIFU hyperthermia. In conclusion, our study demonstrates that depending on the tumor type and treatment parameters, HIFU treatments can enable tumor growth control, immune activation, and chemotherapy in veterinary patient. These findings have significant clinical implications and highlight the potential of HIFU as a promising cancer treatment approach.
Collapse
Affiliation(s)
- Harshini Ashar
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, 169 McElroy Hall, Stillwater, OK, 74078, USA
| | - Akansha Singh
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, 169 McElroy Hall, Stillwater, OK, 74078, USA
| | | | - Tina Neel
- Neel Veterinary Hospital, Oklahoma City, OK, 73127, USA
| | - Sunil More
- Department of Veterinary Pathobiology, Oklahoma State University, Stillwater, OK, 74078, USA
| | - Chenang Liu
- The School of Industrial Engineering & Management, Oklahoma State University, Stillwater, OK, 74078, USA
| | - Danielle Dugat
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, 169 McElroy Hall, Stillwater, OK, 74078, USA
| | - Ashish Ranjan
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, 169 McElroy Hall, Stillwater, OK, 74078, USA.
| |
Collapse
|
7
|
Wong SM, Akbulatov A, Macsemchuk CA, Headrick A, Luo P, Drake JM, Waspe AC. An augmented hybrid multibaseline and referenceless MR thermometry motion compensation algorithm for MRgHIFU hyperthermia. Magn Reson Med 2024; 91:2266-2277. [PMID: 38181187 DOI: 10.1002/mrm.29988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 12/04/2023] [Accepted: 12/08/2023] [Indexed: 01/07/2024]
Abstract
PURPOSE A hybrid principal component analysis and projection onto dipole fields (PCA-PDF) MR thermometry motion compensation algorithm was optimized with atlas image augmentation and validated. METHODS Experiments were conducted on a 3T Philips MRI and Profound V1 Sonalleve high intensity focused ultrasound (high intensity focused ultrasound system. An MR-compatible robot was configured to induce motion on custom gelatin phantoms. Trials with periodic and sporadic motion were introduced on phantoms while hyperthermia was administered. The PCA-PDF algorithm was augmented with a predictive atlas to better compensate for larger sporadic motion. RESULTS During periodic motion, the temperature SD in the thermometry was improved from1 . 1 ± 0 . 1 $$ 1.1\pm 0.1 $$ to0 . 5 ± 0 . 1 ∘ $$ 0.5\pm 0.{1}^{\circ } $$ C with both the original and augmented PCA-PDF application. For large sporadic motion, the augmented atlas improved the motion compensation from the original PCA-PDF correction from8 . 8 ± 0 . 5 $$ 8.8\pm 0.5 $$ to0 . 7 ± 0 . 1 ∘ $$ 0.7\pm 0.{1}^{\circ } $$ C. CONCLUSION The PCA-PDF algorithm improved temperature accuracy to <1°C during periodic motion, but was not able to adequately address sporadic motion. By augmenting the PCA-PDF algorithm, temperature SD during large sporadic motion was also reduced to <1°C, greatly improving the original PCA-PDF algorithm.
Collapse
Affiliation(s)
- Suzanne M Wong
- The Wilfred and Joyce Posluns Centre for Image-Guided Innovation and Theraputic Intervention, The Hospital for Sick Children, Toronto, Ontario, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Arthur Akbulatov
- The Wilfred and Joyce Posluns Centre for Image-Guided Innovation and Theraputic Intervention, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Craig A Macsemchuk
- The Wilfred and Joyce Posluns Centre for Image-Guided Innovation and Theraputic Intervention, The Hospital for Sick Children, Toronto, Ontario, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Andrew Headrick
- The Wilfred and Joyce Posluns Centre for Image-Guided Innovation and Theraputic Intervention, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Phoebe Luo
- The Wilfred and Joyce Posluns Centre for Image-Guided Innovation and Theraputic Intervention, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - James M Drake
- The Wilfred and Joyce Posluns Centre for Image-Guided Innovation and Theraputic Intervention, The Hospital for Sick Children, Toronto, Ontario, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Adam C Waspe
- The Wilfred and Joyce Posluns Centre for Image-Guided Innovation and Theraputic Intervention, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Medical Imaging, University of Toronto, Toronto, Ontario, Canada
- Department of Material Science and Engineering, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
8
|
Lei L, Liu P, Jing W, Wu Z. Advancements in modifying the efficacy of immunotherapies through the thermal effects of nanomaterials. NANO TRANSMED 2023; 2:100022. [DOI: 10.1016/j.ntm.2023.100022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2024]
|
9
|
Liu P, Foiret J, Situ Y, Zhang N, Kare AJ, Wu B, Raie MN, Ferrara KW, Qi LS. Sonogenetic control of multiplexed genome regulation and base editing. Nat Commun 2023; 14:6575. [PMID: 37852951 PMCID: PMC10584809 DOI: 10.1038/s41467-023-42249-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 10/05/2023] [Indexed: 10/20/2023] Open
Abstract
Manipulating gene expression in the host genome with high precision is crucial for controlling cellular function and behavior. Here, we present a precise, non-invasive, and tunable strategy for controlling the expression of multiple endogenous genes both in vitro and in vivo, utilizing ultrasound as the stimulus. By engineering a hyper-efficient dCas12a and effector under a heat shock promoter, we demonstrate a system that can be inducibly activated through thermal energy produced by ultrasound absorption. This system allows versatile thermal induction of gene activation or base editing across cell types, including primary T cells, and enables multiplexed gene activation using a single guide RNA array. In mouse models, localized temperature elevation guided by high-intensity focused ultrasound effectively triggers reporter gene expression in implanted cells. Our work underscores the potential of ultrasound as a clinically viable approach to enhance cell and gene-based therapies via precision genome and epigenome engineering.
Collapse
Affiliation(s)
- Pei Liu
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, School of Medicine, Stanford University, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Josquin Foiret
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Yinglin Situ
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Nisi Zhang
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Aris J Kare
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, School of Medicine, Stanford University, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Bo Wu
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Marina N Raie
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Katherine W Ferrara
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, School of Medicine, Stanford University, Stanford, CA, USA.
| | - Lei S Qi
- Department of Bioengineering, Stanford University, Stanford, CA, USA.
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA.
- Chan Zuckerberg Biohub - San Francisco, San Francisco, CA, USA.
| |
Collapse
|
10
|
Zhang Y, Li Z, Huang Y, Zou B, Xu Y. Amplifying cancer treatment: advances in tumor immunotherapy and nanoparticle-based hyperthermia. Front Immunol 2023; 14:1258786. [PMID: 37869003 PMCID: PMC10587571 DOI: 10.3389/fimmu.2023.1258786] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 09/19/2023] [Indexed: 10/24/2023] Open
Abstract
In the quest for cancer treatment modalities with greater effectiveness, the combination of tumor immunotherapy and nanoparticle-based hyperthermia has emerged as a promising frontier. The present article provides a comprehensive review of recent advances and cutting-edge research in this burgeoning field and examines how these two treatment strategies can be effectively integrated. Tumor immunotherapy, which harnesses the immune system to recognize and attack cancer cells, has shown considerable promise. Concurrently, nanoparticle-based hyperthermia, which utilizes nanotechnology to promote selective cell death by raising the temperature of tumor cells, has emerged as an innovative therapeutic approach. While both strategies have individually shown potential, combination of the two modalities may amplify anti-tumor responses, with improved outcomes and reduced side effects. Key studies illustrating the synergistic effects of these two approaches are highlighted, and current challenges and future prospects in the field are discussed. As we stand on the precipice of a new era in cancer treatment, this review underscores the importance of continued research and collaboration in bringing these innovative treatments from the bench to the bedside.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Radiation Oncology, Division of Thoracic Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Zheng Li
- Department of Radiation Oncology, Division of Thoracic Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ying Huang
- College of Management, Sichuan Agricultural University, Chengdu, China
| | - Bingwen Zou
- Department of Radiation Oncology, Division of Thoracic Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yong Xu
- Department of Radiation Oncology, Division of Thoracic Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
11
|
Kim S, Ahn JH, Jeong DI, Yang M, Jeong JH, Choi YE, Kim HJ, Han Y, Karmakar M, Ko HJ, Cho HJ. Alum-tuned hyaluronic acid-based hydrogel with immune checkpoint inhibition for immunophoto therapy of cancer. J Control Release 2023; 362:1-18. [PMID: 37595669 DOI: 10.1016/j.jconrel.2023.08.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 07/25/2023] [Accepted: 08/13/2023] [Indexed: 08/20/2023]
Abstract
Alum-crosslinked hyaluronic acid-dopamine (HD) hydrogel containing indocyanine green (ICG) with anti-programmed cell death-1 (PD-1) antibody (Ab) administration was developed for immunophoto therapy of cancer. Alum modulates the rheological characteristics of hydrogel for enabling syringe injection, shear-thinning feature, and slower biodegradation. In addition, alum in HD-based hydrogel provided CD8+ T cell-mediated immune responses for cancer therapy. ICG in the hydrogel under near-infrared (NIR) light exposure may induce hyperthermia and generate singlet oxygen for selective cancer cell killing. HD/alum/ICG hydrogel injection with NIR laser irradiation elevated PD-1 level in CD8+ T cells. Administration of PD-1 Ab aiming at highly expressed PD-1 in T cells may amplify the anticancer efficacies of HD/alum/ICG hydrogel along with NIR laser. HD/alum/ICG hydrogel with NIR light may have both CD8+ T cell-linked immune responses and ICG-related photodynamic/photothermal effects. Additional injection of immune checkpoint inhibitor can ultimately suppress primary and distant tumor growth by combination with those therapeutic actions.
Collapse
Affiliation(s)
- Sungyun Kim
- Department of Pharmacy, College of Pharmacy, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Jae-Hee Ahn
- Department of Pharmacy, College of Pharmacy, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Da In Jeong
- Department of Pharmacy, College of Pharmacy, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Mingyu Yang
- Department of Pharmacy, College of Pharmacy, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Jae-Hyeon Jeong
- Department of Pharmacy, College of Pharmacy, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Yeoung Eun Choi
- Department of Pharmacy, College of Pharmacy, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Hyun Jin Kim
- Department of Pharmacy, College of Pharmacy, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Youngjoo Han
- Department of Pharmacy, College of Pharmacy, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Mrinmoy Karmakar
- Department of Pharmacy, College of Pharmacy, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Hyun-Jeong Ko
- Department of Pharmacy, College of Pharmacy, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea; Kangwon Institute of Inclusive Technology, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea.
| | - Hyun-Jong Cho
- Department of Pharmacy, College of Pharmacy, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea; Kangwon Institute of Inclusive Technology, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea.
| |
Collapse
|
12
|
Kumar PPP, Lim DK. Photothermal Effect of Gold Nanoparticles as a Nanomedicine for Diagnosis and Therapeutics. Pharmaceutics 2023; 15:2349. [PMID: 37765317 PMCID: PMC10534847 DOI: 10.3390/pharmaceutics15092349] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 09/05/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
Gold nanoparticles (AuNPs) have received great attention for various medical applications due to their unique physicochemical properties. AuNPs with tunable optical properties in the visible and near-infrared regions have been utilized in a variety of applications such as in vitro diagnostics, in vivo imaging, and therapeutics. Among the applications, this review will pay more attention to recent developments in diagnostic and therapeutic applications based on the photothermal (PT) effect of AuNPs. In particular, the PT effect of AuNPs has played an important role in medical applications utilizing light, such as photoacoustic imaging, photon polymerase chain reaction (PCR), and hyperthermia therapy. First, we discuss the fundamentals of the optical properties in detail to understand the background of the PT effect of AuNPs. For diagnostic applications, the ability of AuNPs to efficiently convert absorbed light energy into heat to generate enhanced acoustic waves can lead to significant enhancements in photoacoustic signal intensity. Integration of the PT effect of AuNPs with PCR may open new opportunities for technological innovation called photonic PCR, where light is used to enable fast and accurate temperature cycling for DNA amplification. Additionally, beyond the existing thermotherapy of AuNPs, the PT effect of AuNPs can be further applied to cancer immunotherapy. Controlled PT damage to cancer cells triggers an immune response, which is useful for obtaining better outcomes in combination with immune checkpoint inhibitors or vaccines. Therefore, this review examines applications to nanomedicine based on the PT effect among the unique optical properties of AuNPs, understands the basic principles, the advantages and disadvantages of each technology, and understands the importance of a multidisciplinary approach. Based on this, it is expected that it will help understand the current status and development direction of new nanoparticle-based disease diagnosis methods and treatment methods, and we hope that it will inspire the development of new innovative technologies.
Collapse
Affiliation(s)
| | - Dong-Kwon Lim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea;
- Department of Integrative Energy Engineering, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
- Brain Science Institute, Korea Institute of Science and Technology (KIST), 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| |
Collapse
|
13
|
Zhang Y, Lu X, Ji H, Zheng L, Chen G, Qian Y. Effects of Deep Hyperthermia Combined with Intraperitoneal Chemotherapy on Liver-Kidney Function, Immune Function, and Long-Term Survival in Patients with Abdominal Metastases. Emerg Med Int 2023; 2023:5878402. [PMID: 37125381 PMCID: PMC10147530 DOI: 10.1155/2023/5878402] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/18/2022] [Accepted: 03/29/2023] [Indexed: 05/02/2023] Open
Abstract
Objectives To analyze the effects of deep hyperthermia combined with intraperitoneal chemotherapy on liver-kidney function, immune function, and long-term survival in patients with abdominal metastases. Methods A total of 88 patients with abdominal metastases confirmed in the hospital were enrolled as the research objects between August 2018 and August 2021. They were randomly divided into control group (n = 44) and observation group (n = 44). The control group was treated with intraperitoneal chemotherapy, while observation group was additionally treated with deep hyperthermia. The general clinical data of patients were recorded. The short-term and long-term curative effects were evaluated. The occurrence of side effects in both groups was recorded. Before and after treatment, levels of alanine transaminase (ALT) and aspartate transaminase (AST) were detected by full-automatic biochemical analyzer. The level of blood urea nitrogen (BUN) was detected by the urease electrode method. The level of serum creatinine (Scr) was detected by the picric acid method. The levels of CD3 +, CD4 +, CD8 +, and NK cells were detected by BD FACSCalibur flow cytometer. Results There was no significant difference in clinical data between the two groups (P > 0.05). In the observation group, ORR was significantly higher than that in the control group (54.55% vs 29.55%) (P < 0.05), OS was significantly longer than that in the control group (P < 0.05), and median survival time and mPFS were longer than those in the control group. After treatment, the levels of ALT, AST, BUN, and Scr were significantly increased in the control group (P < 0.05), but there was no significant difference in peripheral blood CD3 +, CD4 +, and CD4 +/CD8 + ratio or count of NK cells before and after treatment (P > 0.05). Before and after treatment, there was no significant difference in the levels of ALT, AST, BUN, and Scr in the observation group (P > 0.05). After treatment, peripheral blood CD3 +, CD4 +, and CD4 +/CD8 + ratio and count of NK cells were all increased in the observation group, significantly higher than those in the control group (P < 0.05). The incidence of chemotherapy side effects in the observation group was significantly lower than that in the control group (P < 0.05). Conclusion The short-term and long-term curative effects of deep hyperthermia combined with intraperitoneal chemotherapy are good on patients with intraperitoneal metastases, with less damage to liver-kidney function. It is beneficial to enhance immune function of patients, with mild side effects.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Oncology, Affiliated Hai'an Hospital of Nantong University, Nantong City, Jiangsu 226600, China
| | - Xiaomin Lu
- Department of Oncology, Affiliated Hai'an Hospital of Nantong University, Nantong City, Jiangsu 226600, China
| | - Haoming Ji
- Department of Oncology, Affiliated Hai'an Hospital of Nantong University, Nantong City, Jiangsu 226600, China
| | - Liangfeng Zheng
- Cancer Central Laboratory, Affiliated Hai'an Hospital of Nantong University, Nantong City, Jiangsu 226600, China
| | - Guodong Chen
- Department of Oncology, Affiliated Hai'an Hospital of Nantong University, Nantong City, Jiangsu 226600, China
| | - Ye Qian
- Department of Oncology, Affiliated Hai'an Hospital of Nantong University, Nantong City, Jiangsu 226600, China
| |
Collapse
|
14
|
Regenold M, Wang X, Kaneko K, Bannigan P, Allen C. Harnessing immunotherapy to enhance the systemic anti-tumor effects of thermosensitive liposomes. Drug Deliv Transl Res 2023; 13:1059-1073. [PMID: 36577832 DOI: 10.1007/s13346-022-01272-w] [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] [Accepted: 11/29/2022] [Indexed: 12/29/2022]
Abstract
Chemotherapy plays an important role in debulking tumors in advance of surgery and/or radiotherapy, tackling residual disease, and treating metastatic disease. In recent years many promising advanced drug delivery strategies have emerged that offer more targeted delivery approaches to chemotherapy treatment. For example, thermosensitive liposome-mediated drug delivery in combination with localized mild hyperthermia can increase local drug concentrations resulting in a reduction in systemic toxicity and an improvement in local disease control. However, the majority of solid tumor-associated deaths are due to metastatic spread. A therapeutic approach focused on a localized target area harbors the risk of overlooking and undertreating potential metastatic spread. Previous studies reported systemic, albeit limited, anti-tumor effects following treatment with thermosensitive liposomal chemotherapy and localized mild hyperthermia. This work explores the systemic treatment capabilities of a thermosensitive liposome formulation of the vinca alkaloid vinorelbine in combination with mild hyperthermia in an immunocompetent murine model of rhabdomyosarcoma. This treatment approach was found to be highly effective at heated, primary tumor sites. However, it demonstrated limited anti-tumor effects in secondary, distant tumors. As a result, the addition of immune checkpoint inhibition therapy was pursued to further enhance the systemic anti-tumor effect of this treatment approach. Once combined with immune checkpoint inhibition therapy, a significant improvement in systemic treatment capability was achieved. We believe this is one of the first studies to demonstrate that a triple combination of thermosensitive liposomes, localized mild hyperthermia, and immune checkpoint inhibition therapy can enhance the systemic treatment capabilities of thermosensitive liposomes.
Collapse
Affiliation(s)
- Maximilian Regenold
- Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, ON, M5S 3M2, Canada
| | - Xuehan Wang
- Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, ON, M5S 3M2, Canada
| | - Kan Kaneko
- Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, ON, M5S 3M2, Canada
| | - Pauric Bannigan
- Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, ON, M5S 3M2, Canada
| | - Christine Allen
- Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, ON, M5S 3M2, Canada.
| |
Collapse
|
15
|
Abstract
Immunotherapy has revolutionized the treatment of patients with cancer. However, promoting antitumour immunity in patients with tumours that are resistant to these therapies remains a challenge. Thermal therapies provide a promising immune-adjuvant strategy for use with immunotherapy, mostly owing to the capacity to reprogramme the tumour microenvironment through induction of immunogenic cell death, which also promotes the recruitment of endogenous immune cells. Thus, thermal immunotherapeutic strategies for various cancers are an area of considerable research interest. In this Review, we describe the role of the various thermal therapies and provide an update on attempts to combine these with immunotherapies in clinical trials. We also provide an overview of the preclinical development of various thermal immuno-nanomedicines, which are capable of combining thermal therapies with various immunotherapy strategies in a single therapeutic platform. Finally, we discuss the challenges associated with the clinical translation of thermal immuno-nanomedicines and emphasize the importance of multidisciplinary and inter-professional collaboration to facilitate the optimal translation of this technology from bench to bedside.
Collapse
|
16
|
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 2023; 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] [MESH Headings] [Grants] [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
|
17
|
Slotman DJ, Bartels MMTJ, Ferrer CJ, Bos C, Bartels LW, Boomsma MF, Phernambucq ECJ, Nijholt IM, Morganti AG, Siepe G, Buwenge M, Grüll H, Bratke G, Yeo SY, Blanco Sequeiros R, Minn H, Huhtala M, Napoli A, De Felice F, Catalano C, Bazzocchi A, Gasperini C, Campanacci L, Simões Corrêa Galendi J, Müller D, Braat MNGJA, Moonen C, Verkooijen HM. Focused Ultrasound and RadioTHERapy for non-invasive palliative pain treatment in patients with bone metastasis: a study protocol for the three armed randomized controlled FURTHER trial. Trials 2022; 23:1061. [PMID: 36582001 PMCID: PMC9798627 DOI: 10.1186/s13063-022-06942-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 11/17/2022] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Cancer-induced bone pain (CIBP), caused by bone metastases, is a common complication of cancer and strongly impairs quality of life (QoL). External beam radiotherapy (EBRT) is the current standard of care for treatment of CIBP. However, approximately 45% of patients have no adequate pain response after EBRT. Magnetic resonance image-guided high-intensity focused ultrasound (MR-HIFU) may improve pain palliation in this patient population. The main objective of this trial was to compare MR-HIFU, EBRT, and MR-HIFU + EBRT for the palliative treatment of bone metastases. METHODS/DESIGN The FURTHER trial is an international multicenter, three-armed randomized controlled trial. A total of 216 patients with painful bone metastases will be randomized in a 1:1:1 ratio to receive EBRT only, MR-HIFU only, or combined treatment with EBRT followed by MR-HIFU. During a follow-up period of 6 months, patients will be contacted at eight time points to retrieve information about their level of pain, QoL, and the occurrence of (serious) adverse events. The primary outcome of the trial is pain response at 14 days after start of treatment. Secondary outcomes include pain response at 14 days after trial enrolment, pain scores (daily until the 21st day and at 4, 6, 12 and 24 weeks), toxicity, adverse events, QoL, and survival. Cost-effectiveness and cost-utility analysis will be conducted. DISCUSSION The FURTHER trial aims to evaluate the effectiveness and cost-effectiveness of MR-HIFU-alone or in combination with EBRT-compared to EBRT to relieve CIBP. The trial will be performed in six hospitals in four European countries, all of which are partners in the FURTHER consortium. TRIAL REGISTRATION The FURTHER trial is registered under the Netherlands Trials Register number NL71303.041.19 and ClinicalTrials.gov registration number NCT04307914. Date of trial registration is 13-01-2020.
Collapse
Affiliation(s)
- Derk J. Slotman
- grid.7692.a0000000090126352Division of Imaging and Oncology, University Medical Centre Utrecht, Utrecht, The Netherlands ,grid.452600.50000 0001 0547 5927Department of Radiology, Isala Hospital, Zwolle, The Netherlands
| | - Marcia M. T. J. Bartels
- grid.7692.a0000000090126352Division of Imaging and Oncology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Cyril J. Ferrer
- grid.7692.a0000000090126352Division of Imaging and Oncology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Clemens Bos
- grid.7692.a0000000090126352Division of Imaging and Oncology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Lambertus W. Bartels
- grid.7692.a0000000090126352Division of Imaging and Oncology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Martijn F. Boomsma
- grid.7692.a0000000090126352Division of Imaging and Oncology, University Medical Centre Utrecht, Utrecht, The Netherlands ,grid.452600.50000 0001 0547 5927Department of Radiology, Isala Hospital, Zwolle, The Netherlands
| | - Erik C. J. Phernambucq
- grid.452600.50000 0001 0547 5927Department of Radiation Oncology, Isala Hospital, Zwolle, The Netherlands
| | - Ingrid M. Nijholt
- grid.452600.50000 0001 0547 5927Department of Radiology, Isala Hospital, Zwolle, The Netherlands
| | - Alessio G. Morganti
- grid.6292.f0000 0004 1757 1758DIMES, Alma Mater Studiorum - Bologna University, Bologna, Italy ,grid.6292.f0000 0004 1757 1758Radiation Oncology, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| | - Giambattista Siepe
- grid.6292.f0000 0004 1757 1758Radiation Oncology, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| | - Milly Buwenge
- grid.6292.f0000 0004 1757 1758DIMES, Alma Mater Studiorum - Bologna University, Bologna, Italy
| | - Holger Grüll
- grid.6190.e0000 0000 8580 3777Institute of Diagnostic and Interventional Radiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Grischa Bratke
- grid.6190.e0000 0000 8580 3777Institute of Diagnostic and Interventional Radiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Sin Yuin Yeo
- grid.6190.e0000 0000 8580 3777Institute of Diagnostic and Interventional Radiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Roberto Blanco Sequeiros
- grid.410552.70000 0004 0628 215XDepartment of Radiology, Turku University Hospital, Turku, Finland
| | - Heikki Minn
- grid.1374.10000 0001 2097 1371Department of Oncology, University of Turku and Turku University Hospital, Turku, Finland
| | - Mira Huhtala
- grid.1374.10000 0001 2097 1371Department of Oncology, University of Turku and Turku University Hospital, Turku, Finland
| | - Alessandro Napoli
- grid.7841.aDepartment of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, Rome, Italy
| | - Francesca De Felice
- grid.7841.aDepartment of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, Rome, Italy
| | - Carlo Catalano
- grid.7841.aDepartment of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, Rome, Italy
| | - Alberto Bazzocchi
- grid.419038.70000 0001 2154 6641Diagnostic and Interventional Radiology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Chiara Gasperini
- grid.419038.70000 0001 2154 6641Diagnostic and Interventional Radiology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Laura Campanacci
- grid.419038.70000 0001 2154 66413Rd Orthopaedic and Traumatologic Clinic Prevalently Oncologic, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Julia Simões Corrêa Galendi
- grid.6190.e0000 0000 8580 3777Institute of Health Economics and Clinical Epidemiology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Dirk Müller
- grid.6190.e0000 0000 8580 3777Institute of Health Economics and Clinical Epidemiology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Manon N. G. J. A. Braat
- grid.7692.a0000000090126352Division of Imaging and Oncology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Chrit Moonen
- grid.7692.a0000000090126352Division of Imaging and Oncology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Helena M. Verkooijen
- grid.7692.a0000000090126352Division of Imaging and Oncology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | | |
Collapse
|
18
|
Dias AMM, Courteau A, Bellaye PS, Kohli E, Oudot A, Doulain PE, Petitot C, Walker PM, Decréau R, Collin B. Superparamagnetic Iron Oxide Nanoparticles for Immunotherapy of Cancers through Macrophages and Magnetic Hyperthermia. Pharmaceutics 2022; 14:2388. [PMID: 36365207 PMCID: PMC9694944 DOI: 10.3390/pharmaceutics14112388] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 10/21/2022] [Accepted: 10/25/2022] [Indexed: 07/30/2023] Open
Abstract
Cancer immunotherapy has tremendous promise, but it has yet to be clinically applied in a wider variety of tumor situations. Many therapeutic combinations are envisaged to improve their effectiveness. In this way, strategies capable of inducing immunogenic cell death (e.g., doxorubicin, radiotherapy, hyperthermia) and the reprogramming of the immunosuppressive tumor microenvironment (TME) (e.g., M2-to-M1-like macrophages repolarization of tumor-associated macrophages (TAMs)) are particularly appealing to enhance the efficacy of approved immunotherapies (e.g., immune checkpoint inhibitors, ICIs). Due to their modular construction and versatility, iron oxide-based nanomedicines such as superparamagnetic iron oxide nanoparticles (SPIONs) can combine these different approaches in a single agent. SPIONs have already shown their safety and biocompatibility and possess both drug-delivery (e.g., chemotherapy, ICIs) and magnetic capabilities (e.g., magnetic hyperthermia (MHT), magnetic resonance imaging). In this review, we will discuss the multiple applications of SPIONs in cancer immunotherapy, focusing on their theranostic properties to target TAMs and to generate MHT. The first section of this review will briefly describe immune targets for NPs. The following sections will deal with the overall properties of SPIONs (including MHT). The last section is dedicated to the SPION-induced immune response through its effects on TAMs and MHT.
Collapse
Affiliation(s)
- Alexandre M. M. Dias
- Centre George-François Leclerc, Service de Médecine Nucléaire, Plateforme d’Imagerie et de Radiothérapie Précliniques, 1 rue du Professeur Marion, 21079 Dijon, France
| | - Alan Courteau
- Centre George-François Leclerc, Service de Médecine Nucléaire, Plateforme d’Imagerie et de Radiothérapie Précliniques, 1 rue du Professeur Marion, 21079 Dijon, France
- ImViA Laboratory, EA 7535, University of Burgundy, 21000 Dijon, France
| | - Pierre-Simon Bellaye
- Centre George-François Leclerc, Service de Médecine Nucléaire, Plateforme d’Imagerie et de Radiothérapie Précliniques, 1 rue du Professeur Marion, 21079 Dijon, France
- UMR INSERM/uB/AGROSUP 1231, Labex LipSTIC, Faculty of Health Sciences, Université de Bourgogne Franche-Comté, 21079 Dijon, France
| | - Evelyne Kohli
- UMR INSERM/uB/AGROSUP 1231, Labex LipSTIC, Faculty of Health Sciences, Université de Bourgogne Franche-Comté, 21079 Dijon, France
- University Hospital Centre François Mitterrand, 21000 Dijon, France
| | - Alexandra Oudot
- Centre George-François Leclerc, Service de Médecine Nucléaire, Plateforme d’Imagerie et de Radiothérapie Précliniques, 1 rue du Professeur Marion, 21079 Dijon, France
| | | | - Camille Petitot
- Centre George-François Leclerc, Service de Médecine Nucléaire, Plateforme d’Imagerie et de Radiothérapie Précliniques, 1 rue du Professeur Marion, 21079 Dijon, France
| | - Paul-Michael Walker
- Centre George-François Leclerc, Service de Médecine Nucléaire, Plateforme d’Imagerie et de Radiothérapie Précliniques, 1 rue du Professeur Marion, 21079 Dijon, France
- ImViA Laboratory, EA 7535, University of Burgundy, 21000 Dijon, France
- University Hospital Centre François Mitterrand, 21000 Dijon, France
| | - Richard Decréau
- Institut de Chimie Moléculaire de l’Université de Bourgogne, UMR CNRS/uB 6302, Université de Bourgogne Franche-Comté, 21079 Dijon, France
| | - Bertrand Collin
- Centre George-François Leclerc, Service de Médecine Nucléaire, Plateforme d’Imagerie et de Radiothérapie Précliniques, 1 rue du Professeur Marion, 21079 Dijon, France
- Institut de Chimie Moléculaire de l’Université de Bourgogne, UMR CNRS/uB 6302, Université de Bourgogne Franche-Comté, 21079 Dijon, France
| |
Collapse
|
19
|
Kok HP, van Rhoon GC, Herrera TD, Overgaard J, Crezee J. Biological modeling in thermoradiotherapy: present status and ongoing developments toward routine clinical use. Int J Hyperthermia 2022; 39:1126-1140. [PMID: 35998930 DOI: 10.1080/02656736.2022.2113826] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022] Open
Abstract
Biological modeling for anti-cancer treatments using mathematical models can be very supportive in gaining more insight into dynamic processes responsible for cellular response to treatment, and predicting, evaluating and optimizing therapeutic effects of treatment. This review presents an overview of the current status of biological modeling for hyperthermia in combination with radiotherapy (thermoradiotherapy). Various distinct models have been proposed in the literature, with varying complexity; initially aiming to model the effect of hyperthermia alone, and later on to predict the effect of the combined thermoradiotherapy treatment. Most commonly used models are based on an extension of the linear-quadratic (LQ)-model enabling an easy translation to radiotherapy where the LQ model is widely used. Basic predictions of cell survival have further progressed toward 3 D equivalent dose predictions, i.e., the radiation dose that would be needed without hyperthermia to achieve the same biological effect as the combined thermoradiotherapy treatment. This approach, with the use of temperature-dependent model parameters, allows theoretical evaluation of the effectiveness of different treatment strategies in individual patients, as well as in patient cohorts. This review discusses the significant progress that has been made in biological modeling for hyperthermia combined with radiotherapy. In the future, when adequate temperature-dependent LQ-parameters will be available for a large number of tumor sites and normal tissues, biological modeling can be expected to be of great clinical importance to further optimize combined treatments, optimize clinical protocols and guide further clinical studies.
Collapse
Affiliation(s)
- H P Kok
- Amsterdam UMC Location University of Amsterdam, Radiation Oncology, Amsterdam, The Netherlands.,Cancer Center Amsterdam, Treatment and Quality of Life, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - G C van Rhoon
- Department of Radiation Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands.,Department of Radiation Science and Technology, Delft University of Technology, Delft, The Netherlands
| | - T D Herrera
- Amsterdam UMC Location University of Amsterdam, Radiation Oncology, Amsterdam, The Netherlands.,Cancer Center Amsterdam, Treatment and Quality of Life, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - J Overgaard
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - J Crezee
- Amsterdam UMC Location University of Amsterdam, Radiation Oncology, Amsterdam, The Netherlands.,Cancer Center Amsterdam, Treatment and Quality of Life, Cancer Biology and Immunology, Amsterdam, The Netherlands
| |
Collapse
|
20
|
Tumor Temperature: Friend or Foe of Virus-Based Cancer Immunotherapy. Biomedicines 2022; 10:biomedicines10082024. [PMID: 36009571 PMCID: PMC9405776 DOI: 10.3390/biomedicines10082024] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/16/2022] [Accepted: 08/16/2022] [Indexed: 11/17/2022] Open
Abstract
The temperature of a solid tumor is often dissimilar to baseline body temperature and, compared to healthy tissues, may be elevated, reduced, or a mix of both. The temperature of a tumor is dependent on metabolic activity and vascularization and can change due to tumor progression, treatment, or cancer type. Despite the need to function optimally within temperature-variable tumors, oncolytic viruses (OVs) are primarily tested at 37 °C in vitro. Furthermore, animal species utilized to test oncolytic viruses, such as mice, dogs, cats, and non-human primates, poorly recapitulate the temperature profile of humans. In this review, we discuss the importance of temperature as a variable for OV immunotherapy of solid tumors. Accumulating evidence supports that the temperature sensitivity of OVs lies on a spectrum, with some OVs likely hindered but others enhanced by elevated temperatures. We suggest that in vitro temperature sensitivity screening be performed for all OVs destined for the clinic to identify potential hinderances or benefits with regard to elevated temperature. Furthermore, we provide recommendations for the clinical use of temperature and OVs.
Collapse
|
21
|
Forcing the Antitumor Effects of HSPs Using a Modulated Electric Field. Cells 2022; 11:cells11111838. [PMID: 35681533 PMCID: PMC9180583 DOI: 10.3390/cells11111838] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 05/25/2022] [Accepted: 05/31/2022] [Indexed: 12/10/2022] Open
Abstract
The role of Heat Shock Proteins (HSPs) is a “double-edged sword” with regards to tumors. The location and interactions of HSPs determine their pro- or antitumor activity. The present review includes an overview of the relevant functions of HSPs, which could improve their antitumor activity. Promoting the antitumor processes could assist in the local and systemic management of cancer. We explore the possibility of achieving this by manipulating the electromagnetic interactions within the tumor microenvironment. An appropriate electric field may select and affect the cancer cells using the electric heterogeneity of the tumor tissue. This review describes the method proposed to effect such changes: amplitude-modulated radiofrequency (amRF) applied with a 13.56 MHz carrier frequency. We summarize the preclinical investigations of the amRF on the HSPs in malignant cells. The preclinical studies show the promotion of the expression of HSP70 on the plasma membrane, participating in the immunogenic cell death (ICD) pathway. The sequence of guided molecular changes triggers innate and adaptive immune reactions. The amRF promotes the secretion of HSP70 also in the extracellular matrix. The extracellular HSP70 accompanied by free HMGB1 and membrane-expressed calreticulin (CRT) form damage-associated molecular patterns encouraging the dendritic cells’ maturing for antigen presentation. The process promotes killer T-cells. Clinical results demonstrate the potential of this immune process to trigger a systemic effect. We conclude that the properly applied amRF promotes antitumor HSP activity, and in situ, it could support the tumor-specific immune effects produced locally but acting systemically for disseminated cells and metastatic lesions.
Collapse
|
22
|
Combined Therapy with Dacarbazine and Hyperthermia Induces Cytotoxicity in A375 and MNT-1 Melanoma Cells. Int J Mol Sci 2022; 23:ijms23073586. [PMID: 35408947 PMCID: PMC8998307 DOI: 10.3390/ijms23073586] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/17/2022] [Accepted: 03/21/2022] [Indexed: 01/16/2023] Open
Abstract
Melanoma is a drug-resistant cancer, representing a serious challenge in cancer treatment. Dacarbazine (DTIC) is the standard drug in metastatic melanoma treatment, despite the poor results. Hyperthermia has been proven to potentiate chemotherapy. Hence, this work analyzed the combined action of hyperthermia and DTIC on A375 and MNT-1 cell lines. First, temperatures between 40 °C and 45 °C were tested. The effect of DTIC on cell viability was also investigated after exposures of 24, 48, and 72 h. Then, cells were exposed to 43 °C and to the respective DTIC IC10 or IC20 of each time exposure. Overall, hyperthermia reduced cell viability, however, 45 °C caused an excessive cell death (>90%). Combinational treatment revealed that hyperthermia potentiates DTIC’s effect, but it is dependent on the concentration and temperature used. Also, it has different mechanisms from the treatments alone, delaying A375 cells at the G2/M phase and MNT-1 cells at the S and G2/M phases. Intracellular reactive oxygen species (ROS) levels increased after treatment with hyperthermia, but the combined treatment showed no additional differences. Also, hyperthermia highly increased the number of A375 early apoptotic cells. These results suggest that combining hyperthermia and DTIC should be more explored to improve melanoma treatment.
Collapse
|
23
|
Garrett J, Metzger E, Dewhirst MW, Pollok KE, Turchi JJ, Le Poole IC, Couch K, Lew L, Sinn A, Zaleski JM, Dynlacht JR. Characterization and initial demonstration of in vivo efficacy of a novel heat-activated metalloenediyne anti-cancer agent. Int J Hyperthermia 2022; 39:405-413. [PMID: 35236209 PMCID: PMC9612397 DOI: 10.1080/02656736.2021.2024280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Background: Enediynes are anti-cancer agents that are highly cytotoxic due to their propensity for low thermal activation of radical generation. The diradical intermediate produced from Bergman cyclization of the enediyne moiety may induce DNA damage and cell lethality. The cytotoxicity of enediynes and difficulties in controlling their thermal cyclization has limited their clinical use. We recently showed that enediyne toxicity at 37 °C can be mitigated by metallation, but cytotoxic effects of ‘metalloenediynes’ on cultured tumor cells are potentiated by hyperthermia. Reduction of cytotoxicity at normothermia suggests metalloenediynes will have a large therapeutic margin, with cell death occurring primarily in the heated tumor. Based on our previous in vitro findings, FeSO4-PyED, an Fe co-factor complex of (Z)-N,N׳-bis[1-pyridin-2-yl-meth-(E)-ylidene]oct-4-ene-2,6-diyne-1,8-diamine, was prioritized for further in vitro and in vivo testing in normal human melanocytes and melanoma cells. Methods: Clonogenic survival, apopotosis and DNA binding assays were used to determine mechanisms of enhancement of FeSO4-PyED cytotoxicity by hyperthermia. A murine human melanoma xenograft model was used to assess in vivo efficacy of FeSO4-PyED at 37 or 42.5 °C. Results: FeSO4-PyED is a DNA-binding compound. Enhancement of FeSO4-PyED cytotoxicity by hyperthermia in melanoma cells was due to Bergman cyclization, diradical formation, and increased apoptosis. Thermal enhancement, however, was not observed in melanocytes. FeSO4-PyED inhibited tumor growth when melanomas were heated during drug treatment, without inducing normal tissue damage. Conclusion: By leveraging the unique thermal activation properties of metalloenediynes, we propose that localized moderate hyperthermia can be used to confine the cytotoxicity of these compounds to tumors, while sparing normal tissue.
Collapse
Affiliation(s)
- Joy Garrett
- Department of Radiation Oncology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Erin Metzger
- Department of Chemistry, Indiana University, Bloomington, IN, USA
| | - Mark W Dewhirst
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC, USA
| | - Karen E Pollok
- In Vivo Therapeutics Core, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - John J Turchi
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Kira Couch
- Department of Radiation Oncology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Logan Lew
- Department of Radiation Oncology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Anthony Sinn
- In Vivo Therapeutics Core, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Joseph R Dynlacht
- Department of Radiation Oncology, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
24
|
Zhou L, Zhang Z, Nice E, Huang C, Zhang W, Tang Y. Circadian rhythms and cancers: the intrinsic links and therapeutic potentials. J Hematol Oncol 2022; 15:21. [PMID: 35246220 PMCID: PMC8896306 DOI: 10.1186/s13045-022-01238-y] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 02/16/2022] [Indexed: 02/07/2023] Open
Abstract
The circadian rhythm is an evolutionarily conserved time-keeping system that comprises a wide variety of processes including sleep-wake cycles, eating-fasting cycles, and activity-rest cycles, coordinating the behavior and physiology of all organs for whole-body homeostasis. Acute disruption of circadian rhythm may lead to transient discomfort, whereas long-term irregular circadian rhythm will result in the dysfunction of the organism, therefore increasing the risks of numerous diseases especially cancers. Indeed, both epidemiological and experimental evidence has demonstrated the intrinsic link between dysregulated circadian rhythm and cancer. Accordingly, a rapidly increasing understanding of the molecular mechanisms of circadian rhythms is opening new options for cancer therapy, possibly by modulating the circadian clock. In this review, we first describe the general regulators of circadian rhythms and their functions on cancer. In addition, we provide insights into the mechanisms underlying how several types of disruption of the circadian rhythm (including sleep-wake, eating-fasting, and activity-rest) can drive cancer progression, which may expand our understanding of cancer development from the clock perspective. Moreover, we also summarize the potential applications of modulating circadian rhythms for cancer treatment, which may provide an optional therapeutic strategy for cancer patients.
Collapse
Affiliation(s)
- Li Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Zhe Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Edouard Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, 3800, Australia
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Wei Zhang
- Mental Health Center and Psychiatric Laboratory, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Yong Tang
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Acupuncture and Chronobiology Laboratory of Sichuan Province, Chengdu, 610075, China.
| |
Collapse
|
25
|
Neoantigen Cancer Vaccines: Generation, Optimization, and Therapeutic Targeting Strategies. Vaccines (Basel) 2022; 10:vaccines10020196. [PMID: 35214655 PMCID: PMC8877108 DOI: 10.3390/vaccines10020196] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/21/2022] [Accepted: 01/23/2022] [Indexed: 12/30/2022] Open
Abstract
Alternatives to conventional cancer treatments are highly sought after for high-risk malignancies that have a poor response to established treatment modalities. With research advancing rapidly in the past decade, neoantigen-based immunotherapeutic approaches represent an effective and highly tolerable therapeutic option. Neoantigens are tumor-specific antigens that are not expressed in normal cells and possess significant immunogenic potential. Several recent studies have described the conceptual framework and methodologies to generate neoantigen-based vaccines as well as the formulation of appropriate clinical trials to advance this approach for patient care. This review aims to describe some of the key studies in the recent literature in this rapidly evolving field and summarize the current advances in neoantigen identification and selection, vaccine generation and delivery, and the optimization of neoantigen-based therapeutic strategies, including the early data from pivotal clinical studies.
Collapse
|
26
|
Kumar PPP, Lim DK. Gold-Polymer Nanocomposites for Future Therapeutic and Tissue Engineering Applications. Pharmaceutics 2021; 14:70. [PMID: 35056967 PMCID: PMC8781750 DOI: 10.3390/pharmaceutics14010070] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 12/11/2021] [Accepted: 12/14/2021] [Indexed: 12/16/2022] Open
Abstract
Gold nanoparticles (AuNPs) have been extensively investigated for their use in various biomedical applications. Owing to their biocompatibility, simple surface modifications, and electrical and unique optical properties, AuNPs are considered promising nanomaterials for use in in vitro disease diagnosis, in vivo imaging, drug delivery, and tissue engineering applications. The functionality of AuNPs may be further expanded by producing hybrid nanocomposites with polymers that provide additional functions, responsiveness, and improved biocompatibility. Polymers may deliver large quantities of drugs or genes in therapeutic applications. A polymer alters the surface charges of AuNPs to improve or modulate cellular uptake efficiency and their biodistribution in the body. Furthermore, designing the functionality of nanocomposites to respond to an endo- or exogenous stimulus, such as pH, enzymes, or light, may facilitate the development of novel therapeutic applications. In this review, we focus on the recent progress in the use of AuNPs and Au-polymer nanocomposites in therapeutic applications such as drug or gene delivery, photothermal therapy, and tissue engineering.
Collapse
Affiliation(s)
| | - Dong-Kwon Lim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Korea;
- Department of Integrative Energy Engineering, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Korea
| |
Collapse
|
27
|
Das P, Ganguly S, Margel S, Gedanken A. Tailor made magnetic nanolights: fabrication to cancer theranostics applications. NANOSCALE ADVANCES 2021; 3:6762-6796. [PMID: 36132370 PMCID: PMC9419279 DOI: 10.1039/d1na00447f] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 10/12/2021] [Indexed: 05/14/2023]
Abstract
Nanoparticles having magnetic and fluorescent properties could be considered as a gift to materials scientists due to their unique magneto-optical qualities. Multiple component particles can overcome challenges related with a single component and unveil bifunctional/multifunctional features that can enlarge their applications in diagnostic imaging agents and therapeutic delivery vehicles. Bifunctional nanoparticles that have both luminescent and magnetic features are termed as magnetic nanolights. Herein, we present recent progress of magneto-fluorescent nanoparticles (quantum dots based magnetic nanoparticles, Janus particles, and heterocrystalline fluorescent magnetic materials), comprehensively describing fabrication strategies, types, and biomedical applications. In this review, our aim is not only to encompass the preparation strategies of these special types of magneto-fluorescent nanomaterials but also their extensive applications in bioimaging techniques, cancer therapy (targeted and hyperthermic), and sustained release of active agents (drugs, proteins, antibodies, hormones, enzymes, growth factors).
Collapse
Affiliation(s)
- Poushali Das
- Bar-Ilan Institute for Nanotechnology and Advanced Materials (BINA), Bar-Ilan University Ramat-Gan 5290002 Israel
- Departments of Chemistry, Bar-Ilan University Ramat-Gan 5290002 Israel
| | - Sayan Ganguly
- Bar-Ilan Institute for Nanotechnology and Advanced Materials (BINA), Bar-Ilan University Ramat-Gan 5290002 Israel
- Departments of Chemistry, Bar-Ilan University Ramat-Gan 5290002 Israel
| | - Shlomo Margel
- Bar-Ilan Institute for Nanotechnology and Advanced Materials (BINA), Bar-Ilan University Ramat-Gan 5290002 Israel
- Departments of Chemistry, Bar-Ilan University Ramat-Gan 5290002 Israel
| | - Aharon Gedanken
- Bar-Ilan Institute for Nanotechnology and Advanced Materials (BINA), Bar-Ilan University Ramat-Gan 5290002 Israel
- Departments of Chemistry, Bar-Ilan University Ramat-Gan 5290002 Israel
| |
Collapse
|
28
|
Transformable vesicles for cancer immunotherapy. Adv Drug Deliv Rev 2021; 179:113905. [PMID: 34331988 DOI: 10.1016/j.addr.2021.113905] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/22/2021] [Accepted: 07/24/2021] [Indexed: 02/06/2023]
Abstract
Immunotherapy that utilizes the human immune system to fight cancer represents a revolutionary method for cancer treatment. Immunotherapeutic agents that trigger the immune response should be carefully delivered to the desired site to maximize immunotherapy effectiveness and minimize side effects. Vesicles offer the possibility of encapsulating both hydrophilic and hydrophobic drugs and thus serve as a promising delivery tool. As multiple irreconcilable requirements exist at different transport stages, developing vesicles transformable in response to given stimuli is of great significance. In this review, we first introduced various vesicle types used for immunotherapy. Furthermore, the typical stimuli that trigger vesicle transformation and the usually generated transformation styles were described. Focusing on three aspects of antigen-presenting cell (APC)/T cell activation, tumor microenvironment (TME) amelioration, and immunogenic cell death (ICD)-induced immunotherapy, we reviewed recently reported transformable vesicles for tumor treatment. Finally, we put forward possible directions for future research and clinical translation.
Collapse
|
29
|
Cheng B, Bing C, Chu TH, Alzahrani S, Pichardo S, Pike GB. Simultaneous Localized Brain Mild Hyperthermia and Blood-Brain Barrier Opening via Feedback-Controlled Transcranial MR-guided Focused Ultrasound and Microbubbles. IEEE Trans Biomed Eng 2021; 69:1880-1888. [PMID: 34813464 DOI: 10.1109/tbme.2021.3130164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
OBJECTIVE Non-invasive methods to enhance drug delivery and efficacy in the brain have been pursued for decades. Focused ultrasound hyperthermia (HT) combined with thermosensitive therapeutics have been demonstrated promising in enhancing local drug delivery to solid tumors. We hypothesized that the presence of microbubbles (MBs) combined with transcranial MR-guided focused ultrasound (MRgFUS) could be used to reduce the ultrasound power required for HT while simultaneously increasing drug delivery by locally opening the blood-brain barrier (BBB). METHODS Transcranial HT (42 C, 10 min) was performed in wild-type mice using a small animal MRgFUS system incorporated into a 9.4T Bruker MR scanner, with infusions of saline or Definity MBs with doses of 20 or 100 l/kg/min (denoted as MB-20 and MB-100). MR thermometry data was continuously acquired as feedback for the ultrasound controller during the procedure. RESULTS Spatiotemporally precise transcranial HT was achieved in both saline and MB groups. A significant ultrasound power reduction (-45.7%, p = 0.006) was observed in the MB-20 group compared to saline. Localized BBB opening was achieved in MB groups confirmed by CE-T1w MR images. There were no structural abnormalities, edema, hemorrhage, or acute microglial activation in all groups, confirmed by T2w MR imaging and histology. CONCLUSION Our investigations showed that it is feasible and safe to achieve spatiotemporally precise brain HT at significantly reduced power and simultaneous localized BBB opening via transcranial MRgFUS and MBs. SIGNIFICANCE This study provides a new synergistic brain drug delivery method with clinical translation potential.
Collapse
|
30
|
ROS Pleiotropy in Melanoma and Local Therapy with Physical Modalities. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6816214. [PMID: 34777692 PMCID: PMC8580636 DOI: 10.1155/2021/6816214] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/06/2021] [Accepted: 10/11/2021] [Indexed: 12/15/2022]
Abstract
Metabolic energy production naturally generates unwanted products such as reactive oxygen species (ROS), causing oxidative damage. Oxidative damage has been linked to several pathologies, including diabetes, premature aging, neurodegenerative diseases, and cancer. ROS were therefore originally anticipated as an imperative evil, a product of an imperfect system. More recently, however, the role of ROS in signaling and tumor treatment is increasingly acknowledged. This review addresses the main types, sources, and pathways of ROS in melanoma by linking their pleiotropic roles in antioxidant and oxidant regulation, hypoxia, metabolism, and cell death. In addition, the implications of ROS in various physical therapy modalities targeting melanoma, such as radiotherapy, electrochemotherapy, hyperthermia, photodynamic therapy, and medical gas plasma, are also discussed. By including ROS in the main picture of melanoma skin cancer and as an integral part of cancer therapies, a greater understanding of melanoma cell biology is presented, which ultimately may elucidate additional clues on targeting therapy resistance of this most deadly form of skin cancer.
Collapse
|
31
|
Brummelhuis ISG, Simons M, Lindner LH, Kort S, de Jong S, Hossann M, Witjes JA, Oosterwijk E. DPPG 2-based thermosensitive liposomes as drug delivery system for effective muscle-invasive bladder cancer treatment in vivo. Int J Hyperthermia 2021; 38:1415-1424. [PMID: 34581259 DOI: 10.1080/02656736.2021.1983038] [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] [Indexed: 12/12/2022] Open
Abstract
PURPOSE Recommended treatments for muscle-invasive bladder cancer (MIBC) come with considerable morbidity. Hyperthermia (HT) triggered drug release from phosphatidylglycerol-based thermosensitive liposomes (DPPG2-TSL) might prevent surgical bladder removal and toxicity from systemic chemotherapy. We aimed to assess the efficacy of DPPG2-TSL with HT in a syngeneic orthotopic rat urothelial carcinoma model. METHODS A total of 191 female Fischer F344 rats were used. Bladder tumors were initiated by inoculation of AY-27 cells and tumor-bearing rats were selected with cystoscopy and semi-randomized over treatment groups. On days 5 and 8, animals were treated with DOX in different treatment modalities: intravenous (iv) DPPG2-TSL-DOX with HT, iv free DOX without HT, intravesical DOX without HT, intravesical DOX with HT or no treatment (control group), respectively. Animals were euthanized on day 14 and complete tumor response was assessed by histopathological evaluation. RESULTS Iv DPPG2-TSL-DOX + HT resulted in a favorable rate of animals with complete tumor response (70%), compared to iv free DOX (18%, p = .02), no treatment (0%, p = .001), and intravesical DOX with (43%, p = .35) or without HT (50%, p = .41). All rats receiving intravesical DOX with HT and 24% of rats treated with DPPG2-TSL-DOX containing the same DOX dose with HT had to be euthanized before day 14 because of substantial bodyweight loss, which was associated with dilated ureters urine retention in a few rats. CONCLUSION Treatment with DPPG2-TSL-DOX combined with intravesical HT outperformed systemic and intravesical DOX in vivo. There might be a role for DPPG2-TSL encapsulating chemotherapeutics in the treatment of MIBC in the future.
Collapse
Affiliation(s)
- Iris S G Brummelhuis
- Department of Urology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Michiel Simons
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Lars H Lindner
- Department of Medicine III, University Hospital LMU Munich, Munich, Germany
| | - Simone Kort
- Department of Medicine III, University Hospital LMU Munich, Munich, Germany
| | - Sytse de Jong
- Department of Cardiothoracic Surgery, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - J Alfred Witjes
- Department of Urology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Egbert Oosterwijk
- Department of Urology, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
32
|
Sawai H, Ueno S, Yamaguchi Y, Suzuki Y, Murata A, Suganuma E, Yamamoto K, Kuzuya H, Koide S, Kurimoto M, Yanagi T, Koide H, Kamiya A. Hyperthermia with Chemotherapy for Unresectable Gastric Cancer in a Patient with a Vagus Nerve Stimulator Implant: A Case Report. AMERICAN JOURNAL OF CASE REPORTS 2021; 22:e931564. [PMID: 34400601 PMCID: PMC8380855 DOI: 10.12659/ajcr.931564] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 07/06/2021] [Accepted: 06/30/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Radiofrequency (RF) hyperthermia is commonly used as an adjunct to established treatment modalities such as chemotherapy and radiotherapy for the management of cancer patients. This case report aims to introduce the use of hyperthermia, in combination with chemotherapy, for the treatment of unresectable gastric cancer in a patient implanted with a vagus nerve stimulator (VNS). CASE REPORT A 55-year-old man with dermatomyositis, laryngeal squamous cell carcinoma in situ and double synchronous gastric cancer was found to have unresectable gastric disease during surgery despite neoadjuvant chemotherapy. Postoperatively, he received chemotherapy with RF hyperthermia. The patient had a VNS implant to treat epileptic seizures. VNS failure due to RF hyperthermia was an area of significant concern, and the procedures were completed with a full preparation to manage epileptic seizures in the event of its anticipated occurrence. Twenty-one thermotherapies were performed over 21 weeks. After 3 courses of S-1 chemotherapy (12 weeks) with RF hyperthermia without any adverse events, the regimen was changed to S-1+ CDDP combination chemotherapy (SP) and RF hyperthermia. The patient continued to receive treatment with a decrease in the size of the primary gastric tumors as well as lymph node metastases, without major adverse events, until he died due to disseminated disease. CONCLUSIONS We report the first case of unresectable gastric cancer with VNS implants in which chemo-hyperthermal therapy was safe and successful. This case report highlights the importance of providing a multidisciplinary treatment with appropriate measures for patients with intractable cancer who have received special treatments for underlying comorbidities.
Collapse
Affiliation(s)
- Hirozumi Sawai
- Department of Surgery, Narita Memorial Hospital, Toyohashi, Aichi, Japan
| | - Shuhei Ueno
- Department of Surgery, Narita Memorial Hospital, Toyohashi, Aichi, Japan
| | - Yoshimi Yamaguchi
- Radiological Technology, Department of Medical Technique, Narita Memorial Hospital, Toyohashi, Aichi, Japan
| | - Yuka Suzuki
- Radiological Technology, Department of Medical Technique, Narita Memorial Hospital, Toyohashi, Aichi, Japan
| | - Akemi Murata
- Radiological Technology, Department of Medical Technique, Narita Memorial Hospital, Toyohashi, Aichi, Japan
| | - Enami Suganuma
- Radiological Technology, Department of Medical Technique, Narita Memorial Hospital, Toyohashi, Aichi, Japan
| | - Kazuya Yamamoto
- Radiological Technology, Department of Medical Technique, Narita Memorial Hospital, Toyohashi, Aichi, Japan
| | - Hiromasa Kuzuya
- Department of Surgery, Narita Memorial Hospital, Toyohashi, Aichi, Japan
| | - Shuji Koide
- Department of Surgery, Narita Memorial Hospital, Toyohashi, Aichi, Japan
| | - Masaaki Kurimoto
- Department of Surgery, Narita Memorial Hospital, Toyohashi, Aichi, Japan
| | - Takeshi Yanagi
- Department of Proton, Narita Memorial Proton Center, Toyohashi, Aichi, Japan
| | - Hajime Koide
- Department of Surgery, Narita Memorial Hospital, Toyohashi, Aichi, Japan
| | - Atsushi Kamiya
- Department of Surgery, Narita Memorial Hospital, Toyohashi, Aichi, Japan
| |
Collapse
|
33
|
Zubair M, Adams MS, Diederich CJ. Deployable ultrasound applicators for endoluminal delivery of volumetric hyperthermia. Int J Hyperthermia 2021; 38:1188-1204. [PMID: 34376103 DOI: 10.1080/02656736.2021.1936216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
PURPOSE To investigate the design of an endoluminal deployable ultrasound applicator for delivering volumetric hyperthermia to deep tissue sites as a possible adjunct to radiation and chemotherapy. METHOD This study considers an ultrasound applicator consisting of two tubular transducers situated at the end of a catheter assembly, encased within a distensible conical shaped balloon-based reflector that redirects acoustic energy distally into the tissue. The applicator assembly can be inserted endoluminally or laparoscopically in a compact form and expanded after delivery to the target site. Comprehensive acoustic and biothermal simulations and parametric studies were employed in generalized 3D and patient-specific pancreatic head and body tumor models to characterize the acoustic performance and evaluate heating capabilities of the applicator by investigating the device at a range of operating frequencies, tissue acoustic and thermal properties, transducer configurations, power modulation, applicator positioning, and by analyzing the resultant 40, 41, and 43 °C isothermal volumes and penetration depth of the heating volume. Intensity distributions and volumetric temperature contours were calculated to define moderate hyperthermia boundaries. RESULTS Parametric studies demonstrated the frequency selection to control volume and depth of therapeutic heating from 62 to 22 cm3 and 4 to 2.6 cm as frequency ranges from 1 MHz to 4.7 MHz, respectively. Width of the heating profile tracks closely with the aperture. Water cooling within the reflector balloon was effective in controlling temperature to 37 °C maximum within the luminal wall. Patient-specific studies indicated that applicators with extended OD in the range of 3.6-6.2 cm with 0.5-1 cm long and 1 cm OD transducers can heat volumes of 1.1-7 cm3, 3-26 cm3, and 3.3-37.4 cm3 of pancreatic body and head tumors above 43, 41, and 40 °C, respectively. CONCLUSION In silico studies demonstrated the feasibility of combining endoluminal ultrasound with an integrated expandable balloon reflector for delivering volumetric hyperthermia in regions adjacent to body lumens and cavities.
Collapse
Affiliation(s)
- Muhammad Zubair
- Thermal Therapy Research Group, Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, USA
| | - Matthew S Adams
- Thermal Therapy Research Group, Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, USA
| | - Chris J Diederich
- Thermal Therapy Research Group, Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
34
|
Day NB, Wixson WC, Shields CW. Magnetic systems for cancer immunotherapy. Acta Pharm Sin B 2021; 11:2172-2196. [PMID: 34522583 PMCID: PMC8424374 DOI: 10.1016/j.apsb.2021.03.023] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/05/2021] [Accepted: 02/25/2021] [Indexed: 02/06/2023] Open
Abstract
Immunotherapy is a rapidly developing area of cancer treatment due to its higher specificity and potential for greater efficacy than traditional therapies. Immune cell modulation through the administration of drugs, proteins, and cells can enhance antitumoral responses through pathways that may be otherwise inhibited in the presence of immunosuppressive tumors. Magnetic systems offer several advantages for improving the performance of immunotherapies, including increased spatiotemporal control over transport, release, and dosing of immunomodulatory drugs within the body, resulting in reduced off-target effects and improved efficacy. Compared to alternative methods for stimulating drug release such as light and pH, magnetic systems enable several distinct methods for programming immune responses. First, we discuss how magnetic hyperthermia can stimulate immune cells and trigger thermoresponsive drug release. Second, we summarize how magnetically targeted delivery of drug carriers can increase the accumulation of drugs in target sites. Third, we review how biomaterials can undergo magnetically driven structural changes to enable remote release of encapsulated drugs. Fourth, we describe the use of magnetic particles for targeted interactions with cellular receptors for promoting antitumor activity. Finally, we discuss translational considerations of these systems, such as toxicity, clinical compatibility, and future opportunities for improving cancer treatment.
Collapse
Key Words
- BW, body weight
- Biomaterials
- CpG, cytosine-phosphate-guanine
- DAMP, damage associated molecular pattern
- Drug delivery
- EPR, enhanced permeability and retention
- FFR, field free region
- HS-TEX, heat-stressed tumor cell exosomes
- HSP, heat shock protein
- ICD, immunogenic cell death
- IVIS, in vivo imaging system
- Immunotherapy
- MICA, MHC class I-related chain A
- MPI, magnetic particle imaging
- Magnetic hyperthermia
- Magnetic nanoparticles
- Microrobotics
- ODNs, oligodeoxynucleotides
- PARP, poly(adenosine diphosphate-ribose) polymerase
- PDMS, polydimethylsiloxane
- PEG, polyethylene glycol
- PLGA, poly(lactic-co-glycolic acid)
- PNIPAM, poly(N-isopropylacrylamide)
- PVA, poly(vinyl alcohol)
- SDF, stromal cell derived-factor
- SID, small implantable device
- SLP, specific loss power
Collapse
Affiliation(s)
- Nicole B Day
- Department of Chemical & Biological Engineering, University of Colorado, Boulder, CO 80303, USA
| | - William C Wixson
- Department of Chemical & Biological Engineering, University of Colorado, Boulder, CO 80303, USA
| | - C Wyatt Shields
- Department of Chemical & Biological Engineering, University of Colorado, Boulder, CO 80303, USA
| |
Collapse
|
35
|
Liu X, Li KW, Yang R, Geng LS. Review of Deep Learning Based Automatic Segmentation for Lung Cancer Radiotherapy. Front Oncol 2021; 11:717039. [PMID: 34336704 PMCID: PMC8323481 DOI: 10.3389/fonc.2021.717039] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 06/21/2021] [Indexed: 12/14/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related mortality for males and females. Radiation therapy (RT) is one of the primary treatment modalities for lung cancer. While delivering the prescribed dose to tumor targets, it is essential to spare the tissues near the targets-the so-called organs-at-risk (OARs). An optimal RT planning benefits from the accurate segmentation of the gross tumor volume and surrounding OARs. Manual segmentation is a time-consuming and tedious task for radiation oncologists. Therefore, it is crucial to develop automatic image segmentation to relieve radiation oncologists of the tedious contouring work. Currently, the atlas-based automatic segmentation technique is commonly used in clinical routines. However, this technique depends heavily on the similarity between the atlas and the image segmented. With significant advances made in computer vision, deep learning as a part of artificial intelligence attracts increasing attention in medical image automatic segmentation. In this article, we reviewed deep learning based automatic segmentation techniques related to lung cancer and compared them with the atlas-based automatic segmentation technique. At present, the auto-segmentation of OARs with relatively large volume such as lung and heart etc. outperforms the organs with small volume such as esophagus. The average Dice similarity coefficient (DSC) of lung, heart and liver are over 0.9, and the best DSC of spinal cord reaches 0.9. However, the DSC of esophagus ranges between 0.71 and 0.87 with a ragged performance. In terms of the gross tumor volume, the average DSC is below 0.8. Although deep learning based automatic segmentation techniques indicate significant superiority in many aspects compared to manual segmentation, various issues still need to be solved. We discussed the potential issues in deep learning based automatic segmentation including low contrast, dataset size, consensus guidelines, and network design. Clinical limitations and future research directions of deep learning based automatic segmentation were discussed as well.
Collapse
Affiliation(s)
- Xi Liu
- School of Physics, Beihang University, Beijing, China
| | - Kai-Wen Li
- School of Physics, Beihang University, Beijing, China
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Medicine and Engineering, Key Laboratory of Big Data-Based Precision Medicine, Ministry of Industry and Information Technology, Beihang University, Beijing, China
| | - Ruijie Yang
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, China
| | - Li-Sheng Geng
- School of Physics, Beihang University, Beijing, China
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Medicine and Engineering, Key Laboratory of Big Data-Based Precision Medicine, Ministry of Industry and Information Technology, Beihang University, Beijing, China
- Beijing Key Laboratory of Advanced Nuclear Materials and Physics, Beihang University, Beijing, China
- School of Physics and Microelectronics, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
36
|
Tranberg KG. Local Destruction of Tumors and Systemic Immune Effects. Front Oncol 2021; 11:708810. [PMID: 34307177 PMCID: PMC8298109 DOI: 10.3389/fonc.2021.708810] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 06/23/2021] [Indexed: 12/22/2022] Open
Abstract
Current immune-based therapies signify a major advancement in cancer therapy; yet, they are not effective in the majority of patients. Physically based local destruction techniques have been shown to induce immunologic effects and are increasingly used in order to improve the outcome of immunotherapies. The various local destruction methods have different modes of action and there is considerable variation between the different techniques with respect to the ability and frequency to create a systemic anti-tumor immunologic effect. Since the abscopal effect is considered to be the best indicator of a relevant immunologic effect, the present review focused on the tissue changes associated with this effect in order to find determinants for a strong immunologic response, both when local destruction is used alone and combined with immunotherapy. In addition to the T cell-inflammation that was induced by all methods, the analysis indicated that it was important for an optimal outcome that the released antigens were not destroyed, tumor cell death was necrotic and tumor tissue perfusion was at least partially preserved allowing for antigen presentation, immune cell trafficking and reduction of hypoxia. Local treatment with controlled low level hyperthermia met these requisites and was especially prone to result in abscopal immune activity on its own.
Collapse
|
37
|
Hannon G, Tansi FL, Hilger I, Prina‐Mello A. The Effects of Localized Heat on the Hallmarks of Cancer. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202000267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Gary Hannon
- Nanomedicine and Molecular Imaging Group Trinity Translational Medicine Institute Dublin 8 Ireland
- Laboratory of Biological Characterization of Advanced Materials (LBCAM), Trinity Translational Medicine Institute Trinity College Dublin Dublin 8 Ireland
| | - Felista L. Tansi
- Department of Experimental Radiology, Institute of Diagnostic and Interventional Radiology Jena University Hospital—Friedrich Schiller University Jena Am Klinikum 1 07740 Jena Germany
| | - Ingrid Hilger
- Department of Experimental Radiology, Institute of Diagnostic and Interventional Radiology Jena University Hospital—Friedrich Schiller University Jena Am Klinikum 1 07740 Jena Germany
| | - Adriele Prina‐Mello
- Nanomedicine and Molecular Imaging Group Trinity Translational Medicine Institute Dublin 8 Ireland
- Laboratory of Biological Characterization of Advanced Materials (LBCAM), Trinity Translational Medicine Institute Trinity College Dublin Dublin 8 Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre, CRANN Institute Trinity College Dublin Dublin 2 Ireland
| |
Collapse
|
38
|
Abstract
PURPOSE OF REVIEW To provide an overview of recently published articles covering melanoma management of the head and neck region. RECENT FINDINGS Melanoma management represents a rapidly evolving field. The advent of immunotherapy has led to significant changes in diagnosis, treatment, and surveillance for these patients. Invasive interventions including completion lymph node dissection have been largely replaced with increased surveillance driven by robust data showing no significant difference in overall survival. Studies have explored various treatment regimens that offer improved outcomes with the least adverse events, with a recent trend towards neoadjuvant therapy. Research has also shifted towards better understanding genetics and biomarkers that influence response to these medications. The best means to both identify and monitor these changes is being explored. As our understanding of this complex disease process continues to grow, prognosis in patients suffering with melanoma should continue to improve. SUMMARY The expansion of immunotherapy use in melanoma management has led to significantly improved prognosis in diagnosed patients. Present research is largely focused on better understanding the ideal patient populations, dosing, and surveillance for these therapies. Data from these studies will be crucial in better staging and treating patients with melanoma.
Collapse
|
39
|
Guo J, Zeng H, Liu Y, Shi X, Liu Y, Liu C, Chen Y. Multicomponent thermosensitive lipid complexes enhance desmoplastic tumor therapy through boosting anti-angiogenesis and synergistic strategy. Int J Pharm 2021; 601:120533. [PMID: 33781886 DOI: 10.1016/j.ijpharm.2021.120533] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 03/08/2021] [Accepted: 03/22/2021] [Indexed: 01/05/2023]
Abstract
Currently, the chemotherapy drugs-loaded thermosensitive liposomes have not shown an over standard of clinical effects compared to preclinical trials. In addition to the limiting factors of clinical trial design and heating device, abnormal angiogenesis in desmoplastic tumor is a key factor for unexpected clinical efficacy. Malformed tumor vasculature may result in reduced vascular transport and the heterogeneous distribution of thermosensitive liposomes in tumor. Here, we report an anti-angiogenesis strategy through hypoxia-inducible factors (HIF)-1α-vascular endothelial growth factor (VEGF) axis based on icaritin and coix seed oil dual loaded multicomponent thermosensitive lipid complexes (IC-ML). IC-ML could downregulate the HIF-1α expression in HepG2 cells with a synergetic antitumor effect. In addition, HepG2 + LX-2 cells co-cultured 3D tumor spheres administered IC-ML showed the strongest penetration and inhibition of growth. Accordingly, IC-ML displayed improved tumor penetration and superior synergistic antitumor efficacy with HIF-1α-VEGF downregulation in vivo under mild hyperthermia. The improvement of antitumor efficacy of IC-ML comes from the anti-angiogenesis strategy and comprehensive tumor microenvironment remodeling, including depletion of cancer-associated fibroblasts as well as inhibition of M2-type tumor associated macrophage infiltration in desmoplastic tumor. This study proposes a novel multicomponent synergistic antitumor strategy to improve the therapeutic potential of thermosensitive lipid complexes for hepatocellular carcinoma.
Collapse
Affiliation(s)
- Jian Guo
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Jiangsu Provincial Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Huating Zeng
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Jiangsu Provincial Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Yimin Liu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Jiangsu Provincial Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Xinmeng Shi
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Jiangsu Provincial Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Yuping Liu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Jiangsu Provincial Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Congyan Liu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Jiangsu Provincial Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Yan Chen
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Jiangsu Provincial Academy of Traditional Chinese Medicine, Nanjing 210028, China.
| |
Collapse
|
40
|
Mancilla-Galindo J, Galindo-Sevilla N. Exploring the rationale for thermotherapy in COVID-19. Int J Hyperthermia 2021; 38:202-212. [PMID: 33682604 DOI: 10.1080/02656736.2021.1883127] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Increased transmissibility of the pandemic severe acute respiratory coronavirus 2 (SARS-CoV-2) has been noted to occur at lower ambient temperatures. This is seemingly related to a better replication of most respiratory viruses, including SARS-CoV-2, at lower-than-core body temperatures (i.e., 33 °C vs 37 °C). Also, intrinsic characteristics of SARS-CoV-2 make it a heat-susceptible pathogen. Thermotherapy has successfully been used to combat viral infections in plants which could otherwise result in great economic losses; 90% of viruses causing infections in plants are positive-sense single-stranded ribonucleic acid (+ssRNA) viruses, a characteristic shared by SARS-CoV-2. Thus, it is possible to envision the use of heat-based interventions (thermotherapy or mild-temperature hyperthermia) in patients with COVID-19 for which moderate cycles (every 8-12 h) of mild-temperature hyperthermia (1-2 h) have been proposed. However, there are potential safety and mechanistic concerns which could limit the use of thermotherapy only to patients with mild-to-moderate COVID-19 to prevent disease progression rather than to treat patients who have already progressed to severe-to-critical COVID-19. Here, we review the characteristics of SARS-CoV-2 which make it a heat-susceptible virus, potential host mechanisms which could be enhanced at higher temperatures to aid viral clearance, and how thermotherapy could be investigated as a modality of treatment in patients with COVID-19 while taking into consideration potential risks.
Collapse
Affiliation(s)
- Javier Mancilla-Galindo
- Facultad de Medicina, División de Investigación, Unidad de Investigación UNAM-INC, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Norma Galindo-Sevilla
- Departamento de Infectología e Inmunología, Instituto Nacional de Perinatología, Mexico City, Mexico
| |
Collapse
|
41
|
Nagata T, Kanamori M, Sekine S, Arai M, Moriyama M, Fujii T. Clinical study of modulated electro-hyperthermia for advanced metastatic breast cancer. Mol Clin Oncol 2021; 14:103. [PMID: 33796292 PMCID: PMC8010507 DOI: 10.3892/mco.2021.2265] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 03/05/2021] [Indexed: 11/06/2022] Open
Abstract
Modulated electro-hyperthermia (mEHT) is a new treatment modality developed to overcome the problems associated with traditional hyperthermia; mEHT uses a precise impedance-matched system and modulated radiofrequency current flow to malignant tumors. It selects the malignant cells based on their biophysical differences, due to their high metabolic rate, individual (autonomic) behavior and membrane status. The aim of the present study was to report the outcomes of mEHT in the treatment of advanced breast cancer. mEHT was examined in 10 patients with advanced metastatic breast cancer and recurrent disease, who were considered incurable by standard therapy protocols. Of the 10 patients, partial response was achieved in 3, disease stability in 3, and progressive disease in 4; however, their quality of life was improved based on their subjective reports. No adverse effects were observed in any of the 10 patients. The present study demonstrated the feasibility of mEHT as a possible therapy for advanced breast cancer cases when standard therapies fail. Moreover, mEHT had no side effects and may be combined with various treatments for long-term therapy.
Collapse
Affiliation(s)
- Takuya Nagata
- Department of Surgery, Toho University Ohashi Medical Center, Tokyo 153-8515, Japan
| | - Masahiko Kanamori
- Department of Human Science, University of Toyama, Toyama 930-0194, Japan
| | - Shinichi Sekine
- Department of Surgery, Kamiichi General Hospital, Toyama 930-0391, Japan
| | - Mie Arai
- Department of Surgery, Toyama Nishi General Hospital, Toyama 939-2716, Japan
| | - Makoto Moriyama
- Department of Surgery, Tomei Atsugi Hospital, Kanagawa 243-8571, Japan
| | - Tsutomu Fujii
- Department of Science and Surgery, University of Toyama, Toyama 930-0194, Japan
| |
Collapse
|
42
|
Vancsik T, Máthé D, Horváth I, Várallyaly AA, Benedek A, Bergmann R, Krenács T, Benyó Z, Balogh A. Modulated Electro-Hyperthermia Facilitates NK-Cell Infiltration and Growth Arrest of Human A2058 Melanoma in a Xenograft Model. Front Oncol 2021; 11:590764. [PMID: 33732640 PMCID: PMC7959784 DOI: 10.3389/fonc.2021.590764] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 01/13/2021] [Indexed: 11/13/2022] Open
Abstract
Modulated electro-hyperthermia (mEHT), induced by 13.56 MHz radiofrequency, has been demonstrated both in preclinical and clinical studies to efficiently induce tumor damage and complement other treatment modalities. Here, we used a mouse xenograft model of human melanoma (A2058) to test mEHT (~42°C) both alone and combined with NK-cell immunotherapy. A single 30 min shot of mEHT resulted in significant tumor damage due to induced stress, marked by high hsp70 expression followed by significant upregulation of cleaved/activated caspase-3 and p53. When mEHT was combined with either primary human NK cells or the IL-2 independent NK-92MI cell line injected subcutaneously, the accumulation of NK cells was observed at the mEHT pretreated melanoma nodules but not at the untreated controls. mEHT induced the upregulation of the chemoattractant CXCL11 and increased the expression of the matrix metalloproteinase MMP2 which could account for the NK-cell attraction into the treated melanoma. In conclusion, mEHT monotherapy of melanoma xenograft tumors induced irreversible heat and cell stress leading to caspase dependent apoptosis to be driven by p53. mEHT could support the intratumoral attraction of distantly injected NK-cells, contributed by CXCL11 and MMP2 upregulation, resulting in an additive tumor destruction and growth inhibition. Therefore, mEHT may offer itself as a good partner for immunotherapy.
Collapse
Affiliation(s)
- Tamás Vancsik
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Domokos Máthé
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Ildikó Horváth
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | | | - Anett Benedek
- Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Ralf Bergmann
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Tibor Krenács
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Zoltán Benyó
- Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Andrea Balogh
- Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
| |
Collapse
|
43
|
Carneiro MW, Brancato L, Wylleman B, van Zwol E, Conings L, Vueghs P, Gorbaslieva I, Van den Bossche J, Rudenko O, Janicot M, Bogers JP. Safety evaluation of long-term temperature controlled whole-body thermal treatment in female Aachen minipig. Int J Hyperthermia 2021; 38:165-175. [PMID: 33576280 DOI: 10.1080/02656736.2021.1876256] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Objective: Thermal treatment (TT), defined as treatment using supra-physiological body temperatures (39-45 C), somewhat resembles fever in terms of temperature range, one of the first natural barriers for the body to fight exposure to external pathogens. Methods: Whole-body thermal treatment (WBTT) consists of heating up the complete body to a temperature range of 39 to 45 C. Despite the recognized therapeutic potential of hyperthermia, the broad clinical use of WBTT has been limited by safety issues related to medical devices and procedures used to achieve WBTT, in particular adequate control of the body temperature. To circumvent this, a sophisticated medical device was developed, allowing long-term temperature controlled WBTT (41.5 C for up to 8 h). Technical feasibility and tolerability of the WBTT procedure (including complete anesthesia) were tested using female Aachen minipig. Optical fiber temperature sensors inserted in multiple organs were used and demonstrated consistent monitoring and control of different organs temperature over an extended period of time. Results: Clinical evaluation of the animals before, during and after treatment revealed minor clinical parameter changes, but all of them were clinically acceptable. These changes were limited and reversible, and the animals remained healthy throughout the whole procedure and follow-up. In addition, histopathological analysis of selected key organs showed no thermal treatment-related changes. Conclusion: It was concluded that WBTT (41.5 C for up to 8 h) was well tolerated and safe in female Aachen minipigs. Altogether, data supports the safe clinical use of the WBTT medical device and protocol, enabling its implementation into human patients suffering from life-threatening diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - John-Paul Bogers
- ElmediX NV, Mechelen, Belgium.,Laboratory for Cell Biology and Histology, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
44
|
Stephen ZR, Zhang M. Recent Progress in the Synergistic Combination of Nanoparticle-Mediated Hyperthermia and Immunotherapy for Treatment of Cancer. Adv Healthc Mater 2021; 10:e2001415. [PMID: 33236511 PMCID: PMC8034553 DOI: 10.1002/adhm.202001415] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 10/11/2020] [Indexed: 02/06/2023]
Abstract
Immunotherapy has demonstrated great clinical success in certain cancers, driven primarily by immune checkpoint blockade and adoptive cell therapies. Immunotherapy can elicit strong, durable responses in some patients, but others do not respond, and to date immunotherapy has demonstrated success in only a limited number of cancers. To address this limitation, combinatorial approaches with chemo- and radiotherapy have been applied in the clinic. Extensive preclinical evidence suggests that hyperthermia therapy (HT) has considerable potential to augment immunotherapy with minimal toxicity. This progress report will provide a brief overview of immunotherapy and HT approaches and highlight recent progress in the application of nanoparticle (NP)-based HT in combination with immunotherapy. NPs allow for tumor-specific targeting of deep tissue tumors while potentially providing more even heating. NP-based HT increases tumor immunogenicity and tumor permeability, which improves immune cell infiltration and creates an environment more responsive to immunotherapy, particularly in solid tumors.
Collapse
Affiliation(s)
- Zachary R Stephen
- Department of Materials Science and Engineering, University of Washington, Seattle, WA, 98195, USA
| | - Miqin Zhang
- Department of Materials Science and Engineering, Department of Neurological Surgery, University of Washington, Seattle, WA, 98195, USA
| |
Collapse
|
45
|
Krenacs T, Meggyeshazi N, Forika G, Kiss E, Hamar P, Szekely T, Vancsik T. Modulated Electro-Hyperthermia-Induced Tumor Damage Mechanisms Revealed in Cancer Models. Int J Mol Sci 2020; 21:E6270. [PMID: 32872532 PMCID: PMC7504298 DOI: 10.3390/ijms21176270] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/22/2020] [Accepted: 08/24/2020] [Indexed: 12/18/2022] Open
Abstract
The benefits of high-fever range hyperthermia have been utilized in medicine from the Ancient Greek culture to the present day. Amplitude-modulated electro-hyperthermia, induced by a 13.56 MHz radiofrequency current (mEHT, or Oncothermia), has been an emerging means of delivering loco-regional clinical hyperthermia as a complementary of radiation-, chemo-, and molecular targeted oncotherapy. This unique treatment exploits the metabolic shift in cancer, resulting in elevated oxidative glycolysis (Warburg effect), ion concentration, and electric conductivity. These promote the enrichment of electric fields and induce heat (controlled at 42 °C), as well as ion fluxes and disequilibrium through tumor cell membrane channels. By now, accumulating preclinical studies using in vitro and in vivo models of different cancer types have revealed details of the mechanism and molecular background of the oncoreductive effects of mEHT monotherapy. These include the induction of DNA double-strand breaks, irreversible heath and cell stress, and programmed cells death; the upregulation of molecular chaperones and damage (DAMP) signaling, which may contribute to a secondary immunogenic tumor cell death. In combination therapies, mEHT proved to be a good chemosensitizer through increasing drug uptake and tumor reductive effects, as well as a good radiosensitizer by downregulating hypoxia-related target genes. Recently, immune stimulation or intratumoral antigen-presenting dendritic cell injection have been able to extend the impact of local mEHT into a systemic "abscopal" effect. The complex network of pathways emerging from the published mEHT experiments has not been overviewed and arranged yet into a framework to reveal links between the pieces of the "puzzle". In this paper, we review the mEHT-related damage mechanisms published in tumor models, which may allow some geno-/phenotype treatment efficiency correlations to be exploited both in further research and for more rational clinical treatment planning when mEHT is involved in combination therapies.
Collapse
Affiliation(s)
- Tibor Krenacs
- Department of Pathology and Experimental Cancer Research, Semmelweis University, H-1085 Budapest, Hungary; (N.M.); (G.F.); (T.S.)
| | - Nora Meggyeshazi
- Department of Pathology and Experimental Cancer Research, Semmelweis University, H-1085 Budapest, Hungary; (N.M.); (G.F.); (T.S.)
| | - Gertrud Forika
- Department of Pathology and Experimental Cancer Research, Semmelweis University, H-1085 Budapest, Hungary; (N.M.); (G.F.); (T.S.)
| | - Eva Kiss
- Institute of Oncology at 1st Department of Internal Medicine, Semmelweis University, H-1083 Budapest, Hungary;
| | - Peter Hamar
- Institute of Translational Medicine, Semmelweis University, H-1094 Budapest, Hungary; (P.H.); (T.V.)
| | - Tamas Szekely
- Department of Pathology and Experimental Cancer Research, Semmelweis University, H-1085 Budapest, Hungary; (N.M.); (G.F.); (T.S.)
| | - Tamas Vancsik
- Institute of Translational Medicine, Semmelweis University, H-1094 Budapest, Hungary; (P.H.); (T.V.)
| |
Collapse
|
46
|
Datta NR, Kok HP, Crezee H, Gaipl US, Bodis S. Integrating Loco-Regional Hyperthermia Into the Current Oncology Practice: SWOT and TOWS Analyses. Front Oncol 2020; 10:819. [PMID: 32596144 PMCID: PMC7303270 DOI: 10.3389/fonc.2020.00819] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 04/27/2020] [Indexed: 12/14/2022] Open
Abstract
Moderate hyperthermia at temperatures between 40 and 44°C is a multifaceted therapeutic modality. It is a potent radiosensitizer, interacts favorably with a host of chemotherapeutic agents, and, in combination with radiotherapy, enforces immunomodulation akin to “in situ tumor vaccination.” By sensitizing hypoxic tumor cells and inhibiting repair of radiotherapy-induced DNA damage, the properties of hyperthermia delivered together with photons might provide a tumor-selective therapeutic advantage analogous to high linear energy transfer (LET) neutrons, but with less normal tissue toxicity. Furthermore, the high LET attributes of hyperthermia thermoradiobiologically are likely to enhance low LET protons; thus, proton thermoradiotherapy would mimic 12C ion therapy. Hyperthermia with radiotherapy and/or chemotherapy substantially improves therapeutic outcomes without enhancing normal tissue morbidities, yielding level I evidence reported in several randomized clinical trials, systematic reviews, and meta-analyses for various tumor sites. Technological advancements in hyperthermia delivery, advancements in hyperthermia treatment planning, online invasive and non-invasive MR-guided thermometry, and adherence to quality assurance guidelines have ensured safe and effective delivery of hyperthermia to the target region. Novel biological modeling permits integration of hyperthermia and radiotherapy treatment plans. Further, hyperthermia along with immune checkpoint inhibitors and DNA damage repair inhibitors could further augment the therapeutic efficacy resulting in synthetic lethality. Additionally, hyperthermia induced by magnetic nanoparticles coupled to selective payloads, namely, tumor-specific radiotheranostics (for both tumor imaging and radionuclide therapy), chemotherapeutic drugs, immunotherapeutic agents, and gene silencing, could provide a comprehensive tumor-specific theranostic modality akin to “magic (nano)bullets.” To get a realistic overview of the strength (S), weakness (W), opportunities (O), and threats (T) of hyperthermia, a SWOT analysis has been undertaken. Additionally, a TOWS analysis categorizes future strategies to facilitate further integration of hyperthermia with the current treatment modalities. These could gainfully accomplish a safe, versatile, and cost-effective enhancement of the existing therapeutic armamentarium to improve outcomes in clinical oncology.
Collapse
Affiliation(s)
- Niloy R Datta
- Centre for Radiation Oncology KSA-KSB, Kantonsspital Aarau, Aarau, Switzerland
| | - H Petra Kok
- Department of Radiation Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Hans Crezee
- Department of Radiation Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Udo S Gaipl
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Stephan Bodis
- Centre for Radiation Oncology KSA-KSB, Kantonsspital Aarau, Aarau, Switzerland
| |
Collapse
|
47
|
Omata D, Unga J, Suzuki R, Maruyama K. Lipid-based microbubbles and ultrasound for therapeutic application. Adv Drug Deliv Rev 2020; 154-155:236-244. [PMID: 32659255 DOI: 10.1016/j.addr.2020.07.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 06/11/2020] [Accepted: 07/07/2020] [Indexed: 12/12/2022]
Abstract
Microbubbles with diagnostic ultrasound have had a long history of use in the medical field. In recent years, the therapeutic application of the combination of microbubbles and ultrasound, called sonoporation, has received increased attention as microbubble oscillation or collapse close to various barriers in the body was recognized to potentially open those barriers, increasing drug transport across them. In this review, we aimed to describe the development of lipid-stabilized microbubbles equipped with functions, such as long circulation and drug loading, and the therapeutic application of sonoporation for tumor-targeted therapy, brain-targeted therapy, and immunotherapy. We also attempted to discuss the current status of the field and potential future developments.
Collapse
|