1
|
Ödén J, Eriksson K, Pavoni B, Crezee H, Kok HP. A Novel Framework for Thermoradiotherapy Treatment Planning. Int J Radiat Oncol Biol Phys 2024; 119:1530-1544. [PMID: 38387812 DOI: 10.1016/j.ijrobp.2024.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 01/24/2024] [Accepted: 02/08/2024] [Indexed: 02/24/2024]
Abstract
PURPOSE Thermoradiotherapy combines radiation therapy with hyperthermia to increase therapeutic effectiveness. Currently, both modalities are optimized separately and in state-of-the-art research the enhanced therapeutic effect is evaluated using equivalent radiation dose in 2-Gy fractions (EQD2). This study proposes a novel thermoradiotherapy treatment planning framework with voxelwise EQD2 radiation therapy optimizing including thermal radiosensitization and direct thermal cytotoxicity. METHODS AND MATERIALS To demonstrate proof-of-concept of the planning framework, 3 strategies consisting of 20 radiation therapy fractions were planned for 4 prostate cancer cases with substantially different temperature distributions: (1) Conventional radiation therapy plan of 60 Gy combined with 4 hyperthermia sessions (RT60 + HT), (2) standalone uniform dose escalation to 68 Gy without hyperthermia (RT68), and (3) uniform target EQD2 that maximizes the tumor control probability (TCP) accounting for voxelwise thermal effects of 4 hyperthermia sessions without increasing normal tissue doses (RTHT + HT). Assessment included dose, EQD2, TCP, and rectal normal tissue complication probability (NTCP), alongside robustness analyses for TCP and NTCP against parameter uncertainties. RESULTS The estimated TCP of around 76% for RT60 without hyperthermia was increased to an average of 85.9% (range, 81.3%-90.5%) for RT60 + HT, 92.5% (92.4%-92.5%) for RT68, and 94.4% (91.7%-96.6%) for RTHT + HT. The corresponding averaged rectal NTCPs were 8.7% (7.9%-10.0%), 14.9% (13.8%-17.1%), and 8.4% (7.5%-9.7%), respectively. RT68 and RTHT + HT exhibited slightly enhanced TCP robustness against parameter uncertainties compared with RT60 + HT, and RT68 presented higher and less robust rectal NTCP values compared with the other planning strategies. CONCLUSIONS This study introduces an innovative thermoradiotherapy planning approach, integrating thermal effects into EQD2-based radiation therapy optimization. Results demonstrate an ability to achieve enhanced and uniform target EQD2 and TCP across various temperature distributions without elevating normal tissue EQD2 or NTCP compared with conventional methods. Although promising for improving clinical outcomes, realizable enhancements depend on accurate tumor- and tissue-specific data and precise quantification of hyperthermic effects, which are seamlessly integrable in the planning framework as they emerge.
Collapse
Affiliation(s)
- Jakob Ödén
- RaySearch Laboratories AB, Stockholm, Sweden.
| | | | | | - Hans Crezee
- Department of Radiation Oncology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands; Cancer Center Amsterdam, Treatment and Quality of Life, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - H Petra Kok
- Department of Radiation Oncology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands; Cancer Center Amsterdam, Treatment and Quality of Life, Cancer Biology and Immunology, Amsterdam, The Netherlands
| |
Collapse
|
2
|
Taghizadeh S, Shvydka D, Shan A, Mian OY, Parsai EI. Optimization and experimental characterization of the innovative thermo-brachytherapy seed for prostate cancer treatment. Med Phys 2024; 51:839-853. [PMID: 38159297 DOI: 10.1002/mp.16920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 11/20/2023] [Accepted: 12/07/2023] [Indexed: 01/03/2024] Open
Abstract
BACKGROUND Adjuvant administration of hyperthermia (HT) with radiation therapy in the treatment of cancer has been extensively studied in the past five decades. Concurrent use of the two modalities leads to both complementary and synergetic enhancements in tumor management, but presents a practical challenge. Their simultaneous administration using the same implantable thermo-brachytherapy (TB) seed source has been established theoretically through magnetically mediated heat induction with ferromagnetic materials. Careful consideration, however, showed that regular ferromagnetic alloys lack the required conductivity to generate enough power through eddy current to overcome heat dissipation due to blood perfusion at clinically measured rates. PURPOSE We characterized the TB implant that combines a sealed radioactive source with a ferrimagnetic ceramic (ferrite) core, serving as a self-regulating HT source when placed in an alternating electromagnetic field. To increase the heat production and uniformity of temperature distribution the empty spacers between radioisotope seeds were replaced by hyperthermia-only (HT-only) seeds. METHODS The heat generation due to eddy currents circulating in the seed's thin metal shell, surrounding the core, depends drastically on the core permeability. We identified a soft ferrite material (MnZnFe 2 O 4 $\rm MnZnFe_2O_4$ ) as the best candidate for the core, owing to its high permeability, the HT-range Curie temperature, adjustable through material composition, and a sharp Curie transition, leading to heat self-regulation, with no invasive thermometry required. The core permeability as a function of temperature was calculated based on measured resistor-inductor (RL) circuit parameters and material B-H curves. The thickness of the shell was optimized separately for TB and HT-only seeds, having slightly different dimensions. Heat generation was calculated using the power versus temperature approximation. Finally, the temperature distribution for a realistic prostate LDR brachytherapy plan was modeled with COMSOL Multiphysics for a set of blood perfusion rates found in the literature. RESULTS The small size of the investigated ferrite core samples resulted in demagnetization significantly decreasing the relative permeability from its intrinsic value of ∼5000 to about 11 in the range of magnetic field amplitude and frequency values relevant to HT. The power generated by the seed dropped sharply as the shell thickness deviated from the optimal value. The optimized TB and HT-only seeds generated 45 and 267 mW power, respectively, providing a HT source sufficient for >90% volume coverage even for the highest blood perfusion rates. The toxicity of the surrounding normal tissues was minimal due to the rapid temperature fall off within a few millimeters distance from a seed. CONCLUSIONS The investigated TB and HT-only seed prototypes were shown to provide sufficient power for the concurrent administration of radiation and HT. In addition to being used as a source for both radiation and heat at the onset of cancer therapy, these implanted seeds would be available for treatment intensification in the setting of salvage brachytherapy for locally radiorecurrent disease, possibly as a sensitizer to systemic therapies or as a modulator of the immune response, without another invasive procedure. Experimentally determined parameters of the ferrite material cores provided in this study establish a mechanistic foundation for future pre-clinical and clinical validation studies.
Collapse
Affiliation(s)
- Somayeh Taghizadeh
- Department of Radiation Oncology, The University of Toledo Health Science Campus, Toledo, Ohio, USA
- Department of Physics and Astronomy, The University of Toledo, Toledo, Ohio, USA
| | - Diana Shvydka
- Department of Radiation Oncology, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Ambalanath Shan
- Department of Physics and Astronomy, The University of Toledo, Toledo, Ohio, USA
| | - Omar Y Mian
- Department of Radiation Oncology, Cleveland Clinic Taussig Cancer Center, Cleveland, Ohio, USA
| | - E Ishmael Parsai
- Department of Radiation Oncology, The University of Toledo Health Science Campus, Toledo, Ohio, USA
- Department of Physics and Astronomy, The University of Toledo, Toledo, Ohio, USA
| |
Collapse
|
3
|
Montazersaheb P, Pishgahzadeh E, Jahani VB, Farahzadi R, Montazersaheb S. Magnetic nanoparticle-based hyperthermia: A prospect in cancer stem cell tracking and therapy. Life Sci 2023; 323:121714. [PMID: 37088411 DOI: 10.1016/j.lfs.2023.121714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 04/10/2023] [Accepted: 04/18/2023] [Indexed: 04/25/2023]
Abstract
Tumor heterogeneity is a major problem in cancer treatment. Cancer stem cells (CSCs) are a subpopulation of tumor masses that produce proliferating and quiescent cells. Under stress-related conditions, quiescent cells are capable of repopulating tumor masses. Consequently, many attempts have been made to identify, isolate, and eradicate CSCs from various tumors. Research has found that quiescent CSCs are less susceptible to conventional therapy than bulk cancer cells. This could be due to reduced cell cycling and increased DNA repair capacity of these cells. Indeed, disease progression is temporarily suppressed by eliminating fast-proliferating tumor cells and sparing quiescent CSCs lead to cancer relapse. Among all the available therapeutic modalities for cancer treatment, hyperthermia uses moderate heat to kill tumor cells. Nanoparticle-based platforms have the potential to deposit heat locally and selectively with the simultaneous activation of nanoparticles as heat transducers. Over the past few decades, magnetic nanoparticles (MNPs) have been widely investigated in the biomedical field. Magnetic hyperthermia therapy (MHT) is a promising therapeutic approach in which MNPs are delivered directly through targeting (systemic) or by direct injection into a tumor under exposure to an alternating magnetic field (AMF). Heat is generated by the MNPs subjected to AMF at a frequency of 100 kHz. Despite the widespread use of MHT alone or in combination therapies, its effectiveness in targeting CSCs remains unclear. This review discusses various types of MHT and their related mechanisms in cancer therapy, particularly concerning the eradication of CSCs.
Collapse
Affiliation(s)
- Parsa Montazersaheb
- Department of Materials Engineering, Institute of Mechanical Engineering, University of Tabriz, Tabriz, Iran
| | - Elahe Pishgahzadeh
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Vahid Bayrami Jahani
- School of Metallurgy and Materials Engineering, Iran University of Science and Technology (IUST), Narmak, Tehran, Iran
| | - Raheleh Farahzadi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Soheila Montazersaheb
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
4
|
Wang T, Zhou W, Man X, Li W. Local hyperthermia as a good treatment option for hands and feet warts in children: Case series report. Dermatol Ther 2022; 35:e15936. [PMID: 36227625 DOI: 10.1111/dth.15936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 09/20/2022] [Accepted: 10/12/2022] [Indexed: 11/28/2022]
Affiliation(s)
- Tingting Wang
- Department of Dermatology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Weifang Zhou
- Department of Dermatology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoyong Man
- Department of Dermatology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Li
- Department of Dermatology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
5
|
Le Guevelou J, Chirila ME, Achard V, Guillemin PC, Lorton O, Uiterwijk JWE, Dipasquale G, Salomir R, Zilli T. Combined hyperthermia and radiotherapy for prostate cancer: a systematic review. Int J Hyperthermia 2022; 39:547-556. [PMID: 35313781 DOI: 10.1080/02656736.2022.2053212] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Optimization of treatment strategies for prostate cancer patients treated with curative radiation therapy (RT) represents one of the major challenges for the radiation oncologist. Dose escalation or combination of RT with systemic therapies is used to improve tumor control in patients with unfavorable prostate cancer, at the risk of increasing rates and severity of treatment-related toxicities. Elevation of temperature to a supra-physiological level has been shown to both increase tumor oxygenation and reduce DNA repair capabilities. Thus, hyperthermia (HT) combined with RT represents a compelling treatment strategy to improve the therapeutic ratio in prostate cancer patients. The aim of the present systematic review is to report on preclinical and clinical evidence supporting the combination of HT and RT for prostate cancer, discussing future applications and developments of this combined treatment.
Collapse
Affiliation(s)
- Jennifer Le Guevelou
- Division of Radiation Oncology, Geneva University Hospital, Geneva, Switzerland.,Faculty of Medicine, Geneva University, Geneva, Switzerland
| | - Monica Emilia Chirila
- Division of Radiation Oncology, Geneva University Hospital, Geneva, Switzerland.,Amethyst Radiotherapy Centre, Cluj-Napoca, Romania
| | - Vérane Achard
- Division of Radiation Oncology, Geneva University Hospital, Geneva, Switzerland.,Faculty of Medicine, Geneva University, Geneva, Switzerland
| | | | - Orane Lorton
- Department of Radiology and Medical Informatics, Geneva University Hospital, Geneva, Switzerland
| | | | - Giovanna Dipasquale
- Division of Radiation Oncology, Geneva University Hospital, Geneva, Switzerland
| | - Rares Salomir
- Faculty of Medicine, Geneva University, Geneva, Switzerland.,Department of Radiology and Medical Informatics, Geneva University Hospital, Geneva, Switzerland
| | - Thomas Zilli
- Division of Radiation Oncology, Geneva University Hospital, Geneva, Switzerland.,Faculty of Medicine, Geneva University, Geneva, Switzerland
| |
Collapse
|
6
|
Clinical Evidence for Thermometric Parameters to Guide Hyperthermia Treatment. Cancers (Basel) 2022; 14:cancers14030625. [PMID: 35158893 PMCID: PMC8833668 DOI: 10.3390/cancers14030625] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/14/2022] [Accepted: 01/19/2022] [Indexed: 01/01/2023] Open
Abstract
Hyperthermia (HT) is a cancer treatment modality which targets malignant tissues by heating to 40-43 °C. In addition to its direct antitumor effects, HT potently sensitizes the tumor to radiotherapy (RT) and chemotherapy (CT), thereby enabling complete eradication of some tumor entities as shown in randomized clinical trials. Despite the proven efficacy of HT in combination with classic cancer treatments, there are limited international standards for the delivery of HT in the clinical setting. Consequently, there is a large variability in reported data on thermometric parameters, including the temperature obtained from multiple reference points, heating duration, thermal dose, time interval, and sequence between HT and other treatment modalities. Evidence from some clinical trials indicates that thermal dose, which correlates with heating time and temperature achieved, could be used as a predictive marker for treatment efficacy in future studies. Similarly, other thermometric parameters when chosen optimally are associated with increased antitumor efficacy. This review summarizes the existing clinical evidence for the prognostic and predictive role of the most important thermometric parameters to guide the combined treatment of RT and CT with HT. In conclusion, we call for the standardization of thermometric parameters and stress the importance for their validation in future prospective clinical studies.
Collapse
|
7
|
Nakahara S, Ohguri T, Kakinouchi S, Itamura H, Morisaki T, Tani S, Yahara K, Fujimoto N. Intensity-Modulated Radiotherapy with Regional Hyperthermia for High-Risk Localized Prostate Carcinoma. Cancers (Basel) 2022; 14:cancers14020400. [PMID: 35053562 PMCID: PMC8774016 DOI: 10.3390/cancers14020400] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 12/25/2021] [Accepted: 01/11/2022] [Indexed: 02/04/2023] Open
Abstract
Background: The purpose of this study was to evaluate the efficacy and toxicity of adding regional hyperthermia to intensity-modulated radiotherapy (IMRT) plus neoadjuvant androgen deprivation therapy (ADT) for high-risk localized prostate carcinoma. Methods: Data from 121 consecutive patients with high-risk prostate carcinoma who were treated with IMRT were retrospectively analyzed. The total planned dose of IMRT was 76 Gy in 38 fractions for all patients; hyperthermia was used in 70 of 121 patients. Intra-rectal temperatures at the prostate level were measured to evaluate thermal dose. Results: Median number of heating sessions was five and the median total thermal dose of CEM43T90 was 7.5 min. Median follow-up duration was 64 months. Addition of hyperthermia to IMRT predicted better clinical relapse-free survival. Higher thermal dose with CEM43T90 (>7 min) predicted improved biochemical disease-free survival. The occurrence of acute and delayed toxicity ≥Grade 2 was not significantly different between patients with or without hyperthermia. Conclusions: IMRT plus regional hyperthermia represents a promising approach with acceptable toxicity for high-risk localized prostate carcinoma. Further studies are needed to verify the efficacy of this combined treatment.
Collapse
Affiliation(s)
- Sota Nakahara
- Department of Therapeutic Radiology, University Hospital of Occupational and Environmental Health, Kitakyushu 807-8555, Japan; (S.N.); (S.K.); (H.I.); (T.M.); (S.T.)
| | - Takayuki Ohguri
- Department of Therapeutic Radiology, University Hospital of Occupational and Environmental Health, Kitakyushu 807-8555, Japan; (S.N.); (S.K.); (H.I.); (T.M.); (S.T.)
- Correspondence: ; Tel.: +81-93-691-7264; Fax: +81-93-692-0249
| | - Sho Kakinouchi
- Department of Therapeutic Radiology, University Hospital of Occupational and Environmental Health, Kitakyushu 807-8555, Japan; (S.N.); (S.K.); (H.I.); (T.M.); (S.T.)
| | - Hirohide Itamura
- Department of Therapeutic Radiology, University Hospital of Occupational and Environmental Health, Kitakyushu 807-8555, Japan; (S.N.); (S.K.); (H.I.); (T.M.); (S.T.)
| | - Takahiro Morisaki
- Department of Therapeutic Radiology, University Hospital of Occupational and Environmental Health, Kitakyushu 807-8555, Japan; (S.N.); (S.K.); (H.I.); (T.M.); (S.T.)
| | - Subaru Tani
- Department of Therapeutic Radiology, University Hospital of Occupational and Environmental Health, Kitakyushu 807-8555, Japan; (S.N.); (S.K.); (H.I.); (T.M.); (S.T.)
| | - Katuya Yahara
- Department of Radiotherapy, Kurashiki Medical Center, Kurashiki 710-8522, Japan;
| | - Naohiro Fujimoto
- Department of Urology, University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan;
| |
Collapse
|
8
|
Androulakis I, Mestrom RMC, Christianen MEMC, Kolkman-Deurloo IKK, van Rhoon GC. Design of the novel ThermoBrachy applicators enabling simultaneous interstitial hyperthermia and high dose rate brachytherapy. Int J Hyperthermia 2021; 38:1660-1671. [PMID: 34814784 DOI: 10.1080/02656736.2021.2005160] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
OBJECTIVE In High Dose Rate Brachytherapy for prostate cancer there is a need for a new way of increasing cancer cell kill in combination with a stable dose to the organs at risk. In this study, we propose a novel ThermoBrachy applicator that offers the unique ability to apply interstitial hyperthermia while simultaneously serving as an afterloading catheter for high dose rate brachytherapy for prostate cancer. This approach achieves a higher thermal enhancement ratio than in sequential application of radiation and hyperthermia and has the potential to decrease the overall treatment time. METHODS The new applicator uses the principle of capacitively coupled electrodes. We performed a proof of concept experiment to demostrate the feasibility of the proposed applicator. Moreover, we used electromagnetic and thermal simulations to evaluate the power needs and temperature homogeneity in different tissues. Furthermore we investigated whether dynamic phase and amplitude adaptation can be used to improve longitudinal temperature control. RESULTS Simulations demonstrate that the electrodes achieve good temperature homogeneity in a homogenous phantom when following current applicator spacing guidelines. Furthermore, we demonstrate that by dynamic phase and amplitude adaptation provides a great advancement for further adaptability of the heating pattern. CONCLUSIONS This newly designed ThermoBrachy applicator has the potential to revise the interest in interstitial thermobrachytherapy, since the simultaneous application of radiation and hyperthermia enables maximum thermal enhancement and at maximum efficiency for patient and organization.
Collapse
Affiliation(s)
- Ioannis Androulakis
- Department of Radiotherapy, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Rob M C Mestrom
- Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | | | | | - Gerard C van Rhoon
- Department of Radiotherapy, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| |
Collapse
|
9
|
Zhang X, Landgraf L, Bailis N, Unger M, Jochimsen TH, Melzer A. Image-Guided High-Intensity Focused Ultrasound, A Novel Application for Interventional Nuclear Medicine? J Nucl Med 2021; 62:1181-1188. [PMID: 34088775 PMCID: PMC8882895 DOI: 10.2967/jnumed.120.256230] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 05/05/2021] [Indexed: 12/25/2022] Open
Abstract
Image-guided high-intensity focused ultrasound (HIFU) has been increasingly used in medicine over the past few decades, and several systems for such have become commercially available. HIFU has passed regulatory approval around the world for the ablation of various solid tumors, the treatment of neurologic diseases, and the palliative management of bone metastases. The mechanical and thermal effects of focused ultrasound provide a possibility for histotripsy, supportive radiation therapy, and targeted drug delivery. The integration of imaging modalities into HIFU systems allows for precise temperature monitoring and accurate treatment planning, increasing the safety and efficiency of treatment. Preclinical and clinical results have demonstrated the potential of image-guided HIFU to reduce adverse effects and increase the quality of life postoperatively. Interventional nuclear image-guided HIFU is an attractive noninvasive option for the future.
Collapse
Affiliation(s)
- Xinrui Zhang
- Innovation Center Computer Assisted Surgery (ICCAS), University of Leipzig, Leipzig, Germany
| | - Lisa Landgraf
- Innovation Center Computer Assisted Surgery (ICCAS), University of Leipzig, Leipzig, Germany
| | - Nikolaos Bailis
- Department of Diagnostic and Interventional Radiology, University of Leipzig, Leipzig, Germany
| | - Michael Unger
- Innovation Center Computer Assisted Surgery (ICCAS), University of Leipzig, Leipzig, Germany
| | - Thies H Jochimsen
- Department of Nuclear Medicine, Leipzig University Hospital, Leipzig, Germany; and
| | - Andreas Melzer
- Innovation Center Computer Assisted Surgery (ICCAS), University of Leipzig, Leipzig, Germany;
- Institute of Medical Science and Technology (IMSaT), University of Dundee, Dundee, Scotland
| |
Collapse
|
10
|
Bienia A, Wiecheć-Cudak O, Murzyn AA, Krzykawska-Serda M. Photodynamic Therapy and Hyperthermia in Combination Treatment-Neglected Forces in the Fight against Cancer. Pharmaceutics 2021; 13:1147. [PMID: 34452108 PMCID: PMC8399393 DOI: 10.3390/pharmaceutics13081147] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/26/2021] [Accepted: 07/16/2021] [Indexed: 12/24/2022] Open
Abstract
Cancer is one of the leading causes of death in humans. Despite the progress in cancer treatment, and an increase in the effectiveness of diagnostic methods, cancer is still highly lethal and very difficult to treat in many cases. Combination therapy, in the context of cancer treatment, seems to be a promising option that may allow minimizing treatment side effects and may have a significant impact on the cure. It may also increase the effectiveness of anti-cancer therapies. Moreover, combination treatment can significantly increase delivery of drugs to cancerous tissues. Photodynamic therapy and hyperthermia seem to be ideal examples that prove the effectiveness of combination therapy. These two kinds of therapy can kill cancer cells through different mechanisms and activate various signaling pathways. Both PDT and hyperthermia play significant roles in the perfusion of a tumor and the network of blood vessels wrapped around it. The main goal of combination therapy is to combine separate mechanisms of action that will make cancer cells more sensitive to a given therapeutic agent. Such an approach in treatment may contribute toward increasing its effectiveness, optimizing the cancer treatment process in the future.
Collapse
Affiliation(s)
| | | | | | - Martyna Krzykawska-Serda
- Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Kraków, Poland; (A.B.); (O.W.-C.); (A.A.M.)
| |
Collapse
|
11
|
Zhu L, Huang Y, Lam D, Gach HM, Zoberi I, Hallahan DE, Grigsby PW, Chen H, Altman MB. Targetability of cervical cancer by magnetic resonance-guided high-intensity focused ultrasound (MRgHIFU)-mediated hyperthermia (HT) for patients receiving radiation therapy. Int J Hyperthermia 2021; 38:498-510. [PMID: 33757406 DOI: 10.1080/02656736.2021.1895330] [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: 12/30/2022] Open
Abstract
PURPOSE To evaluate the targetability of late-stage cervical cancer by magnetic resonance-guided high-intensity focused ultrasound (MRgHIFU)-induced hyperthermia (HT) as an adjuvant to radiation therapy (RT). METHODS Seventy-nine cervical cancer patients (stage IIIB-IVA) who received RT with lesions visible on positron emission tomography-computed tomography (PET-CT) were retrospectively analyzed for targetability using a commercially-available HT-capable MRgHIFU system. Targetability was assessed for both primary targets and/or any metastatic lymph nodes using both posterior (supine) and anterior (prone) patient setups relative to the transducer. Thirty-four different angles of rotation along subjects' longitudinal axis were analyzed. Targetability was categorized as: (1) Targetable with/without minimal intervention; (2) Not targetable. To determine if any factors could be used for prospective screening of patients, potential associations between demographic/anatomical factors and targetability were analyzed. RESULTS 72.15% primary tumors and 33.96% metastatic lymph nodes were targetable from at least one angle. 49.37% and 39.24% of primary tumors could be targeted with patient laying in supine and prone positions, respectively. 25°-30° rotation and 0° rotation had the highest rate of the posterior and anterior targetability, respectively. The ventral depth of the tumor and its distance to the coccyx were statistically correlated with the anterior and posterior targetability, respectively. CONCLUSION Most late-stage cervical cancer primaries were targetable by MRgHIFU HT requiring either no/minimal intervention. A rotation of 0° or 25°-30° relative to the transducer might benefit anterior and posterior targetability, respectively. Certain demographic/anatomic parameters might be useful in screening patients for treatability.
Collapse
Affiliation(s)
- Lifei Zhu
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Yi Huang
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, MO, USA
| | - Dao Lam
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, MO, USA
| | - H Michael Gach
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA.,Department of Radiation Oncology, Washington University in St. Louis, St. Louis, MO, USA.,Department of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Imran Zoberi
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, MO, USA.,Siteman Comprehensive Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Dennis E Hallahan
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA.,Department of Radiation Oncology, Washington University in St. Louis, St. Louis, MO, USA.,Siteman Comprehensive Cancer Center, Washington University in St. Louis, St. Louis, MO, USA.,Institute of Clinical and Translational Sciences, Washington University in St. Louis, St. Louis, MO, USA
| | - Perry W Grigsby
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, MO, USA.,Siteman Comprehensive Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Hong Chen
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA.,Department of Radiation Oncology, Washington University in St. Louis, St. Louis, MO, USA.,Siteman Comprehensive Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Michael B Altman
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA.,Department of Radiation Oncology, Washington University in St. Louis, St. Louis, MO, USA.,Siteman Comprehensive Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
12
|
Zhang X, Bobeica M, Unger M, Bednarz A, Gerold B, Patties I, Melzer A, Landgraf L. Focused ultrasound radiosensitizes human cancer cells by enhancement of DNA damage. Strahlenther Onkol 2021; 197:730-743. [PMID: 33885910 PMCID: PMC8292237 DOI: 10.1007/s00066-021-01774-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 03/23/2021] [Indexed: 12/19/2022]
Abstract
Purpose High-intensity focused ultrasound (HIFU/FUS) has expanded as a noninvasive quantifiable option for hyperthermia (HT). HT in a temperature range of 40–47 °C (thermal dose CEM43 ≥ 25) could work as a sensitizer to radiation therapy (RT). Here, we attempted to understand the tumor radiosensitization effect at the cellular level after a combination treatment of FUS+RT. Methods An in vitro FUS system was developed to induce HT at frequencies of 1.147 and 1.467 MHz. Human head and neck cancer (FaDU), glioblastoma (T98G), and prostate cancer (PC-3) cells were exposed to FUS in ultrasound-penetrable 96-well plates followed by single-dose X‑ray irradiation (10 Gy). Radiosensitizing effects of FUS were investigated by cell metabolic activity (WST‑1 assay), apoptosis (annexin V assay, sub-G1 assay), cell cycle phases (propidium iodide staining), and DNA double-strand breaks (γH2A.X assay). Results The FUS intensities of 213 (1.147 MHz) and 225 W/cm2 (1.467 MHz) induced HT for 30 min at mean temperatures of 45.20 ± 2.29 °C (CEM43 = 436 ± 88) and 45.59 ± 1.65 °C (CEM43 = 447 ± 79), respectively. FUS improves the effect of RT significantly by reducing metabolic activity in T98G cells 48 h (RT: 96.47 ± 8.29%; FUS+RT: 79.38 ± 14.93%; p = 0.012) and in PC-3 cells 72 h (54.20 ± 10.85%; 41.01 ± 11.17%; p = 0.016) after therapy, but not in FaDu cells. Mechanistically, FUS+RT leads to increased apoptosis and enhancement of DNA double-strand breaks compared to RT alone in T98G and PC-3 cells. Conclusion Our in vitro findings demonstrate that FUS has good potential to sensitize glioblastoma and prostate cancer cells to RT by mainly enhancing DNA damage. Supplementary Information The online version of this article (10.1007/s00066-021-01774-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xinrui Zhang
- Innovation Center Computer Assisted Surgery (ICCAS), University of Leipzig, Semmelweisstr. 14, Haus 14, Leipzig, 04103, Germany.
| | - Mariana Bobeica
- Institute for Medical Science and Technology (IMSaT), University of Dundee, Wilson House, 1 Wurzburg Loan, Dundee MediPark, Dundee, DD2 1FD, UK.,Extreme Light Infrastructure - Nuclear Physics ELI-NP, "Horia Hulubei" National Institute for Physics and Nuclear Engineering, 30 Reactorului Street, Bucharest-Magurele, 077125, Romania
| | - Michael Unger
- Innovation Center Computer Assisted Surgery (ICCAS), University of Leipzig, Semmelweisstr. 14, Haus 14, Leipzig, 04103, Germany
| | - Anastasia Bednarz
- Innovation Center Computer Assisted Surgery (ICCAS), University of Leipzig, Semmelweisstr. 14, Haus 14, Leipzig, 04103, Germany
| | - Bjoern Gerold
- Institute for Medical Science and Technology (IMSaT), University of Dundee, Wilson House, 1 Wurzburg Loan, Dundee MediPark, Dundee, DD2 1FD, UK.,Theraclion, 102 Rue Etienne Dolet, Malakoff, 92240, France
| | - Ina Patties
- Innovation Center Computer Assisted Surgery (ICCAS), University of Leipzig, Semmelweisstr. 14, Haus 14, Leipzig, 04103, Germany.,Department of Radiation Oncology, University of Leipzig, Stephanstr. 9a, Leipzig, 04103, Germany
| | - Andreas Melzer
- Innovation Center Computer Assisted Surgery (ICCAS), University of Leipzig, Semmelweisstr. 14, Haus 14, Leipzig, 04103, Germany. .,Institute for Medical Science and Technology (IMSaT), University of Dundee, Wilson House, 1 Wurzburg Loan, Dundee MediPark, Dundee, DD2 1FD, UK.
| | - Lisa Landgraf
- Innovation Center Computer Assisted Surgery (ICCAS), University of Leipzig, Semmelweisstr. 14, Haus 14, Leipzig, 04103, Germany
| |
Collapse
|
13
|
Mathematical model for the thermal enhancement of radiation response: thermodynamic approach. Sci Rep 2021; 11:5503. [PMID: 33750833 PMCID: PMC7970926 DOI: 10.1038/s41598-021-84620-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 02/15/2021] [Indexed: 02/08/2023] Open
Abstract
Radiotherapy can effectively kill malignant cells, but the doses required to cure cancer patients may inflict severe collateral damage to adjacent healthy tissues. Recent technological advances in the clinical application has revitalized hyperthermia treatment (HT) as an option to improve radiotherapy (RT) outcomes. Understanding the synergistic effect of simultaneous thermoradiotherapy via mathematical modelling is essential for treatment planning. We here propose a theoretical model in which the thermal enhancement ratio (TER) relates to the cell fraction being radiosensitised by the infliction of sublethal damage through HT. Further damage finally kills the cell or abrogates its proliferative capacity in a non-reversible process. We suggest the TER to be proportional to the energy invested in the sensitisation, which is modelled as a simple rate process. Assuming protein denaturation as the main driver of HT-induced sublethal damage and considering the temperature dependence of the heat capacity of cellular proteins, the sensitisation rates were found to depend exponentially on temperature; in agreement with previous empirical observations. Our findings point towards an improved definition of thermal dose in concordance with the thermodynamics of protein denaturation. Our predictions well reproduce experimental in vitro and in vivo data, explaining the thermal modulation of cellular radioresponse for simultaneous thermoradiotherapy.
Collapse
|
14
|
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: 36] [Impact Index Per Article: 9.0] [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
|
15
|
Jose J, Kumar R, Harilal S, Mathew GE, Parambi DGT, Prabhu A, Uddin MS, Aleya L, Kim H, Mathew B. Magnetic nanoparticles for hyperthermia in cancer treatment: an emerging tool. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:19214-19225. [PMID: 31884543 DOI: 10.1007/s11356-019-07231-2] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Accepted: 12/02/2019] [Indexed: 05/07/2023]
Abstract
Cancer remains as the major cause of death worldwide. The main reason why available therapies fail is that a vicious cycle in established which initiates multiple pathways and recurrence after metastasis. Hyperthermic treatment, which involves heating tumor tissues to a moderate temperature of 40-43 °C, has emerged as an effective strategy for treating tumors. This method is highly efficient at destroying tumor cells and does not induce the side effects of conventional cancer treatments. On the other hand, hyperthermic treatment method can be co-administered with conventional treatments. Nanotechnology had created huge opportunities in almost all areas of research, including the field of hyperthermic treatment. The utilization of magnetic nanoparticles (MNPs) offers functionalities not possible using conventional magnetic materials. In this review, we detail recent developments and applications of MNPs for hyperthermic treatment and discuss future possibilities.
Collapse
Affiliation(s)
- Jobin Jose
- Department of Pharmaceutics, NGSM Institute of Pharmaceutical Science, NITTE Deemed to be University, Mangalore, 575018, India
| | - Rajesh Kumar
- Kerala University of Health Sciences, Thrissur, Kerala, 680596, India
| | - Seetha Harilal
- Kerala University of Health Sciences, Thrissur, Kerala, 680596, India
| | | | | | - Ankitha Prabhu
- Department of Pharmaceutics, NGSM Institute of Pharmaceutical Science, NITTE Deemed to be University, Mangalore, 575018, India
| | - Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh
- Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | - Lotfi Aleya
- Chrono-Environment Laboratory, CNRS-6249, Bourgogne Franche-Comte University, Besancon, France
| | - Hoon Kim
- Department of Pharmacy, and Research Institute of Life Pharmaceutical Sciences, Sunchon National University, Suncheon, 57922, Republic of Korea.
| | - Bijo Mathew
- Department of Pharmaceutical Chemistry, Division of Drug Design and Medicinal Chemistry Research Lab, Ahalia School of Pharmacy, Palakkad, Kerala, 678557, India.
| |
Collapse
|
16
|
Mitxelena-Iribarren O, Campisi J, Martínez de Apellániz I, Lizarbe-Sancha S, Arana S, Zhukova V, Mujika M, Zhukov A. Glass-coated ferromagnetic microwire-induced magnetic hyperthermia for in vitro cancer cell treatment. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 106:110261. [DOI: 10.1016/j.msec.2019.110261] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 09/25/2019] [Accepted: 09/25/2019] [Indexed: 10/25/2022]
|
17
|
Zhu L, Altman MB, Laszlo A, Straube W, Zoberi I, Hallahan DE, Chen H. Ultrasound Hyperthermia Technology for Radiosensitization. ULTRASOUND IN MEDICINE & BIOLOGY 2019; 45:1025-1043. [PMID: 30773377 PMCID: PMC6475527 DOI: 10.1016/j.ultrasmedbio.2018.12.007] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 11/28/2018] [Accepted: 12/17/2018] [Indexed: 05/08/2023]
Abstract
Hyperthermia therapy (HT) raises tissue temperature to 40-45°C for up to 60 min. Hyperthermia is one of the most potent sensitizers of radiation therapy (RT). Ultrasound-mediated HT for radiosensitization has been used clinically since the 1960s. Recently, magnetic resonance-guided high-intensity focused ultrasound (MRgHIFU), which has been approved by the United States Food and Drug Administration for thermal ablation therapy, has been adapted for HT. With emerging clinical trials using MRgHIFU HT for radiosensitization, there is a pressing need to review the ultrasound HT technology. The objective of this review is to overview existing HT technology, summarize available ultrasound HT devices, evaluate clinical studies combining ultrasound HT with RT and discuss challenges and future directions.
Collapse
Affiliation(s)
- Lifei Zhu
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, Missouri, USA
| | - Michael B Altman
- Department of Radiation Oncology, Washington University in Saint Louis, Saint Louis, Missouri, USA
| | - Andrei Laszlo
- Department of Radiation Oncology, Washington University in Saint Louis, Saint Louis, Missouri, USA
| | - William Straube
- Department of Radiation Oncology, Washington University in Saint Louis, Saint Louis, Missouri, USA
| | - Imran Zoberi
- Department of Radiation Oncology, Washington University in Saint Louis, Saint Louis, Missouri, USA
| | - Dennis E Hallahan
- Department of Radiation Oncology, Washington University in Saint Louis, Saint Louis, Missouri, USA
| | - Hong Chen
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, Missouri, USA; Department of Radiation Oncology, Washington University in Saint Louis, Saint Louis, Missouri, USA.
| |
Collapse
|
18
|
Cohen J, Anvari A, Samanta S, Poirier Y, Soman S, Alexander A, Ranjbar M, Pavlovic R, Zodda A, Jackson IL, Mahmood J, Vujaskovic Z, Sawant A. Mild hyperthermia as a localized radiosensitizer for deep-seated tumors: investigation in an orthotopic prostate cancer model in mice. Br J Radiol 2019; 92:20180759. [PMID: 30673305 PMCID: PMC6541201 DOI: 10.1259/bjr.20180759] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 01/02/2019] [Accepted: 01/16/2019] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE: Non-ablative or mild hyperthermia (HT) has been shown in preclinical (and clinical) studies as a localized radiosensitizer that enhances the tumoricidal effects of radiation. Most preclinical in vivo HT studies use subcutaneous tumor models which do not adequately represent clinical conditions (e.g. proximity of normal/critical organs) or replicate the tumor microenvironment-both of which are important factors for eventual clinical translation. The purpose of this work is to demonstrate proof-of-concept of locoregional radiosensitization with superficially applied, radiofrequency (RF)-induced HT in an orthotopic mouse model of prostate cancer. METHODS: In a 4-arm study, 40 athymic male nude mice were inoculated in the prostate with luciferase-transfected human prostate cancer cells (PC3). Tumor volumes were allowed to reach 150-250 mm3 (as measured by ultrasound) following which, mice were randomized into (i) control (no intervention); (ii) HT alone; (iii) RT alone; and (iv) HT + RT. RF-induced HT was administered (Groups ii and iv) using the Oncotherm LAB EHY-100 device to achieve a target temperature of 41 °C in the prostate. RT was administered ~30 min following HT, using an image-guided small animal radiotherapy research platform. In each case, a dual arc plan was used to deliver 12 Gy to the target in a single fraction. One animal from each cohort was euthanized on Day 10 or 11 after treatment for caspase-9 and caspase-3 Western blot analysis. RESULTS: The inoculation success rate was 89%. Mean tumor size at randomization (~16 days post-inoculation) was ~189 mm3 . Following the administration of RT and HT, mean tumor doubling times in days were: control = 4.2; HT = 4.5; RT = 30.4; and HT + RT = 33.4. A significant difference (p = 0.036) was noted between normalized nadir volumes for the RT alone (0.76) and the HT + RT (0.40) groups. Increased caspase-3 expression was seen in the combination treatment group compared to the other treatment groups. CONCLUSION: These early results demonstrate the successful use of external mild HT as a localized radiosensitizer for deep-seated tumors. ADVANCES IN KNOWLEDGE: We successfully demonstrated the feasibility of administering external mild HT in an orthotopic tumor model and demonstrated preclinical proof-of-concept of HT-based localized radiosensitization in prostate cancer radiotherapy.
Collapse
Affiliation(s)
- Justin Cohen
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Akbar Anvari
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Santanu Samanta
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Yannick Poirier
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Sandrine Soman
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Allen Alexander
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Maida Ranjbar
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Ramilda Pavlovic
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Andrew Zodda
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Isabel L Jackson
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Javed Mahmood
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Zeljko Vujaskovic
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Amit Sawant
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
19
|
Dobšíček Trefná H, Schmidt M, van Rhoon GC, Kok HP, Gordeyev SS, Lamprecht U, Marder D, Nadobny J, Ghadjar P, Abdel-Rahman S, Kukiełka AM, Strnad V, Hurwitz MD, Vujaskovic Z, Diederich CJ, Stauffer PR, Crezee J. Quality assurance guidelines for interstitial hyperthermia. Int J Hyperthermia 2019; 36:277-294. [PMID: 30676101 DOI: 10.1080/02656736.2018.1564155] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Quality assurance (QA) guidelines are essential to provide uniform execution of clinical hyperthermia treatments and trials. This document outlines the clinical and technical consequences of the specific properties of interstitial heat delivery and specifies recommendations for hyperthermia administration with interstitial techniques. Interstitial hyperthermia aims at tumor temperatures in the 40-44 °C range as an adjunct to radiation or chemotherapy. The clinical part of this document imparts specific clinical experience of interstitial heat delivery to various tumor sites as well as recommended interstitial hyperthermia workflow and procedures. The second part describes technical requirements for quality assurance of current interstitial heating equipment including electromagnetic (radiative and capacitive) and ultrasound heating techniques. Detailed instructions are provided on characterization and documentation of the performance of interstitial hyperthermia applicators to achieve reproducible hyperthermia treatments of uniform high quality. Output power and consequent temperature rise are the key parameters for characterization of applicator performance in these QA guidelines. These characteristics determine the specific maximum tumor size and depth that can be heated adequately. The guidelines were developed by the ESHO Technical Committee with participation of senior STM members and members of the Atzelsberg Circle.
Collapse
Affiliation(s)
- H Dobšíček Trefná
- a Department of Electrical Engineering , Chalmers University of Technology , Göteborg , Sweden
| | - M Schmidt
- b Department of Radiation Oncology , University Hospital Erlangen , Erlangen , Germany
| | - G C van Rhoon
- c Department of Radiation Oncology , Erasmus MC Cancer Institute , Rotterdam , the Netherlands
| | - H P Kok
- d Department of Radiation Oncology, Cancer Center Amsterdam , Amsterdam UMC, University of Amsterdam , Amsterdam , the Netherlands
| | - S S Gordeyev
- e Department of Colorectal Oncology , N.N.Blokhin Russian Cancer Research Center , Moscow, Russia
| | - U Lamprecht
- f Radioonkologische Klinik , Universitätsklinikum Tübingen , Tübingen , Germany
| | - D Marder
- g Kantonsspital Aarau , Radio-Onkologie-Zentrum KSA-KSB , Aarau , Switzerland
| | - J Nadobny
- h Klinik für Radioonkologie und Strahlentherapie , Charité Universitätsmedizin Berlin , Berlin , Germany
| | - P Ghadjar
- h Klinik für Radioonkologie und Strahlentherapie , Charité Universitätsmedizin Berlin , Berlin , Germany
| | - S Abdel-Rahman
- i Klinikum der Universität München-Campus Grosshadern , München , Germany
| | - A M Kukiełka
- j Department of Radiation Oncology , Centrum Diagnostyki i Terapii Onkologicznej Nu-Med , Zamość , Poland
| | - V Strnad
- b Department of Radiation Oncology , University Hospital Erlangen , Erlangen , Germany
| | - M D Hurwitz
- k Department of Radiation Oncology , Thomas Jefferson University , Philadelphia , PA , USA
| | - Z Vujaskovic
- l Department of Radiation Oncology , University of Maryland Baltimore , Baltimore , MD , USA
| | - C J Diederich
- m Department of Radiation Oncology , University of California , San Francisco , CA , USA
| | - P R Stauffer
- k Department of Radiation Oncology , Thomas Jefferson University , Philadelphia , PA , USA
| | - J Crezee
- d Department of Radiation Oncology, Cancer Center Amsterdam , Amsterdam UMC, University of Amsterdam , Amsterdam , the Netherlands
| |
Collapse
|
20
|
Zhu L, Partanen A, Talcott MR, Gach HM, Greco SC, Henke LE, Contreras JA, Zoberi I, Hallahan DE, Chen H, Altman MB. Feasibility and safety assessment of magnetic resonance-guided high-intensity focused ultrasound (MRgHIFU)-mediated mild hyperthermia in pelvic targets evaluated using an in vivo porcine model. Int J Hyperthermia 2019; 36:1147-1159. [PMID: 31752562 PMCID: PMC7105895 DOI: 10.1080/02656736.2019.1685684] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 10/02/2019] [Accepted: 10/23/2019] [Indexed: 12/23/2022] Open
Abstract
Purpose: To evaluate the feasibility and assess safety parameters of magnetic resonance-guided high-intensity focused ultrasound (MRgHIFU)-mediated hyperthermia (HT; heating to 40-45 °C) in various pelvic targets in a porcine model in vivo.Methods: Thirteen HT treatments were performed in six pigs with a commercial MRgHIFU system (Sonalleve V2, Profound Medical Inc., Mississauga, Canada) to muscle adjacent to the ventral/dorsal bladder wall and uterus to administer 42 °C (±1°) for 30 min (±5%) using an 18-mm target diameter and 100 W power. Feasibility was assessed using accuracy, uniformity, and MR-thermometry performance-based metrics. Safety parameters were assessed for tissues in the targets and beam-path by contrast-enhanced MRI, gross-pathology and histopathology.Results: Across all HT sessions, the mean difference between average temperature (Tavg) and the target temperature within the target region-of-interest (tROI, the cross-section of the heated volume at focal depth) was 0.51 ± 0.33 °C. Within the tROI, the temperature standard deviation averaged 1.55 ± 0.31 °C, the average 30-min Tavg variation was 0.80 ± 0.17 °C, and the maximum difference between Tavg and the 10th- or 90th-percentile temperature averaged 2.01 ± 0.44 °C. The average time to reach ≥41 °C and cool to ≤40 °C within the tROI at the beginning and end of treatment was 47.25 ± 27.47 s and 66.37 ± 62.68 s, respectively. Compared to unheated controls, no abnormally-perfused tissue or permanent damage was evident in the MR images, gross pathology or histological analysis.Conclusions: MRgHIFU-mediated HT is feasible and safety assessment is satisfactory for treating an array of clinically-mimicking pelvic geometries in a porcine model in vivo, implying the technique may have utility in treating pelvic targets in human patients.
Collapse
Affiliation(s)
- Lifei Zhu
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, 63130, USA
| | - Ari Partanen
- Clinical Science, Profound Medical Inc., Mississauga, Ontario, Canada
| | - Michael R. Talcott
- Division of Comparative Medicine, Washington University in St. Louis, St. Louis, Missouri, 63110, USA
| | - H. Michael Gach
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, 63130, USA
- Department of Radiology, Washington University in St. Louis, St. Louis, Missouri, 63108, USA
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, Missouri, 63110, USA
| | - Suellen C. Greco
- Division of Comparative Medicine, Washington University in St. Louis, St. Louis, Missouri, 63110, USA
| | - Lauren E. Henke
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, Missouri, 63110, USA
| | - Jessika A. Contreras
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, Missouri, 63110, USA
| | - Imran Zoberi
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, Missouri, 63110, USA
| | - Dennis E. Hallahan
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, Missouri, 63110, USA
| | - Hong Chen
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, 63130, USA
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, Missouri, 63110, USA
| | - Michael B. Altman
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, 63130, USA
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, Missouri, 63110, USA
| |
Collapse
|
21
|
Ozhinsky E, Salgaonkar VA, Diederich CJ, Rieke V. MR thermometry-guided ultrasound hyperthermia of user-defined regions using the ExAblate prostate ablation array. J Ther Ultrasound 2018; 6:7. [PMID: 30123506 PMCID: PMC6088423 DOI: 10.1186/s40349-018-0115-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 07/26/2018] [Indexed: 01/21/2023] Open
Abstract
Background Hyperthermia therapy (HT) has shown to be an effective adjuvant to radiation, chemotherapy, and immunotherapy. In order to be safe and effective, delivery of HT requires maintenance of target tissue temperature within a narrow range (40-44 °C) for 30-60 min, which necessitates conformal heat delivery and accurate temperature monitoring. The goal of this project was to develop an MR thermometry-guided hyperthermia delivery platform based upon the ExAblate prostate array that would achieve uniform stable heating over large volumes within the prostate, while allowing the user to precisely control the power deposition patterns and shape of the region of treatment (ROT). Methods The HT platform incorporates an accelerated multi-slice real time MR thermometry pulse sequence and reconstruction pipeline. Temperature uniformity over a large contiguous area was achieved by multi-point temperature sampling with multi-focal feedback power control. The hyperthermia delivery system was based on an InSightec ExAblate 2100 prostate focused ultrasound ablation system, and HeartVista's RTHawk real-time MRI system integrated with a 3 T MRI scanner. The integrated system was evaluated in experiments with a tissue-mimicking phantom for prolonged exposures with a target temperature increase of 7 °C from baseline. Results Five various shapes of the region of treatment, defined on a 5 × 5 grid (35 × 35 mm, 11-25 focal spots per shape), were implemented to evaluate the performance of the system. MR temperature images, acquired after steady state was reached, showed different patterns of heating that closely matched the prescribed regions. Temperature uncertainty of the thermometry acquisition was 0.5 °C. The time to reach the target temperature (2:58-7:44 min) depended on the chosen ROT shape and on the distance from transducer to focal plane. Pre-cooling with circulating water helped to reduce near-field heating. Conclusions We have implemented a real-time MR thermometry-guided system for hyperthermia delivery within user-defined regions with the ExAblate prostate array and evaluated it in phantom experiments for different shapes and focal depths. Our results demonstrate the feasibility of using a commercially available endorectal FUS transducer to perform spatially-conformal hyperthermia therapy and could lead to a new set of exciting applications for these devices.
Collapse
Affiliation(s)
- Eugene Ozhinsky
- 1Department of Radiology and Biomedical Imaging, University of California San Francisco, 185 Berry Street, Suite 350, Box 0946, San Francisco, CA 94107 USA
| | - Vasant A Salgaonkar
- 2Department of Radiation Oncology, University of California San Francisco, 2340 Sutter Street, S331, Box 1708, San Francisco, CA 94115 USA
| | - Chris J Diederich
- 2Department of Radiation Oncology, University of California San Francisco, 2340 Sutter Street, S331, Box 1708, San Francisco, CA 94115 USA
| | - Viola Rieke
- 1Department of Radiology and Biomedical Imaging, University of California San Francisco, 185 Berry Street, Suite 350, Box 0946, San Francisco, CA 94107 USA
| |
Collapse
|
22
|
He MH, Chen L, Zheng T, Tu Y, He Q, Fu HL, Lin JC, Zhang W, Shu G, He L, Yuan ZX. Potential Applications of Nanotechnology in Urological Cancer. Front Pharmacol 2018; 9:745. [PMID: 30038573 PMCID: PMC6046453 DOI: 10.3389/fphar.2018.00745] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 06/19/2018] [Indexed: 01/16/2023] Open
Abstract
Nowadays, the potential scope of nanotechnology in uro-oncology (cancers of the prostate, bladder, and kidney) is broad, ranging from drug delivery, prevention, and diagnosis to treatment. Novel drug delivery methods using magnetic nanoparticles, gold nanoparticles, and polymeric nanoparticles have been investigated in prostate cancer. Additionally, renal cancer treatment may be profoundly influenced by applications of nanotechnology principles. Various nanoparticle-based strategies for kidney cancer therapy have been proposed. Partly due to the dilution of drug concentrations by urine production, causing inadequate drug delivery to tumor cells in the treatment of bladder cancer, various multifunctional bladder-targeted nanoparticles have been developed to enhance therapeutic efficiency. In each of these cancer research fields, nanotechnology has shown several advantages over widely used traditional methods. Different types of nanoparticles improve the solubility of poorly soluble drugs, and multifunctional nanoparticles have good specificity toward prostate, renal, and bladder cancer. Moreover, nanotechnology can also combine with other novel technologies to further enhance effectivity. As our understanding of nanotechnologies grows, additional opportunities to improve the diagnosis and treatment of urological cancer are excepted to arise. In this review, we focus on nanotechnologies with potential applications in urological cancer therapy and highlight clinical areas that would benefit from nanoparticle therapy.
Collapse
Affiliation(s)
- Ming-Hui He
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Li Chen
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Ting Zheng
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yu Tu
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Qian He
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Hua-Lin Fu
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Ju-Chun Lin
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Wei Zhang
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Gang Shu
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Lili He
- College of Pharmacy, Southwest Minzu University, Chengdu, China
| | - Zhi-Xiang Yuan
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
23
|
Abstract
Over the past few decades there is growing appreciation for the role of chemotherapy in treatment of prostate cancer. Initial successful phase III randomized trials in castration resistant prostate cancer (CRPC) have led to additional successful trials in earlier presentations of disease. Given the established role of radiation in management of locally advanced disease and demonstrated efficacy of taxanes in treatment of prostate cancer, optimal combination of radiation and chemotherapy in this patient population has garnered increased attention. Successful phase III trials in this space have given additional stimulus to further exploring combination therapy with radiation. New directions include assessment of additional chemotherapeutic agents including cabazitaxel and PARP inhibitors as well as personalization of therapy with use of genomic testing and other emerging markers to guide therapeutic decisions.
Collapse
Affiliation(s)
- Mark David Hurwitz
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
24
|
Wang HX, Yang Y, Guo H, Hou DD, Zheng S, Hong YX, Cai YF, Huo W, Qi RQ, Zhang L, Chen HD, Gao XH. HSPB1 deficiency sensitizes melanoma cells to hyperthermia induced cell death. Oncotarget 2018; 7:67449-67462. [PMID: 27626679 PMCID: PMC5341888 DOI: 10.18632/oncotarget.11894] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 08/22/2016] [Indexed: 12/04/2022] Open
Abstract
Hyperthermia has shown clinical potency as a single agent or as adjuvant to other therapies in cancer treatment. However, thermotolerance induced by thermosensitive genes such as the heat shock proteins can limit the efficacy of hyperthermic treatment. In the present study, we identified HSPB1 (HSP27) is hyperthermically inducible or endogenously highly expressed in both murine and human melanoma cell lines. We used a siRNA strategy to reduce HSPB1 levels and showed increased intolerance to hyperthermia via reduced cell viability and/or proliferation of cells. In the investigation of underlying mechanisms, we found knock down of HSPB1 further increased the proportion of apoptotic cells in hyperthermic treated melanoma cells when compared with either single agent alone, and both agents leaded to cell cycle arrest at G0/G1 or G2/M phases. We concluded that hyperthermia combined with silencing of HSPB1 enhanced cell death and resulted in failure to thrive in melanoma cell lines, implying the potential clinical utility of hyperthermia in combination with HSPB1 inhibition in cancer treatment.
Collapse
Affiliation(s)
- He-Xiao Wang
- Department of Dermatology, The First Hospital of China Medical University, Shenyang 110001, P.R. China
| | - Yang Yang
- Department of Dermatology, The First Hospital of China Medical University, Shenyang 110001, P.R. China
| | - Hao Guo
- Department of Dermatology, The First Hospital of China Medical University, Shenyang 110001, P.R. China
| | - Dian-Dong Hou
- Department of Dermatology, The First Hospital of China Medical University, Shenyang 110001, P.R. China
| | - Song Zheng
- Department of Dermatology, The First Hospital of China Medical University, Shenyang 110001, P.R. China
| | - Yu-Xiao Hong
- Department of Dermatology, The First Hospital of China Medical University, Shenyang 110001, P.R. China
| | - Yun-Fei Cai
- Department of Dermatology, The First Hospital of China Medical University, Shenyang 110001, P.R. China
| | - Wei Huo
- Department of Dermatology, The First Hospital of China Medical University, Shenyang 110001, P.R. China
| | - Rui-Qun Qi
- Department of Dermatology, The First Hospital of China Medical University, Shenyang 110001, P.R. China
| | - Li Zhang
- Department of Dermatology, The First Hospital of China Medical University, Shenyang 110001, P.R. China
| | - Hong-Duo Chen
- Department of Dermatology, The First Hospital of China Medical University, Shenyang 110001, P.R. China
| | - Xing-Hua Gao
- Department of Dermatology, The First Hospital of China Medical University, Shenyang 110001, P.R. China
| |
Collapse
|
25
|
Kouloulias V, Nikolakopoulou A, Karanasiou I, Antypas C, Armpilia C, Uzunoglou N. Documentation of a New Intracavitary Applicator for Transrectal Hyperthermia Designed for Prostate Cancer Cases: A Phantom Study. J Med Phys 2018; 43:141-145. [PMID: 29962693 PMCID: PMC6020625 DOI: 10.4103/jmp.jmp_67_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Concerning clinical trials, intracavitary hyperthermia has already shown antitumor activity and has a potential role in the treatment of prostate cancer. The aim of this study was to document a new intracavitary applicator operating at 433 MHz, designed for transrectal hyperthermia, as well as to assess the specific absorption rate (SAR) distributions in terms of temperature measurements in a soft-tissue phantom. The microwave applicator consists of a dipole-type λ/2, a reflector, and the cooling system. The applicator was placed into a soft-tissue gel-phantom box that was mimicking the dielectric properties of the normal tissue. A calibrated thermometer was implanted inside the phantom at specific locations, to calculate temperature distributions. The maximum value of the SAR was 108 W/kg on the surface's central area at the footprint of the antenna, while the penetration depth was at around 3 cm. Our experimental measurements confirmed the role of the reflector concerning the directivity in a certain area and non icotropic, by means of protecting normal tissues around the prostate. The SAR experimental measurements showed that our applicator might be used effectively as a treatment device for prostate cancer, demonstrating a clear advantage over other similar transrectal devices.
Collapse
Affiliation(s)
- Vassilis Kouloulias
- Department of Electrical and Computer Engineering, National Technical University of Athens, Athens, Greece,2nd Department of Radiology, Radiotherapy Unit, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Aggeliki Nikolakopoulou
- 1st Department of Radiology, Medical Physics Unit, Medical School, National and Kapodistrian University of Athens, Athens, Greece,Address for correspondence: Dr. Aggeliki Nikolakopoulou, 1st Department of Radiology, Aretaieion Hospital, Vas Sofias 76 Ave., Athens 11528, Greece. E-mail:
| | - Irene Karanasiou
- Department of Electrical and Computer Engineering, National Technical University of Athens, Athens, Greece
| | - Christos Antypas
- 1st Department of Radiology, Medical Physics Unit, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Christina Armpilia
- 1st Department of Radiology, Medical Physics Unit, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos Uzunoglou
- Department of Electrical and Computer Engineering, National Technical University of Athens, Athens, Greece
| |
Collapse
|
26
|
de Andrade Mello P, Bian S, Savio LEB, Zhang H, Zhang J, Junger W, Wink MR, Lenz G, Buffon A, Wu Y, Robson SC. Hyperthermia and associated changes in membrane fluidity potentiate P2X7 activation to promote tumor cell death. Oncotarget 2017; 8:67254-67268. [PMID: 28978031 PMCID: PMC5620171 DOI: 10.18632/oncotarget.18595] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 05/22/2017] [Indexed: 12/19/2022] Open
Abstract
Extracellular ATP (eATP) accumulation within the tumor microenvironment (TME) has the potential to activate purinergic signaling. The eATP evoked signaling effects bolster antitumor immune responses while exerting direct cytotoxicity on tumor cells and vascular endothelial cells, mediated at least in part through P2X7 receptors. Approaches to augment purinergic signaling in TME e.g. by ectonucleotidase CD39 blockade, and/or boosting P2X7 functional responses, might be used as immunomodulatory therapies in cancer treatment. In this study, we delineated the translatable strategy of hyperthermia to demonstrate impacts on P2X7 responsiveness to eATP. Hyperthermia (40°C) was noted to enhance eATP-mediated cytotoxicity on MCA38 colon cancer cells. Increased membrane fluidity induced by hyperthermia boosted P2X7 functionality, potentiating pore opening and modulating downstream AKT/PRAS40/mTOR signaling events. When combined with cisplatin or mitomycin C, hyperthermia and eATP together markedly potentiate cancer cell death. Our data indicate that clinically tolerable hyperthermia with modulated P2X7-purinergic signaling will boost efficacy of conventional cancer treatments.
Collapse
Affiliation(s)
- Paola de Andrade Mello
- Laboratório de Análises Bioquímicas e Citológicas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard University, Boston, MA, USA
| | - Shu Bian
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard University, Boston, MA, USA
- Department of Gastroenterology, Tianjin Union Medical Center, Tianjin, P.R. China
| | - Luiz Eduardo Baggio Savio
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard University, Boston, MA, USA
- Programa de Imunobiologia, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Haohai Zhang
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard University, Boston, MA, USA
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P.R. China
| | - Jingping Zhang
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard University, Boston, MA, USA
| | - Wolfgang Junger
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard University, Boston, MA, USA
| | - Márcia Rosângela Wink
- Laboratório de Biologia Celular, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - Guido Lenz
- Departamento de Biofísica e Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Andréia Buffon
- Laboratório de Análises Bioquímicas e Citológicas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Yan Wu
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard University, Boston, MA, USA
| | - Simon Christopher Robson
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard University, Boston, MA, USA
| |
Collapse
|
27
|
Peeken JC, Vaupel P, Combs SE. Integrating Hyperthermia into Modern Radiation Oncology: What Evidence Is Necessary? Front Oncol 2017; 7:132. [PMID: 28713771 PMCID: PMC5492395 DOI: 10.3389/fonc.2017.00132] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 06/06/2017] [Indexed: 12/13/2022] Open
Abstract
Hyperthermia (HT) is one of the hot topics that have been discussed over decades. However, it never made its way into primetime. The basic biological rationale of heat to enhance the effect of radiation, chemotherapeutic agents, and immunotherapy is evident. Preclinical work has confirmed this effect. HT may trigger changes in perfusion and oxygenation as well as inhibition of DNA repair mechanisms. Moreover, there is evidence for immune stimulation and the induction of systemic immune responses. Despite the increasing number of solid clinical studies, only few centers have included this adjuvant treatment into their repertoire. Over the years, abundant prospective and randomized clinical data have emerged demonstrating a clear benefit of combined HT and radiotherapy for multiple entities such as superficial breast cancer recurrences, cervix carcinoma, or cancers of the head and neck. Regarding less investigated indications, the existing data are promising and more clinical trials are currently recruiting patients. How do we proceed from here? Preclinical evidence is present. Multiple indications benefit from additional HT in the clinical setting. This article summarizes the present evidence and develops ideas for future research.
Collapse
Affiliation(s)
- Jan C Peeken
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München, München, Germany
| | - Peter Vaupel
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München, München, Germany
| | - Stephanie E Combs
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München, München, Germany.,Department of Radiation Sciences (DRS), Institute of Innovative Radiotherapy (iRT), Helmholtz Zentrum München, Neuherberg, Germany
| |
Collapse
|
28
|
Janati Esfahani A, Mahdavi SR, Shiran MB, Khoei S. The Role of Radiofrequency Hyperthermia in The Radiosensitization of A Human Prostate Cancer Cell Line. CELL JOURNAL 2017; 19:86-95. [PMID: 28580312 PMCID: PMC5448328 DOI: 10.22074/cellj.2017.4460] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Accepted: 10/17/2016] [Indexed: 11/24/2022]
Abstract
Objective This study evaluated enhanced induced DNA damages and apoptosis of a
spheroid culture of DU145 prostate cancer cells treated by a combination of radiofrequency
hyperthermia (RF HT) with radiation treatment (RT) from an external radiotherapy
machine compared to RT alone.
Materials and Methods In this experimental study, DU145 cells were cultured as spheroids
until they reached 300 µm in diameter. We exposed these cultures to either: RF HT for 90 minutes
at 43˚C originated from a Celsius TCS system, RF HT followed by RT at doses of 2 Gy
or 4 Gy (15 MV energy) with 15-minute interval, or RT alone at the above mentioned doses.
The trypan blue exclusion assay, alkaline comet assay, and annexin V/PI flow cytometry
were performed to measure cell viability, the amount of DNA damage in an individual cell
as the tail moment, and percentage of induced cell apoptosis in response to treatments
explained.
Results We calculated the thermal enhancement factor (TEF) for the combined treatment
regime. RF HT followed by the 4 Gy dose of RT resulted in minimum viability (85.33
± 1.30%), the highest tail moment (1.98 ± 0.18), and highest percentage of apoptotic cells
(64.48 ± 3.40%) compared to the other treatments. The results of the TEF assay were
2.54 from the comet assay and 2.33 according to flow cytometry.
Conclusion The present data suggest that combined treatment of mega voltage X-rays
and RF HT can result in significant radiosensitization of prostate cancer cells.
Collapse
Affiliation(s)
- Azam Janati Esfahani
- Department of Medical Physics, Iran University of Medical Sciences, Tehran, Iran
| | - Seied Rabi Mahdavi
- Radiation Biology Research Center, Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Samideh Khoei
- Department of Medical Physics, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
29
|
Mahdavi SR, Janati Esfahani A, Shiran MB, Khoei S, Estiri N. Enhanced DNA Damages of Human Prostate Cancer Cells Induced by Radiofrequency Capacitive Hyperthermia Pre- and Post X-rays: 6 MV versus 15 MV. CELL JOURNAL 2017; 19:79-85. [PMID: 28580311 PMCID: PMC5448316 DOI: 10.22074/cellj.2017.4749] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 12/31/2016] [Indexed: 01/18/2023]
Abstract
Objective This study aimed to determine the effect of 13.56 MHz radiofrequency (RF)
capacitive hyperthermia (HT) on radiosensivity of human prostate cancer cells pre and
post X-ray radiation treatment (RT).
Materials and Methods In this experimental study, the human prostate cancer cell line
DU145 was cultured as 300 µm diameter spheroids. We divided the spheroids into group I:
control, group II: HT at 43˚C for 30 minutes (HT), group III: 4 Gy irradiation with 6 MV X-ray [RT
(6 MV)], group IV: 4 Gy irradiation with 15 MV X-ray [RT (15 MV)], group V: HT+RT (6 MV),
group VI: HT+RT (15 MV), group VII: RT (6 MV)+HT, and group VIII: RT (15 MV)+HT. The alkaline
comet assay was used to assess DNA damages in terms of tail moment (TM). Thermal
enhancement factor (TEF) was obtained for the different treatment combinations.
Results Mean TM increased with increasing photon energy. Group II had significantly greater TM compared to group I. Groups III and IV also had significantly higher TM
compared to group I. Significant differences in TM existed between groups V, VII, and III
(P<0.05). We observed significant differences in TM between groups VI, VIII, and IV. TEF
values demonstrated that enhanced response to radiation was more pronounced in group
V compared to the other combined treatments.
Conclusion Our results suggest that HT applied before RT leads to higher radiosensivity
compared to after RT. HT at 43˚C for 30 minutes added to 6 MV X-ray causes higher
enhancement of radiation compared to 15 MV X-ray.
Collapse
Affiliation(s)
- Seied Rabi Mahdavi
- Radiation Biology Research Center, Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Azam Janati Esfahani
- Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Bagher Shiran
- Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Samideh Khoei
- Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Nader Estiri
- Department of Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
30
|
Huang H, Yu K, Mohammadi A, Karanthanasis E, Godley A, Yu JS. It's Getting Hot in Here: Targeting Cancer Stem-like Cells with Hyperthermia. JOURNAL OF STEM CELL AND TRANSPLANTATION BIOLOGY 2017; 2:113. [PMID: 30542674 PMCID: PMC6287629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Cancer stem-like cells (CSCs) are a subset of cancer cells that are resistant to conventional radiotherapy and chemotherapy. As such, CSCs have been recognized as playing a large role in tumor initiation and recurrence. Although hyperthermia is broadly used in cancer treatment either alone or in combination with radio- or chemo-therapy, its potential to target CSCs is not well understood. In this review, we discuss different types of hyperthermia and potential mechanisms of action in cancer treatment, particularly in regards to killing CSCs.
Collapse
Affiliation(s)
- Haidong Huang
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, NE30, Cleveland, OH 44195, USA
| | - Kevin Yu
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, NE30, Cleveland, OH 44195, USA
| | - Alireza Mohammadi
- Department of Neurosurgery, Rose Ella Burckhardt Brain tumor and Neuro-oncology Center, Neurological Institute, Cleveland Clinic, 9500 Euclid Avenue, CA51, Cleveland, OH 44195, USA,Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, 9500 Euclid Avenue, CA51, Cleveland, OH 44195, USA
| | - Efstathios Karanthanasis
- Department of Biomedical Engineering, Case Western Reserve University, 1900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Andrew Godley
- Department of Radiation Oncology, Taussig Cancer Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, CA50, OH 44195, USA
| | - Jennifer S. Yu
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, NE30, Cleveland, OH 44195, USA,Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, 9500 Euclid Avenue, CA51, Cleveland, OH 44195, USA,Department of Radiation Oncology, Taussig Cancer Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, CA50, OH 44195, USA,Corresponding Author: Jennifer S. Yu, Department of Radiation Oncology, Department of Stem Cell Biology and Regenerative Medicine, Cleveland Clinic, 9500 Euclid Avenue, CA50, Cleveland, OH 44195, USA, Tel: 216-445-9799; Fax: 216-445-1068,
| |
Collapse
|
31
|
Kukiełka AM, Hetnał M, Bereza K. Evaluation of tolerance and toxicity of high-dose-rate brachytherapy boost combined with interstitial hyperthermia for prostate cancer. Int J Hyperthermia 2016; 32:324-30. [PMID: 27056204 DOI: 10.3109/02656736.2015.1132339] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Purpose The aim of this retrospective study was to evaluate the tolerance and early as well as late toxicity of high dose rate brachytherapy (HDRBT) boost combined with interstitial hyperthermia (IHT) in patients treated for prostate cancer. Material and methods Between January 2011 and June 2013 76 patients diagnosed with prostate cancer received treatment consisting of external beam radiotherapy (EBRT), followed by a HDRBT boost combined with IHT. IHT was performed before each brachytherapy fraction. Results The median follow-up time was 26.3 months (range 7-43 months). Early genitourinary (GU) grade 1 and 2 toxicities were common, but only two patients (2.6%) experienced acute urinary retention requiring temporary catheterisation (grade 2 toxicity). No grade 3 or 4 genitourinary or gastrointestinal toxicities were observed. In the group analysed, 59 of 76 patients had follow-up times longer than 18 months. The incidence of grade 2 late toxicity in the group studied did not exceed 23.7%. There were no late grade 2 or higher complications from the gastrointestinal tract. Conclusions The tolerance of HDRBT boost combined with IHT is good. The profile and the percentage of early and late complications are acceptable.
Collapse
Affiliation(s)
- Andrzej Marek Kukiełka
- a Amethyst Radiotherapy Centre, Ludwik Rydygier Memorial Hospital , Kraków , Poland ;,b Department of Radiotherapy , Maria Skłodowska Curie Memorial Centre of Oncology , Kraków Division , Poland
| | - Marcin Hetnał
- a Amethyst Radiotherapy Centre, Ludwik Rydygier Memorial Hospital , Kraków , Poland
| | - Krzysztof Bereza
- a Amethyst Radiotherapy Centre, Ludwik Rydygier Memorial Hospital , Kraków , Poland ;,c Gynecology and Oncology Clinic , Collegium Medicum Jagiellonian University , Kraków , Poland
| |
Collapse
|
32
|
Jia R, Wang T, Jiang Q, Wang Z, Song C, Ding B. Self-Assembled DNA Nanostructures for Drug Delivery. CHINESE J CHEM 2016. [DOI: 10.1002/cjoc.201500838] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
33
|
Okada T, Niiyama E, Uto K, Aoyagi T, Ebara M. Inactivated Sendai Virus (HVJ-E) Immobilized Electrospun Nanofiber for Cancer Therapy. MATERIALS (BASEL, SWITZERLAND) 2015; 9:E12. [PMID: 28787810 PMCID: PMC5456544 DOI: 10.3390/ma9010012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 12/17/2015] [Accepted: 12/17/2015] [Indexed: 01/29/2023]
Abstract
Inactivated Hemagglutinating Virus of Japan Envelope (HVJ-E) was immobilized on electrospun nanofibers of poly(ε-caprolactone) by layer-by-layer (LbL) assembly technique. The precursor LbL film was first constructed with poly-L-lysine and alginic acid via electrostatic interaction. Then the HVJ-E particles were immobilized on the cationic PLL outermost surface. The HVJ-E adsorption was confirmed by surface wettability test, scanning laser microscopy, scanning electron microscopy, and confocal laser microscopy. The immobilized HVJ-E particles were released from the nanofibers under physiological condition. In vitro cytotoxic assay demonstrated that the released HVJ-E from nanofibers induced cancer cell deaths. This surface immobilization technique is possible to perform on anti-cancer drug incorporated nanofibers that enables the fibers to show chemotherapy and immunotherapy simultaneously for an effective eradication of tumor cells in vivo.
Collapse
Affiliation(s)
- Takaharu Okada
- Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki 305-8577, Japan.
- Biomaterials Unit, Nano-Life Field, International Center for Materials Nanoarchitectonics (WPI-MANA), National Institute for Materials Science (NIMS), 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan.
- Japan Society for the Promotion of Science (JSPS), 8 Ichibancho, Chiyoda-ku, Tokyo 102-0083, Japan.
| | - Eri Niiyama
- Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki 305-8577, Japan.
- Biomaterials Unit, Nano-Life Field, International Center for Materials Nanoarchitectonics (WPI-MANA), National Institute for Materials Science (NIMS), 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan.
| | - Koichiro Uto
- Biomaterials Unit, Nano-Life Field, International Center for Materials Nanoarchitectonics (WPI-MANA), National Institute for Materials Science (NIMS), 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan.
| | - Takao Aoyagi
- Biomaterials Unit, Nano-Life Field, International Center for Materials Nanoarchitectonics (WPI-MANA), National Institute for Materials Science (NIMS), 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan.
| | - Mitsuhiro Ebara
- Biomaterials Unit, Nano-Life Field, International Center for Materials Nanoarchitectonics (WPI-MANA), National Institute for Materials Science (NIMS), 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan.
- Graduate School of Tokyo University of Science, 6-3-1 Niijuku, Katsushika-ku, Tokyo 125-8585, Japan.
| |
Collapse
|
34
|
Hauser AK, Wydra RJ, Stocke NA, Anderson KW, Hilt JZ. Magnetic nanoparticles and nanocomposites for remote controlled therapies. J Control Release 2015; 219:76-94. [PMID: 26407670 PMCID: PMC4669063 DOI: 10.1016/j.jconrel.2015.09.039] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 09/19/2015] [Indexed: 12/17/2022]
Abstract
This review highlights the state-of-the-art in the application of magnetic nanoparticles (MNPs) and their composites for remote controlled therapies. Novel macro- to nano-scale systems that utilize remote controlled drug release due to actuation of MNPs by static or alternating magnetic fields and magnetic field guidance of MNPs for drug delivery applications are summarized. Recent advances in controlled energy release for thermal therapy and nanoscale energy therapy are addressed as well. Additionally, studies that utilize MNP-based thermal therapy in combination with other treatments such as chemotherapy or radiation to enhance the efficacy of the conventional treatment are discussed.
Collapse
Affiliation(s)
- Anastasia K Hauser
- Department of Chemical and Materials Engineering, University of Kentucky, Lexington, KY 40506, USA
| | - Robert J Wydra
- Department of Chemical and Materials Engineering, University of Kentucky, Lexington, KY 40506, USA
| | - Nathanael A Stocke
- Department of Chemical and Materials Engineering, University of Kentucky, Lexington, KY 40506, USA
| | - Kimberly W Anderson
- Department of Chemical and Materials Engineering, University of Kentucky, Lexington, KY 40506, USA
| | - J Zach Hilt
- Department of Chemical and Materials Engineering, University of Kentucky, Lexington, KY 40506, USA.
| |
Collapse
|
35
|
Bellissimo F, Pinzone MR, Cacopardo B, Nunnari G. Diagnostic and therapeutic management of hepatocellular carcinoma. World J Gastroenterol 2015; 21:12003-12021. [PMID: 26576088 PMCID: PMC4641121 DOI: 10.3748/wjg.v21.i42.12003] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 08/03/2015] [Accepted: 09/30/2015] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is an increasing health problem, representing the second cause of cancer-related mortality worldwide. The major risk factor for HCC is cirrhosis. In developing countries, viral hepatitis represent the major risk factor, whereas in developed countries, the epidemic of obesity, diabetes and nonalcoholic steatohepatitis contribute to the observed increase in HCC incidence. Cirrhotic patients are recommended to undergo HCC surveillance by abdominal ultrasounds at 6-mo intervals. The current diagnostic algorithms for HCC rely on typical radiological hallmarks in dynamic contrast-enhanced imaging, while the use of α-fetoprotein as an independent tool for HCC surveillance is not recommended by current guidelines due to its low sensitivity and specificity. Early diagnosis is crucial for curative treatments. Surgical resection, radiofrequency ablation and liver transplantation are considered the cornerstones of curative therapy, while for patients with more advanced HCC recommended options include sorafenib and trans-arterial chemo-embolization. A multidisciplinary team, consisting of hepatologists, surgeons, radiologists, oncologists and pathologists, is fundamental for a correct management. In this paper, we review the diagnostic and therapeutic management of HCC, with a focus on the most recent evidences and recommendations from guidelines.
Collapse
|
36
|
Bing C, Nofiele J, Staruch R, Ladouceur-Wodzak M, Chatzinoff Y, Ranjan A, Chopra R. Localised hyperthermia in rodent models using an MRI-compatible high-intensity focused ultrasound system. Int J Hyperthermia 2015; 31:813-22. [PMID: 26540488 DOI: 10.3109/02656736.2015.1094833] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
PURPOSE Localised hyperthermia in rodent studies is challenging due to the small target size. This study describes the development and characterisation of an MRI-compatible high-intensity focused ultrasound (HIFU) system to perform localised mild hyperthermia treatments in rodent models. MATERIAL AND METHODS The hyperthermia platform consisted of an MRI-compatible small animal HIFU system, focused transducers with sector-vortex lenses, a custom-made receive coil, and means to maintain systemic temperatures of rodents. The system was integrated into a 3T MR imager. Control software was developed to acquire images, process temperature maps, and adjust output power using a proportional-integral-derivative feedback control algorithm. Hyperthermia exposures were performed in tissue-mimicking phantoms and in a rodent model (n = 9). During heating, an ROI was assigned in the heated region for temperature control and the target temperature was 42 °C; 30 min mild hyperthermia treatment followed by a 10-min cooling procedure was performed on each animal. RESULTS 3D-printed sector-vortex lenses were successful at creating annular focal regions which enables customisation of the heating volume. Localised mild hyperthermia performed in rats produced a mean ROI temperature of 42.1 ± 0.3 °C. The T10 and T90 percentiles were 43.2 ± 0.4 °C and 41.0 ± 0.3 °C, respectively. For a 30-min treatment, the mean time duration between 41-45 °C was 31.1 min within the ROI. CONCLUSIONS The MRI-compatible HIFU system was successfully adapted to perform localised mild hyperthermia treatment in rodent models. A target temperature of 42 °C was well-maintained in a rat thigh model for 30 min.
Collapse
Affiliation(s)
- Chenchen Bing
- a Department of Radiology , University of Texas Southwestern Medical Center , Dallas , Texas
| | - Joris Nofiele
- a Department of Radiology , University of Texas Southwestern Medical Center , Dallas , Texas
| | - Robert Staruch
- a Department of Radiology , University of Texas Southwestern Medical Center , Dallas , Texas .,b Clinical Sites Research Program, Philips Research , Briarcliff Manor , New York
| | | | - Yonatan Chatzinoff
- c Applied Research Center, University of Texas at Dallas , Dallas , Texas
| | - Ashish Ranjan
- d Center of Veterinary Health Sciences, Oklahoma State University , Stillwater , Oklahoma , USA , and
| | - Rajiv Chopra
- a Department of Radiology , University of Texas Southwestern Medical Center , Dallas , Texas .,e Advanced Imaging Research Center, University of Texas Southwestern Medical Center , Dallas , Texas
| |
Collapse
|
37
|
Mallory M, Gogineni E, Jones GC, Greer L, Simone CB. Therapeutic hyperthermia: The old, the new, and the upcoming. Crit Rev Oncol Hematol 2015; 97:56-64. [PMID: 26315383 DOI: 10.1016/j.critrevonc.2015.08.003] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 07/09/2015] [Accepted: 08/04/2015] [Indexed: 01/12/2023] Open
Abstract
Hyperthermia has long been used for cancer treatment, either alone or in combination with chemotherapy, radiation therapy, or both. Its efficacy and versatility continue to be well demonstrated in randomized trials across a number of primary cancers, but barriers to its widespread adoption persist including effective delivery and verification systems. This article describes hyperthermia, details its biological mechanisms of action and immunological effects, and summarizes select preclinical data and key clinical trials combining hyperthermia with standard cancer treatments. Current challenges and emerging technologies that have the potential to make this translational therapy more accessible to a greater number of patients are also described.
Collapse
Affiliation(s)
- Matthew Mallory
- Virginia Commonwealth University, Internal Medicine, Richmond, VA 23298, USA.
| | - Emile Gogineni
- New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY 11568, USA.
| | - Guy C Jones
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, MD, 20892, USA.
| | - Lester Greer
- Department of Radiation Oncology, Fort Belvoir Community Hospital, 9300 DeWitt Loop Fort Belvoir, 22060, VA, USA.
| | - Charles B Simone
- Department of Radiation Oncology, University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia PA 19104, USA.
| |
Collapse
|
38
|
Müller AC, Zips D, Heinrich V, Lamprecht U, Voigt O, Burock S, Budach V, Wust P, Ghadjar P. Regional hyperthermia and moderately dose-escalated salvage radiotherapy for recurrent prostate cancer. Protocol of a phase II trial. Radiat Oncol 2015; 10:138. [PMID: 26152590 PMCID: PMC4493985 DOI: 10.1186/s13014-015-0442-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 06/20/2015] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Current studies on salvage radiotherapy (sRT) investigate timing, dose-escalation and anti-hormonal treatment (ADT) for recurrent prostate cancer. These approaches could either be limited by radiation-related susceptibility of the anastomosis or by suspected side-effects of long-term ADT. A phase II protocol was developed to investigate the benefit and tolerability of regional hyperthermia with moderately dose-escalated radiotherapy. METHODS The study hypothesis is that radio-thermotherapy is a safe and feasible salvage treatment modality. The primary endpoint is safety measured by frequency of grade 3+ genitourinary (GU) and gastrointestinal (GI) adverse events (AE) according to Common Toxicity Criteria (CTC) version 4. Feasibility is defined by number of hyperthermia treatments (n ≥ 7) and feasibility of radiotherapy according to protocol. Target volume delineation is performed according to the EORTC guidelines. Radiation treatment is administered with single doses of 2 Gy 5×/week to a total dose of 70 Gy. Regional hyperthermia is given 2×/week to a total of 10 treatments. RESULTS European centres participate in the phase II trial using intensity modulated RT (IMRT) or volumetric modulated arc technique (VMAT). The initiating centres were participants of the SAKK 09/10 study, where the same patient criteria and target volume definition (mandatory successful performed dummy run) were applied insuring a high standardisation of the study procedures. CONCLUSIONS The introduced phase II study implements highly precise image-guided radiotherapy and regional hyperthermia. If the phase II study is found to be safe and feasible, a multicenter phase III study is planned to test whether the addition of regional hyperthermia to dose-intensified sRT improves biochemical control.
Collapse
Affiliation(s)
- Arndt-Christian Müller
- Department of Radiation Oncology, Eberhard Karls University, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany.
| | - Daniel Zips
- Department of Radiation Oncology, Eberhard Karls University, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany.
| | - Vanessa Heinrich
- Department of Radiation Oncology, Eberhard Karls University, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany.
| | - Ulf Lamprecht
- Department of Radiation Oncology, Eberhard Karls University, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany.
| | - Otilia Voigt
- Department of Radiation Oncology, Eberhard Karls University, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany.
| | - Susen Burock
- Department of Radiation Oncology, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.
| | - Volker Budach
- Department of Radiation Oncology, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.
| | - Peter Wust
- Department of Radiation Oncology, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.
| | - Pirus Ghadjar
- Department of Radiation Oncology, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.
| |
Collapse
|
39
|
Datta NR, Ordóñez SG, Gaipl US, Paulides MM, Crezee H, Gellermann J, Marder D, Puric E, Bodis S. Local hyperthermia combined with radiotherapy and-/or chemotherapy: recent advances and promises for the future. Cancer Treat Rev 2015; 41:742-53. [PMID: 26051911 DOI: 10.1016/j.ctrv.2015.05.009] [Citation(s) in RCA: 290] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2015] [Revised: 05/16/2015] [Accepted: 05/20/2015] [Indexed: 02/08/2023]
Abstract
Hyperthermia, one of the oldest forms of cancer treatment involves selective heating of tumor tissues to temperatures ranging between 39 and 45°C. Recent developments based on the thermoradiobiological rationale of hyperthermia indicate it to be a potent radio- and chemosensitizer. This has been further corroborated through positive clinical outcomes in various tumor sites using thermoradiotherapy or thermoradiochemotherapy approaches. Moreover, being devoid of any additional significant toxicity, hyperthermia has been safely used with low or moderate doses of reirradiation for retreatment of previously treated and recurrent tumors, resulting in significant tumor regression. Recent in vitro and in vivo studies also indicate a unique immunomodulating prospect of hyperthermia, especially when combined with radiotherapy. In addition, the technological advances over the last decade both in hardware and software have led to potent and even safer loco-regional hyperthermia treatment delivery, thermal treatment planning, thermal dose monitoring through noninvasive thermometry and online adaptive temperature modulation. The review summarizes the outcomes from various clinical studies (both randomized and nonrandomized) where hyperthermia is used as a thermal sensitizer of radiotherapy and-/or chemotherapy in various solid tumors and presents an overview of the progresses in loco-regional hyperthermia. These recent developments, supported by positive clinical outcomes should merit hyperthermia to be incorporated in the therapeutic armamentarium as a safe and an effective addendum to the existing oncological treatment modalities.
Collapse
Affiliation(s)
- N R Datta
- Centre of Radiation Oncology, KSA-KSB, Kantonsspital Aarau, Aarau, Switzerland.
| | - S Gómez Ordóñez
- Centre of Radiation Oncology, KSA-KSB, Kantonsspital Aarau, Aarau, Switzerland.
| | - U S Gaipl
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany.
| | - M M Paulides
- Department of Radiation Oncology, Hyperthermia Unit, Erasmus MC Cancer Institute, Rotterdam, The Netherlands.
| | - H Crezee
- Department of Radiation Oncology, Academic Medical Centre, University of Amsterdam, The Netherlands.
| | - J Gellermann
- Praxis/Zentrum für Strahlentherapie und Radioonkologie, Janusz-Korczak-Str. 12, 12627 Berlin, Germany.
| | - D Marder
- Centre of Radiation Oncology, KSA-KSB, Kantonsspital Aarau, Aarau, Switzerland.
| | - E Puric
- Centre of Radiation Oncology, KSA-KSB, Kantonsspital Aarau, Aarau, Switzerland.
| | - S Bodis
- Centre of Radiation Oncology, KSA-KSB, Kantonsspital Aarau, Aarau, Switzerland; Department of Radiation Oncology, University Hospital Zurich, Switzerland.
| |
Collapse
|
40
|
Kudzia R, Nahajowski D, Kukiełka AM, Dąbrowski T, Dybek D, Brandys P, Waligórski MPR. Does microwave interstitial hyperthermia prior to high-dose-rate brachytherapy change prostate volume or therapy plan parameters? Int J Hyperthermia 2015; 31:568-73. [PMID: 25885417 DOI: 10.3109/02656736.2015.1033481] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
PURPOSE In this prospective preliminary study we evaluated changes of prostate volume and changes of brachytherapy treatment plan parameters due to interstitial hyperthermia (IHT) applied prior to high-dose-rate brachytherapy (HDRBT), compared to our standard HDRBT procedure. MATERIAL AND METHODS In a group of 60 consecutive patients with prostate adenocarcinoma, 30 were treated with HDRBT alone and 30 with IHT preceding HDRBT. Prior to catheter implantation, a 'virtual' treatment plan (VP) was complied, a 'live' plan (LP) was prepared before patient irradiation, and a 'post' plan (PP) was drawn up after completing the irradiation procedure. In each plan, based on transrectal ultrasound images, the contours of the prostate, urethra, and rectum were delineated and the respective volumes and dose-volume histogram parameters were evaluated. These parameters, established for the LP, were then compared with those of the PP. RESULTS Changes in prostate volume and in parameters of the treatment plans were observed, but differences between the two patient groups were not statistically significant. For all 60 patients treated, the average prostate volume in the VP was 32 cm(3), in the LP 41 cm(3), and the PP 43 cm(3). Average values of relative changes in the therapy planning parameters between LP and PP were for the prostate D90 -5.7%, V100 -5.6%, V200 -13.2%, for the urethra D0, 1 cm(3) -1.6%, and for rectum D2 cm(3) 0%. CONCLUSION Hyperthermia prior to HDRBT does not significantly change the volume of the prostate and there is no need to perform the new treatment plan after the hyperthermia session.
Collapse
Affiliation(s)
- Roksana Kudzia
- Department of Medical Physics, Maria Skłodowska Curie Memorial Centre of Oncology , Kraków Division , Kraków
| | | | | | | | | | | | | |
Collapse
|
41
|
Li D, Kang J, Golas BJ, Yeung VW, Madoff DC. Minimally invasive local therapies for liver cancer. Cancer Biol Med 2015; 11:217-36. [PMID: 25610708 PMCID: PMC4296086 DOI: 10.7497/j.issn.2095-3941.2014.04.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 10/28/2014] [Indexed: 12/11/2022] Open
Abstract
Primary and metastatic liver tumors are an increasing global health problem, with hepatocellular carcinoma (HCC) now being the third leading cause of cancer-related mortality worldwide. Systemic treatment options for HCC remain limited, with Sorafenib as the only prospectively validated agent shown to increase overall survival. Surgical resection and/or transplantation, locally ablative therapies and regional or locoregional therapies have filled the gap in liver tumor treatments, providing improved survival outcomes for both primary and metastatic tumors. Minimally invasive local therapies have an increasing role in the treatment of both primary and metastatic liver tumors. For patients with low volume disease, these therapies have now been established into consensus practice guidelines. This review highlights technical aspects and outcomes of commonly utilized, minimally invasive local therapies including laparoscopic liver resection (LLR), radiofrequency ablation (RFA), microwave ablation (MWA), high-intensity focused ultrasound (HIFU), irreversible electroporation (IRE), and stereotactic body radiation therapy (SBRT). In addition, the role of combination treatment strategies utilizing these minimally invasive techniques is reviewed.
Collapse
Affiliation(s)
- David Li
- 1 Department of Radiology, Division of Interventional Radiology, New York-Presbyterian Hospital/Weill Cornell Medical Center, New York, NY 10065, USA ; 2 Department of Medicine, NYU Langone Medical Center, New York, NY 10016, USA ; 3 Flushing Radiation Oncology Services, Flushing, New York, NY 11354, USA ; 4 Department of Surgery, Division of Surgical Oncology, New York-Presbyterian Hospital/Weill Cornell Medical Center New York, New York, NY 10065, USA
| | - Josephine Kang
- 1 Department of Radiology, Division of Interventional Radiology, New York-Presbyterian Hospital/Weill Cornell Medical Center, New York, NY 10065, USA ; 2 Department of Medicine, NYU Langone Medical Center, New York, NY 10016, USA ; 3 Flushing Radiation Oncology Services, Flushing, New York, NY 11354, USA ; 4 Department of Surgery, Division of Surgical Oncology, New York-Presbyterian Hospital/Weill Cornell Medical Center New York, New York, NY 10065, USA
| | - Benjamin J Golas
- 1 Department of Radiology, Division of Interventional Radiology, New York-Presbyterian Hospital/Weill Cornell Medical Center, New York, NY 10065, USA ; 2 Department of Medicine, NYU Langone Medical Center, New York, NY 10016, USA ; 3 Flushing Radiation Oncology Services, Flushing, New York, NY 11354, USA ; 4 Department of Surgery, Division of Surgical Oncology, New York-Presbyterian Hospital/Weill Cornell Medical Center New York, New York, NY 10065, USA
| | - Vincent W Yeung
- 1 Department of Radiology, Division of Interventional Radiology, New York-Presbyterian Hospital/Weill Cornell Medical Center, New York, NY 10065, USA ; 2 Department of Medicine, NYU Langone Medical Center, New York, NY 10016, USA ; 3 Flushing Radiation Oncology Services, Flushing, New York, NY 11354, USA ; 4 Department of Surgery, Division of Surgical Oncology, New York-Presbyterian Hospital/Weill Cornell Medical Center New York, New York, NY 10065, USA
| | - David C Madoff
- 1 Department of Radiology, Division of Interventional Radiology, New York-Presbyterian Hospital/Weill Cornell Medical Center, New York, NY 10065, USA ; 2 Department of Medicine, NYU Langone Medical Center, New York, NY 10016, USA ; 3 Flushing Radiation Oncology Services, Flushing, New York, NY 11354, USA ; 4 Department of Surgery, Division of Surgical Oncology, New York-Presbyterian Hospital/Weill Cornell Medical Center New York, New York, NY 10065, USA
| |
Collapse
|
42
|
Kulkarni VM, Bodas D, Paknikar KM. Lanthanum strontium manganese oxide (LSMO) nanoparticles: a versatile platform for anticancer therapy. RSC Adv 2015. [DOI: 10.1039/c5ra02731d] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Multiple uses of LSMO nanoparticles in anticancer therapy.
Collapse
Affiliation(s)
| | - Dhananjay Bodas
- Center for Nanobioscience
- Agharkar Research Institute
- Pune 411004
- India
| | | |
Collapse
|
43
|
Salgaonkar VA, Prakash P, Rieke V, Ozhinsky E, Plata J, Kurhanewicz J, Hsu ICJ, Diederich CJ. Model-based feasibility assessment and evaluation of prostate hyperthermia with a commercial MR-guided endorectal HIFU ablation array. Med Phys 2014; 41:033301. [PMID: 24593742 DOI: 10.1118/1.4866226] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
PURPOSE Feasibility of targeted and volumetric hyperthermia (40-45 °C) delivery to the prostate with a commercial MR-guided endorectal ultrasound phased array system, designed specifically for thermal ablation and approved for ablation trials (ExAblate 2100, Insightec Ltd.), was assessed through computer simulations and tissue-equivalent phantom experiments with the intention of fast clinical translation for targeted hyperthermia in conjunction with radiotherapy and chemotherapy. METHODS The simulations included a 3D finite element method based biothermal model, and acoustic field calculations for the ExAblate ERUS phased array (2.3 MHz, 2.3 × 4.0 cm(2), ∼1000 channels) using the rectangular radiator method. Array beamforming strategies were investigated to deliver protracted, continuous-wave hyperthermia to focal prostate cancer targets identified from representative patient cases. Constraints on power densities, sonication durations and switching speeds imposed by ExAblate hardware and software were incorporated in the models. Preliminary experiments included beamformed sonications in tissue mimicking phantoms under MR temperature monitoring at 3 T (GE Discovery MR750W). RESULTS Acoustic intensities considered during simulation were limited to ensure mild hyperthermia (Tmax < 45 °C) and fail-safe operation of the ExAblate array (spatial and time averaged acoustic intensity ISATA < 3.4 W/cm(2)). Tissue volumes with therapeutic temperature levels (T > 41 °C) were estimated. Numerical simulations indicated that T > 41 °C was calculated in 13-23 cm(3) volumes for sonications with planar or diverging beam patterns at 0.9-1.2 W/cm(2), in 4.5-5.8 cm(3) volumes for simultaneous multipoint focus beam patterns at ∼0.7 W/cm(2), and in ∼6.0 cm(3) for curvilinear (cylindrical) beam patterns at 0.75 W/cm(2). Focused heating patterns may be practical for treating focal disease in a single posterior quadrant of the prostate and diffused heating patterns may be useful for heating quadrants, hemigland volumes or even bilateral targets. Treatable volumes may be limited by pubic bone heating. Therapeutic temperatures were estimated for a range of physiological parameters, sonication duty cycles and rectal cooling. Hyperthermia specific phasing patterns were implemented on the ExAblate prostate array and continuous-wave sonications (∼0.88 W/cm(2), 15 min) were performed in tissue-mimicking material with real-time MR-based temperature imaging (PRFS imaging at 3.0 T). Shapes of heating patterns observed during experiments were consistent with simulations. CONCLUSIONS The ExAblate 2100, designed specifically for thermal ablation, can be controlled for delivering continuous hyperthermia in prostate while working within operational constraints.
Collapse
Affiliation(s)
- Vasant A Salgaonkar
- Thermal Therapy Research Group, Radiation Oncology, University of California San Francisco, 1600 Divisadero Street, Suite H-1031, San Francisco, California 94143
| | - Punit Prakash
- Department of Electrical and Computer Engineering, Kansas State University, 2077 Rathbone Hall, Manhattan, Kansas 66506
| | - Viola Rieke
- Department of Radiology and Biomedical Imaging, University of California San Francisco, 505 Parnassus Avenue, San Francisco, California 94143
| | - Eugene Ozhinsky
- Department of Radiology and Biomedical Imaging, University of California San Francisco, 505 Parnassus Avenue, San Francisco, California 94143
| | - Juan Plata
- Department of Radiology, Stanford University, 1201 Welch Road, Stanford, California 94305
| | - John Kurhanewicz
- Department of Radiology and Biomedical Imaging, University of California San Francisco, 505 Parnassus Avenue, San Francisco, California 94143
| | - I-C Joe Hsu
- Thermal Therapy Research Group, Radiation Oncology, University of California San Francisco, 1600 Divisadero Street, Suite H-1031, San Francisco, California 94143
| | - Chris J Diederich
- Thermal Therapy Research Group, Radiation Oncology, University of California San Francisco, 1600 Divisadero Street, Suite H-1031, San Francisco, California 94143
| |
Collapse
|
44
|
Hurwitz M, Stauffer P. Hyperthermia, radiation and chemotherapy: the role of heat in multidisciplinary cancer care. Semin Oncol 2014; 41:714-29. [PMID: 25499632 DOI: 10.1053/j.seminoncol.2014.09.014] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The compelling biologic basis for combining hyperthermia with modern cancer therapies including radiation and chemotherapy was first appreciated nearly half a century ago. Hyperthermia complements radiation as conditions contributing to radio-resistance generally enhance sensitivity to heat and sensitizing effects occur through increased perfusion/tumor oxygenation and alteration of cellular death pathways. Chemosensitization with hyperthermia is dependent on the particular mechanism of effect for each agent with synergistic effects noted for several commonly used agents. Clinically, randomized trials have demonstrated benefit including survival with the addition of hyperthermia to radiation or chemotherapy in treatment of a wide range of malignancies. Improvements in treatment delivery techniques, streamlined logistics, and greater understanding of the relationship of thermal dosimetry to treatment outcomes continue to facilitate wider clinical implementation. Evolving applications include thermal enhancement of immunotherapy, targeted drug delivery and application of principals of thermal biology towards integration of thermal ablation into multimodality oncologic care.
Collapse
Affiliation(s)
- Mark Hurwitz
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA.
| | - Paul Stauffer
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA
| |
Collapse
|
45
|
Sun J, Guo M, Pang H, Qi J, Zhang J, Ge Y. Treatment of malignant glioma using hyperthermia. Neural Regen Res 2014; 8:2775-82. [PMID: 25206588 PMCID: PMC4145998 DOI: 10.3969/j.issn.1673-5374.2013.29.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Accepted: 07/20/2013] [Indexed: 12/28/2022] Open
Abstract
Thirty pathologically diagnosed patients with grade III–IV primary or recurrent malignant glioma (tumor diameter 3–7 cm) were randomly divided into two groups. The control group underwent conventional radiotherapy and chemotherapy. In the hyperthermia group, primary cases received hyperthermia treatment, and patients with recurrent tumors were treated with hyperthermia in com-bination with radiotherapy and chemotherapy. Hyperthermia treatment was administered using a 13.56-MHz radio frequency hyperthermia device. Electrodes were inserted into the tumor with the aid of a CT-guided stereotactic apparatus and heat was applied for 1 hour. During 3 months after hyperthermia, patients were evaluated with head CT or MRI every month. Gliomas in the hyper-thermia group exhibited growth retardation or growth termination. Necrosis was evident in 80% of the heated tumor tissue and there was a decrease in tumor diameter. Our findings indicate that ra-dio frequency hyperthermia has a beneficial effect in the treatment of malignant glioma.
Collapse
Affiliation(s)
- Jiahang Sun
- Department of Neurosurgery, the Second Affiliated Hospital of Harbin Medical University, Harbin 150000, Heilongjiang Province, China
| | - Mian Guo
- Department of Neurosurgery, the Second Affiliated Hospital of Harbin Medical University, Harbin 150000, Heilongjiang Province, China
| | - Hengyuan Pang
- Department of Neurosurgery, the Second Affiliated Hospital of Harbin Medical University, Harbin 150000, Heilongjiang Province, China
| | - Jingtao Qi
- Department of Neurosurgery, the Second Affiliated Hospital of Harbin Medical University, Harbin 150000, Heilongjiang Province, China
| | - Jinwei Zhang
- Department of Neurosurgery, the Second Affiliated Hospital of Harbin Medical University, Harbin 150000, Heilongjiang Province, China
| | - Yunlong Ge
- Department of Neurosurgery, the Second Affiliated Hospital of Harbin Medical University, Harbin 150000, Heilongjiang Province, China
| |
Collapse
|
46
|
Zaorsky NG, Den RB, Doyle LA, Dicker AP, Hurwitz MD. Combining theoretical potential and advanced technology in high-dose rate brachytherapy boost therapy for prostate cancer. Expert Rev Med Devices 2014; 10:751-63. [PMID: 24195459 DOI: 10.1586/17434440.2013.841347] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
External beam radiation therapy (EBRT) combined with brachytherapy (BT) is an attractive treatment option for select patients with clinically localized prostate cancer. Either low- or high-dose rate BT may be combined with EBRT ('LDR-BT boost,' 'HDR-BT boost,' respectively). HDR-BT boost has potential theoretical benefits over LDR-BT boost or external beam radiation therapy monotherapy in terms of radiobiology, radiophysics and patient convenience. Based on prospective studies in this review, freedom from biochemical failure (FFBF) rates at 5 years for low-, intermediate- and high-risk patients have generally been 85-100%, 68-97%, 63-85%, respectively; late Radiotherapy and Oncology Group Grades 3 and 4 genitourinary and gastrointestinal toxicities are seen in <8% of patients. HDR-BT boost is now a relatively well-established treatment modality for certain intermediate-risk and high-risk prostate cancer patients, though limitations exist in drawing conclusions from the currently published studies.
Collapse
Affiliation(s)
- Nicholas G Zaorsky
- Department of Radiation Oncology, Kimmel Cancer Center, Jefferson Medical College of Thomas Jefferson University, Philadelphia, PA, USA
| | | | | | | | | |
Collapse
|
47
|
Li D, Kang J, Madoff DC. Locally ablative therapies for primary and metastatic liver cancer. Expert Rev Anticancer Ther 2014; 14:931-45. [PMID: 24746315 DOI: 10.1586/14737140.2014.911091] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Locally ablative therapies have an increasing role in the effective multidisciplinary approach towards the treatment of both primary and metastatic liver tumors. In patients who are not considered surgical candidates and have low volume disease, these therapies have now become established into consensus practice guidelines. A large range of therapeutic options exist including percutaneous ethanol injection (PEI), radiofrequency ablation (RFA), microwave ablation (MWA), cryoablation, percutaneous laser ablation (PLA), irreversible electroporation (IRE), stereotactic body radiation therapy (SBRT) and high intensity focused ultrasound (HIFU); each having benefits and drawbacks. The greatest body of evidence supporting clinical utility in the liver currently exists for RFA, with PEI having fallen out of favor. MWA, IRE, SBRT and HIFU are relatively nascent technologies, and outcomes data supporting their use is promising. Future directions of ablative therapies include tandem approaches to improve efficacy in the treatment of liver tumors.
Collapse
Affiliation(s)
- David Li
- Department of Radiology, Division of Interventional Radiology, New York-Presbyterian Hospital/Weill Cornell Medical Center, New York, NY, USA
| | | | | |
Collapse
|
48
|
Schlesinger D, Benedict S, Diederich C, Gedroyc W, Klibanov A, Larner J. MR-guided focused ultrasound surgery, present and future. Med Phys 2014; 40:080901. [PMID: 23927296 DOI: 10.1118/1.4811136] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
MR-guided focused ultrasound surgery (MRgFUS) is a quickly developing technology with potential applications across a spectrum of indications traditionally within the domain of radiation oncology. Especially for applications where focal treatment is the preferred technique (for example, radiosurgery), MRgFUS has the potential to be a disruptive technology that could shift traditional patterns of care. While currently cleared in the United States for the noninvasive treatment of uterine fibroids and bone metastases, a wide range of clinical trials are currently underway, and the number of publications describing advances in MRgFUS is increasing. However, for MRgFUS to make the transition from a research curiosity to a clinical standard of care, a variety of challenges, technical, financial, clinical, and practical, must be overcome. This installment of the Vision 20∕20 series examines the current status of MRgFUS, focusing on the hurdles the technology faces before it can cross over from a research technique to a standard fixture in the clinic. It then reviews current and near-term technical developments which may overcome these hurdles and allow MRgFUS to break through into clinical practice.
Collapse
Affiliation(s)
- David Schlesinger
- Department of Radiation Oncology, University of Virginia, Charlottesville, Virginia 22908, USA.
| | | | | | | | | | | |
Collapse
|
49
|
Kok HP, Crezee J, Franken NA, Stalpers LJ, Barendsen GW, Bel A. Quantifying the Combined Effect of Radiation Therapy and Hyperthermia in Terms of Equivalent Dose Distributions. Int J Radiat Oncol Biol Phys 2014; 88:739-45. [DOI: 10.1016/j.ijrobp.2013.11.212] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Revised: 11/08/2013] [Accepted: 11/10/2013] [Indexed: 01/13/2023]
|
50
|
Devun F, Biau J, Huerre M, Croset A, Sun JS, Denys A, Dutreix M. Colorectal cancer metastasis: the DNA repair inhibitor Dbait increases sensitivity to hyperthermia and improves efficacy of radiofrequency ablation. Radiology 2013; 270:736-46. [PMID: 24475822 DOI: 10.1148/radiol.13130805] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE To assess the usefulness of combining hyperthermia with a DNA repair inhibitor (double-strand break bait [Dbait]) and its potential application to radiofrequency ablation (RFA) in a preclinical model of human colorectal cancer. MATERIALS AND METHODS The local ethics committee of animal experimentation approved all investigations. First, the relevance was assessed by studying the survival of four human colorectal adenocarcinoma cell cultures after 1 hour of hyperthermia at 41°C or 43°C with or without Dbait. Human colon adenocarcinoma cells (HT-29) were grafted subcutaneously into nude mice (n = 111). When tumors reached approximately 500 mm(3), mice were treated with Dbait alone (n = 20), sublethal RFA (n = 21), three different Dbait schemes and sublethal RFA (n = 52), or a sham treatment (n = 18). RFA was performed to ablate the tumor center alone. To elucidate antitumor mechanisms, 39 mice were sacrificed for blinded pathologic analysis, including assessment of DNA damage, cell proliferation, and tumor necrosis. Others were monitored for tumor growth and survival. Analyses of variance and log-rank tests were used to evaluate differences. RESULTS When associated with mild hyperthermia, Dbait induced cytotoxicity in all tested colon cancer cell lines. Sublethal RFA or Dbait treatment alone moderately improved survival (median, 40 days vs 28 days for control; P = .0005) but combination treatment significantly improved survival (median, 84 days vs 40 days for RFA alone, P = .0004), with approximately half of the animals showing complete tumor responses. Pathologic studies showed that the Dbait and RFA combination strongly enhances DNA damage and coagulation areas in tumors. CONCLUSION Combining Dbait with RFA sensitizes the tumor periphery to mild hyperthermia and increases RFA antitumor efficacy.
Collapse
Affiliation(s)
- Flavien Devun
- From the Department of Research, Institut Curie, Bat 11215 Georges Clémenceau, Orsay 91405, France (F.D., M.H., A.C., M.D.); CNRS, Unit UMR3347, Orsay, France (M.D.); INSERM, Unit U1021, Orsay, France (M.D.); DNA Therapeutics, Evry, France (F.D., A.C., J.S.S.); Department of Radiotherapy, Centre Jean Perrin, Clermont-Ferrand, France (J.B.); Department of Pathology, Institut Pasteur, Paris, France (M.H.); Muséum National d'Histoire Naturelle USM503, Paris, France (J.S.S.); Université Paris-Sud, Paris, France (M.D.); and Department of Radiology and Interventional Radiology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland (A.D.)
| | | | | | | | | | | | | |
Collapse
|