51
|
Ren G, Ju H, Wu Y, Song H, Ma X, Ge M, Qiu W, Chen Y, He Y, Zhuang Q, Meng J, Guo W. A multicenter randomized phase II trial of hyperthermia combined with TPF induction chemotherapy compared with TPF induction chemotherapy in locally advanced resectable oral squamous cell carcinoma. Int J Hyperthermia 2021; 38:939-947. [PMID: 34134574 DOI: 10.1080/02656736.2021.1937714] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Hyperthermia has been reported to cause cancer stage regression, thus providing surgical opportunities in patients with unresectable tumors and improving the quality of life of patients by preserving certain organs. METHODS A prospective open-label phase II trial was conducted to evaluate the efficacy of hyperthermia combined with induction chemotherapy in patients with locally advanced resectable oral squamous cell carcinoma (OSCC). Patients received hyperthermia combined with two cycles of 5-fluorouracil, cisplatin, and docetaxel (TPF) induction chemotherapy regimens or TPF induction chemotherapy alone, followed by radical surgery with postoperative radiotherapy. The primary endpoint was the clinical response rate of the induction chemotherapy. The secondary endpoints were overall survival (OS), disease-free survival (DFS), and toxicity. RESULTS A total of 120 patients were enrolled, and 115 patients were included in the clinical response analysis. The clinical response rate was significantly higher in the experimental arm than in the control arm (65.45% vs. 40.00%, p = 0.0088). There were no unexpected toxicities, and hyperthermia and induction chemotherapy did not increase the perioperative morbidity rate. Moreover, there was a significant improvement in DFS, but no significant difference in OS between the two arms. In the subgroup analysis, increased OS and DFS rates were associated with patients with favorable clinical response after induction chemotherapy in the total population, experimental arm, and control arm. CONCLUSIONS Our study demonstrates that hyperthermia combined with induction chemotherapy is associated with a high response rate and provides a new treatment option for patients with resectable stage III or IVA OSCC.
Collapse
Affiliation(s)
- Guoxin Ren
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, China
| | - Houyu Ju
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, China
| | - Yunteng Wu
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, China
| | - Hao Song
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, China
| | - Xuhui Ma
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, China
| | - Minghua Ge
- Department of Head and Neck Surgery, Zhejiang Cancer Hospital, Hangzhou, China
| | - Weiliu Qiu
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, China
| | - Yazhu Chen
- Biomedical Instrument Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai Med-X Engineering Center for Medical Equipment and Technology, Shanghai Jiao Tong University, Shanghai, China
| | - Yue He
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, China
| | - Qianwei Zhuang
- Department of Oromaxillofacial Head and Neck Surgery, College of Medicine, Affiliated Xuzhou Hospital, Southeast University, Xuzhou, China
| | - Jian Meng
- Department of Oromaxillofacial Head and Neck Surgery, College of Medicine, Affiliated Xuzhou Hospital, Southeast University, Xuzhou, China
| | - Wei Guo
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, China
| |
Collapse
|
52
|
Wu CC, Hsu YT, Chang CL. Hyperthermic intraperitoneal chemotherapy enhances antitumor effects on ovarian cancer through immune-mediated cancer stem cell targeting. Int J Hyperthermia 2021; 38:1013-1022. [PMID: 34192990 DOI: 10.1080/02656736.2021.1945688] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
PURPOSE We aimed to determine the effects and possible mechanisms of hyperthermic intraperitoneal chemotherapy (HIPEC) in targeting ovarian cancer stem-like cells (CSCs). METHODS Murine ovarian cancer cell lines presenting CSC surface markers were grown intraperitoneally in both immunocompetent and immunodeficient mice, which were then treated by intraperitoneal hyperthermia with the chemotherapeutic agents: paclitaxel and cisplatin. Tumor growth was measured by non-invasive luminescent imaging. Intraperitoneal immune cells, such as CD4+, CD8+ T cells, macrophages, and dendritic cells, were evaluated through flow cytometry analysis. RESULTS Combined hyperthermia and chemotherapy exhibited an efficient therapeutic effect in the immunocompetent mice. However, a similar effect was not observed in the immunodeficient mice. Intraperitoneal hyperthermia increased the number of Intraperitoneal macrophages and dendritic cells that were lost due to chemotherapy. Compared with ovarian cancer bulk cells, CSCs were more susceptible to phagocytosis by macrophages. CONCLUSION We demonstrated that the superior therapeutic efficacy and reduced proportion of CSCs associated with intraperitoneal hyperthermic chemotherapy were immune-related. Hyperthermia recruits the phagocytes that target surviving CSCs after chemotherapy. These results provide a novel mechanism for the efficacy of HIPEC in treating ovarian cancer.
Collapse
Affiliation(s)
- Chao-Chih Wu
- Departmental of Medical Research, MacKay Memorial Hospital, Taipei City, Taiwan.,MacKay Junior College of Medicine, Nursing, and Management, New Taipei City, Taiwan
| | - Yun-Ting Hsu
- Departmental of Medical Research, MacKay Memorial Hospital, Taipei City, Taiwan.,MacKay Junior College of Medicine, Nursing, and Management, New Taipei City, Taiwan
| | - Chih-Long Chang
- Departmental of Medical Research, MacKay Memorial Hospital, Taipei City, Taiwan.,Department of Obstetrics and Gynecology, MacKay Memorial Hospital, Taipei City, Taiwan.,Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
| |
Collapse
|
53
|
Hannon G, Tansi FL, Hilger I, Prina‐Mello A. The Effects of Localized Heat on the Hallmarks of Cancer. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202000267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Gary Hannon
- Nanomedicine and Molecular Imaging Group Trinity Translational Medicine Institute Dublin 8 Ireland
- Laboratory of Biological Characterization of Advanced Materials (LBCAM), Trinity Translational Medicine Institute Trinity College Dublin Dublin 8 Ireland
| | - Felista L. Tansi
- Department of Experimental Radiology, Institute of Diagnostic and Interventional Radiology Jena University Hospital—Friedrich Schiller University Jena Am Klinikum 1 07740 Jena Germany
| | - Ingrid Hilger
- Department of Experimental Radiology, Institute of Diagnostic and Interventional Radiology Jena University Hospital—Friedrich Schiller University Jena Am Klinikum 1 07740 Jena Germany
| | - Adriele Prina‐Mello
- Nanomedicine and Molecular Imaging Group Trinity Translational Medicine Institute Dublin 8 Ireland
- Laboratory of Biological Characterization of Advanced Materials (LBCAM), Trinity Translational Medicine Institute Trinity College Dublin Dublin 8 Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre, CRANN Institute Trinity College Dublin Dublin 2 Ireland
| |
Collapse
|
54
|
Yi X, Duan QY, Wu FG. Low-Temperature Photothermal Therapy: Strategies and Applications. RESEARCH (WASHINGTON, D.C.) 2021; 2021:9816594. [PMID: 34041494 PMCID: PMC8125200 DOI: 10.34133/2021/9816594] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 03/01/2021] [Indexed: 12/13/2022]
Abstract
Although photothermal therapy (PTT) with the assistance of nanotechnology has been considered as an indispensable strategy in the biomedical field, it still encounters some severe problems that need to be solved. Excessive heat can induce treated cells to develop thermal resistance, and thus, the efficacy of PTT may be dramatically decreased. In the meantime, the uncontrollable diffusion of heat can pose a threat to the surrounding healthy tissues. Recently, low-temperature PTT (also known as mild PTT or mild-temperature PTT) has demonstrated its remarkable capacity of conquering these obstacles and has shown excellent performance in bacterial elimination, wound healing, and cancer treatments. Herein, we summarize the recently proposed strategies for achieving low-temperature PTT based on nanomaterials and introduce the synthesis, characteristics, and applications of these nanoplatforms. Additionally, the combination of PTT and other therapeutic modalities for defeating cancers and the synergistic cancer therapeutic effect of the combined treatments are discussed. Finally, the current limitations and future directions are proposed for inspiring more researchers to make contributions to promoting low-temperature PTT toward more successful preclinical and clinical disease treatments.
Collapse
Affiliation(s)
- Xiulin Yi
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing 210096, China
| | - Qiu-Yi Duan
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing 210096, China
| | - Fu-Gen Wu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing 210096, China
| |
Collapse
|
55
|
Carter TJ, Agliardi G, Lin FY, Ellis M, Jones C, Robson M, Richard-Londt A, Southern P, Lythgoe M, Zaw Thin M, Ryzhov V, de Rosales RTM, Gruettner C, Abdollah MRA, Pedley RB, Pankhurst QA, Kalber TL, Brandner S, Quezada S, Mulholland P, Shevtsov M, Chester K. Potential of Magnetic Hyperthermia to Stimulate Localized Immune Activation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2005241. [PMID: 33734595 DOI: 10.1002/smll.202005241] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 01/20/2021] [Indexed: 05/27/2023]
Abstract
Magnetic hyperthermia (MH) harnesses the heat-releasing properties of superparamagnetic iron oxide nanoparticles (SPIONs) and has potential to stimulate immune activation in the tumor microenvironment whilst sparing surrounding normal tissues. To assess feasibility of localized MH in vivo, SPIONs are injected intratumorally and their fate tracked by Zirconium-89-positron emission tomography, histological analysis, and electron microscopy. Experiments show that an average of 49% (21-87%, n = 9) of SPIONs are retained within the tumor or immediately surrounding tissue. In situ heating is subsequently generated by exposure to an externally applied alternating magnetic field and monitored by thermal imaging. Tissue response to hyperthermia, measured by immunohistochemical image analysis, reveals specific and localized heat-shock protein expression following treatment. Tumor growth inhibition is also observed. To evaluate the potential effects of MH on the immune landscape, flow cytometry is used to characterize immune cells from excised tumors and draining lymph nodes. Results show an influx of activated cytotoxic T cells, alongside an increase in proliferating regulatory T cells, following treatment. Complementary changes are found in draining lymph nodes. In conclusion, results indicate that biologically reactive MH is achievable in vivo and can generate localized changes consistent with an anti-tumor immune response.
Collapse
Affiliation(s)
- Thomas J Carter
- UCL Cancer Institute, University College London (UCL), Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK
| | - Giulia Agliardi
- UCL Cancer Institute, University College London (UCL), Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK
| | - Fang-Yu Lin
- UCL Healthcare Biomagnetics Laboratory, 21 Albermarle Street, London, W1S 4BS, UK
| | - Matthew Ellis
- Division of Neuropathology, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, London, WC1N 3BG, UK
- Cancer Sciences Unit, Cancer Research UK Centre, University of Southampton, Somers Building, Southampton, SO16 6YD, UK
| | - Clare Jones
- School of Biomedical Engineering and Imaging Sciences, King's College London (KCL), St Thomas' Hospital, London, SE1 7EH, UK
| | - Mathew Robson
- UCL Cancer Institute, University College London (UCL), Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK
| | - Angela Richard-Londt
- Division of Neuropathology, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Paul Southern
- UCL Healthcare Biomagnetics Laboratory, 21 Albermarle Street, London, W1S 4BS, UK
- Resonant Circuits Limited (RCL), London, W1S 4BS, UK
| | - Mark Lythgoe
- Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, WC1E 6DD, UK
| | - May Zaw Thin
- Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, WC1E 6DD, UK
| | - Vyacheslav Ryzhov
- NRC "Kurchatov Institute", Petersburg Nuclear Physics Institute, Gatchina, 188300, Russia
| | - Rafael T M de Rosales
- School of Biomedical Engineering and Imaging Sciences, King's College London (KCL), St Thomas' Hospital, London, SE1 7EH, UK
| | - Cordula Gruettner
- Micromod Partikeltechnologie GmbH, Friedrich-Barnewitz-Str. 4, Rostock, D-18119, Germany
| | - Maha R A Abdollah
- UCL Cancer Institute, University College London (UCL), Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, The British University in Egypt (BUE), El Shorouk City, Misr- Ismalia Desert Road, 11873, Cairo, Egypt
| | - R Barbara Pedley
- UCL Cancer Institute, University College London (UCL), Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK
| | - Quentin A Pankhurst
- UCL Healthcare Biomagnetics Laboratory, 21 Albermarle Street, London, W1S 4BS, UK
- Resonant Circuits Limited (RCL), London, W1S 4BS, UK
| | - Tammy L Kalber
- Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, WC1E 6DD, UK
| | - Sebastian Brandner
- Division of Neuropathology, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Sergio Quezada
- UCL Cancer Institute, University College London (UCL), Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK
| | - Paul Mulholland
- UCL Cancer Institute, University College London (UCL), Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK
| | - Maxim Shevtsov
- NRC "Kurchatov Institute", Petersburg Nuclear Physics Institute, Gatchina, 188300, Russia
- Technical University of Munich, Klinikum Rechts der Isar, Ismaninger str. 22, Munich, 81675, Germany
| | - Kerry Chester
- UCL Cancer Institute, University College London (UCL), Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK
| |
Collapse
|
56
|
Gani C, Lamprecht U, Ziegler A, Moll M, Gellermann J, Heinrich V, Wenz S, Fend F, Königsrainer A, Bitzer M, Zips D. Deep regional hyperthermia with preoperative radiochemotherapy in locally advanced rectal cancer, a prospective phase II trial. Radiother Oncol 2021; 159:155-160. [PMID: 33741467 DOI: 10.1016/j.radonc.2021.03.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 03/07/2021] [Accepted: 03/08/2021] [Indexed: 12/25/2022]
Abstract
PURPOSE The goal of the present study was to investigate the effect of deep regional hyperthermia on early and long-term oncological outcomes in the context of preoperative radiochemotherapy in rectal cancer. METHODS In this prospective phase II trial, patients with locally advanced rectal cancer were treated with 5-fluorouracil based preoperative radiochemotherapy with 50.4 Gy in 28 fractions. Deep regional hyperthermia was scheduled twice weekly. Pathological tumor regression was scored according to the Dworak regression system. The primary endpoint was pathological complete response (pCR). Further endpoints were local control (LC), distant control (DC), disease-free survival (DFS) and overall survival (OS). Hyperthermia was defined as feasible if 70% of patients received at least eight treatments. Quality of life was assessed at follow-up by the EORTC-QLQ-C30 and QLQ-CR29 questionnaires. Time to event data was analyzed according to Kaplan-Meier based on first-events. The study was registered on clinicaltrials.gov (NCT02353858). RESULTS From 2012 until 2017, 78 patients were recruited. Median follow-up was 54 months. Based on magnetic resonance imaging, the mesorectal fascia was involved or threatened in 60% of the patients. Compliance with radiotherapy was 99%, 91% received both cycles of chemotherapy and 77% had eight or more hyperthermia treatments. Median time from the end of radiotherapy to surgery was 6.7 weeks. A pathological complete response was reported in 14% of the patients, 50% had either Dworak 4 (complete regression) or Dworak 3 regression (scattered tumor cells only). Three year estimates for OS, DFS, LC and DC were 94%, 81%, 96% and 87%. Patients with higher hyperthermia related cumulative temperatures showed stronger tumor regression. Global health status based on EORTC-QLQ-C30 was comparable with data from the general population. CONCLUSION Deep regional hyperthermia was feasible, did not compromise standard treatments and resulted in promising long-term oncological outcomes and QoL.
Collapse
Affiliation(s)
- Cihan Gani
- University Hospital Tübingen, Department of Radiation Oncology, Germany; German Cancer Research Center (DKFZ) Heidelberg and German Consortium for Translational Cancer Research (DKTK), Partner Site Tübingen, Germany.
| | - Ulf Lamprecht
- University Hospital Tübingen, Department of Radiation Oncology, Germany
| | - Alexander Ziegler
- Department of Internal Medicine, Oncology/Hematology, Gastroenterology, Hospital Esslingen GmbH, Germany
| | - Matthias Moll
- Department of Radiation Oncology, Medical University of Vienna, Wien, Austria
| | | | - Vanessa Heinrich
- University Hospital Tübingen, Department of Radiation Oncology, Germany
| | - Svetlana Wenz
- Institute of Pathology and Neuropathology, Eberhard-Karls University, Tuebingen, Germany
| | - Falko Fend
- German Cancer Research Center (DKFZ) Heidelberg and German Consortium for Translational Cancer Research (DKTK), Partner Site Tübingen, Germany; Institute of Pathology and Neuropathology, Eberhard-Karls University, Tuebingen, Germany
| | - Alfred Königsrainer
- University Hospital Tübingen, Department of General, Visceral and Transplant Surgery, Germany
| | - Michael Bitzer
- University Hospital Tübingen, Department of Internal Medicine I, Germany
| | - Daniel Zips
- University Hospital Tübingen, Department of Radiation Oncology, Germany; German Cancer Research Center (DKFZ) Heidelberg and German Consortium for Translational Cancer Research (DKTK), Partner Site Tübingen, Germany
| |
Collapse
|
57
|
Datta NR, Marder D, Datta S, Meister A, Puric E, Stutz E, Rogers S, Eberle B, Timm O, Staruch M, Riesterer O, Bodis S. Quantification of thermal dose in moderate clinical hyperthermia with radiotherapy: a relook using temperature-time area under the curve (AUC). Int J Hyperthermia 2021; 38:296-307. [PMID: 33627018 DOI: 10.1080/02656736.2021.1875060] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Thermal dose in clinical hyperthermia reported as cumulative equivalent minutes (CEM) at 43 °C (CEM43) and its variants are based on direct thermal cytotoxicity assuming Arrhenius 'break' at 43 °C. An alternative method centered on the actual time-temperature plot during each hyperthermia session and its prognostic feasibility is explored. METHODS AND MATERIALS Patients with bladder cancer treated with weekly deep hyperthermia followed by radiotherapy were evaluated. From intravesical temperature (T) recordings obtained every 10 secs, the area under the curve (AUC) was computed for each session for T > 37 °C (AUC > 37 °C) and T ≥ 39 °C (AUC ≥ 39 °C). These along with CEM43, CEM43(>37 °C), CEM43(≥39 °C), Tmean, Tmin and Tmax were evaluated for bladder tumor control. RESULTS Seventy-four hyperthermia sessions were delivered in 18 patients (median: 4 sessions/patient). Two patients failed in the bladder. For both individual and summated hyperthermia sessions, the Tmean, CEM43, CEM43(>37 °C), CEM43(≥39 °C), AUC > 37 °C and AUC ≥ 39 °C were significantly lower in patients who had a local relapse. Individual AUC ≥ 39 °C for patients with/without local bladder failure were 105.9 ± 58.3 °C-min and 177.9 ± 58.0 °C-min, respectively (p = 0.01). Corresponding summated AUC ≥ 39 °C were 423.7 ± 27.8 °C-min vs. 734.1 ± 194.6 °C-min (p < 0.001), respectively. The median AUC ≥ 39 °C for each hyperthermia session in patients with bladder tumor control was 190 °C-min. CONCLUSION AUC ≥ 39 °C for each hyperthermia session represents the cumulative time-temperature distribution at clinically defined moderate hyperthermia in the range of 39 °C to 45 °C. It is a simple, mathematically computable parameter without any prior assumptions and appears to predict treatment outcome as evident from this study. However, its predictive ability as a thermal dose parameter merits further evaluation in a larger patient cohort.
Collapse
Affiliation(s)
- Niloy R Datta
- Department of Radiation Oncology KSA-KSB, Kantonsspital Aarau, Aarau, Switzerland
| | - Dietmar Marder
- Department of Radiation Oncology KSA-KSB, Kantonsspital Aarau, Aarau, Switzerland
| | - Sneha Datta
- Animal Production and Health Laboratory, Joint FAO/IAEA Division of Nuclear Techniques in Food and Agriculture, Department of Nuclear Sciences and Applications, International Atomic Energy Agency (IAEA), Vienna, Austria
| | - Andreas Meister
- Department of Radiation Oncology KSA-KSB, Kantonsspital Aarau, Aarau, Switzerland
| | - Emsad Puric
- Department of Radiation Oncology KSA-KSB, Kantonsspital Aarau, Aarau, Switzerland
| | - Emanuel Stutz
- Department of Radiation Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Susanne Rogers
- Department of Radiation Oncology KSA-KSB, Kantonsspital Aarau, Aarau, Switzerland
| | - Brigitte Eberle
- Department of Radiation Oncology KSA-KSB, Kantonsspital Aarau, Aarau, Switzerland
| | - Olaf Timm
- Department of Radiation Oncology KSA-KSB, Kantonsspital Aarau, Aarau, Switzerland
| | - Michal Staruch
- Department of Radiation Oncology KSA-KSB, Kantonsspital Aarau, Aarau, Switzerland
| | - Oliver Riesterer
- Department of Radiation Oncology KSA-KSB, Kantonsspital Aarau, Aarau, Switzerland.,Department of Radiation Oncology, University Hospital Zurich, Switzerland
| | - Stephan Bodis
- Department of Radiation Oncology KSA-KSB, Kantonsspital Aarau, Aarau, Switzerland.,Department of Radiation Oncology, University Hospital Zurich, Switzerland
| |
Collapse
|
58
|
van der Horst A, Kok HP, Crezee J. Effect of gastrointestinal gas on the temperature distribution in pancreatic cancer hyperthermia treatment planning. Int J Hyperthermia 2021; 38:229-240. [PMID: 33602033 DOI: 10.1080/02656736.2021.1882709] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
PURPOSE In pancreatic cancer treatment, hyperthermia can be added to increase efficacy of chemo- and/or radiotherapy. Gas in stomach, intestines and colon is often in close proximity to the target volume. We investigated the impact of variations in gastrointestinal gas (GG) on temperature distributions during simulated hyperthermia treatment (HT). METHODS We used sets of one CT and eight cone-beam CT (CBCT) scans obtained prior to/during fractionated image-guided radiotherapy in four pancreatic cancer patients. In Plan2Heat, we simulated locoregional heating by an ALBA-4D phased array radiofrequency system and calculated temperature distributions for (i) the segmented CT (sCT), (ii) sCT with GG replaced by muscle (sCT0), (iii) sCT0 with eight different GG distributions as visible on CBCT inserted (sCTCBCT). We calculated cumulative temperature-volume histograms for the clinical target volume (CTV) for all ten temperature distributions for each patient and investigated the relationship between GG volume and change in ΔT50 (temperature increase at 50% of CTV volume). We determined location and volume of normal tissue receiving a high thermal dose. RESULTS GG volume on CBCT varied greatly (9-991 cm3). ΔT50 increased for increasing GG volume; maximum ΔT50 difference per patient was 0.4-0.6 °C. The risk for GG-associated treatment-limiting hot spots appeared low. Normal tissue high-temperature regions mostly occurred anteriorly; their volume and maximum temperature showed moderate positive correlations with GG volume, while fat-muscle interfaces were associated with higher risks for hot spots. CONCLUSIONS Considerable changes in volume and position of gastrointestinal gas can occur and are associated with clinically relevant tumor temperature differences.
Collapse
Affiliation(s)
- Astrid van der Horst
- Department of Radiation Oncology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - H Petra Kok
- Department of Radiation Oncology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Johannes Crezee
- Department of Radiation Oncology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
59
|
Nguyen HTM, Katta N, Widman JA, Takematsu E, Feng X, Torres-Hurtado SA, Betancourt T, Baker AB, Suggs LJ, Milner TE, Tunnell JW. Laser nanobubbles induce immunogenic cell death in breast cancer. NANOSCALE 2021; 13:3644-3653. [PMID: 33538275 PMCID: PMC8710258 DOI: 10.1039/d0nr06587k] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Recent advances in immunotherapy have highlighted a need for therapeutics that initiate immunogenic cell death in tumors to stimulate the body's immune response to cancer. This study examines whether laser-generated bubbles surrounding nanoparticles ("nanobubbles") induce an immunogenic response for cancer treatment. A single nanosecond laser pulse at 1064 nm generates micron-sized bubbles surrounding gold nanorods in the cytoplasm of breast cancer cells. Cell death occurred in cells treated with nanorods and irradiated, but not in cells with irradiation treatment alone. Cells treated with nanorods and irradiation had increased damage-associated molecular patterns (DAMPs), including increased expression of chaperone proteins human high mobility group box 1 (HMGB1), adenosine triphosphate (ATP), and heat shock protein 70 (HSP70). This enhanced expression of DAMPs led to the activation of dendritic cells. Overall, this treatment approach is a rapid and highly specific method to eradicate tumor cells with simultaneous immunogenic cell death signaling, showing potential as a combination strategy for immunotherapy.
Collapse
Affiliation(s)
- Hieu T M Nguyen
- Department of Biomedical Engineering, The University of Texas at Austin, TX, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
60
|
Nanoparticle-Mediated Heating: A Theoretical Study for Photothermal Treatment and Photo Immunotherapy. Bioanalysis 2021. [DOI: 10.1007/978-3-030-78338-9_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
61
|
Multifunctional Gold Nanostars for Sensitive Detection, Photothermal Treatment and Immunotherapy of Brain Tumor. Bioanalysis 2021. [DOI: 10.1007/978-3-030-78338-9_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
62
|
Ruan Q, Ding D, Wang B, He C, Ren X, Feng Z, Pang Z, Wang J, Zhang X, Tang H, Wang J, He Q, Lei Z, Liao Q, Luo J, Cui S. A multi-institutional retrospective study of hyperthermic plus intravesical chemotherapy versus intravesical chemotherapy treatment alone in intermediate and high risk nonmuscle-invasive bladder cancer. Cancer Biol Med 2021; 18:308-317. [PMID: 33628603 PMCID: PMC7877165 DOI: 10.20892/j.issn.2095-3941.2020.0125] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 07/10/2020] [Indexed: 11/11/2022] Open
Abstract
Objective: To compare the efficacy and safety of hyperthermic intravesical chemotherapy (HIVEC) and intravesical chemotherapy (IVEC) in patients with intermediate and high risk nonmuscle-invasive bladder cancer (NMIBC) after transurethral resection. Methods: We included 560 patients diagnosed with primary or recurrent NMIBC between April 2009 and December 2015 at 1 of 6 tertiary centers. We matched 364 intermediate or high risk cases and divided them into 2 groups: the HIVEC+IVEC group [chemohyperthermia (CHT) composed of 3 consecutive sessions followed by intravesical instillation without hyperthermia] and the IVEC group (intravesical instillation without hyperthermia). The data were recorded in the database. The primary endpoint was 2-year recurrence-free survival (RFS) in all NMIBC patients (n = 364), whereas the secondary endpoints were the assessment of radical cystectomy (RC) and 5-year overall survival (OS). Results: There was a significant difference in the 2-year RFS between the two groups in all patients (n = 364; HIVEC+IVEC: 82.42% vs. IVEC: 74.18%, P = 0.038). Compared with the IVEC group, the HIVEC+IVEC group had a lower incidence of RC (P = 0.0274). However, the 5-year OS was the same between the 2 groups (P = 0.1434). Adverse events (AEs) occurred in 32.7% of all patients, but none of the events was serious (grades 3–4). No difference in the incidence or severity of AEs between each treatment modality was observed. Conclusions: This retrospective study showed that HIVEC+IVEC had a higher 2-year RFS and a lower incidence of RC than IVEC therapy in intermediate and high risk NMIBC patients. Both treatments were well-tolerated in a similar manner.
Collapse
Affiliation(s)
- Qiang Ruan
- Department of Abdominal Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou 510095, China.,The First Affiliated Hospital of Jinan University, the First Clinical Medical College of Jinan University, Guangzhou 510630, China
| | - Degang Ding
- Department of Urinary Surgery, Henan Provincial People's Hospital, Zhengzhou 450003, China
| | - Bin Wang
- Department of Urinary Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou 510095, China
| | - Chaohong He
- Department of Urinary Surgery, Henan Cancer Hospital, Zhengzhou 450008, China
| | - Xuequn Ren
- Department of General Surgery, Huaihe Hospital of Henan University, Kaifeng 475000, China
| | - Zhenhua Feng
- Department of Urinary Surgery, Gaozhou People's Hospital, Maoming 525200, China
| | - Zhigang Pang
- Department of General Surgery, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450014, China
| | - Jin Wang
- Department of Abdominal Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou 510095, China
| | - Xiangliang Zhang
- Department of Abdominal Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou 510095, China
| | - Hongsheng Tang
- Department of Abdominal Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou 510095, China
| | - Jiahong Wang
- Department of Abdominal Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou 510095, China
| | - Qingjun He
- Department of Abdominal Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou 510095, China
| | - Ziying Lei
- Department of Abdominal Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou 510095, China
| | - Quanxing Liao
- Department of Abdominal Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou 510095, China
| | - Jiali Luo
- Department of Abdominal Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou 510095, China
| | - Shuzhong Cui
- Department of Abdominal Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou 510095, China
| |
Collapse
|
63
|
Vo-Dinh T. The New Frontier in Medicine at the Convergence of Nanotechnology and Immunotherapy. Bioanalysis 2021. [DOI: 10.1007/978-3-030-78338-9_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
64
|
Vo-Dinh T, Inman BA, Maccarini P, Palmer GM, Liu Y, Etienne W. Plasmonic Gold Nanostars for Immuno Photothermal Nanotherapy to Treat Cancers and Induce Long-Term Immunity. Bioanalysis 2021. [DOI: 10.1007/978-3-030-78338-9_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
65
|
Nanoparticle Systems Applied for Immunotherapy in Various Treatment Modalities. Bioanalysis 2021. [DOI: 10.1007/978-3-030-78338-9_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
66
|
Terasaki A, Kurokawa H, Ito H, Komatsu Y, Matano D, Terasaki M, Bando H, Hara H, Matsui H. Elevated Production of Mitochondrial Reactive Oxygen Species via Hyperthermia Enhanced Cytotoxic Effect of Doxorubicin in Human Breast Cancer Cell Lines MDA-MB-453 and MCF-7. Int J Mol Sci 2020; 21:ijms21249522. [PMID: 33333736 PMCID: PMC7765207 DOI: 10.3390/ijms21249522] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/04/2020] [Accepted: 12/10/2020] [Indexed: 01/04/2023] Open
Abstract
Hyperthermia (HT) treatment is a noninvasive cancer therapy, often used with radiation therapy and chemotherapy. Compared with 37 °C, 42 °C is mild heat stress for cells and produces reactive oxygen species (ROS) from mitochondria. To involve subsequent intracellular accumulation of DOX, we have previously reported that the expression of ATP-binding cassette sub-family G member 2 (ABCG2), an exporter of doxorubicin (DOX), was suppressed by a larger amount of intracellular mitochondrial ROS. We then hypothesized that the additive effect of HT and chemotherapy would be induced by the downregulation of ABCG2 expression via intracellular ROS increase. We used human breast cancer cell lines, MCF-7 and MDA-MB-453, incubated at 37 °C or 42 °C for 1 h to clarify this hypothesis. Intracellular ROS production after HT was detected via electron spin resonance (ESR), and DOX cytotoxicity was calculated. Additionally, ABCG2 expression in whole cells was analyzed using Western blotting. We confirmed that the ESR signal peak with HT became higher than that without HT, indicating that the intracellular ROS level was increased by HT. ABCG2 expression was downregulated by HT, and cells were injured after DOX treatment. DOX cytotoxicity enhancement with HT was considered a result of ABCG2 expression downregulation via the increase of ROS production. HT increased intracellular ROS production and downregulated ABCG2 protein expression, leading to cell damage enhancement via DOX.
Collapse
Affiliation(s)
- Azusa Terasaki
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki 305-8577, Japan; (A.T.); (Y.K.); (D.M.)
- Department of Breast-Thyroid-Endocrine Surgery, University of Tsukuba Hospital, Ibaraki 305-8577, Japan
| | - Hiromi Kurokawa
- Faculty of Medicine, University of Tsukuba, Ibaraki 305-8577, Japan; (H.K.); (H.I.)
| | - Hiromu Ito
- Faculty of Medicine, University of Tsukuba, Ibaraki 305-8577, Japan; (H.K.); (H.I.)
- Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-0065, Japan
| | - Yoshiki Komatsu
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki 305-8577, Japan; (A.T.); (Y.K.); (D.M.)
| | - Daisuke Matano
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki 305-8577, Japan; (A.T.); (Y.K.); (D.M.)
| | - Masahiko Terasaki
- Division of Gastroenterology, Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan; (M.T.); (H.M.)
| | - Hiroko Bando
- Division of Breast and Endocrine Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki 305-8577, Japan;
- Correspondence: ; Tel.: +81-29-853-3341
| | - Hisato Hara
- Division of Breast and Endocrine Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki 305-8577, Japan;
| | - Hirofumi Matsui
- Division of Gastroenterology, Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan; (M.T.); (H.M.)
| |
Collapse
|
67
|
Grimberg DC, Shah A, Tan WP, Etienne W, Spasojevic I, Inman BA. Hyperthermia Improves Solubility of Intravesical Chemotherapeutic Agents. Bladder Cancer 2020; 6:461-470. [PMID: 36118287 PMCID: PMC9441059 DOI: 10.3233/blc-200350] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 08/24/2020] [Indexed: 11/15/2022]
Abstract
BACKGROUND: Nearly 70% of all new cases of bladder cancer are non-muscle invasive disease, the treatment for which includes transurethral resection followed by intravesical therapy. Unfortunately, recurrence rates approach 50% in part due to poor intravesical drug delivery. Hyperthermia is frequently used as an adjunct to intravesical chemotherapy to improve drug delivery and response to treatment. OBJECTIVE: To assess the solubility profile of intravesical chemotherapies under varying conditions of pH and temperature. METHODS: Using microplate laser nephelometry we measured the solubility of three intravesical chemotherapy agents (mitomycin C, gemcitabine, and cisplatin) at varying physical conditions. Drugs were assessed at room temperature (23°C), body temperature (37°C), and 43°C, the temperature used for hyperthermic intravesical treatments. To account for variations in urine pH, solubility was also investigated at pH 4.00, 6.00, and 8.00. RESULTS: Heat incrementally increased the solubility of all three drugs studied. Conversely, pH largely did not impact solubility aside for gemcitabine which showed slightly reduced solubility at pH 8.00 versus 6.00 or 4.00. Mitomycin C at the commonly used 2.0 mg/mL was insoluble at room temperature, but soluble at both 37 and 43°C. CONCLUSIONS: Hyperthermia as an adjunct to intravesical treatment would improve drug solubility, and likely drug delivery as some current regimens are insoluble without heat. Improvements in solubility also allow for testing of alternative administration regimens to improve drug delivery or tolerability. Further studies are needed to confirm that improvements in solubility result in increased drug delivery.
Collapse
Affiliation(s)
- Dominic C. Grimberg
- Division of Urology, Duke Prostate and Urologic Cancer Center, Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| | - Ankeet Shah
- Division of Urology, Duke Prostate and Urologic Cancer Center, Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| | - Wei Phin Tan
- Division of Urology, Duke Prostate and Urologic Cancer Center, Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| | - Wiguins Etienne
- Division of Urology, Duke Prostate and Urologic Cancer Center, Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| | - Ivan Spasojevic
- Department of Medicine, Duke Prostate and Urologic Cancer Center, Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| | - Brant A. Inman
- Division of Urology, Duke Prostate and Urologic Cancer Center, Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
68
|
Meng Y, Hynynen K, Lipsman N. Applications of focused ultrasound in the brain: from thermoablation to drug delivery. Nat Rev Neurol 2020; 17:7-22. [PMID: 33106619 DOI: 10.1038/s41582-020-00418-z] [Citation(s) in RCA: 208] [Impact Index Per Article: 41.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/16/2020] [Indexed: 02/07/2023]
Abstract
Focused ultrasound (FUS) is a disruptive medical technology, and its implementation in the clinic represents the culmination of decades of research. Lying at the convergence of physics, engineering, imaging, biology and neuroscience, FUS offers the ability to non-invasively and precisely intervene in key circuits that drive common and challenging brain conditions. The actions of FUS in the brain take many forms, ranging from transient blood-brain barrier opening and neuromodulation to permanent thermoablation. Over the past 5 years, we have seen a dramatic expansion of indications for and experience with FUS in humans, with a resultant exponential increase in academic and public interest in the technology. Applications now span the clinical spectrum in neurological and psychiatric diseases, with insights still emerging from preclinical models and human trials. In this Review, we provide a comprehensive overview of therapeutic ultrasound and its current and emerging indications in the brain. We examine the potential impact of FUS on the landscape of brain therapies as well as the challenges facing further advancement and broader adoption of this promising minimally invasive therapeutic alternative.
Collapse
Affiliation(s)
- Ying Meng
- Division of Neurosurgery, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Sunnybrook Research Institute, Hurvitz Brain Sciences Program, Harquail Centre for Neuromodulation, Toronto, ON, Canada.,Institute of Medical Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Kullervo Hynynen
- Sunnybrook Research Institute, Toronto, ON, Canada.,Department of Medical Biophysics and Institute of Biomaterials & Biomedical Engineering (IBBME), University of Toronto, Toronto, ON, Canada
| | - Nir Lipsman
- Division of Neurosurgery, Sunnybrook Health Sciences Centre, Toronto, ON, Canada. .,Sunnybrook Research Institute, Hurvitz Brain Sciences Program, Harquail Centre for Neuromodulation, Toronto, ON, Canada. .,Institute of Medical Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
69
|
Regional Hyperthermia Enhances Mesenchymal Stem Cell Recruitment to Tumor Stroma: Implications for Mesenchymal Stem Cell-Based Tumor Therapy. Mol Ther 2020; 29:788-803. [PMID: 33068779 DOI: 10.1016/j.ymthe.2020.10.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 09/22/2020] [Accepted: 10/12/2020] [Indexed: 12/13/2022] Open
Abstract
The tropism of mesenchymal stem cells (MSCs) for tumors forms the basis for their use as delivery vehicles for the tumor-specific transport of therapeutic genes, such as the theranostic sodium iodide symporter (NIS). Hyperthermia is used as an adjuvant for various tumor therapies and has been proposed to enhance leukocyte recruitment. Here, we describe the enhanced recruitment of adoptively applied NIS-expressing MSCs to tumors in response to regional hyperthermia. Hyperthermia (41°C, 1 h) of human hepatocellular carcinoma cells (HuH7) led to transiently increased production of immunomodulatory factors. MSCs showed enhanced chemotaxis to supernatants derived from heat-treated cells in a 3D live-cell tracking assay and was validated in vivo in subcutaneous HuH7 mouse xenografts. Cytomegalovirus (CMV)-NIS-MSCs were applied 6-48 h after or 24-48 h before hyperthermia treatment. Using 123I-scintigraphy, thermo-stimulation (41°C, 1 h) 24 h after CMV-NIS-MSC injection resulted in a significantly increased uptake of 123I in heat-treated tumors compared with controls. Immunohistochemical staining and real-time PCR confirmed tumor-selective, temperature-dependent MSC migration. Therapeutic efficacy was significantly enhanced by combining CMV-NIS-MSC-mediated 131I therapy with regional hyperthermia. We demonstrate here for the first time that hyperthermia can significantly boost tumoral MSC recruitment, thereby significantly enhancing therapeutic efficacy of MSC-mediated NIS gene therapy.
Collapse
|
70
|
Issels RD, Lindner LH, von Bergwelt-Baildon M, Lang P, Rischpler C, Diem H, Mosetter B, Eckl J, Schendel DJ, Salat C, Stötzer O, Burdach S, von Luettichau-Teichert I, Handgretinger R, Neumann J, Kirchner T, Steiger K, Boxberg M, Mansmann U, Multhoff G, Noessner E. Systemic antitumor effect by regional hyperthermia combined with low-dose chemotherapy and immunologic correlates in an adolescent patient with rhabdomyosarcoma - a case report. Int J Hyperthermia 2020; 37:55-65. [PMID: 31918587 DOI: 10.1080/02656736.2019.1709666] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Introduction: An abscopal effect is a clinical observation whereby a local treatment is associated with regression of metastatic cancer at a site distant from the primary location of treatment. Here, we describe the clinical systemic effect induced by regional hyperthermia combined with low-dose chemotherapy and provide immunologic correlates.Case presentation: A 15-year-old patient had been diagnosed with alveolar rhabdomyosarcoma (ARMS). All previous treatment options failed in the patient including haploidentical stem cell transplantation and donor lymphocyte infusion. The patient presented with local and metastatic disease, and upon admission, underwent regional hyperthermia combined with low-dose chemotherapy. Immediately following therapy severe skin reactions were observed. Skin biopsies revealed an intraepithelial lymphocytic infiltration dominated by CD3+/CD8+ T cells with a regular network of dendritic cells. Clinical images compared before and during sequential treatment cycles showed complete metabolic response of the local tumor for more than 10 months of therapy. In addition, metastases completely regressed although they were not direct targets of regional hyperthermia. The systemic effect was associated with enhanced frequency of NK cells and T cells expressing the lectin-like natural-killer group 2 D activating receptor (NKG2D), an increase of the CD56bright subset of NK cells, as well as an increase of effector/memory and effector CD8+ and CD4+ T cells in the blood while the percentage of CD25+FOXP3+ regulatory T cells declined.Conclusions: Regional hyperthermia combined with low-dose chemotherapy had the potential to create a systemic effect which was associated with activation of NK cells and T cells.
Collapse
Affiliation(s)
- Rolf D Issels
- Department of Medicine III, University Hospital, Munich, Germany
| | - Lars H Lindner
- Department of Medicine III, University Hospital, Munich, Germany
| | | | - Peter Lang
- Department of General Pediatrics, Hematology/Oncology, University Children's Hospital, Tuebingenthe, Germany
| | - Christoph Rischpler
- Department of Nuclear Medicine, Technical University Munich, Munich, Germany
| | - Heinz Diem
- Laboratory for Hematological Diagnostics, Gauting, Germany
| | - Barbara Mosetter
- Immunoanalytics Research Group - Tissue Control of Immunocytes & Core Facility, Helmholtz Center, Munich, Germany
| | | | | | - Christoph Salat
- Medical Center for Hematology and Oncology Munich, Munich, Germany
| | - Oliver Stötzer
- Medical Center for Hematology and Oncology Munich, Munich, Germany
| | - Stefan Burdach
- Department of Pediatrics and Children's Cancer Research Center, Technical University of Munich, Munich, Germany
| | | | - Rupert Handgretinger
- Department of General Pediatrics, Hematology/Oncology, University Children's Hospital, Tuebingenthe, Germany
| | | | | | - Katja Steiger
- Institute of Pathology, Technical University Munich, Munich, Germany
| | - Melanie Boxberg
- Institute of Pathology, Technical University Munich, Munich, Germany
| | - Ulrich Mansmann
- Institute of Biostatistics and Epidemiology, LMU, Munich, Germany
| | - Gabriele Multhoff
- Radiation Immune-Oncology Group, Center for Translational Cancer Research, Technical University Munich, Munich, Germany
| | - Elfriede Noessner
- Immunoanalytics Research Group - Tissue Control of Immunocytes & Core Facility, Helmholtz Center, Munich, Germany
| |
Collapse
|
71
|
Zhu L, Lam D, Pacia CP, Gach HM, Partanen A, Talcott MR, Greco SC, Zoberi I, Hallahan DE, Chen H, Altman MB. Characterization of magnetic resonance-guided high-intensity focused ultrasound (MRgHIFU)-induced large-volume hyperthermia in deep and superficial targets in a porcine model. Int J Hyperthermia 2020; 37:1159-1173. [DOI: 10.1080/02656736.2020.1825836] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Lifei Zhu
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Dao Lam
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Christopher Pham Pacia
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - H. Michael Gach
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, Missouri, USA
- Department of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA
- Siteman Comprehensive Cancer Center, St. Louis, St. Louis, Missouri, 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, USA
| | - Suellen C. Greco
- Division of Comparative Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Imran Zoberi
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, Missouri, USA
- Siteman Comprehensive Cancer Center, St. Louis, St. Louis, Missouri, USA
| | - Dennis E. Hallahan
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, Missouri, USA
- Siteman Comprehensive Cancer Center, St. Louis, St. Louis, Missouri, USA
| | - Hong Chen
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, Missouri, USA
- Siteman Comprehensive Cancer Center, St. Louis, St. Louis, Missouri, USA
| | - Michael B. Altman
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, Missouri, USA
- Siteman Comprehensive Cancer Center, St. Louis, St. Louis, Missouri, USA
| |
Collapse
|
72
|
Rahban D, Doostan M, Salimi A. Cancer Therapy; Prospects for Application of Nanoparticles for Magnetic-Based Hyperthermia. Cancer Invest 2020; 38:507-521. [PMID: 32870068 DOI: 10.1080/07357907.2020.1817482] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Hyperthermic therapy is defined as increasing the temperature of tumor tissues to 40-43 °C that has been effective approach for destroying malignant cells in the field of cancer therapy. Recent line of research has applied different approaches along with hyperthermic treatment to obtain high efficiency and little side effects. Magnetic nanoparticle-based hyperthermia has demonstrated an improved functionality in targeting malignant cells and implement their therapeutic role by heating the tumor cells. Here in this review article, we clarify the diverse aspects of magnetic nanoparticles in the treatment of cancer.
Collapse
Affiliation(s)
- Dariuosh Rahban
- Nanobiotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mahtab Doostan
- Nanobiotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ali Salimi
- Nanobiotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
73
|
Zhu L, Xu Z, Wu Y, Liu P, Qian J, Yu S, Xia B, Lai J, Ma S, Wu Z. Prophylactic chemotherapeutic hyperthermic intraperitoneal perfusion reduces peritoneal metastasis in gastric cancer: a retrospective clinical study. BMC Cancer 2020; 20:827. [PMID: 32867714 PMCID: PMC7461269 DOI: 10.1186/s12885-020-07339-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 08/25/2020] [Indexed: 02/07/2023] Open
Abstract
Background Peritoneal metastasis is the most frequent failure in gastric cancer. This study evaluated the role of prophylactic chemotherapeutic hyperthermic intraperitoneal perfusion (CHIP) in patients after D2 dissection. Methods Gastric cancer patients after D2 dissection were enrolled in this study. Patients received either chemotherapy (IV group) or CHIP (CHIP group). Sites of recurrence or metastasis, disease-free survival (DFS), overall survival (OS) and adverse events were evaluated. Results Twenty-two patients received CHIP treatment, and 21 patients received chemotherapy alone. The median DFS time was 24.5 and 36.5 months in the IV group and CHIP group (P = 0.044), respectively. The median OS time was 33.1 months in the IV group and not reached in the CHIP group (P = 0.037). We also found that CHIP could reduce the total recurrence/metastasis rate, especially that of peritoneal metastasis. In the subgroup analysis, DFS and OS were both superior in deficient mismatch repair (dMMR) patients than in proficient MMR (pMMR) patients. Conclusion This hypothesis-generating study indicates that CHIP might be feasible for gastric cancer patients after D2 resection.
Collapse
Affiliation(s)
- Lucheng Zhu
- Department of Radiotherapy, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou Cancer Hospital, Hangzhou, 310002, P.R. China.,Department of Oncology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, P.R. China
| | - Zhizheng Xu
- Department of oncology, Changxing people's hospital, Huzhou, 313100, P.R. China
| | - Yajun Wu
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, 310053, P.R. China
| | - Pengyuan Liu
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, 310053, P.R. China
| | - Jianing Qian
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, 310053, P.R. China
| | - Shuhuan Yu
- Department of Oncology, Zhejiang Hospital, Hangzhou, Zhejiang, 310013, P.R. China
| | - Bing Xia
- Department of Radiotherapy, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou Cancer Hospital, Hangzhou, 310002, P.R. China.,Department of Oncology, Jiande Second People's Hospital, Zhejiang, 311604, P.R. China
| | - Jianjun Lai
- Department of Oncology, Zhejiang Hospital, Hangzhou, Zhejiang, 310013, P.R. China
| | - Shenglin Ma
- Department of Oncology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, P.R. China.
| | - Zhibing Wu
- Department of Oncology, Zhejiang Hospital, Hangzhou, Zhejiang, 310013, P.R. China.
| |
Collapse
|
74
|
Ghasemi M, Sivaloganathan S. A computational study of combination HIFU-chemotherapy as a potential means of overcoming cancer drug resistance. Math Biosci 2020; 329:108456. [PMID: 32841615 DOI: 10.1016/j.mbs.2020.108456] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 08/14/2020] [Accepted: 08/14/2020] [Indexed: 11/18/2022]
Abstract
The application of local hyperthermia, particularly in conjunction with other treatment strategies (like chemotherapy and radiotherapy) has been known to be a useful means of enhancing tumor treatment outcomes. However, to our knowledge, there has been no mathematical model designed to capture the impact of the combination of hyperthermia and chemotherapies on tumor growth and control. In this study, we propose a nonlinear Partial Differential Equation (PDE) model which describes the tumor response to chemotherapy, and use the model to study the effects of hyperthermia on the response of prototypical tumor to the generic chemotherapeutic agent. Ultrasound energy is delivered to the tumor through High Intensity Focused Ultrasound (HIFU), as a noninvasive technique to elevate the tumor temperature in a controlled manner. The proposed tumor growth model is coupled with the nonlinear density dependent Westervelt and Penne's bio-heat equations, used to calculate the net delivered energy and temperature of the tumor and its surrounding normal tissue. The tumor is assumed to be composed of two species: drug-sensitive and drug-resistant. The central assumption underlying our model is that the drug-resistant species is converted to a drug-sensitive type when the tumor temperature is elevated above a certain threshold temperature. The "in silico" results obtained, confirm that hyperthermia can result in less aggressive tumor development and emphasize the importance of designing an optimized thermal dose strategy. Furthermore, our results suggest that increasing the length of the on/off cycle of the transducer is an efficient approach to treatment scheduling in the sense of optimizing tumor eradication.
Collapse
Affiliation(s)
- Maryam Ghasemi
- Department of Applied Mathematics, Univ. Waterloo, Waterloo, ON, Canada, N2L 3G1.
| | | |
Collapse
|
75
|
Sharma D, Cartar H, Law N, Giles A, Farhat G, Oelze M, Czarnota GJ. Optimization of microbubble enhancement of hyperthermia for cancer therapy in an in vivo breast tumour model. PLoS One 2020; 15:e0237372. [PMID: 32797049 PMCID: PMC7428078 DOI: 10.1371/journal.pone.0237372] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 07/24/2020] [Indexed: 11/18/2022] Open
Abstract
We have demonstrated that exposing human breast tumour xenografts to ultrasound-stimulated microbubbles enhances tumour cell death and vascular disruption resulting from hyperthermia treatment. The aim of this study was to investigate the effect of varying the hyperthermia and ultrasound-stimulated microbubbles treatment parameters in order to optimize treatment bioeffects. Human breast cancer (MDA-MB-231) tumour xenografts in severe combined immunodeficiency (SCID) mice were exposed to varying microbubble concentrations (0%, 0.1%, 1% or 3% v/v) and ultrasound sonication durations (0, 1, 3 or 5 min) at 570 kPa peak negative pressure and central frequency of 500 kHz. Five hours later, tumours were immersed in a 43°C water bath for varying hyperthermia treatment durations (0, 10, 20, 30, 40, 50 or 60 minutes). Results indicated a significant increase in tumour cell death reaching 64 ± 5% with combined treatment compared to 11 ± 3% and 26 ± 5% for untreated and USMB-only treated tumours, respectively. A similar but opposite trend was observed in the vascular density of the tumours receiving the combined treatment. Optimal treatment parameters were found to consist of 40 minutes of heat with low power ultrasound treatment microbubble parameters of 1 minute of sonification and a 1% microbubble concentration.
Collapse
Affiliation(s)
- Deepa Sharma
- Imaging Research and Physical Sciences, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
- Departments of Medical Biophysics, and Radiation Oncology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Holliday Cartar
- Imaging Research and Physical Sciences, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Niki Law
- Imaging Research and Physical Sciences, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Anoja Giles
- Imaging Research and Physical Sciences, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Golnaz Farhat
- Imaging Research and Physical Sciences, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Michael Oelze
- Department of Electrical and Computer Engineering, Beckman Institute, University of Chicago Illinois at Urbana Champaign, Illinois, United States of America
| | - Gregory J. Czarnota
- Imaging Research and Physical Sciences, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
- Departments of Medical Biophysics, and Radiation Oncology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
76
|
Srinivasan ES, Sankey EW, Grabowski MM, Chongsathidkiet P, Fecci PE. The intersection between immunotherapy and laser interstitial thermal therapy: a multipronged future of neuro-oncology. Int J Hyperthermia 2020; 37:27-34. [PMID: 32672126 PMCID: PMC11229985 DOI: 10.1080/02656736.2020.1746413] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 03/04/2020] [Accepted: 03/15/2020] [Indexed: 10/23/2022] Open
Abstract
The rise of immunotherapy (IT) in oncological treatment has greatly improved outcomes in a number of disease states. However, its use in tumors of the central nervous system (CNS) remains limited for multiple reasons related to the unique immunologic tumor microenvironment. As such, it is valuable to consider the intersection of IT with additional treatment methods that may improve access to the CNS and effectiveness of existing IT modalities. One such combination is the pairing of IT with localized hyperthermia (HT) generated through technologies such as laser interstitial thermal therapy (LITT). The wide-ranging immunomodulatory effects of localized and whole-body HT have been investigated for some time. Hyperthermia has demonstrated immunostimulatory effects at the level of tumor cells, immune cells, and the broader environment governing potential immune surveillance. A thorough understanding of these effects as well as the current and upcoming investigations of such in combination with IT is important in considering the future directions of neuro-oncology.
Collapse
Affiliation(s)
- Ethan S Srinivasan
- Department of Neurosurgery, Duke University School of Medicine, Durham, NC, USA
| | - Eric W Sankey
- Department of Neurosurgery, Duke University School of Medicine, Durham, NC, USA
- Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
| | | | | | - Peter E Fecci
- Department of Neurosurgery, Duke University School of Medicine, Durham, NC, USA
- Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
77
|
Soetaert F, Korangath P, Serantes D, Fiering S, Ivkov R. Cancer therapy with iron oxide nanoparticles: Agents of thermal and immune therapies. Adv Drug Deliv Rev 2020; 163-164:65-83. [PMID: 32603814 PMCID: PMC7736167 DOI: 10.1016/j.addr.2020.06.025] [Citation(s) in RCA: 201] [Impact Index Per Article: 40.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 05/19/2020] [Accepted: 06/23/2020] [Indexed: 12/12/2022]
Abstract
Significant research and preclinical investment in cancer nanomedicine has produced several products, which have improved cancer care. Nevertheless, there exists a perception that cancer nanomedicine 'has not lived up to its promise' because the number of approved products and their clinical performance are modest. Many of these analyses do not consider the long clinical history and many clinical products developed from iron oxide nanoparticles. Iron oxide nanoparticles have enjoyed clinical use for about nine decades demonstrating safety, and considerable clinical utility and versatility. FDA-approved applications of iron oxide nanoparticles include cancer diagnosis, cancer hyperthermia therapy, and iron deficiency anemia. For cancer nanomedicine, this wealth of clinical experience is invaluable to provide key lessons and highlight pitfalls in the pursuit of nanotechnology-based cancer therapeutics. We review the clinical experience with systemic liposomal drug delivery and parenteral therapy of iron deficiency anemia (IDA) with iron oxide nanoparticles. We note that the clinical success of injectable iron exploits the inherent interaction between nanoparticles and the (innate) immune system, which designers of liposomal drug delivery seek to avoid. Magnetic fluid hyperthermia, a cancer therapy that harnesses magnetic hysteresis heating is approved for treating humans only with iron oxide nanoparticles. Despite its successful demonstration to enhance overall survival in clinical trials, this nanotechnology-based thermal medicine struggles to establish a clinical presence. We review the physical and biological attributes of this approach, and suggest reasons for barriers to its acceptance. Finally, despite the extensive clinical experience with iron oxide nanoparticles new and exciting research points to surprising immune-modulating potential. Recent data demonstrate the interactions between immune cells and iron oxide nanoparticles can induce anti-tumor immune responses. These present new and exciting opportunities to explore additional applications with this venerable technology. Clinical applications of iron oxide nanoparticles present poignant case studies of the opportunities, complexities, and challenges in cancer nanomedicine. They also illustrate the need for revised paradigms and multidisciplinary approaches to develop and translate nanomedicines into clinical cancer care.
Collapse
Affiliation(s)
- Frederik Soetaert
- Department of Electrical Energy, Metals, Mechanical Constructions and Systems, Ghent University, Belgium; Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Preethi Korangath
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - David Serantes
- Department of Applied Physics and Instituto de Investigacións Tecnolóxicas, Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain
| | - Steven Fiering
- Geisel School of Medicine, Dartmouth College, Lebanon, NH 03756, USA
| | - Robert Ivkov
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Oncology, Sidney Kimmel Comprehensive Cancer Centre, School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA; Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore 21218, USA; Department of Mechanical Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore 21218, USA.
| |
Collapse
|
78
|
Datta NR, Kok HP, Crezee H, Gaipl US, Bodis S. Integrating Loco-Regional Hyperthermia Into the Current Oncology Practice: SWOT and TOWS Analyses. Front Oncol 2020; 10:819. [PMID: 32596144 PMCID: PMC7303270 DOI: 10.3389/fonc.2020.00819] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 04/27/2020] [Indexed: 12/14/2022] Open
Abstract
Moderate hyperthermia at temperatures between 40 and 44°C is a multifaceted therapeutic modality. It is a potent radiosensitizer, interacts favorably with a host of chemotherapeutic agents, and, in combination with radiotherapy, enforces immunomodulation akin to “in situ tumor vaccination.” By sensitizing hypoxic tumor cells and inhibiting repair of radiotherapy-induced DNA damage, the properties of hyperthermia delivered together with photons might provide a tumor-selective therapeutic advantage analogous to high linear energy transfer (LET) neutrons, but with less normal tissue toxicity. Furthermore, the high LET attributes of hyperthermia thermoradiobiologically are likely to enhance low LET protons; thus, proton thermoradiotherapy would mimic 12C ion therapy. Hyperthermia with radiotherapy and/or chemotherapy substantially improves therapeutic outcomes without enhancing normal tissue morbidities, yielding level I evidence reported in several randomized clinical trials, systematic reviews, and meta-analyses for various tumor sites. Technological advancements in hyperthermia delivery, advancements in hyperthermia treatment planning, online invasive and non-invasive MR-guided thermometry, and adherence to quality assurance guidelines have ensured safe and effective delivery of hyperthermia to the target region. Novel biological modeling permits integration of hyperthermia and radiotherapy treatment plans. Further, hyperthermia along with immune checkpoint inhibitors and DNA damage repair inhibitors could further augment the therapeutic efficacy resulting in synthetic lethality. Additionally, hyperthermia induced by magnetic nanoparticles coupled to selective payloads, namely, tumor-specific radiotheranostics (for both tumor imaging and radionuclide therapy), chemotherapeutic drugs, immunotherapeutic agents, and gene silencing, could provide a comprehensive tumor-specific theranostic modality akin to “magic (nano)bullets.” To get a realistic overview of the strength (S), weakness (W), opportunities (O), and threats (T) of hyperthermia, a SWOT analysis has been undertaken. Additionally, a TOWS analysis categorizes future strategies to facilitate further integration of hyperthermia with the current treatment modalities. These could gainfully accomplish a safe, versatile, and cost-effective enhancement of the existing therapeutic armamentarium to improve outcomes in clinical oncology.
Collapse
Affiliation(s)
- Niloy R Datta
- Centre for Radiation Oncology KSA-KSB, Kantonsspital Aarau, Aarau, Switzerland
| | - H Petra Kok
- Department of Radiation Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Hans Crezee
- Department of Radiation Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Udo S Gaipl
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Stephan Bodis
- Centre for Radiation Oncology KSA-KSB, Kantonsspital Aarau, Aarau, Switzerland
| |
Collapse
|
79
|
Stoetzer O, Di Gioia D, Issels RD, Abdel-Rahman S, Mansmann U, Lindner LH, Gluz O, Würstlein R, Braun M, Hamann M, Edler von Koch F, Harbeck N, Salat C. Post-Neoadjuvant Gemcitabine and Cisplatin with Regional Hyperthermia for Patients with Triple-Negative Breast Cancer and Non-pCR after Neoadjuvant Chemotherapy: A Single-Institute Experience. Breast Care (Basel) 2020; 16:173-180. [PMID: 34012372 DOI: 10.1159/000507473] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 03/24/2020] [Indexed: 12/31/2022] Open
Abstract
Background Patients with triple-negative primary breast cancer (TNBC) who have residual invasive carcinoma after neoadjuvant chemotherapy have poor prognosis. Proven adjuvant approaches to reduce the risk of recurrence and improve outcome in patients with non-pathological complete response (non-pCR) are limited. Methods From our institutional registry, a consecutive case series of patients with operable, unilateral, primary invasive noninflammatory early TNBC of stage I-IIIB and pathologically verified residual cancer cells (no pathological complete response) after neoadjuvant chemotherapy underwent adjuvant treatment with gemcitabine plus cisplatin combined with regional hyperthermia. For quality assurance, we analyzed feasibility, efficacy, and toxicity of all treated patients. Outcome was evaluated for the entire group of patients as well as for the subgroups of patients with or without lymph node involvement at baseline (cN0/ cN+). Results From August 2012 to January 2019, we offered this treatment to 53 patients at our center as part of routine care. The median follow-up was 38 months. The majority of patients (64.2%) had cT2 tumors at baseline. Twenty-four patients (45%) were clinically node positive as evaluated by sonography. Thirty-nine patients (74%) had grade 3, and 14 patients (26%) had grade 2 tumors. Forty-one patients (76%) showed a regression grade 1 according to Sinn. Patients received a median of six treatment cycles of gemcitabine and cisplatin (range 1-6) combined with 12 applications of regional hyperthermia (median 12, range 2-12). Disease-free survival (DFS) at 3 years was 57.5%. In patients with no lymph node involvement at baseline (cN0), DFS at 3 years was significantly higher than in initially node-positive (cN+) patients (80 vs. 31%; p = 0.001). Overall survival (OS) at 3 years was 81.6%. In patients with no lymph node involvement at baseline (cN0), OS at 3 years was significantly higher than in node-positive (cN+) patients (93 vs. 70.4%; p = 0.02). Overall, grade 3/4 toxicities were leukopenia (38%), thrombocytopenia (4%), and anemia (4%). Conclusion After standard neoadjuvant chemotherapy containing anthracycline plus cyclophosphamide followed by taxanes, addition of adjuvant gemcitabine plus cisplatin in combination with regional hyperthermia was safe and effective in TNBC patients with non-pCR.
Collapse
Affiliation(s)
- Oliver Stoetzer
- Medical Center for Hematology and Oncology MVZ, Munich, Germany
| | - Dorit Di Gioia
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
| | - Rolf Dieter Issels
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
| | - Sultan Abdel-Rahman
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
| | - Ulrich Mansmann
- Institute of Epidemiology and Biostatistics, LMU Munich, Munich, Germany
| | | | - Oleg Gluz
- West German Study Group, Mönchengladbach, Germany
| | - Rachel Würstlein
- West German Study Group, Mönchengladbach, Germany.,Breast Center, Department of Obstetrics and Gynecology, and Comprehensive Cancer Center (CCC Munich LMU), University Hospital, LMU Munich, Munich, Germany
| | | | | | | | - Nadia Harbeck
- West German Study Group, Mönchengladbach, Germany.,Breast Center, Department of Obstetrics and Gynecology, and Comprehensive Cancer Center (CCC Munich LMU), University Hospital, LMU Munich, Munich, Germany
| | - Christoph Salat
- Medical Center for Hematology and Oncology MVZ, Munich, Germany
| |
Collapse
|
80
|
Duval KEA, Vernice NA, Wagner RJ, Fiering SN, Petryk JD, Lowry GJ, Tau SS, Yin J, Houde GR, Chaudhry AS, Hoopes PJ. Immunogenetic effects of low dose (CEM43 30) magnetic nanoparticle hyperthermia and radiation in melanoma cells. Int J Hyperthermia 2020; 36:37-46. [PMID: 31795829 PMCID: PMC6943912 DOI: 10.1080/02656736.2019.1627433] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Objective: In this in vitro study we have used an RNA quantification technique, nanoString, and a conventional protein analysis technique (Western Blot) to assess the genetic and protein expression of B16 murine melanoma cells following a modest magnetic nanoparticle hyperthermia (mNPH) dose equivalent to 30 minutes @ 43°C (CEM43 30) and/or a clinically relevant 8 Gy radiation dose. Methods: Melanoma cells with mNPs(2.5 μg Fe/106 cells) were pelleted and exposed to an alternating magnetic field (AMF) to generate the targeted thermal dose. Thermal dose was accurately monitored by a fiber optic probe and automatically maintained at CEM43 30. All cells were harvested 24 hours after treatment. Results: The mNPH dose demonstrated notable elevations in the thermotolerance/immunogenic HSP70 gene and a number of chemoattractant and toll-like receptor gene pathways. The 8 Gy dose also upregulated a number of important immune and cytotoxic genetic and protein pathways. However, the mNPH/radiation combination was the most effective stimulator of a wide variety of immune and cytotoxic genes including HSP70, cancer regulating chemokines CXCL10, CXCL11, the T-cell trafficking chemokine CXCR3, innate immune activators TLR3, TLR4, the MDM2 and mTOR negative regulator of p53, the pro-apoptotic protein PUMA, and the cell death receptor Fas. Importantly a number of the genetic changes were accurately validated by protein expression changes, i.e., HSP70, p-mTOR, p-MDM2. Conclusion: These results not only show that low dose mNPH and radiation independently increase the expression of important immune and cytotoxic genes but that the effect is greatly enhanced when they are used in combination.
Collapse
Affiliation(s)
- Kayla E A Duval
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | | | - Robert J Wagner
- Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | | | - James D Petryk
- Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | | | - Steven S Tau
- Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | - John Yin
- Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | - Georgia R Houde
- Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | | | - P Jack Hoopes
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA.,Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| |
Collapse
|
81
|
Oei AL, Korangath P, Mulka K, Helenius M, Coulter JB, Stewart J, Velarde E, Crezee J, Simons B, Stalpers LJA, Kok HP, Gabrielson K, Franken NAP, Ivkov R. Enhancing the abscopal effect of radiation and immune checkpoint inhibitor therapies with magnetic nanoparticle hyperthermia in a model of metastatic breast cancer. Int J Hyperthermia 2020; 36:47-63. [PMID: 31795835 PMCID: PMC7017719 DOI: 10.1080/02656736.2019.1685686] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Purpose: Enhancing immune responses in triple negative breast cancers (TNBCs) remains a challenge. Our study aimed to determine whether magnetic iron oxide nanoparticle (MION) hyperthermia (HT) can enhance abscopal effects with radiotherapy (RT) and immune checkpoint inhibitors (IT) in a metastatic TNBC model.Methods: One week after implanting 4T1-luc cells into the mammary glands of BALB/c mice, tumors were treated with RT (3 × 8 Gy)±local HT, mild (HTM, 43 °C/20 min) or partially ablative (HTAbl, 45 °C/5 min plus 43 °C/15 min),±IT with anti-PD-1 and anti-CTLA-4 antibodies (both 4 × 10 mg/kg, i.p.). Tumor growth was measured daily. Two weeks after treatment, lungs and livers were harvested for histopathology evaluation of metastases.Results: Compared to untreated controls, all treatment groups demonstrated a decreased tumor volume; however, when compared against surgical resection, only RT + HTM+IT, RT + HTAbl+IT and RT + HTAbl had similar or smaller tumors. These cohorts showed more infiltration of CD3+ T-lymphocytes into the primary tumor. Tumor growth effects were partially reversed with T-cell depletion. Combinations that proved most effective for primary tumors generated modest reductions in numbers of lung metastases. Conversely, numbers of lung metastases showed potential to increase following HT + IT treatment, particularly when compared to RT. Compared to untreated controls, there was no improvement in survival with any treatment.Conclusions: Single-fraction MION HT added to RT + IT improved local tumor control and recruitment of CD3+ T-lymphocytes, with only a modest effect to reduce lung metastases and no improvement in overall survival. HT + IT showed potential to increase metastatic dissemination to lungs.
Collapse
Affiliation(s)
- Arlene L Oei
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, University of Amsterdam, Amsterdam, The Netherlands.,Department of Radiation Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Preethi Korangath
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kathleen Mulka
- Department of Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mikko Helenius
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jonathan B Coulter
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jacqueline Stewart
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Esteban Velarde
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Johannes Crezee
- Department of Radiation Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Brian Simons
- Department of Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lukas J A Stalpers
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, University of Amsterdam, Amsterdam, The Netherlands.,Department of Radiation Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - H Petra Kok
- Department of Radiation Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Kathleen Gabrielson
- Department of Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nicolaas A P Franken
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, University of Amsterdam, Amsterdam, The Netherlands.,Department of Radiation Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Robert Ivkov
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Mechanical Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA.,Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
82
|
Payne M, Bossmann SH, Basel MT. Direct treatment versus indirect: Thermo-ablative and mild hyperthermia effects. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 12:e1638. [PMID: 32352660 DOI: 10.1002/wnan.1638] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 03/02/2020] [Accepted: 04/07/2020] [Indexed: 11/11/2022]
Abstract
Hyperthermia is a rapidly growing field in cancer therapy and many advances have been made in understanding and applying the mechanisms of hyperthermia. Secondary effects of hyperthermia have been increasingly recognized as important in therapeutic effects and multiple studies have started to elucidate their implications for treatment. Immune effects have especially been recognized as important in the efficacy of hyperthermia treatment of cancer. Both thermo-ablative and mild hyperthermia activate the immune system, but mild hyperthermia seems to be more effective at doing so. This may suggest that mild hyperthermia has some advantages over thermo-ablative hyperthermia and research into immune effects of mild hyperthermia should continue. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Therapeutic Approaches and Drug Discovery > Emerging Technologies Implantable Materials and Surgical Technologies > Nanoscale Tools and Techniques in Surgery.
Collapse
Affiliation(s)
- Macy Payne
- Department of Chemistry, Kansas State University, Manhattan, Kansas, USA
| | - Stefan H Bossmann
- Department of Chemistry, Kansas State University, Manhattan, Kansas, USA
| | - Matthew T Basel
- Department of Anatomy & Physiology, Kansas State University, Manhattan, Kansas, USA
| |
Collapse
|
83
|
Hader M, Savcigil DP, Rosin A, Ponfick P, Gekle S, Wadepohl M, Bekeschus S, Fietkau R, Frey B, Schlücker E, Gaipl US. Differences of the Immune Phenotype of Breast Cancer Cells after Ex Vivo Hyperthermia by Warm-Water or Microwave Radiation in a Closed-Loop System Alone or in Combination with Radiotherapy. Cancers (Basel) 2020; 12:cancers12051082. [PMID: 32349284 PMCID: PMC7281749 DOI: 10.3390/cancers12051082] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/17/2020] [Accepted: 04/18/2020] [Indexed: 12/20/2022] Open
Abstract
The treatment of breast cancer by radiotherapy can be complemented by hyperthermia. Little is known about how the immune phenotype of tumor cells is changed thereby, also in terms of a dependence on the heating method. We developed a sterile closed-loop system, using either a warm-water bath or a microwave at 2.45 GHz to examine the impact of ex vivo hyperthermia on cell death, the release of HSP70, and the expression of immune checkpoint molecules (ICMs) on MCF-7 and MDA-MB-231 breast cancer cells by multicolor flow cytometry and ELISA. Heating was performed between 39 and 44 °C. Numerical process simulations identified temperature distributions. Additionally, irradiation with 2 × 5 Gy or 5 × 2 Gy was applied. We observed a release of HSP70 after hyperthermia at all examined temperatures and independently of the heating method, but microwave heating was more effective in cell killing, and microwave heating with and without radiotherapy increased subsequent HSP70 concentrations. Adding hyperthermia to radiotherapy, dynamically or individually, affected the expression of the ICM PD-L1, PD-L2, HVEM, ICOS-L, CD137-L, OX40-L, CD27-L, and EGFR on breast cancer cells. Well-characterized pre-clinical heating systems are mandatory to screen the immune phenotype of tumor cells in clinically relevant settings to define immune matrices for therapy adaption.
Collapse
Affiliation(s)
- Michael Hader
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (M.H.); (D.P.S.); (R.F.); (B.F.)
- Chair for Ceramic Materials Engineering, Keylab Glass Technology, University of Bayreuth, 95447 Bayreuth, Germany; (A.R.); (P.P.)
| | - Deniz Pinar Savcigil
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (M.H.); (D.P.S.); (R.F.); (B.F.)
| | - Andreas Rosin
- Chair for Ceramic Materials Engineering, Keylab Glass Technology, University of Bayreuth, 95447 Bayreuth, Germany; (A.R.); (P.P.)
| | - Philipp Ponfick
- Chair for Ceramic Materials Engineering, Keylab Glass Technology, University of Bayreuth, 95447 Bayreuth, Germany; (A.R.); (P.P.)
| | - Stephan Gekle
- Biofluid Simulations and Modeling, Fachbereich Physik, University of Bayreuth, 95447 Bayreuth, Germany;
| | | | - Sander Bekeschus
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology, Felix-Hausdorff-Str. 2, 17489 Greifswald, Germany;
| | - Rainer Fietkau
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (M.H.); (D.P.S.); (R.F.); (B.F.)
| | - Benjamin Frey
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (M.H.); (D.P.S.); (R.F.); (B.F.)
| | - Eberhard Schlücker
- Department of Chemical and Biological Engineering, Institute of Process Machinery and Systems Engineering (iPAT), Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany;
| | - Udo S. Gaipl
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (M.H.); (D.P.S.); (R.F.); (B.F.)
- Correspondence: ; Tel.: +49-9131-8544-258; Fax: +49-9131-8539-335
| |
Collapse
|
84
|
Sharma A, Rudek MA, Korangath P, Bunz F, Ivkov R. For HIPEC, synergistic effects of hyperthermia and doxorubicin are optimal when simultaneously combined. Int J Hyperthermia 2020; 37:346-348. [PMID: 32270728 DOI: 10.1080/02656736.2020.1750714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Affiliation(s)
- Anirudh Sharma
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michelle A Rudek
- Department of Medicine, Division of Clinical Pharmacology, Johns Hopkins University, Baltimore, MD, USA
| | - Preethi Korangath
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Fred Bunz
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert Ivkov
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, USA.,Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA.,Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
85
|
Datta NR, Bodis S. Hyperthermia with photon radiotherapy is thermoradiobiologically analogous to neutrons for tumors without enhanced normal tissue toxicity. Int J Hyperthermia 2020; 36:1073-1078. [PMID: 31709846 DOI: 10.1080/02656736.2019.1679895] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
The depth dose profiles of photons mirror those of fast neutrons. However, in contrast to the high linear energy transfer (LET) characteristics of neutrons; photons exhibit low LET features. Hyperthermia (HT) inhibits the repair of radiation-induced DNA damage and is cytotoxic to the radioresistant hypoxic tumor cells. Thus, thermoradiobiologically, HT simulates high LET radiation with photons. At temperatures of 39-45 °C, the physiological vasodilation allows rapid heat dissipation from normal tissues. On the contrary, the chaotic and relatively rigid tumor vasculature results in heat retention leading to higher intratumoural temperatures. Consequently, the high LET attributes of HT with photon radiations are mostly limited to the confines of the heated tumor while the normothermic normal tissues would be irradiated with low LET photons. HT thereby augments photon therapy by conferring therapeutic advantages of high LET radiations to the tumors akin to neutrons, while the 'heat-sink' effect spares the normal tissues from thermal radiosensitization. Thus, photon thermoradiotherapy imparts radiobiological advantages selectively to tumors analogous to neutrons without exaggerating normal tissue morbidities. The later has been the major concern with clinical fast neutron beam therapy. Outcomes reported from several clinical trials in diverse tumor sites add testimony to the enhanced therapeutic efficacy of photon thermoradiotherapy.
Collapse
Affiliation(s)
- Niloy Ranjan Datta
- Department of Radiation Oncology, KSA-KSB, Kantonsspital Aarau, Aarau, Switzerland
| | - Stephan Bodis
- Department of Radiation Oncology, KSA-KSB, Kantonsspital Aarau, Aarau, Switzerland.,Department of Radiation Oncology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
86
|
Paulides M, Dobsicek Trefna H, Curto S, Rodrigues D. Recent technological advancements in radiofrequency- andmicrowave-mediated hyperthermia for enhancing drug delivery. Adv Drug Deliv Rev 2020; 163-164:3-18. [PMID: 32229271 DOI: 10.1016/j.addr.2020.03.004] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 03/20/2020] [Accepted: 03/24/2020] [Indexed: 12/23/2022]
Abstract
Hyperthermia therapy is a potent enhancer of chemotherapy and radiotherapy. In particular, microwave (MW) and radiofrequency (RF) hyperthermia devices provide a variety of heating approaches that can treat most cancers regardless the size. This review introduces the physics of MW/RF hyperthermia, the current state-of-the-art systems for both localized and regional heating, and recent advancements in hyperthermia treatment guidance using real-time computational simulations and magnetic resonance thermometry. Clinical trials involving RF/MW hyperthermia as adjuvant for chemotherapy are also presented per anatomical site. These studies favor the use of adjuvant hyperthermia since it significantly improves curative and palliative clinical outcomes. The main challenge of hyperthermia is the distribution of state-of-the-art heating systems. Nevertheless, we anticipate that recent technology advances will expand the use of hyperthermia to chemotherapy centers for enhanced drug delivery. These new technologies hold great promise not only for (image-guided) perfusion modulation and sensitization for cytotoxic drugs, but also for local delivery of various compounds using thermosensitive liposomes.
Collapse
|
87
|
Minnaar CA, Kotzen JA, Ayeni OA, Vangu MDT, Baeyens A. Potentiation of the Abscopal Effect by Modulated Electro-Hyperthermia in Locally Advanced Cervical Cancer Patients. Front Oncol 2020; 10:376. [PMID: 32266151 PMCID: PMC7105641 DOI: 10.3389/fonc.2020.00376] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 03/04/2020] [Indexed: 11/19/2022] Open
Abstract
Background: A Phase III randomized controlled trial investigating the addition of modulated electro-hyperthermia (mEHT) to chemoradiotherapy for locally advanced cervical cancer patients is being conducted in South Africa (Human Research Ethics Committee approval: M1704133; ClincialTrials.gov ID: NCT03332069). Two hundred and ten participants were randomized and 202 participants were eligible for six month local disease control evaluation. Screening 18F-FDG PET/CT scans were conducted and repeated at six months post-treatment. Significant improvement in local control was reported in the mEHT group and complete metabolic resolution (CMR) of extra-pelvic disease was noted in some participants. We report on an analysis of the participants with CMR of disease inside and outside the radiation field. Method: Participants were included in this analysis if nodes outside the treatment field (FDG-uptake SUV>2.5) were visualized on pre-treatment scans and if participants were evaluated by 18F-FDG PET/CT scans at six months post-treatment. Results: One hundred and eight participants (mEHT: HIV-positive n = 25, HIV-negative n = 29; Control Group: HIV-positive n = 26, HIV-negative n = 28) were eligible for analysis. There was a higher CMR of all disease inside and outside the radiation field in the mEHT Group: n = 13 [24.1%] than the control group: n = 3 [5.6%] (Chi squared, Fisher's exact: p = 0.013) with no significant difference in the extra-pelvic response to treatment between the HIV-positive and -negative participants of each group. Conclusion: The CMR of disease outside the radiation field at six months post-treatment provides evidence of an abscopal effect which was significantly associated with the addition of mEHT to treatment protocols. This finding is important as the combined synergistic use of radiotherapy with mEHT could broaden the scope of radiotherapy to include systemic disease.
Collapse
Affiliation(s)
- Carrie Anne Minnaar
- Radiobiology, Department of Radiation Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Jeffrey Allan Kotzen
- Radiation Oncology, Wits Donald Gordon Medical Centre, Johannesburg, South Africa
| | - Olusegun Akinwale Ayeni
- Nuclear Medicine, Department of Radiation Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Mboyo-Di-Tamba Vangu
- Nuclear Medicine, Department of Radiation Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Ans Baeyens
- Radiobiology, Department of Radiation Sciences, University of the Witwatersrand, Johannesburg, South Africa.,Radiobiology, Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| |
Collapse
|
88
|
Notter M, Thomsen AR, Nitsche M, Hermann RM, Wolff HA, Habl G, Münch K, Grosu AL, Vaupel P. Combined wIRA-Hyperthermia and Hypofractionated Re-Irradiation in the Treatment of Locally Recurrent Breast Cancer: Evaluation of Therapeutic Outcome Based on a Novel Size Classification. Cancers (Basel) 2020; 12:cancers12030606. [PMID: 32155740 PMCID: PMC7139693 DOI: 10.3390/cancers12030606] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/02/2020] [Accepted: 03/03/2020] [Indexed: 02/06/2023] Open
Abstract
Effective tumor control in patients suffering from unresectable locally recurrent breast cancer (LRBC) in pre-irradiated areas can be achieved by re-irradiation combined with superficial hyperthermia. Using this combined modality, total re-irradiation dose and toxicity can be significantly reduced compared to conventionally fractionated treatment schedules with total doses of 60–66 Gy. Applying contact-free, thermography-controlled water-filtered infrared-A superficial hyperthermia, immediately followed by hypofractionated re-irradiation, consisting of 4 Gy once per week up to a total dose of 20 Gy, resulted in high overall response rates even in large-sized tumors. Comparability of clinical data between different combined Hyperthermia (HT)/Radiotherapy (RT) treatment schedules is impeded by the highly individual characteristics of this disease. Tumor size, ranging from microscopic disease and small lesions to large-sized cancer en cuirasse, is described as one of the most important prognostic factors. However, in clinical studies and analyses of LRBC, tumor size has so far been reported in a very heterogeneous way. Therefore, we suggest a novel, simple and feasible size classification (rClasses 0–IV). Applying this classification for the evaluation of 201 patients with pre-irradiated LRBC allowed for a stratification into distinct prognostic groups.
Collapse
Affiliation(s)
- Markus Notter
- Department of Radiation Oncology, Lindenhofspital Bern, 3012 Bern, Switzerland; (M.N.); (K.M.)
| | - Andreas R. Thomsen
- Department of Radiation Oncology, Medical Center, University of Freiburg, 79106 Freiburg, Germany; (A.R.T.); (A.-L.G.)
- German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Mirko Nitsche
- Center for Radiotherapy and Radiooncology Bremen and Westerstede, 28239 Bremen, Germany; (M.N.); (R.M.H.)
| | - Robert M. Hermann
- Center for Radiotherapy and Radiooncology Bremen and Westerstede, 28239 Bremen, Germany; (M.N.); (R.M.H.)
| | - Hendrik A. Wolff
- Department of Radiology, Nuclear Medicine and Radiotherapy, Radiology Munich, 80333 Munich, Germany; (H.A.W.); (G.H.)
- Department of Radiation Oncology, Medical Center, University of Regensburg, 93053 Regensburg, Germany
| | - Gregor Habl
- Department of Radiology, Nuclear Medicine and Radiotherapy, Radiology Munich, 80333 Munich, Germany; (H.A.W.); (G.H.)
- Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Karin Münch
- Department of Radiation Oncology, Lindenhofspital Bern, 3012 Bern, Switzerland; (M.N.); (K.M.)
| | - Anca-L. Grosu
- Department of Radiation Oncology, Medical Center, University of Freiburg, 79106 Freiburg, Germany; (A.R.T.); (A.-L.G.)
- German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Peter Vaupel
- Department of Radiation Oncology, Medical Center, University of Freiburg, 79106 Freiburg, Germany; (A.R.T.); (A.-L.G.)
- German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Correspondence: ; Tel.: +49-171-124-0073
| |
Collapse
|
89
|
Cheng Y, Weng S, Yu L, Zhu N, Yang M, Yuan Y. The Role of Hyperthermia in the Multidisciplinary Treatment of Malignant Tumors. Integr Cancer Ther 2020; 18:1534735419876345. [PMID: 31522574 PMCID: PMC7242805 DOI: 10.1177/1534735419876345] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Hyperthermia is often used in combination with chemotherapy and radiotherapy for
cancer treatment. Recently, immunotherapy has become a popular research area,
breaking exciting new ground with concurrent immunotherapy and hyperthermia.
Much evidence has demonstrated the effectiveness of multidisciplinary
synergistic therapy, and the underlying mechanism has been gradually explored.
In this review, we focus on the mechanism of various cancer treatments in the
current literature and recent advances in hyperthermia. Additionally, we review
clinical studies of hyperthermia combined with other therapies in the previous
10 years and propose future prospects for hyperthermia in multidisciplinary
synergistic therapy.
Collapse
Affiliation(s)
- Yi Cheng
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Shanshan Weng
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Linzhen Yu
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Ning Zhu
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Mengyuan Yang
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Ying Yuan
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| |
Collapse
|
90
|
Eleftheriou K, Kaminari A, Panagiotaki KN, Sideratou Z, Zachariadis M, Anastassopoulou J, Tsiourvas D. A combination drug delivery system employing thermosensitive liposomes for enhanced cell penetration and improved in vitro efficacy. Int J Pharm 2020; 574:118912. [PMID: 31809858 DOI: 10.1016/j.ijpharm.2019.118912] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 11/15/2019] [Accepted: 11/27/2019] [Indexed: 02/06/2023]
Abstract
Drug-loaded thermosensitive liposomes are investigated as drug delivery systems in combination with local mild hyperthermia therapy due to their capacity to release their cargo at a specific temperature range (40-42 °C). Additional benefit can be achieved by the development of such systems that combine two different anticancer drugs, have cell penetration properties and, when heated, release their drug payload in a controlled fashion. To this end, liposomes were developed incorporating at low concentration (5 mol%) a number of monoalkylether phosphatidylcholine lipids, encompassing the platelet activating factor, PAF, and its analogues that induce thermoresponsiveness and have anticancer biological activity. These thermoresponsive liposomes were efficiently (>90%) loaded with doxorubicin (DOX), and their thermal properties, stability and drug release were investigated both at 37 ◦C and at elevated temperatures. In vitro studies of the most advantageous liposomal formulation containing the methylated PAF derivative (methyl-PAF, edelfosine), an established antitumor agent, were performed on human prostate cancer cell lines. This system exhibits controlled release of DOX at 40-42 °C, enhanced cell uptake due to the presence of methyl-PAF, and improved cell viability inhibition due to the combined action of both medications.
Collapse
Affiliation(s)
- Kleopatra Eleftheriou
- Institute of Nanoscience and Nanotechnology, NCSR ''Demokritos", 15310 Aghia Paraskevi, Greece
| | - Archontia Kaminari
- Institute of Nanoscience and Nanotechnology, NCSR ''Demokritos", 15310 Aghia Paraskevi, Greece
| | - Katerina N Panagiotaki
- Institute of Nanoscience and Nanotechnology, NCSR ''Demokritos", 15310 Aghia Paraskevi, Greece
| | - Zili Sideratou
- Institute of Nanoscience and Nanotechnology, NCSR ''Demokritos", 15310 Aghia Paraskevi, Greece
| | - Michael Zachariadis
- Institute of Biosciences and Applications, NCSR ''Demokritos", 15310 Aghia Paraskevi, Greece
| | - Jane Anastassopoulou
- Radiation Chemistry and Biospectroscopy, School of Chemical Engineering, National Technical University of Athens, Athens, Greece
| | - Dimitris Tsiourvas
- Institute of Nanoscience and Nanotechnology, NCSR ''Demokritos", 15310 Aghia Paraskevi, Greece.
| |
Collapse
|
91
|
Immune biological rationales for the design of combined radio- and immunotherapies. Cancer Immunol Immunother 2020; 69:293-306. [PMID: 31953578 PMCID: PMC7000501 DOI: 10.1007/s00262-019-02460-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 12/22/2019] [Indexed: 02/07/2023]
Abstract
Cancer immunotherapies are promising treatments for many forms of cancer. Nevertheless, the response rates to, e.g., immune checkpoint inhibitors (ICI), are still in low double-digit percentage. This calls for further therapy optimization that should take into account combination of immunotherapies with classical tumor therapies such as radiotherapy. By designing multimodal approaches, immune modulatory properties of certain radiation schemes, additional immune modulation by immunotherapy with ICI and hyperthermia, as well as patient stratification based on genetic and immune constitutions have to be considered. In this context, both the tumor and its microenvironment including cells of the innate and adaptive immune system have to be viewed in synopsis. Knowledge of immune activation and immune suppression by radiation is the basis for well-elaborated addition of certain immunotherapies. In this review, the focus is set on additional immune stimulation by hyperthermia and restoration of an immune response by ICI. The impact of radiation dose and fractionation on immune modulation in multimodal settings has to be considered, as the dynamics of the immune response and the timing between radiotherapy and immunotherapy. Another big challenge is the patient stratification that should be based on matrices of biomarkers, taking into account genetics, proteomics, radiomics, and “immunomics”. One key aim is to turn immunological “cold” tumors into “hot” tumors, and to eliminate barriers of immune-suppressed or immune-excluded tumors. Comprehensive knowledge of immune alterations induced by radiation and immunotherapy when being applied together should be utilized for patient-adapted treatment planning and testing of innovative tumor therapies within clinical trials.
Collapse
|
92
|
Faridi P, Bossmann SH, Prakash P. Simulation-based design and characterization of a microwave applicator for MR-guided hyperthermia experimental studies in small animals. Biomed Phys Eng Express 2020; 6:015001. [PMID: 32999735 PMCID: PMC7521833 DOI: 10.1088/2057-1976/ab36dd] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Purpose The objective of this study was to design and characterize a 2.45 GHz microwave hyperthermia applicator for delivering hyperthermia in experimental small animals to 2 - 4 mm diameter targets located 1 - 3 mm from the skin surface, with minimal heating of the surrounding tissue, under 14.1 T MRI real-time monitoring and feedback control. Materials and methods An experimentally validated 3D computational model was employed to design and characterize a non-invasive directional water-cooled microwave hyperthermia applicator. We assessed the effects of: reflector geometry, monopole shape, cooling water temperature, and flow rate on spatial-temperature profiles. The system was integrated with real-time MR thermometry and feedback control to monitor and maintain temperature elevations in the range of 4 - 5 °C at 1 - 3 mm from the applicator surface. The quality of heating was quantified by determining the fraction of the target volume heated to the desired temperature, and the extent of heating in non-targeted regions. Results Model-predicted hyperthermic profiles were in good agreement with experimental measurements (Dice Similarity Coefficient of 0.95 - 0.99). Among the four considered criteria, a reflector aperture angle of 120 °, S-shaped monopole antenna with 0.6 mm displacement, and coolant flow rate of 150 ml/min were selected as the end result of the applicator design. The temperature of circulating water and input power were identified as free variables, allowing considerable flexibility in heating target sizes within varying distances from the applicator surface. 2 - 4 mm diameter targets positioned 1 - 3 mm from the applicator surface were heated to hyperthermic temperatures, with target coverage ratio ranging between 76 - 93 % and 11 - 26 % of non-targeted tissue heated. Conclusion We have designed an experimental platform for MR-guided hyperthermia, incorporating a microwave applicator integrated with temperature-based feedback control to heat deep-seated targets for experimental studies in small animals.
Collapse
Affiliation(s)
- Pegah Faridi
- Department of Electrical and Computer Engineering, Kansas State University, Manhattan, KS 66506, USA
| | - Stefan H. Bossmann
- Department of Chemistry, Kansas State University, Manhattan, KS 66506, USA
| | - Punit Prakash
- Department of Electrical and Computer Engineering, Kansas State University, Manhattan, KS 66506, USA
| |
Collapse
|
93
|
Oei A, Kok H, Oei S, Horsman M, Stalpers L, Franken N, Crezee J. Molecular and biological rationale of hyperthermia as radio- and chemosensitizer. Adv Drug Deliv Rev 2020; 163-164:84-97. [PMID: 31982475 DOI: 10.1016/j.addr.2020.01.003] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 11/11/2019] [Accepted: 01/20/2020] [Indexed: 12/24/2022]
Abstract
Mild hyperthermia, local heating of the tumour up to temperatures <43 °C, has been clinically applied for almost four decades and has been proven to substantially enhance the effectiveness of both radiotherapy and chemotherapy in treatment of primary and recurrent tumours. Clinical results and mechanisms of action are discussed in this review, including the molecular and biological rationale of hyperthermia as radio- and chemosensitizer as established in in vitro and in vivo experiments. Proven mechanisms include inhibition of different DNA repair processes, (in)direct reduction of the hypoxic tumour cell fraction, enhanced drug uptake, increased perfusion and oxygen levels. All mechanisms show different dose effect relationships and different optimal scheduling with radiotherapy and chemotherapy. Therefore, obtaining the ideal multi-modality treatment still requires elucidation of more detailed data on dose, sequence, duration, and possible synergisms between modalities. A multidisciplinary approach with different modalities including hyperthermia might further increase anti-tumour effects and diminish normal tissue damage.
Collapse
|
94
|
Zheng YM, Chow JM, Chang CL, Chao LH, Whang-Peng J, Lai GM. Hematuria induced by combination regorafenib and hyperthermia – a radiation recall effect. Int J Hyperthermia 2019; 36:1186-1189. [DOI: 10.1080/02656736.2019.1687941] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Affiliation(s)
- Yu-Mei Zheng
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Wan Fang Hospital, Taipei, Taiwan
| | - Jyh-Ming Chow
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Wan Fang Hospital, Taipei, Taiwan
- College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chia-Lun Chang
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Wan Fang Hospital, Taipei, Taiwan
- College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Liang-Hsiao Chao
- Consultant, Medical Physicist, Linden Bioscience Co, Taipei, Taiwan
| | - Jacqueline Whang-Peng
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Wan Fang Hospital, Taipei, Taiwan
- College of Medicine, Taipei Medical University, Taipei, Taiwan
- Cancer Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Taipei Cancer Center, Taipei Medical University, Taipei, Taiwan
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan
| | - Gi-Ming Lai
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Wan Fang Hospital, Taipei, Taiwan
- College of Medicine, Taipei Medical University, Taipei, Taiwan
- Cancer Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Taipei Cancer Center, Taipei Medical University, Taipei, Taiwan
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan
| |
Collapse
|
95
|
Tan WP, Longo TA, Inman BA. Heated Intravesical Chemotherapy: Biology and Clinical Utility. Urol Clin North Am 2019; 47:55-72. [PMID: 31757301 DOI: 10.1016/j.ucl.2019.09.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Non-muscle-invasive bladder cancer can be a challenging disease to manage. In recent years, hyperthermia therapy in conjunction with intravesical therapy has been gaining traction as a treatment option for bladder cancer, especially if Bacillus Calmette-Guerin might not be available. Trials of intravesical chemotherapy with heat are few and there has been considerable heterogeneity between studies. However, multiple new trials have accrued and high-quality data are forthcoming. In this review, we discuss the role of combined intravesical hyperthermia and chemotherapy as a novel approach for the treatment of bladder cancer.
Collapse
Affiliation(s)
- Wei Phin Tan
- Division of Urology, Duke University Medical Center, Durham, NC 27710, USA
| | - Thomas A Longo
- Division of Urology, Duke University Medical Center, Durham, NC 27710, USA
| | - Brant A Inman
- Division of Urology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
96
|
Ma L, Kambe R, Tsuchiya T, Kanegasaki S, Takahashi A. Anti-Metastatic Benefits Produced by Hyperthermia and a CCL3 Derivative. Cancers (Basel) 2019; 11:cancers11111770. [PMID: 31717914 PMCID: PMC6895898 DOI: 10.3390/cancers11111770] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 11/05/2019] [Indexed: 12/12/2022] Open
Abstract
Significant numbers of malignant tumor cells that have spread to surrounding tissues and other distant organs are often too small to be picked up in a diagnostic test, and prevention of even such small metastases should improve patient outcomes. Using a mouse model, we show in this article that intravenous administration of a human CCL3 variant carrying a single amino acid substitution after mild local hyperthermia not only induces tumor growth inhibition at the treated site but also inhibits metastasis. Colon26 adenocarcinoma cells (1 × 105 cells/mouse) were grafted subcutaneously into the right hind leg of syngeneic BALB/c mice and after nine days, when tumor size reached ~11 mm in diameter, the local tumor mass was exposed to high-frequency waves, by which intratumoral temperature was maintained at 42 °C for 30 min. Mice received the CCL3 variant named eMIP (2 μg/mouse/day) intravenously for five consecutive days starting one day after heat treatment. We found that tumor growth in eMIP recipients after hyperthermia was inhibited markedly but no effect was seen in animals treated with either hyperthermia or eMIP alone. Furthermore, the number of lung metastases evaluated at 18 days after hyperthermia treatment was dramatically reduced in animals receiving the combination therapy compared with all other controls. These results encourage future clinical application of this combination therapy.
Collapse
Affiliation(s)
- Liqiu Ma
- Gunma University Heavy Ion Medical Center, Gunma 371-8511, Japan; (L.M.); (R.K.)
- China Institute of Atomic Energy, Beijing 102413, China
| | - Ryosuke Kambe
- Gunma University Heavy Ion Medical Center, Gunma 371-8511, Japan; (L.M.); (R.K.)
| | - Tomoko Tsuchiya
- Research Institute, National Center for Global Health and Medicine, Tokyo 162-8655, Japan; (T.T.); (S.K.)
| | - Shiro Kanegasaki
- Research Institute, National Center for Global Health and Medicine, Tokyo 162-8655, Japan; (T.T.); (S.K.)
| | - Akihisa Takahashi
- Gunma University Heavy Ion Medical Center, Gunma 371-8511, Japan; (L.M.); (R.K.)
- Correspondence: ; Tel.: +81-27-220-7917
| |
Collapse
|
97
|
Guo Y, Ran Y, Wang Z, Cheng J, Cao Y, Yang C, Liu F, Ran H. Magnetic-responsive and targeted cancer nanotheranostics by PA/MR bimodal imaging-guided photothermally triggered immunotherapy. Biomaterials 2019; 219:119370. [DOI: 10.1016/j.biomaterials.2019.119370] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 07/03/2019] [Accepted: 07/17/2019] [Indexed: 12/29/2022]
|
98
|
Zhou J, Li L, Li X, Yu Q, Cui S, Shu K, Liu J, Liu J, Ding D, Du T. Efficacy analysis of a novel thermochemotherapy scheme with pirarubicin for intermediate- and high-risk nonmuscle-invasive bladder cancer: a single-institution nonrandomized concurrent controlled trial. Int J Hyperthermia 2019; 36:868-875. [PMID: 31452420 DOI: 10.1080/02656736.2019.1646929] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Affiliation(s)
- Jun Zhou
- Department of Urology, Henan University People's Hospital, Zhengzhou, P.R. China
| | - Linlin Li
- Department of Urology, Henan Provincial People's Hospital, Zhengzhou, P.R. China
| | - Xing Li
- Department of Urology, Zhengzhou University People's Hospital, Zhengzhou, P.R. China
| | - Qian Yu
- Department of Urology, Henan University People's Hospital, Zhengzhou, P.R. China
| | - Shaowei Cui
- Department of Urology, Henan University People's Hospital, Zhengzhou, P.R. China
| | - Kunpeng Shu
- Department of Urology, Henan Provincial People's Hospital, Zhengzhou, P.R. China
| | - Jianjun Liu
- Department of Urology, Henan University People's Hospital, Zhengzhou, P.R. China
- Department of Urology, Henan Provincial People's Hospital, Zhengzhou, P.R. China
| | - Jie Liu
- Department of Urology, Henan University People's Hospital, Zhengzhou, P.R. China
- Department of Urology, Henan Provincial People's Hospital, Zhengzhou, P.R. China
| | - Degang Ding
- Department of Urology, Henan University People's Hospital, Zhengzhou, P.R. China
- Department of Urology, Henan Provincial People's Hospital, Zhengzhou, P.R. China
- Department of Urology, Zhengzhou University People's Hospital, Zhengzhou, P.R. China
| | - Tao Du
- Department of Urology, Henan University People's Hospital, Zhengzhou, P.R. China
- Department of Urology, Henan Provincial People's Hospital, Zhengzhou, P.R. China
- Department of Urology, Zhengzhou University People's Hospital, Zhengzhou, P.R. China
| |
Collapse
|
99
|
Aghamiri S, Mehrjardi KF, Shabani S, Keshavarz-Fathi M, Kargar S, Rezaei N. Nanoparticle-siRNA: a potential strategy for ovarian cancer therapy? Nanomedicine (Lond) 2019; 14:2083-2100. [PMID: 31368405 DOI: 10.2217/nnm-2018-0379] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Ovarian cancer is one of the most common causes of mortality throughout the world. Unfortunately, chemotherapy has failed to cure advanced cancers developing multidrug resistance (MDR). Moreover, it has critical side effects because of nonspecific toxicity. Thanks to specific silencing of oncogenes and MDR-associated genes, nano-siRNA drugs can be a great help address the limitations of chemotherapy. Here, we review the current advances in nanoparticle-mediated siRNA delivery strategies such as polymeric- and lipid-based systems, rigid nanoparticles and nanoparticles coupled to specific ligand systems. Nanoparticle-based codelivery of anticancer drugs and siRNA targeting various mechanisms of MDR is a cutting-edge strategy for ovarian cancer therapy, which is completely discussed in this review.
Collapse
Affiliation(s)
- Shahin Aghamiri
- Student Research Committee, Department of Medical Biotechnology, School of Advanced Technology in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, 19839-63113, Iran
| | - Keyvan Fallah Mehrjardi
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, 1416753955, Iran.,Cancer Immunology Project (CIP), Universal Scientific Education & Research Network (USERN), Tehran, 1419733151, Iran
| | - Sasan Shabani
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, 1416753955, Iran
| | - Mahsa Keshavarz-Fathi
- Cancer Immunology Project (CIP), Universal Scientific Education & Research Network (USERN), Tehran, 1419733151, Iran.,Students' Scientific Research Center, School of Medicine, Tehran University of Medical Sciences, Tehran, 1416753955, Iran
| | - Saeed Kargar
- Department of Life Science Engineering, Faculty of New Sciences & Technologies, University of Tehran, Tehran, 1417466191, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, 1419733151, Iran
| |
Collapse
|
100
|
Merten R, Ott O, Haderlein M, Bertz S, Hartmann A, Wullich B, Keck B, Kühn R, Rödel CM, Weiss C, Gall C, Uter W, Fietkau R. Long-Term Experience of Chemoradiotherapy Combined with Deep Regional Hyperthermia for Organ Preservation in High-Risk Bladder Cancer (Ta, Tis, T1, T2). Oncologist 2019; 24:e1341-e1350. [PMID: 31292267 PMCID: PMC6975936 DOI: 10.1634/theoncologist.2018-0280] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 02/11/2019] [Indexed: 01/19/2023] Open
Abstract
This article reports on the different treatments for bladder cancer and related effects on frequency of bladder preservation, disease‐free survival, and overall survival, with a focus on the efficacy and safety of chemoradiotherapy combined with regional deep hyperthermia of high‐risk bladder cancer after transurethral resection of bladder tumor. Background. The aim of this study was to evaluate the efficacy and safety of chemoradiotherapy (RCT) combined with regional deep hyperthermia (RHT) of high‐risk bladder cancer after transurethral resection of bladder tumor (TUR‐BT). Materials and methods. Between 1982 and 2016, 369 patients with pTa, pTis, pT1, and pT2 cN0–1 cM0 bladder cancer were treated with a multimodal treatment after TUR‐BT. All patients received radiotherapy (RT) of the bladder and regional lymph nodes. RCT was administered to 215 patients, RCT + RHT was administered to 79 patients, and RT was used in 75 patients. Treatment response was evaluated 4–6 weeks after treatment with TUR‐BT. Results. Complete response (CR) overall was 83% (290/351), and in treatment groups was RT 68% (45/66), RCT 86% (178/208), and RCT + RHT 87% (67/77). CR was significantly improved by concurrent RCT compared with RT (odds ratio [OR], 2.32; 95% confidence interval [CI], 1.05–5.12; p = .037), less influenced by hyperthermia (OR, 2.56; 95% CI, 0.88–8.00; p = .092). Overall survival (OS) after RCT was superior to RT (hazard ratio [HR], 0.7; 95% CI, 0.50–0.99; p = .045). Five‐year OS from unadjusted Kaplan‐Meier estimates was RCT 64% versus RT 45%. Additional RHT increased 5‐year OS to 87% (HR, 0.32; 95% CI, 0.18–0.58; p = .0001). RCT + RHT compared with RCT showed a significantly better bladder‐preservation rate (HR, 0.13; 95% CI, 0.03–0.56; p = .006). Median follow‐up was 71 months. The median number of RHT sessions was five. Conclusion. The multimodal treatment consisted of a maximal TUR‐BT followed by RT; concomitant platinum‐based chemotherapy combined with RHT in patients with high‐grade bladder cancer improves local control, bladder‐preservation rate, and OS. It offers a promising alternative to surgical therapies like radical cystectomy. Implications for Practice. Radical cystectomy with appropriate lymph node dissection has long represented the standard of care for muscle‐invasive bladder cancer in medically fit patients, despite many centers reporting excellent long‐term results for bladder preserving strategies. This retrospective analysis compares different therapeutic modalities in bladder‐preservation therapy. The results of this study show that multimodal treatment consisting of maximal transurethral resection of bladder tumor followed by radiotherapy, concomitant platinum‐based chemotherapy combined with regional deep hyperthermia in patients with Ta, Tis, T1–2 bladder carcinomas improves local control, bladder‐preservation rate, and survival. More importantly, these findings offer a promising alternative to surgical therapies like radical cystectomy. The authors hope that, in the future, closer collaboration between urologists and radiotherapists will further improve treatments and therapies for the benefit of patients.
Collapse
Affiliation(s)
- Ricarda Merten
- Department of Radiation Oncology, Universitätsklinikum Erlangen Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Oliver Ott
- Department of Radiation Oncology, Universitätsklinikum Erlangen Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Marlen Haderlein
- Department of Radiation Oncology, Universitätsklinikum Erlangen Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Simone Bertz
- Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Arndt Hartmann
- Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Bernd Wullich
- Department of Urology and Pediatric Urology, Universitätsklinikum Erlangen Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Bastian Keck
- Department of Urology and Pediatric Urology, Universitätsklinikum Erlangen Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Reinhard Kühn
- Department of Urology, Martha Maria Medical Center, Nuremberg, Germany
| | - Claus Michael Rödel
- Department of Radiotherapy and Oncology, Universitätsklinikum Frankfurt, Frankfurt, Germany
| | - Christian Weiss
- Department of Radiation Oncology, Klinikum Darmstadt GmbH, Darmstadt, Germany
| | - Christine Gall
- Department of Medical Informatics, Biometry and Epidemiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Wolfgang Uter
- Department of Medical Informatics, Biometry and Epidemiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Rainer Fietkau
- Department of Radiation Oncology, Universitätsklinikum Erlangen Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|