1
|
Scharr MV, Damm F, Krahl P, Dieper A, Veltsista PD, Hansch A, Beck M, Gerster D, Giovannelli AC, Bullinger L, Zips D, Ghadjar P. Review of preclinical data on hyperthermia treatment in lymphomas and its potential for clinical application. Int J Hyperthermia 2024; 41:2418427. [PMID: 39489511 DOI: 10.1080/02656736.2024.2418427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/12/2024] [Accepted: 10/14/2024] [Indexed: 11/05/2024] Open
Abstract
INTRODUCTION Hyperthermia (HT) at temperatures between 39 °C and 44 °C is utilized as an adjunctive cancer therapy, serving as potent radio- and chemosensitizer. Its effectiveness in treating solid malignancies has been well established. This raises the question of whether HT can also benefit patients with nonsolid tumors, such as lymphomas. OBJECTIVE To provide an overview of the current literature on research involving the use of HT in the treatment of lymphomas. MATERIAL AND METHODS This systematic literature review was conducted following the PRISMA guidelines. For this purpose, a MeSH-term-defined literature search on MEDLINE (Pubmed) and Embase (Ovid) was conducted from June 25 to June 28, 2024. Included were in vitro studies on lymphoma cell lines and preclinical studies on animal models with lymphoma that were both treated with HT as monotherapy or HT in combination with another treatment, and studies on patients with lymphoma. Excluded were studies that used thermal ablation and hyperthermic perfusions. RESULTS Thirty-nine studies were included, predominantly in vitro studies (n = 32) or studies on animal models (n = 5). The in vitro studies utilized HT either as monotherapy (n = 6), with substances that enhance HT efficacy (n = 18) or as a sensitizer for other treatments (n = 8). Additionally, two clinical case reports on the treatment of lymphoma patients were included. CONCLUSIONS In vitro results suggest that HT can have anticancer effects on lymphoma cells and may enhance existing treatments. These findings are supported by in vivo studies and case reports. However, additional clinical data are needed before translation into the clinic can be implemented.
Collapse
Affiliation(s)
- Moritz V Scharr
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin corporate member of Free University Berlin and Humboldt University Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Frederik Damm
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Hematology, Oncology and Cancer Immunology, Charité - Charité - Universitätsmedizin Berlin corporate member of Free University Berlin and Humboldt University Berlin, Berlin, Germany
| | - Paul Krahl
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin corporate member of Free University Berlin and Humboldt University Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Anna Dieper
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin corporate member of Free University Berlin and Humboldt University Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Paraskevi D Veltsista
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin corporate member of Free University Berlin and Humboldt University Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Alexander Hansch
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin corporate member of Free University Berlin and Humboldt University Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Marcus Beck
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin corporate member of Free University Berlin and Humboldt University Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dominik Gerster
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin corporate member of Free University Berlin and Humboldt University Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Anna-Chiara Giovannelli
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin corporate member of Free University Berlin and Humboldt University Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lars Bullinger
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Hematology, Oncology and Cancer Immunology, Charité - Charité - Universitätsmedizin Berlin corporate member of Free University Berlin and Humboldt University Berlin, Berlin, Germany
| | - Daniel Zips
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin corporate member of Free University Berlin and Humboldt University Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Pirus Ghadjar
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin corporate member of Free University Berlin and Humboldt University Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
2
|
Boulouta A, Kyriazoglou A, Kotsantis I, Economopoulou P, Anastasiou M, Pantazopoulos A, Kyrkasiadou M, Moutafi M, Gavrielatou N, Zazas E, Caglar C, Nixon I, Tolia M, Kavourakis G, Psyrri A. Pathologic complete response in patients with localized soft tissue sarcoma treated with neoadjuvant therapy and its correlation with clinical outcomes: A systematic review. Cancer Treat Rev 2024; 130:102820. [PMID: 39216184 DOI: 10.1016/j.ctrv.2024.102820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 08/22/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
Soft tissue sarcomas (STS), comprising approximately 1% of adult solid malignancies, are primarily treated with surgery, with the choice of perioperative treatment being a challenging and highly individualized decision. Clinical trials assessing neoadjuvant modalities in STS predominantly use clinical outcomes or radiologic response as endpoints, with pathologic complete response (pCR) not being employed as a designated study endpoint. Our systematic review aimed to assess the rates of pCR in clinical trials of different neoadjuvant modalities for STS and its correlation with patient clinical outcomes. 23 phase I, II and III studies were included, from which data regarding rates of pCR with each treatment, as well as correlation of pCR with clinical outcomes were retrieved. In 16 trials that assessed pCR, the percentage of patients who achieved a pCR ranged from 8 to 58%. Most of these trials did not aim to establish an association between pCR and clinical outcomes. However, among those that did investigate this correlation, a positive association was identified between pCR and both 5-year disease-specific survival (DSS) and 5-year overall survival (OS). While pCR serves as a crucial marker guiding treatment decisions in other neoplasms like triple negative breast cancer and urothelial cancer, it is not yet used in a similar setting for STS. Our findings indicate variability in patients achieving pCR across different neoadjuvant treatments for STS and a possible positive correlation with patient outcomes. Consequently, we propose considering pCR as a surrogate endpoint in future prospective trials for STS.
Collapse
Affiliation(s)
- A Boulouta
- Second Department of Internal Medicine, Oncology Unit, Attikon University Hospital, Athens, Greece.
| | - A Kyriazoglou
- Second Department of Internal Medicine, Oncology Unit, Attikon University Hospital, Athens, Greece
| | - I Kotsantis
- Second Department of Internal Medicine, Oncology Unit, Attikon University Hospital, Athens, Greece
| | - P Economopoulou
- Second Department of Internal Medicine, Oncology Unit, Attikon University Hospital, Athens, Greece
| | - M Anastasiou
- Second Department of Internal Medicine, Oncology Unit, Attikon University Hospital, Athens, Greece
| | - A Pantazopoulos
- Second Department of Internal Medicine, Oncology Unit, Attikon University Hospital, Athens, Greece
| | - M Kyrkasiadou
- Second Department of Internal Medicine, Oncology Unit, Attikon University Hospital, Athens, Greece
| | - M Moutafi
- Second Department of Internal Medicine, Oncology Unit, Attikon University Hospital, Athens, Greece
| | - N Gavrielatou
- Second Department of Internal Medicine, Oncology Unit, Attikon University Hospital, Athens, Greece
| | - E Zazas
- Second Department of Internal Medicine, Oncology Unit, Attikon University Hospital, Athens, Greece
| | - C Caglar
- Second Department of Internal Medicine, Oncology Unit, Attikon University Hospital, Athens, Greece
| | - I Nixon
- Department of Clinical Oncology, The Beatson West of Scotland Cancer Center, Glasgow, UK
| | - M Tolia
- Department of Radiotherapy, Faculty of Medicine, School of Health Sciences, University of Crete, Heraklion, Greece
| | - G Kavourakis
- Second Department of Internal Medicine, Oncology Unit, Attikon University Hospital, Athens, Greece
| | - A Psyrri
- Second Department of Internal Medicine, Oncology Unit, Attikon University Hospital, Athens, Greece
| |
Collapse
|
3
|
Zhu W, Pan S, Zhang J, Xu J, Zhang R, Zhang Y, Fu Z, Wang Y, Hu C, Xu Z. The role of hyperthermia in the treatment of tumor. Crit Rev Oncol Hematol 2024:104541. [PMID: 39461607 DOI: 10.1016/j.critrevonc.2024.104541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 09/19/2024] [Accepted: 10/17/2024] [Indexed: 10/29/2024] Open
Abstract
Despite recent advancements in the diagnosis and treatment options for cancer, it remains one of the most serious threats to health. Hyperthermia (HT) has emerged as a highly promising area of research due to its safety and cost-effectiveness. Currently, based on temperature, HT can be categorized into thermal ablation and mild hyperthermia. Thermal ablation involves raising the temperature within the tumor to over 60°C, resulting in direct necrosis in the central region of the tumor. In contrast, mild hyperthermia operates at relatively lower temperatures, typically in the range of 41-45°C, to induce damage to tumor cells. Furthermore, HT also serves as an immune adjuvant strategy in radiotherapy, chemotherapy, and immunotherapy, enhancing the effectiveness of radiotherapy, increasing the uptake of chemotherapy drugs, and reprogramming the tumor microenvironment through the induction of immunogenic cell death, thereby promoting the recruitment of endogenous immune cells. This article reviews the current status and development of hyperthermia, outlines potential mechanisms by which hyperthermia inhibits tumors, describes clinical trial attempts combining hyperthermia with radiotherapy, chemotherapy, and immunotherapy, and discusses the relationship between nanoparticles and hyperthermia.
Collapse
Affiliation(s)
- Weiwei Zhu
- Second clinical medical College,Zhejiang Chinese Medical University, Hangzhou 310053, China; Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Siwei Pan
- Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China; Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Jiaqing Zhang
- Second clinical medical College,Zhejiang Chinese Medical University, Hangzhou 310053, China; Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Jingli Xu
- Second clinical medical College,Zhejiang Chinese Medical University, Hangzhou 310053, China; Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Ruolan Zhang
- Second clinical medical College,Zhejiang Chinese Medical University, Hangzhou 310053, China; Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Yanqiang Zhang
- Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China; Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Zhenjie Fu
- Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China; Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Yuqi Wang
- Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China; Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Can Hu
- Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China; Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou 310022, China.
| | - Zhiyuan Xu
- Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China; Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou 310022, China.
| |
Collapse
|
4
|
Muratoglu R, Gerster D, Nadobny J, Hansch A, Krahl P, Veltsista PD, Beck M, Zips D, Ghadjar P. Comparisons of computer simulations and experimental data for capacitive hyperthermia using different split-phantoms. Int J Hyperthermia 2024; 41:2416999. [PMID: 39428108 DOI: 10.1080/02656736.2024.2416999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 10/07/2024] [Accepted: 10/10/2024] [Indexed: 10/22/2024] Open
Abstract
INTRODUCTION Several positive clinical trials have demonstrated that capacitive hyperthermia (CHT) improves the effectiveness of radiation therapy for the treatment of various cancer entities. However, the ability of CHT to induce significant heating throughout the body is under debate. OBJECTIVES To perform a pilot study involving comparisons of computer simulations and experimental data using different split-phantoms to validate hyperthermia treatment modeling for pre-planning for a clinical CHT system and to investigate the feasibility of split-phantom measurements in capacitive hyperthermia. MATERIALS AND METHODS The CHT system EHY-2030 (Oncotherm, Budapest, Hungary) was used. The system provides two electrode sizes, but only the smaller electrode, indicated as D200 electrode, was investigated in this pilot study. Horizontally and vertically splittable, different multi-slice phantoms with dielectric material properties simulating muscle and electrically low conductive fat were produced and heated. During the heating procedure, temperature-time curves were measured, and thermal images were captured. Specific absorption rate values were derived from the temperature rise (TR) values. Concomitantly, computer field simulations utilizing a detailed CAD-based model of the CHT system were performed using the simulation platform Sim4Life and compared with measurements. RESULTS For the investigated electrode D200 the system power of 75 W was applied, which is half of the maximum power of 150 W and lies in the range of usual values for this electrode applied in patient treatments in our clinic. For 75 W, a heating of 3.6 °C in 6 min in a depth of 1 cm in an agar-based, muscle tissue-equivalent phantom was achieved. The addition of a 1 cm thick, synthetic, low dielectric fat layer reduced the TR up until a depth of 8.5 cm by on average around 38% (from 8.5 cm onwards the absolute local TR is similar, deviations are ≤0.1 °C). In terms of point-to-point absolute SAR comparison (without any normalization), up to a depth of 11 cm in the phantoms central vertical plot, the simulation differs from the measured TR points by on average 25% (ranging from 7% to 36%) for the homogeneous phantom and by on average 43% (ranging from 26% to 60%) for the inhomogeneous phantom. CONCLUSION Computer simulations and experimental data were compared for the CHT system EHY-2030 using the D200 electrode, applying a thermal imaging technique for different vertically splittable phantoms. This pilot study data can be used as a guidance regarding the expected heating for this commonly used electrode size but also to further elucidate the significance of non-thermal anticancer effects. Further studies are needed for different sizes and geometries of electrodes and phantoms.
Collapse
Affiliation(s)
- Rami Muratoglu
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| | - Dominik Gerster
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| | - Jacek Nadobny
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| | - Alexander Hansch
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| | - Paul Krahl
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| | - Paraskevi Danai Veltsista
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| | - Marcus Beck
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| | - Daniel Zips
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| | - Pirus Ghadjar
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
5
|
Qin S, Liu Y, He G, Yang J, Zeng F, Lu Q, Wang M, He B, Song Y. Spatiotemporal Delivery of Dual Nanobodies by Engineered Probiotics to Reverse Tumor Immunosuppression via Targeting Tumor-Derived Exosomes. ACS NANO 2024; 18:26858-26871. [PMID: 39308426 DOI: 10.1021/acsnano.4c08117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/02/2024]
Abstract
The anti-PD-L1 and its bispecific antibodies have exhibited durable antitumor immunity but still elicit immunosuppression mainly caused by tumor-derived exosomes (TDEs), leading to difficulty in clinical transformation. Herein, engineered Escherichia coli Nissle 1917 (EcN) coexpressing anti-PD-L1 and anti-CD9 nanobodies (EcN-Nb) are developed and decorated with zinc-based metal-organic frameworks (MOFs) loaded with indocyanine green (ICG), to generate EcN-Nb-ZIF-8CHO-ICG (ENZC) for spatiotemporal lysis of bacteria for immunotherapy. The tumor-homing hybrid system can specifically release nanobodies in response to near-infrared (NIR) radiation, thereby targeting TDEs and changing their biological distribution, remodeling tumor-associated macrophages to M1 states, activating more effective and cytotoxic T lymphocytes, and finally, leading to the inhibition of tumor proliferation and metastasis. Altogether, the microfluidic-enabled MOF-modified engineered probiotics target TDEs and activate the antitumor immune response in a spatiotemporally manipulated manner, offering promising TDE-targeted immune therapy.
Collapse
Affiliation(s)
- Shurong Qin
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210023, China
| | - Yuta Liu
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210023, China
| | - Guanzhong He
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210023, China
| | - Jingjing Yang
- Department of Biochemistry and Molecular Biology, School of Medicine & Holistic Integrative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Fei Zeng
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210023, China
| | - Qianglan Lu
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210023, China
| | - Meng Wang
- Department of Gastric and Hernia Surgery, Nanjing University Medical School Affiliated Drum Tower Hospital, Nanjing 210023, China
| | - Bangshun He
- Department of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Yujun Song
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210023, China
| |
Collapse
|
6
|
Esaki K, Matsunobu T, Nomoto S, Shimohata Y, Maekawa A, Yoshimoto M. Limb salvage using radical combined hyperthermia and radiotherapy for myxofibrosarcoma of the lower leg in an elderly patient. Int Cancer Conf J 2024; 13:499-503. [PMID: 39398925 PMCID: PMC11464714 DOI: 10.1007/s13691-024-00719-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 08/17/2024] [Indexed: 10/15/2024] Open
Abstract
Soft tissue sarcomas account for only 1.5% of malignant tumors in adults and are therefore challenging to treat. We present a case of myxofibrosarcoma of the lower leg in an 88-year-old woman who successfully responded to combined hyperthermia and radiotherapy. We proposed a below-knee amputation because of the spread of the lesion, but the patient opted for hyperthermia with radiotherapy. One and a half years later, the tumor partially regrew, and the regrown mass was resected with an R0 margin. Unfortunately, the patient developed a surgical site infection immediately after the resection, and a skin ulcer formed. It took about 2 years for conservative treatment to result in complete ulcer epithelization. The patient has been ambulant, and has not experienced any symptoms of local recurrence or metastasis in the two and a half years since the surgery. Although adverse events related to combined hyperthermia and radiotherapy, such as delayed wound healing, should be considered, it could be an option for the treatment of localized soft-tissue sarcoma, especially in elderly patients.
Collapse
Affiliation(s)
- Katsuhiro Esaki
- Department of Orthopedic Surgery, Kyushu Rosai Hospital, 1-1 Sonekitamachi, Kokura Minami-Ku, Kitakyushu, Fukuoka 800-0296 Japan
| | - Tomoya Matsunobu
- Department of Orthopedic Surgery, Kyushu Rosai Hospital, 1-1 Sonekitamachi, Kokura Minami-Ku, Kitakyushu, Fukuoka 800-0296 Japan
| | - Satoshi Nomoto
- Department of Radiology and Oncology, Kyushu Rosai Hospital, Kitakyushu, Fukuoka, Japan
| | - Yumi Shimohata
- Department of Nursing, Certified Nurse in Wound, Ostomy and Continence Nursing, Kyushu Rosai Hospital, Kitakyushu, Fukuoka, Japan
| | - Akira Maekawa
- Department of Orthopedic Surgery, Kyushu Rosai Hospital, 1-1 Sonekitamachi, Kokura Minami-Ku, Kitakyushu, Fukuoka 800-0296 Japan
| | - Masato Yoshimoto
- Department of Orthopedic Surgery, Kyushu Rosai Hospital, 1-1 Sonekitamachi, Kokura Minami-Ku, Kitakyushu, Fukuoka 800-0296 Japan
| |
Collapse
|
7
|
Zhu N, Meng X, Wang Z, Hu Y, Zhao T, Fan H, Niu F, Han J. Radiomics in Diagnosis, Grading, and Treatment Response Assessment of Soft Tissue Sarcomas: A Systematic Review and Meta-analysis. Acad Radiol 2024; 31:3982-3992. [PMID: 38772802 DOI: 10.1016/j.acra.2024.03.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 03/12/2024] [Accepted: 03/22/2024] [Indexed: 05/23/2024]
Abstract
RATIONALE AND OBJECTIVES To evaluate radiomics in soft tissue sarcomas (STSs) for diagnostic accuracy, grading, and treatment response assessment, with a focus on clinical relevance. METHODS In this diagnostic accuracy study, radiomics was applied using multiple MRI sequences and AI classifiers, with histopathological diagnosis as the reference standard. Statistical analysis involved meta-analysis, random-effects model, and Deeks' funnel plot asymmetry test. RESULTS Among 579 unique titles and abstracts, 24 articles were included in the systematic review, with 21 used for meta-analysis. Radiomics demonstrated a pooled sensitivity of 84% (95% CI: 80-87) and specificity of 63% (95% CI: 56-70), AUC of 0.93 for diagnosis, sensitivity of 84% (95% CI: 82-87) and specificity of 73% (95% CI: 68-77), AUC of 0.91 for grading, and sensitivity of 83% (95% CI: 67-94) and specificity of 67% (95% CI: 59-74), AUC of 0.87 for treatment response assessment. CONCLUSION Radiomics exhibits potential for accurate diagnosis, grading, and treatment response assessment in STSs, emphasizing the need for standardization and prospective trials. CLINICAL RELEVANCE STATEMENT Radiomics offers precise tools for STS diagnosis, grading, and treatment response assessment, with implications for optimizing patient care and treatment strategies in this complex malignancy.
Collapse
Affiliation(s)
- Nana Zhu
- The Department of Radiology, Tianjin Hospital, 406 Jiefang Southern Road, Tianjin, China; Graduate School, Tianjin Medical University, Tianjin, China
| | - Xianghong Meng
- The Department of Radiology, Tianjin Hospital, 406 Jiefang Southern Road, Tianjin, China
| | - Zhi Wang
- The Department of Radiology, Tianjin Hospital, 406 Jiefang Southern Road, Tianjin, China; Graduate School, Tianjin Medical University, Tianjin, China.
| | - Yongcheng Hu
- The Department of Radiology, Tianjin Hospital, 406 Jiefang Southern Road, Tianjin, China; Graduate School, Tianjin Medical University, Tianjin, China
| | - Tingting Zhao
- The Department of Radiology, Tianjin Hospital, 406 Jiefang Southern Road, Tianjin, China; Graduate School, Tianjin Medical University, Tianjin, China
| | - Hongxing Fan
- The Department of Radiology, Tianjin Hospital, 406 Jiefang Southern Road, Tianjin, China; Graduate School, Tianjin Medical University, Tianjin, China
| | - Feige Niu
- The Department of Radiology, Tianjin Hospital, 406 Jiefang Southern Road, Tianjin, China; Graduate School, Tianjin Medical University, Tianjin, China
| | - Jun Han
- The Department of Radiology, Tianjin Hospital, 406 Jiefang Southern Road, Tianjin, China; Graduate School, Tianjin University, Tianjin, China
| |
Collapse
|
8
|
Radaelli S, Merlini A, Khan M, Gronchi A. Progress in histology specific treatments in soft tissue sarcoma. Expert Rev Anticancer Ther 2024; 24:845-868. [PMID: 39099398 DOI: 10.1080/14737140.2024.2384584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 07/22/2024] [Indexed: 08/06/2024]
Abstract
INTRODUCTION Soft tissue sarcomas (STS) represent a heterogenous group of rare tumors, primarily treated with surgery. Preoperative radiotherapy is often recommended for extremity high-risk STS. Neoadjuvant chemotherapy, typically based on doxorubicin with ifosfamide, has shown efficacy in limbs and trunk wall STS. Second-line chemotherapy, commonly utilized in the metastatic setting, is mostly histology-driven. Molecular targeted agents are used across various histologies, and although the use of immunotherapy in STS is still in its early stages, there is increasing interest in exploring its potential. AREAS COVERED This article involved an extensive recent search on PubMed. It explored the current treatment landscape for localized and metastatic STS, focusing on the combined use of radiotherapy and chemotherapy for both extremity and retroperitoneal tumors, and with a particular emphasis on the most innovative histopathology driven therapeutic approaches. Additionally, ongoing clinical trials identified via clinicaltrials.gov are included. EXPERT OPINION Recently there have been advancements in the treatment of STS, largely driven by the outcomes of clinical trials. However further research is imperative to comprehend the effect of chemotherapy, targeted therapy and immunotherapy in various STS, as well as to identify biomarkers able to predict which patients are most likely to benefit from these treatments.
Collapse
Affiliation(s)
- Stefano Radaelli
- Sarcoma Service, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Alessandra Merlini
- Department of Oncology, University of Turin, Orbassano, Italy
- Department of Oncology, San Luigi Gonzaga University Hospital, Orbassano, Italy
| | - Misbah Khan
- Surgery, East Sussex NHS Healthcare, East Sussex, UK
| | - Alessandro Gronchi
- Sarcoma Service, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
9
|
Patri S, Thanh NTK, Kamaly N. Magnetic iron oxide nanogels for combined hyperthermia and drug delivery for cancer treatment. NANOSCALE 2024; 16:15446-15464. [PMID: 39113663 DOI: 10.1039/d4nr02058h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
Hyperthermia and chemotherapy represent potential modalities for cancer treatments. However, hyperthermia can be invasive, while chemotherapy drugs often have severe side effects. Recent clinical investigations have underscored the potential synergistic efficacy of combining hyperthermia with chemotherapy, leading to enhanced cancer cell killing. In this context, magnetic iron oxide nanogels have emerged as promising candidates as they can integrate superparamagnetic iron oxide nanoparticles (IONPs), providing the requisite magnetism for magnetic hyperthermia, with the nanogel scaffold facilitating smart drug delivery. This review provides an overview of the synthetic methodologies employed in fabricating magnetic nanogels. Key properties and designs of these nanogels are discussed and challenges for their translation to the clinic and the market are summarised.
Collapse
Affiliation(s)
- Sofia Patri
- Department of Materials, Molecular Sciences Research Hub, Imperial College London, 82 Wood Ln, London W12 0BZ, UK.
| | - Nguyen Thi Kim Thanh
- UCL Healthcare Biomagnetic and Nanomaterials Laboratories, 21 Albemarle Street, London W1S 4BS, UK.
- Biophysic Group, Department of Physics and Astronomy, University College London, London WC1E 6BT, UK
| | - Nazila Kamaly
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, 82 Wood Ln, London W12 0BZ, UK.
| |
Collapse
|
10
|
Sarogni P, Frusca V, Zamborlin A, Giannini N, Menicagli M, Brancato L, Linsalata S, Di Martino F, Gonnelli A, Paiar F, Van den Bossche J, Bogers J, Voliani V. Neoadjuvant Hyperthermia Combined with Hybrid Nanoarchitectures Enhances Chemoradiotherapy Efficacy in Head and Neck Carcinoma. ACS APPLIED MATERIALS & INTERFACES 2024; 16:43272-43282. [PMID: 39126693 DOI: 10.1021/acsami.4c07393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/12/2024]
Abstract
Head and neck squamous cell carcinomas are characterized by a high incidence of recurrence, especially in patients with locally advanced disease. Standard treatment strategies can be associated with severe side effects to healthy tissues that can negatively impact the patient's quality of life. Hyperthermia (HT) is a noninvasive treatment modality that has improved the effectiveness of chemotherapy (CT) and/or radiotherapy (RT) for the management of some solid neoplasms. In this context, the association of this approach with rationally designed nanomaterials may further enhance the treatment outcome. In this study, we demonstrate the enhanced effect of neoadjuvant HT in combination with hybrid nanoarchitectures enclosing a cisplatin prodrug (NAs-CisPt) and RT. All the treatments and their combinations have been fully evaluated by employing standardized chorioallantoic membrane tumor models of HPV-negative head and neck carcinoma. An improved tumor-shrinking effect was observed by the administration of the trimodal treatment (HT/NAs-CisPt/RT), which also highlighted a significant increase in apoptosis. Our findings demonstrate that the combination of HT with nanotechnology-based CT and RT in a certain order enhances the in vivo treatment outcome. On a broader basis, this study paves the way for the next exploration of noninvasive treatment approaches for the clinical management of oral cancer based on innovative strategies.
Collapse
Affiliation(s)
- Patrizia Sarogni
- Center for Nanotechnology Innovation@NEST, Istituto Italiano di Tecnologia, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Valentina Frusca
- Center for Nanotechnology Innovation@NEST, Istituto Italiano di Tecnologia, Piazza San Silvestro 12, 56127 Pisa, Italy
- Scuola Superiore Sant'Anna, Piazza Martiri della Libertà 33, 56127 Pisa, Italy
| | - Agata Zamborlin
- Center for Nanotechnology Innovation@NEST, Istituto Italiano di Tecnologia, Piazza San Silvestro 12, 56127 Pisa, Italy
- NEST-Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Noemi Giannini
- Center for Nanotechnology Innovation@NEST, Istituto Italiano di Tecnologia, Piazza San Silvestro 12, 56127 Pisa, Italy
- Radiation Oncology Unit, Pisa University Hospital "Azienda Ospedaliero-Universitaria Pisana", Via Roma 67, 56126 Pisa, Italy
| | - Michele Menicagli
- Fondazione Pisana per la Scienza ONLUS, via Ferruccio Giovannini 13, S. Giuliano Terme, 56017 Pisa, Italy
| | | | - Stefania Linsalata
- Unit of Medical Physics, Pisa University Hospital "Azienda Ospedaliero-Universitaria Pisana", Via Roma 67, 56126 Pisa, Italy
| | - Fabio Di Martino
- Unit of Medical Physics, Pisa University Hospital "Azienda Ospedaliero-Universitaria Pisana", Via Roma 67, 56126 Pisa, Italy
| | - Alessandra Gonnelli
- Center for Nanotechnology Innovation@NEST, Istituto Italiano di Tecnologia, Piazza San Silvestro 12, 56127 Pisa, Italy
- Radiation Oncology Unit, Pisa University Hospital "Azienda Ospedaliero-Universitaria Pisana", Via Roma 67, 56126 Pisa, Italy
| | - Fabiola Paiar
- Radiation Oncology Unit, Pisa University Hospital "Azienda Ospedaliero-Universitaria Pisana", Via Roma 67, 56126 Pisa, Italy
| | | | - Johannes Bogers
- ElmediX NV, Esperantolaan 4, 3001 Heverlee, Belgium
- Laboratory of Cell Biology and Histology, University of Antwerp, 2610 Antwerp, Belgium
| | - Valerio Voliani
- Center for Nanotechnology Innovation@NEST, Istituto Italiano di Tecnologia, Piazza San Silvestro 12, 56127 Pisa, Italy
- Department of Pharmacy, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Cembrano, 4, 16148 Genoa, Italy
| |
Collapse
|
11
|
Zhang YF, Lu M. Advances in magnetic induction hyperthermia. Front Bioeng Biotechnol 2024; 12:1432189. [PMID: 39161353 PMCID: PMC11331313 DOI: 10.3389/fbioe.2024.1432189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 07/25/2024] [Indexed: 08/21/2024] Open
Abstract
Magnetic induction hyperthermia (MIH), is a technique that has developed rapidly in recent years in the field of tumor thermotherapy. It implants a magnetic heating medium (millimeter-sized heat seeds, micron-sized magnetic particles and nanometer-sized magnetic fluids, etc.) inside the tumor. The material heats up under the induction of an external alternating magnetic field (100-500 kHz), which causes a high temperature zone to rapidly form in the local biological tissues and induces apoptosis in tumor cells. Magnetic induction hyperthermia has the advantages of high safety, strong targeting, repeatable treatment, and the size of the incision during treatment is negligible compared to surgical resection, and is currently used in clinical treatment. However, the millimeter-scale heat seed heating that is typically used in treatments can result in uneven temperatures within the tissue. Common MIH heating devices are bulky and complex in design, and are not easy for medical staff to get their hands on, which are issues that limit the diffusion of MIH. In this view, this paper will discuss the basic theoretical research on MIH and the progress of MIH-related technologies, with a focus on the latest research and development results and research hotspots of nanoscale ferromagnetic media and magnetic heat therapy devices, as well as the validation results and therapeutic efficacy of the new MIH technology on animal experiments and clinical trials. In this paper, it is found that induction heating using magnetic nanoparticles improves the uniformity of the temperature field, and the magneto-thermal properties of nanoscale ferromagnetic materials are significantly improved. The heating device was miniaturized to simplify the operation steps, while the focusing of the magnetic field was locally enhanced. However, there are fewer studies on the biotoxicity aspects of nanomedicines, and the localized alternating magnetic field uniformity used for heating and the safety of the alternating magnetic field after irradiation of the human body have not been sufficiently discussed. Ultimately, the purpose of this paper is to advance research related to magnetic induction thermotherapy that can be applied in clinical treatment.
Collapse
Affiliation(s)
| | - Mai Lu
- Key Laboratory of Opto-Electronic Technology and Intelligent Control of Ministry of Education, Lanzhou Jiaotong University, Lanzhou, China
| |
Collapse
|
12
|
Zhao S, Sun L, Zhou J, Li R, Sun Q, Wang W, Wang D. Advancements in Diagnosis and Multimodal Treatment Strategies for Retroperitoneal Tumors: A Comprehensive Review. Am J Clin Oncol 2024; 47:350-356. [PMID: 38476111 DOI: 10.1097/coc.0000000000001094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2024]
Abstract
Retroperitoneal tumors (RPTs) encompass both benign and malignant entities, constituting ~0.1% to 0.2% of all malignant tumors, of which 70% to 80% manifest malignancy. Predominantly, retroperitoneal sarcomas (RPS) represent the most prevalent subtype among RPT. With over 70 histologic forms identified, liposarcomas and leiomyosarcomas emerge as the primary constituents of RPS. Accurate diagnosis of RPTs necessitates preoperative core-needle biopsy and comprehensive imaging assessment. The current staging protocol for RPS relies on the eighth edition of the American Joint Committee on Cancer/TNM classification. Surgical excision remains the established gold standard for treating RPS. Therapeutic approaches vary according to the underlying pathophysiology. Although chemotherapy and radiotherapy exhibit efficacy in managing metastatic and recurrent unresectable RPS, their role in primary RPS remains unresolved, necessitating further clinical trials for validation. Concurrently, ongoing research explores the potential of targeted therapies and immunotherapy. This literature review aims to provide a comprehensive overview of existing research, delineating diagnostic pathways and optimal therapeutic strategies for RPT.
Collapse
Affiliation(s)
- Shuai Zhao
- Department of General Surgery, Northern Jiangsu People's Hospital, Clinical Teaching Hospital of Medical School, Nanjing University
| | - Longhe Sun
- Department of General Surgery, Northern Jiangsu People's Hospital, Yangzhou
| | - Jiajie Zhou
- Department of General Surgery, Northern Jiangsu People's Hospital, Clinical Teaching Hospital of Medical School, Nanjing University
| | - Ruiqi Li
- Department of General Surgery, Northern Jiangsu People's Hospital, Clinical Teaching Hospital of Medical School, Nanjing University
| | - Qiannan Sun
- Department of General Surgery, Taizhou Fourth People's Hospital
- Yangzhou Key Laboratory of Basic and Clinical Transformation of Digestive and Metabolic Diseases, Yangzhou, China
| | - Wei Wang
- Department of General Surgery, Taizhou Fourth People's Hospital
| | - Daorong Wang
- Department of General Surgery, Northern Jiangsu People's Hospital, Clinical Teaching Hospital of Medical School, Nanjing University
- Department of General Surgery, Taizhou Fourth People's Hospital
- Yangzhou Key Laboratory of Basic and Clinical Transformation of Digestive and Metabolic Diseases, Yangzhou, China
| |
Collapse
|
13
|
Peng J, Partanen A, Pichardo S, Staruch R, Perry K, McGuffin M, Huang Y, Chan KK, Wong S, Czarnota G, Hynynen K, Chu W. Mild hyperthermia with magnetic resonance- guided high intensity focused ultrasound combined with salvage chemoradiation for recurrent rectal cancer. Int J Hyperthermia 2024; 41:2365385. [PMID: 38897584 DOI: 10.1080/02656736.2024.2365385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 06/03/2024] [Indexed: 06/21/2024] Open
Abstract
INTRODUCTION Pelvic recurrences from rectal cancer present a challenging clinical scenario. Hyperthermia represents an innovative treatment option in combination with concurrent chemoradiation to enhance therapeutic effect. We provide the initial results of a prospective single center feasibility study (NCT02528175) for patients undergoing rectal cancer retreatment using concurrent chemoradiation and mild hyperthermia with MR-guided high intensity focused ultrasound (MR-HIFU). METHODS All patients were deemed ineligible for salvage surgery and were evaluated in a multidisciplinary fashion with a surgical oncologist, radiation oncologist and medical oncologist. Radiation was delivered to a dose of 30.6 Gy in 1.8 Gy per fraction with concurrent capecitabine. MR-HIFU was delivered on days 1, 8 and 15 of concurrent chemoradiation. Our primary objective was feasibility and toxicity. RESULTS Six patients (total 11 screened) were treated with concurrent chemoradiation and mild hyperthermia with MR-HIFU. Tumor size varied between 3.1-16.6 cm. Patients spent an average of 228 min in the MRI suite and sonication with the external transducer lasted an average of 35 min. There were no complications on the day of the MR-HIFU procedure and all acute toxicities (no grade >/=3 toxicities) resolved after completion of treatment. There were no late grade >/=3 toxicities. CONCLUSION Mild hyperthermia with MR-HIFU, in combination with concurrent chemoradiation for appropriately selected patients, is safe for localized pelvic recurrences from rectal cancer. The potential for MR-HIFU to be applied in the recurrent setting in rectal cancer treatment requires further technical development and prospective evaluation.
Collapse
Affiliation(s)
- Jonathan Peng
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre and University of Toronto, Toronto, Canada
| | | | - Samuel Pichardo
- Cumming School of Medicine, University of Calgary, Calgary, Canada
| | | | - Kaitlyn Perry
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre and University of Toronto, Toronto, Canada
| | - Merrylee McGuffin
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre and University of Toronto, Toronto, Canada
| | - Yuexi Huang
- Department of Medical Biophysics, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Kelvin Kw Chan
- Department of Medical Oncology, Sunnybrook Health Sciences Centre and University of Toronto, Toronto, Canada
| | - Shun Wong
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre and University of Toronto, Toronto, Canada
| | - Greg Czarnota
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre and University of Toronto, Toronto, Canada
- Department of Medical Biophysics, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Kullervo Hynynen
- Department of Medical Biophysics, Sunnybrook Research Institute, Toronto, ON, Canada
| | - William Chu
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre and University of Toronto, Toronto, Canada
| |
Collapse
|
14
|
Hong J, Wang L, Zheng Q, Cai C, Yang X, Liao Z. The Recent Applications of Magnetic Nanoparticles in Biomedical Fields. MATERIALS (BASEL, SWITZERLAND) 2024; 17:2870. [PMID: 38930238 PMCID: PMC11204782 DOI: 10.3390/ma17122870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 06/05/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024]
Abstract
Magnetic nanoparticles (MNPs) have found extensive application in the biomedical domain due to their enhanced biocompatibility, minimal toxicity, and strong magnetic responsiveness. MNPs exhibit great potential as nanomaterials in various biomedical applications, including disease detection and cancer therapy. Typically, MNPs consist of a magnetic core surrounded by surface modification coatings, such as inorganic materials, organic molecules, and polymers, forming a nucleoshell structure that mitigates nanoparticle agglomeration and enhances targeting capabilities. Consequently, MNPs exhibit magnetic responsiveness in vivo for transportation and therapeutic effects, such as enhancing medical imaging resolution and localized heating at the site of injury. MNPs are utilized for specimen purification through targeted binding and magnetic separation in vitro, thereby optimizing efficiency and expediting the process. This review delves into the distinctive functional characteristics of MNPs as well as the diverse bioactive molecules employed in their surface coatings and their corresponding functionalities. Additionally, the advancement of MNPs in various applications is outlined. Additionally, we discuss the advancements of magnetic nanoparticles in medical imaging, disease treatment, and in vitro assays, and we anticipate the future development prospects and obstacles in this field. The objective is to furnish readers with a thorough comprehension of the recent practical utilization of MNPs in biomedical disciplines.
Collapse
Affiliation(s)
| | | | | | | | | | - Zhenlin Liao
- College of Food Science, South China Agricultural University, Guangzhou 510642, China; (J.H.); (L.W.); (Q.Z.); (C.C.); (X.Y.)
| |
Collapse
|
15
|
Wong SM, Akbulatov A, Macsemchuk CA, Headrick A, Luo P, Drake JM, Waspe AC. An augmented hybrid multibaseline and referenceless MR thermometry motion compensation algorithm for MRgHIFU hyperthermia. Magn Reson Med 2024; 91:2266-2277. [PMID: 38181187 DOI: 10.1002/mrm.29988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 12/04/2023] [Accepted: 12/08/2023] [Indexed: 01/07/2024]
Abstract
PURPOSE A hybrid principal component analysis and projection onto dipole fields (PCA-PDF) MR thermometry motion compensation algorithm was optimized with atlas image augmentation and validated. METHODS Experiments were conducted on a 3T Philips MRI and Profound V1 Sonalleve high intensity focused ultrasound (high intensity focused ultrasound system. An MR-compatible robot was configured to induce motion on custom gelatin phantoms. Trials with periodic and sporadic motion were introduced on phantoms while hyperthermia was administered. The PCA-PDF algorithm was augmented with a predictive atlas to better compensate for larger sporadic motion. RESULTS During periodic motion, the temperature SD in the thermometry was improved from1 . 1 ± 0 . 1 $$ 1.1\pm 0.1 $$ to0 . 5 ± 0 . 1 ∘ $$ 0.5\pm 0.{1}^{\circ } $$ C with both the original and augmented PCA-PDF application. For large sporadic motion, the augmented atlas improved the motion compensation from the original PCA-PDF correction from8 . 8 ± 0 . 5 $$ 8.8\pm 0.5 $$ to0 . 7 ± 0 . 1 ∘ $$ 0.7\pm 0.{1}^{\circ } $$ C. CONCLUSION The PCA-PDF algorithm improved temperature accuracy to <1°C during periodic motion, but was not able to adequately address sporadic motion. By augmenting the PCA-PDF algorithm, temperature SD during large sporadic motion was also reduced to <1°C, greatly improving the original PCA-PDF algorithm.
Collapse
Affiliation(s)
- Suzanne M Wong
- The Wilfred and Joyce Posluns Centre for Image-Guided Innovation and Theraputic Intervention, The Hospital for Sick Children, Toronto, Ontario, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Arthur Akbulatov
- The Wilfred and Joyce Posluns Centre for Image-Guided Innovation and Theraputic Intervention, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Craig A Macsemchuk
- The Wilfred and Joyce Posluns Centre for Image-Guided Innovation and Theraputic Intervention, The Hospital for Sick Children, Toronto, Ontario, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Andrew Headrick
- The Wilfred and Joyce Posluns Centre for Image-Guided Innovation and Theraputic Intervention, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Phoebe Luo
- The Wilfred and Joyce Posluns Centre for Image-Guided Innovation and Theraputic Intervention, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - James M Drake
- The Wilfred and Joyce Posluns Centre for Image-Guided Innovation and Theraputic Intervention, The Hospital for Sick Children, Toronto, Ontario, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Adam C Waspe
- The Wilfred and Joyce Posluns Centre for Image-Guided Innovation and Theraputic Intervention, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Medical Imaging, University of Toronto, Toronto, Ontario, Canada
- Department of Material Science and Engineering, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
16
|
Khaoula M, Sirine B, Sana M, Eya A, Ghada S, Karima M. Primary breast sarcoma: Case report and literature review. Int J Surg Case Rep 2024; 119:109587. [PMID: 38749389 PMCID: PMC11109321 DOI: 10.1016/j.ijscr.2024.109587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 03/22/2024] [Accepted: 03/23/2024] [Indexed: 05/25/2024] Open
Abstract
INTRODUCTION Primary breast sarcoma (PBS), excluding phyllodes tumors, is an extremely rare and heterogeneous group of cancers, accounting for <1 % of all breast cancers. PBS is typically diagnosed in the fifth or sixth decades of life. There are no pathognomonic clinical or radiological features. Diagnosis is generally established through a microbiopsy. For young women, the diagnosis can be confused with a benign pathology. CLINICAL PRESENTATION Our patient was 27 years old, with no family or personal history of breast cancer, which was initially brought to the emergency department with a breast abscess. The clinical diagnosis of abscessed mastitis was made. The patient underwent tissue excision with evacuation of blood clots and necrotic tissue. Histopathological examination revealed a primary breast sarcoma. An MRI showed locally advanced retro-glandular tumor. An abdominal and pelvic CT scan performed showed no evidence of secondary locations. Therefore, the patient was referred for neoadjuvant radiotherapy and chemotherapy. After the third course of chemotherapy, the patient died following cardiogenic shock. DISCUSSION The PBS in younger women is extremely rare. The etiopathogenesis remains undetermined. The clinical and radiological characteristics of PBS mimic breast adenocarcinoma. Mastectomy is the treatment of choice but in the case of locally advanced tumor, the use of neoadjuvant chemotherapy can be indicated. PBS presents a significantly poorer prognosis. CONCLUSION Breast sarcomas are rare malignant tumors for which treatment protocols are not well-established. Further research efforts are needed to improve the understanding and treatment of PBS.
Collapse
Affiliation(s)
- Magdoud Khaoula
- Emergency Department, Maternity And Neonatology Center, Tunis, Tunisia; Faculty of Medicine of Tunis, University of Tunis El Manar, Tunisia.
| | - Bayar Sirine
- Department of Gynecology, Maternity and Neonatology Center, Tunis, Tunisia
| | - Menjli Sana
- Department of Gynecology, Maternity and Neonatology Center, Tunis, Tunisia
| | - Azouz Eya
- Department of Radiology, Rabta Hospital, Tunis, Tunisia
| | - Sahraoui Ghada
- Faculty of Medicine of Tunis, University of Tunis El Manar, Tunisia; Department of Histology, Salah Azaiez Instate, Tunisia
| | - Mrad Karima
- Faculty of Medicine of Tunis, University of Tunis El Manar, Tunisia; Department of Histology, Salah Azaiez Instate, Tunisia
| |
Collapse
|
17
|
Roohani S, Ehret F, Beck M, Veltsista DP, Nadobny J, Zschaeck S, Abdel-Rahman S, Eckert F, Flörcken A, Issels RD, Klöck S, Krempien R, Lindner LH, Notter M, Ott OJ, Pink D, Potkrajcic V, Reichardt P, Riesterer O, Spałek MJ, Stutz E, Wessalowski R, Zilli T, Zips D, Ghadjar P, Kaul D. Regional hyperthermia for soft tissue sarcoma - a survey on current practice, controversies and consensus among 12 European centers. Int J Hyperthermia 2024; 41:2342348. [PMID: 38653548 DOI: 10.1080/02656736.2024.2342348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 04/09/2024] [Indexed: 04/25/2024] Open
Abstract
PURPOSE To analyze the current practice of regional hyperthermia (RHT) for soft tissue sarcoma (STS) at 12 European centers to provide an overview, find consensuses and identify controversies necessary for future guidelines and clinical trials. METHODS In this cross-sectional survey study, a 27-item questionnaire assessing clinical subjects and procedural details on RHT for STS was distributed to 12 European cancer centers for RHT. RESULTS We have identified seven controversies and five consensus points. Of 12 centers, 6 offer both, RHT with chemotherapy (CTX) or with radiotherapy (RT). Two centers only offer RHT with CTX and four centers only offer RHT with RT. All 12 centers apply RHT for localized, high-risk STS of the extremities, trunk wall and retroperitoneum. However, eight centers also use RHT in metastatic STS, five in palliative STS, eight for superficial STS and six for low-grade STS. Pretherapeutic imaging for RHT treatment planning is used by 10 centers, 9 centers set 40-43 °C as the intratumoral target temperature, and all centers use skin detectors or probes in body orifices for thermometry. DISCUSSION There is disagreement regarding the integration of RHT in contemporary interdisciplinary care of STS patients. Many clinical controversies exist that require a standardized consensus guideline and innovative study ideas. At the same time, our data has shown that existing guidelines and decades of experience with the technique of RHT have mostly standardized procedural aspects. CONCLUSIONS The provided results may serve as a basis for future guidelines and inform future clinical trials for RHT in STS patients.
Collapse
Affiliation(s)
- Siyer Roohani
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité, Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité (Junior) Clinician Scientist Program, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Partner site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Felix Ehret
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Partner site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Marcus Beck
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Danai P Veltsista
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Jacek Nadobny
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Sebastian Zschaeck
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité, Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité (Junior) Clinician Scientist Program, Berlin, Germany
| | - Sultan Abdel-Rahman
- Department of Medicine III, University Hospital Munich, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Franziska Eckert
- Department of Radiation Oncology, University Hospital Tübingen, Tübingen, Germany
- Department of Radiation Oncology, AKH, Comprehensive Cancer Center Vienna, Medical University Vienna, Vienna, Austria
| | - Anne Flörcken
- Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Partner site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Rolf D Issels
- Department of Medicine III, University Hospital Munich, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Stephan Klöck
- Department of Radiation Oncology, Lindenhofspital Bern, Bern, Switzerland
| | - Robert Krempien
- Clinic for Radiotherapy, HELIOS Klinikum Berlin-Buch, Berlin, Germany
- MSB Medical School Berlin, Fakultät für Medizin, Berlin, Germany
| | - Lars H Lindner
- Department of Medicine III, University Hospital Munich, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Markus Notter
- Department of Radiation Oncology, Lindenhofspital Bern, Bern, Switzerland
| | - Oliver J Ott
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - Daniel Pink
- Department of Medical Oncology, Helios Klinikum Bad Saarow, Bad Saarow, Germany
- Cinic for Internal Medicine C - Haematology and Oncology, Stem Cell Transplantation and Palliative Care, University Medicine Greifswald, Greifswald, Germany
| | - Vlatko Potkrajcic
- Department of Radiation Oncology, University Hospital Tübingen, Tübingen, Germany
| | - Peter Reichardt
- Department of Medical Oncology, Helios Klinikum Berlin-Buch, and Medical School Berlin, Berlin, Germany
| | - Oliver Riesterer
- Center for Radiation Oncology KSA-KSB, Kantonsspital Aarau, Aarau, Switzerland
| | - Mateusz Jacek Spałek
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
- Department of Radiotherapy I, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Emanuel Stutz
- Department of Radiation Oncology, Inselspital Bern University Hospital, University of Bern, Bern, Switzerland
| | - Rüdiger Wessalowski
- Department of Paediatric Haematology and Oncology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Thomas Zilli
- Department of Radiation Oncology, Oncology Institute of Southern Switzerland (IOSI), EOC, Bellinzona, Switzerland
- Facoltà di Scienze Biomediche, Università Della Svizzera Italiana (USI), Lugano, Switzerland
- Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Daniel Zips
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Partner site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Pirus Ghadjar
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - David Kaul
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Partner site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
18
|
Borghi A, Gronchi A. Extremity and Truncal Soft Tissue Sarcoma: Risk Assessment and Multidisciplinary Management. Semin Radiat Oncol 2024; 34:147-163. [PMID: 38508780 DOI: 10.1016/j.semradonc.2023.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
Extremity and truncal soft tissue sarcomas are a heterogeneous group of rare cancers that arise from mesenchymal tissues. Hence, the adoption of tailored risk assessment and prognostication tools plays a crucial role in optimizing the decision-making for which of the many possible treatment strategies to select. Management of these tumors requires a multidisciplinary strategy, which has seen significant development in recent decades. Surgery has emerged as the primary treatment approach, with the main goal of achieving microscopic negative tumor margins. To reduce the likelihood of local recurrence, loco-regional treatments such as radiation therapy and isolated limb perfusion are often added to the treatment regimen in combination with surgery. This approach also enables surgeons to perform limb-sparing surgery, particularly in cases where a positive tumor margin is expected. Chemotherapy may also provide a further benefit in decreasing the probability of local recurrence or reducing distant metastasis in selected patients. Selecting the optimal treatment strategy for these rare tumors is best accomplished by an experienced multi-disciplinary team.
Collapse
Affiliation(s)
- Alessandra Borghi
- Department of Surgery, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Alessandro Gronchi
- Department of Surgery, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy..
| |
Collapse
|
19
|
Fromm J, Klein A, Kirilova M, Lindner LH, Nachbichler S, Holzapfel BM, Goller SS, Knösel T, Dürr HR. The Effect of chemo- and radiotherapy on tumor necrosis in soft tissue sarcoma- does it influence prognosis? BMC Cancer 2024; 24:303. [PMID: 38448852 PMCID: PMC10916229 DOI: 10.1186/s12885-024-12027-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 02/20/2024] [Indexed: 03/08/2024] Open
Abstract
BACKGROUND Soft tissue sarcomas (STSs) are a heterogeneous group of tumors. Wide surgical resection is standard, often combined with neoadjuvant chemotherapy, radiotherapy, or both. Studies have shown the predictive value of tumor necrosis in bone sarcoma (BS); however, the role of necrosis in STS after neoadjuvant therapies is still unclear. This study aimed to investigate the role of chemo- and radiotherapy in the formation of tumor necrosis and to evaluate the influence of tumor necrosis on overall survival and local recurrence-free survival. Data from BS patients and patients who did not receive neoadjuvant therapy were compared. METHODS A total of 779 patients with STS or BS were treated surgically. In all patients, tumor-specific factors such as type, size, or grading and the type of adjuvant therapy were documented. Local recurrence (LR), the diagnosis of metastatic disease, and survival during follow-up were evaluated. RESULTS A total of 565 patients with STS and 214 with BS were investigated. In STS, 24.1% G1 lesions, 34.1% G2 lesions, and 41.8% G3 lesions were observed. Two hundred twenty-four of the patients with STS and neoadjuvant therapy had either radiotherapy (RTx) (n = 80), chemotherapy (CTx) (n = 93), or both (n = 51). Three hundred forty-one had no neoadjuvant therapy at all. In STS, tumor necrosis after neoadjuvant treatment was significantly higher (53.5%) than in patients without neoadjuvant therapy (15.7%) (p < 0.001). Patients with combined neoadjuvant chemo-/radiotherapy had substantially higher tumor necrosis than those with radiotherapy alone (p = 0.032). There was no difference in tumor necrosis in patients with combined chemo-/radiotherapy and chemotherapy alone (p = 0.4). The mean overall survival for patients with STS was 34.7 months. Tumor necrosis did not influence survival in a subgroup of G2/3 patients. In STS with no neoadjuvant therapy and grading of G2/3, the correlation between necrosis and overall survival was significant (p = 0.0248). There was no significant correlation between local recurrence (LR) and necrosis. CONCLUSION STS shows a broad spectrum of necrosis even without neoadjuvant chemo- or radiotherapy. After CTx or/and RTx necrosis is enhanced and is significantly pronounced with a combination of both. There is a trend toward higher necrosis with CTx than with RTx. Grading substantially influences the necrosis rate, but necrosis in soft-tissue sarcoma following neoadjuvant therapy does not correlate with better survival or a lower local recurrence rate, as in bone sarcomas.
Collapse
Affiliation(s)
- Julian Fromm
- Department of Orthopaedics and Trauma Surgery, Orthopaedic Oncology, Musculoskeletal University Center Munich (MUM), LMU University Hospital, LMU Munich, Marchioninistr. 15, D- 81377, Munich, Germany
- SarKUM, Center of Bone and Soft Tissue Tumors, LMU University Hospital, LMU Munich, München, Germany
| | - Alexander Klein
- Department of Orthopaedics and Trauma Surgery, Orthopaedic Oncology, Musculoskeletal University Center Munich (MUM), LMU University Hospital, LMU Munich, Marchioninistr. 15, D- 81377, Munich, Germany
- SarKUM, Center of Bone and Soft Tissue Tumors, LMU University Hospital, LMU Munich, München, Germany
| | - Maya Kirilova
- Department of Orthopaedics and Trauma Surgery, Orthopaedic Oncology, Musculoskeletal University Center Munich (MUM), LMU University Hospital, LMU Munich, Marchioninistr. 15, D- 81377, Munich, Germany
- SarKUM, Center of Bone and Soft Tissue Tumors, LMU University Hospital, LMU Munich, München, Germany
| | - Lars Hartwin Lindner
- Department of Medicine III, LMU University Hospital, LMU Munich, München, Germany
- SarKUM, Center of Bone and Soft Tissue Tumors, LMU University Hospital, LMU Munich, München, Germany
| | - Silke Nachbichler
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, München, Germany
- SarKUM, Center of Bone and Soft Tissue Tumors, LMU University Hospital, LMU Munich, München, Germany
| | - Boris Michael Holzapfel
- Department of Orthopaedics and Trauma Surgery, Orthopaedic Oncology, Musculoskeletal University Center Munich (MUM), LMU University Hospital, LMU Munich, Marchioninistr. 15, D- 81377, Munich, Germany
- SarKUM, Center of Bone and Soft Tissue Tumors, LMU University Hospital, LMU Munich, München, Germany
| | - Sophia Samira Goller
- Department of Radiology, LMU University Hospital, LMU Munich, München, Germany
- SarKUM, Center of Bone and Soft Tissue Tumors, LMU University Hospital, LMU Munich, München, Germany
| | - Thomas Knösel
- Institute of Pathology, LMU Munich, München, Germany
- SarKUM, Center of Bone and Soft Tissue Tumors, LMU University Hospital, LMU Munich, München, Germany
| | - Hans Roland Dürr
- Department of Orthopaedics and Trauma Surgery, Orthopaedic Oncology, Musculoskeletal University Center Munich (MUM), LMU University Hospital, LMU Munich, Marchioninistr. 15, D- 81377, Munich, Germany.
- SarKUM, Center of Bone and Soft Tissue Tumors, LMU University Hospital, LMU Munich, München, Germany.
| |
Collapse
|
20
|
Mentrup F, Klein A, Lindner LH, Nachbichler S, Holzapfel BM, Albertsmeier M, Knösel T, Dürr HR. Refusal of Adjuvant Therapies and Its Impact on Local Control and Survival in Patients with Bone and Soft Tissue Sarcomas of the Extremities and Trunk. Cancers (Basel) 2024; 16:239. [PMID: 38254731 PMCID: PMC10814158 DOI: 10.3390/cancers16020239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/02/2024] [Accepted: 01/03/2024] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND In soft tissue or bone sarcomas, multimodal therapeutic concepts represent the standard of care. Some patients reject the therapeutic recommendations due to several reasons. The aim of this study was to assess the impact of that rejection on both prognosis and local recurrence. METHODS Between 2012 and 2019, a total of 828 sarcoma patients were surgically treated. Chemotherapy was scheduled as a neoadjuvant, and adjuvant multi-agent therapy was performed following recommendations from an interdisciplinary tumor board. Radiotherapy, if deemed appropriate, was administered either in a neoadjuvant or an adjuvant manner. The recommended type of therapy, patient compliance, and the reasons for refusal were documented. Follow-ups included local recurrences, diagnosis of metastatic disease, and patient mortality. RESULTS Radiotherapy was recommended in 407 (49%) patients. A total of 40 (10%) individuals did not receive radiation. A reduction in overall survival and local recurrence-free survival was evident in those patients who declined radiotherapy. Chemotherapy was advised for 334 (40%) patients, 250 (75%) of whom did receive all recommended cycles. A total of 25 (7%) individuals did receive a partial course while 59 (18%) did not receive any recommended chemotherapy. Overall survival and local recurrence-free survival were reduced in patients refusing chemotherapy. Overall survival was worst for the group of patients who received no chemotherapy due to medical reasons. Refusing chemotherapy for non-medical reasons was seen in 8.8% of patients, and refusal of radiotherapy for non-medical reasons was seen in 4.7% of patients. CONCLUSIONS Divergence from the advised treatment modalities significantly impacted overall survival and local recurrence-free survival across both treatment modalities. There is an imperative need for enhanced physician-patient communication. Reducing treatment times, as achieved with hypofractionated radiotherapy and with therapy in a high-volume sarcoma center, might also have a positive effect on complying with the treatment recommendations.
Collapse
Affiliation(s)
- Franziska Mentrup
- Department of Orthopaedics and Trauma Surgery, Orthopaedic Oncology, Musculoskeletal University Center Munich (MUM), LMU University Hospital, LMU Munich, 81377 München, Germany; (F.M.); (A.K.); (B.M.H.)
- SarKUM, Center of Bone and Soft Tissue Tumors, LMU University Hospital, LMU Munich, 81377 München, Germany; (L.H.L.); (S.N.); (M.A.); (T.K.)
| | - Alexander Klein
- Department of Orthopaedics and Trauma Surgery, Orthopaedic Oncology, Musculoskeletal University Center Munich (MUM), LMU University Hospital, LMU Munich, 81377 München, Germany; (F.M.); (A.K.); (B.M.H.)
- SarKUM, Center of Bone and Soft Tissue Tumors, LMU University Hospital, LMU Munich, 81377 München, Germany; (L.H.L.); (S.N.); (M.A.); (T.K.)
| | - Lars Hartwin Lindner
- SarKUM, Center of Bone and Soft Tissue Tumors, LMU University Hospital, LMU Munich, 81377 München, Germany; (L.H.L.); (S.N.); (M.A.); (T.K.)
- Department of Medicine III, LMU University Hospital, LMU Munich, 81377 München, Germany
| | - Silke Nachbichler
- SarKUM, Center of Bone and Soft Tissue Tumors, LMU University Hospital, LMU Munich, 81377 München, Germany; (L.H.L.); (S.N.); (M.A.); (T.K.)
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, 81377 München, Germany
| | - Boris Michael Holzapfel
- Department of Orthopaedics and Trauma Surgery, Orthopaedic Oncology, Musculoskeletal University Center Munich (MUM), LMU University Hospital, LMU Munich, 81377 München, Germany; (F.M.); (A.K.); (B.M.H.)
- SarKUM, Center of Bone and Soft Tissue Tumors, LMU University Hospital, LMU Munich, 81377 München, Germany; (L.H.L.); (S.N.); (M.A.); (T.K.)
| | - Markus Albertsmeier
- SarKUM, Center of Bone and Soft Tissue Tumors, LMU University Hospital, LMU Munich, 81377 München, Germany; (L.H.L.); (S.N.); (M.A.); (T.K.)
- Department of General, Visceral and Transplantation Surgery, LMU University Hospital, LMU Munich, 81377 München, Germany
| | - Thomas Knösel
- SarKUM, Center of Bone and Soft Tissue Tumors, LMU University Hospital, LMU Munich, 81377 München, Germany; (L.H.L.); (S.N.); (M.A.); (T.K.)
- Institute of Pathology, LMU Munich, 81377 München, Germany
| | - Hans Roland Dürr
- Department of Orthopaedics and Trauma Surgery, Orthopaedic Oncology, Musculoskeletal University Center Munich (MUM), LMU University Hospital, LMU Munich, 81377 München, Germany; (F.M.); (A.K.); (B.M.H.)
- SarKUM, Center of Bone and Soft Tissue Tumors, LMU University Hospital, LMU Munich, 81377 München, Germany; (L.H.L.); (S.N.); (M.A.); (T.K.)
| |
Collapse
|
21
|
de Bree E, Michelakis D, Heretis I, Kontopodis N, Spanakis K, Lagoudaki E, Tolia M, Zografakis-Sfakianakis M, Ioannou C, Mavroudis D. Retroperitoneal Soft Tissue Sarcoma: Emerging Therapeutic Strategies. Cancers (Basel) 2023; 15:5469. [PMID: 38001729 PMCID: PMC10670057 DOI: 10.3390/cancers15225469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/09/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Retroperitoneal soft tissue sarcoma (RPS) is a rare and heterogenous disease for which surgery is the cornerstone of treatment. However, the local recurrence rate is much higher than in soft tissue sarcoma of the extremities since wide resection is usually unfeasible in RPS due to its large size, indistinct tumour borders, anatomical constraints and the thinness of the overlying peritoneum. Local recurrence is the leading cause of death for low-grade RPS, whereas high-grade tumours are prone to distant metastases. In recent decades, the role of emerging therapeutic strategies, such as more extended surgery and (neo)adjuvant treatments to improve oncological outcome in primary localised RPS, has been extensively investigated. In this review, the recent data on the evolving multidisciplinary management of primary localised RPS are comprehensively discussed. The heterogeneity of RPS, with their different histological subtypes and biological behaviour, renders a standard therapeutic 'one-size-fits-all' approach inappropriate, and treatment should be modified according to histological type and malignancy grade. There is sufficient evidence that frontline extended surgery with compartmental resection including all ipsilateral retroperitoneal fat and liberal en bloc resection of adjacent organs and structures, even if they are not macroscopically involved, increases local tumour control in low-grade sarcoma and liposarcoma, but not in leiomyosarcoma for which complete macroscopic resection seems sufficient. Additionally, preoperative radiotherapy is not indicated for all RPSs, but seems to be beneficial in well-differentiated liposarcoma and grade I/II dedifferentiated liposarcoma, and probably in solitary fibrous tumour. Whether neoadjuvant chemotherapy is of benefit in high-grade RPS remains unclear from retrospective data and is subject of the ongoing randomised STRASS 2 trial, from which the results are eagerly awaited. Personalised, histology-tailored multimodality treatment is promising and will likely further evolve as our understanding of the molecular and genetic characteristics within RPS improves.
Collapse
Affiliation(s)
- Eelco de Bree
- Department of Surgical Oncology, Medical School of Crete University Hospital, 71110 Heraklion, Greece;
| | - Dimosthenis Michelakis
- Department of Surgical Oncology, Medical School of Crete University Hospital, 71110 Heraklion, Greece;
| | - Ioannis Heretis
- Department of Urology, Medical School of Crete University Hospital, 71110 Heraklion, Greece;
| | - Nikolaos Kontopodis
- Department of Vascular Surgery, Medical School of Crete University Hospital, 71110 Heraklion, Greece; (N.K.); (C.I.)
| | - Konstantinos Spanakis
- Department of Medical Imaging, Medical School of Crete University Hospital, 71110 Heraklion, Greece;
| | - Eleni Lagoudaki
- Department of Pathology, Medical School of Crete University Hospital, 71110 Heraklion, Greece;
| | - Maria Tolia
- Department of Radiation Oncology, Medical School of Crete University Hospital, 71110 Heraklion, Greece;
| | | | - Christos Ioannou
- Department of Vascular Surgery, Medical School of Crete University Hospital, 71110 Heraklion, Greece; (N.K.); (C.I.)
| | - Dimitrios Mavroudis
- Department of Medical Oncology, Medical School of Crete University Hospital, 71110 Heraklion, Greece;
| |
Collapse
|
22
|
Carrapiço-Seabra C, De Lazzari M, Ameziane A, van Rhoon GC, Dobšícek Trefná H, Curto S. Application of the ESHO-QA guidelines for determining the performance of the LCA superficial hyperthermia heating system. Int J Hyperthermia 2023; 40:2272578. [PMID: 37879635 DOI: 10.1080/02656736.2023.2272578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 10/15/2023] [Indexed: 10/27/2023] Open
Abstract
PURPOSE This study aimed to assess the quality of the lucite cone applicator (LCA), the standard applicator for superficial hyperthermia at the Erasmus MC Cancer Institute, using the most recent quality assurance guidelines, thus verifying their feasibility. MATERIALS AND METHODS The assessment was conducted on each of the six LCAs available for clinical treatments. The temperature distribution was evaluated using an infrared camera across different layers of a fat-muscle mimicking phantom. The maximum temperature increase, thermal effective penetration depth (TEPD), and thermal effective field size (TEFS) were used as quality metrics. The experimental results were validated through comparison with simulated results, using a canonical phantom model and a realistic phantom model segmented from CT imaging. RESULTS A maximum temperature increase above 6 °C at 2 cm depth in the fat-muscle phantom for all the experiments was found. A mean negative difference between simulated and experimental data was of 1.3 °C when using the canonical phantom model. This value decreased to a mean negative difference of 0.4 °C when using the realistic model. Simulated and measured TEPD showed good agreement for both in silico scenarios, while discrepancies were present for TEFS. CONCLUSIONS The LCAs passed all QA guidelines requirements for superficial hyperthermia delivery when used singularly or in an array configuration. A further characterization of parameters such as antenna efficiency and heat transfer coefficients would be beneficial for translating experimental results to simulated values. Implementing the QA guidelines was time-consuming and demanding, requiring careful preparation and correct setup of antenna elements.
Collapse
Affiliation(s)
- Carolina Carrapiço-Seabra
- Department of Radiotherapy, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Mattia De Lazzari
- Department of Electrical Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Abdelali Ameziane
- Department of Radiotherapy, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Gerard C van Rhoon
- Department of Radiotherapy, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Radiation Science and Technology, Faculty of Applied Sciences, Delft University of Technology, Delft, the Netherlands
| | - Hana Dobšícek Trefná
- Department of Electrical Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Sergio Curto
- Department of Radiotherapy, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| |
Collapse
|
23
|
Hohneck AL, Sadikaj L, Heinemann L, Schroeder M, Riess H, Gerhards A, Burkholder I, Heckel-Reusser S, Gottfried J, Hofheinz RD. Patients with Advanced Pancreatic Cancer Treated with Mistletoe and Hyperthermia in Addition to Palliative Chemotherapy: A Retrospective Single-Center Analysis. Cancers (Basel) 2023; 15:4929. [PMID: 37894296 PMCID: PMC10605673 DOI: 10.3390/cancers15204929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 09/29/2023] [Accepted: 10/08/2023] [Indexed: 10/29/2023] Open
Abstract
This retrospective analysis investigated the influence of integrative therapies in addition to palliative chemotherapy in patients with advanced pancreatic cancer, treated at a single institution specialized in integrative oncology between January 2015 and December 2019. In total, 206 consecutive patients were included in the study, whereof 142 patients (68.9%) received palliative chemotherapy (gemcitabine/nab-paclitaxel 33.8%; FOLFIRINOX 35.9%; gemcitabine 30.3%) while the remainder were treated with best supportive and integrative care. Integrative therapies were used in 117 of 142 patients (82.4%) in addition to conventional chemotherapy, whereby mistletoe was used in 117 patients (82.4%) and hyperthermia in 74 patients (52.1%). A total of 107/142 patients (86.3%) died during the observation period, whereby survival times differed significantly depending on the additional use of integrative mistletoe or hyperthermia: chemotherapy alone 8.6 months (95% CI 4.7-15.4), chemotherapy and only mistletoe therapy 11.2 months (95% CI 7.1-14.2), or a combination of chemotherapy with mistletoe and hyperthermia 18.9 months (95% CI 15.2-24.5). While the survival times observed for patients with advanced pancreatic cancer receiving chemotherapy alone are consistent with pivotal phase-III studies and German registry data, we found significantly improved survival using additional mistletoe and/or hyperthermia.
Collapse
Affiliation(s)
- Anna Lena Hohneck
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, 69117 Heidelberg, Germany
- European Center for AngioScience (ECAS), German Center for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, 68167 Mannheim, Germany
| | - Largsi Sadikaj
- Onkologische Praxis Kaiserslautern, 67655 Kaiserslautern, Germany
| | - Lara Heinemann
- Department of Haematology and Oncology, University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, 69117 Heidelberg, Germany (R.-D.H.)
| | | | - Hartmut Riess
- AnthroMed Öschelbronn, Centrum für Integrative Medizin, 75223 Oeschelbronn, Germany; (H.R.)
| | - Annette Gerhards
- AnthroMed Öschelbronn, Centrum für Integrative Medizin, 75223 Oeschelbronn, Germany; (H.R.)
| | - Iris Burkholder
- Department of Nursing and Health, University of Applied Sciences of the Saarland, 66117 Saarbruecken, Germany
| | | | | | - Ralf-Dieter Hofheinz
- Department of Haematology and Oncology, University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, 69117 Heidelberg, Germany (R.-D.H.)
| |
Collapse
|
24
|
Chen L, Zhao D, Ren X, Ren J, Meng X, Fu C, Li X. Shikonin-Loaded Hollow Fe-MOF Nanoparticles for Enhanced Microwave Thermal Therapy. ACS Biomater Sci Eng 2023; 9:5405-5417. [PMID: 37638660 PMCID: PMC10498989 DOI: 10.1021/acsbiomaterials.3c00644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 08/15/2023] [Indexed: 08/29/2023]
Abstract
Microwave (MW) thermal therapy has been widely used for the treatment of cancer in clinics, but it still shows limited efficacy and a high recurrence rate owing to non-selective heat delivery and thermo-resistance. Regulating glycolysis shows great promise to improve MW thermal therapy since glycolysis plays an important role in thermo-resistance, progression, metabolism, and recurrence. Herein, we developed a delivery nanosystem of shikonin (SK)-loaded and hyaluronic acid (HA)-modified hollow Fe-MOF (HFM), HFM@SK@HA, as an efficient glycolysis-meditated agent to improve the efficacy of MW thermal therapy. The HFM@SK@HA nanosystem shows a high SK loading capacity of 31.7 wt %. The loaded SK can be effectively released from the HFM@SK@HA under the stimulation of an acidic tumor microenvironment and MW irradiation, overcoming the intrinsically low solubility and severe toxicity of SK. We also find that the HFM@SK@HA can not only greatly improve the heating effect of MW in the tumor site but also mediate MW-enhancing dynamic therapy efficiency by catalyzing the endogenous H2O2 to generate reactive oxygen species (ROS). As such, the MW irradiation treatment in the presence of HFM@SK@HA in vitro enables a highly improved anti-tumor efficacy due to the combined effect of released SK and generated ROS on inhibiting glycolysis in cancer cells. Our in vivo experiments show that the tumor inhibition rate is up to 94.75% ± 3.63% with no obvious recurrence during the 2 weeks after treatment. This work provides a new strategy for improving the efficacy of MW thermal therapy.
Collapse
Affiliation(s)
- Lufeng Chen
- Department
of Radiation Oncology, First Clinical Medical
School and First Hospital of Shanxi Medical University, No.85 Jiefang Road, Taiyuan City 030001, PR China
| | - Dongming Zhao
- Department
of Radiation Oncology, First Clinical Medical
School and First Hospital of Shanxi Medical University, No.85 Jiefang Road, Taiyuan City 030001, PR China
- Department
of Pathology, Basic Medical School, Shanxi
Medical University, No.56 Xinjian Road, Taiyuan City 030001, PR China
| | - Xiangling Ren
- Laboratory
of Controllable Preparation and Application of Nanomaterials, Technical
Institute of Physics and Chemistry, Chinese
Academy of Sciences, No.29 East Road Zhongguancun, Beijing 100190, PR China
- CAS
Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Jun Ren
- Laboratory
of Controllable Preparation and Application of Nanomaterials, Technical
Institute of Physics and Chemistry, Chinese
Academy of Sciences, No.29 East Road Zhongguancun, Beijing 100190, PR China
- CAS
Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Xianwei Meng
- Laboratory
of Controllable Preparation and Application of Nanomaterials, Technical
Institute of Physics and Chemistry, Chinese
Academy of Sciences, No.29 East Road Zhongguancun, Beijing 100190, PR China
- CAS
Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Changhui Fu
- Laboratory
of Controllable Preparation and Application of Nanomaterials, Technical
Institute of Physics and Chemistry, Chinese
Academy of Sciences, No.29 East Road Zhongguancun, Beijing 100190, PR China
- CAS
Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Xianfeng Li
- Department
of Radiation Oncology, First Clinical Medical
School and First Hospital of Shanxi Medical University, No.85 Jiefang Road, Taiyuan City 030001, PR China
- Department
of Pathology, Basic Medical School, Shanxi
Medical University, No.56 Xinjian Road, Taiyuan City 030001, PR China
| |
Collapse
|
25
|
Drizdal T, van Rhoon GC, Fiser O, Vrba D, van Holthe N, Vrba J, Paulides MM. Assessment of the thermal tissue models for the head and neck hyperthermia treatment planning. J Therm Biol 2023; 115:103625. [PMID: 37429086 DOI: 10.1016/j.jtherbio.2023.103625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 06/09/2023] [Accepted: 06/09/2023] [Indexed: 07/12/2023]
Abstract
PURPOSE To compare different thermal tissue models for head and neck hyperthermia treatment planning, and to assess the results using predicted and measured applied power data from clinical treatments. METHODS Three commonly used temperature models from literature were analysed: "constant baseline", "constant thermal stress" and "temperature dependent". Power and phase data of 93 treatments of 20 head and neck patients treated with the HYPERcollar3D applicator were used. The impact on predicted median temperature T50 inside the target region was analysed with maximum allowed temperature of 44 °C in healthy tissue. The robustness of predicted T50 for the three models against the influence of blood perfusion, thermal conductivity and the assumed hotspot temperature level was analysed. RESULTS We found an average predicted T50 of 41.0 ± 1.3 °C (constant baseline model), 39.9 ± 1.1 °C (constant thermal stress model) and 41.7 ± 1.1 °C (temperature dependent model). The constant thermal stress model resulted in the best agreement between the predicted power (P = 132.7 ± 45.9 W) and the average power measured during the hyperthermia treatments (P = 129.1 ± 83.0 W). CONCLUSION The temperature dependent model predicts an unrealistically high T50. The power values for the constant thermal stress model, after scaling simulated maximum temperatures to 44 °C, matched best to the average measured powers. We consider this model to be the most appropriate for temperature predictions using the HYPERcollar3D applicator, however further studies are necessary for developing of robust temperature model for tissues during heat stress.
Collapse
Affiliation(s)
- Tomas Drizdal
- Hyperthermia Unit, Dept. of Radiation Oncology, Erasmus MC Cancer Institute, Dr. Molewaterplein, 3015 GD, Rotterdam, Rotterdam, the Netherlands; Dept. of Biomedical Technology, Faculty of Biomedical Engineering, Czech Technical University in Prague, Nam. Sitna 3105, 272 01, Kladno, Czech Republic.
| | - Gerard C van Rhoon
- Hyperthermia Unit, Dept. of Radiation Oncology, Erasmus MC Cancer Institute, Dr. Molewaterplein, 3015 GD, Rotterdam, Rotterdam, the Netherlands
| | - Ondrej Fiser
- Dept. of Biomedical Technology, Faculty of Biomedical Engineering, Czech Technical University in Prague, Nam. Sitna 3105, 272 01, Kladno, Czech Republic
| | - David Vrba
- Dept. of Biomedical Technology, Faculty of Biomedical Engineering, Czech Technical University in Prague, Nam. Sitna 3105, 272 01, Kladno, Czech Republic
| | - Netteke van Holthe
- Hyperthermia Unit, Dept. of Radiation Oncology, Erasmus MC Cancer Institute, Dr. Molewaterplein, 3015 GD, Rotterdam, Rotterdam, the Netherlands
| | - Jan Vrba
- Dept. of Biomedical Technology, Faculty of Biomedical Engineering, Czech Technical University in Prague, Nam. Sitna 3105, 272 01, Kladno, Czech Republic
| | - Margarethus M Paulides
- Hyperthermia Unit, Dept. of Radiation Oncology, Erasmus MC Cancer Institute, Dr. Molewaterplein, 3015 GD, Rotterdam, Rotterdam, the Netherlands; Dept. of Electrical Engineering, Eindhoven University of Technology, De Rondom 70, 5612 AP, Eindhoven, the Netherlands
| |
Collapse
|
26
|
Álvarez Álvarez R, Manzano A, Agra Pujol C, Artigas Raventós V, Correa R, Cruz Jurado J, Fernandez JA, Garcia Del Muro X, Gonzalez JA, Hindi N, Lozano Lominchar P, Martínez-Trufero J, Méndez R, Muñoz M, Muñoz Casares C, Orbis Castellanos F, Orellana Fernandez R, Paniagua González M, Redondo A, Valverde Morales C, Asencio JM. Updated Review and Clinical Recommendations for the Diagnosis and Treatment of Patients with Retroperitoneal Sarcoma by the Spanish Sarcoma Research Group (GEIS). Cancers (Basel) 2023; 15:3194. [PMID: 37370803 DOI: 10.3390/cancers15123194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/06/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
Soft tissue sarcomas (STS) are an uncommon and biologically heterogeneous group of tumors arising from mesenchymal cells. The incidence is estimated at five cases per 100,000 people per year. Retroperitoneal sarcomas (RPS) account for 10-15% of all STS, and their management depends on their anatomical characteristics and histotype. Due to their very low incidence, it is recommended that RPS be treated in reference centers and evaluated by an experienced multidisciplinary team (MDT). In Spain, the Spanish Group for Research in Sarcomas (GEIS) brings together experts from various specialties to promote research on sarcomas and improve treatment results. This paper summarizes the GEIS recommendations for the diagnosis, treatment, and follow-up of patients with RPS.
Collapse
Affiliation(s)
- Rosa Álvarez Álvarez
- Medical Oncology Department, Instituto de Investigacion Sanitaria Gregorio Marañon, Hospital Universitario Gregorio Marañon, 28007 Madrid, Spain
| | - Aránzazu Manzano
- Medical Oncology Department, Hospital Universitario Clínico San Carlos, 28040 Madrid, Spain
| | - Carolina Agra Pujol
- Pathology Department, Hospital Universitario Gregorio Marañon, Complutense University, 28007 Madrid, Spain
| | - Vicente Artigas Raventós
- Surgery Department, Hospital Universitario Sant Pau, Universidad Autonoma de Barcelona, 08035 Barcelona, Spain
| | - Raquel Correa
- Radiation Oncology Department, Hospital Virgen de la Victoria, 29010 Malaga, Spain
| | - Josefina Cruz Jurado
- Medical Oncology Department, Hospital Universitario Canarias, 38320 Santa Cruz de Tenerife, Spain
| | - Juan Angel Fernandez
- Sarcoma Multidisciplinary Unit, Surgery Department, Hospital Virgen de la Arrixaca, 30120 Murcia, Spain
| | - Xavier Garcia Del Muro
- Sarcoma Multidisciplinary Unit, Medical Oncology Department, Idibell, Instituto Catalan Oncología Hospitalet, 08908 Barcelona, Spain
| | - Jose Antonio Gonzalez
- Surgery Department, Hospital Universitario Sant Pau, Universidad Autonoma de Barcelona, 08035 Barcelona, Spain
| | - Nadia Hindi
- Medical Oncology Department, Fundacion Jimenez Diaz University Hospital, 28040 Madrid, Spain
- Medical Oncology Department, Hospital General de Villalba, 28400 Madrid, Spain
- Health Research Institute-Fundación Jiménez Díaz (IIS-FJD), Universidad Autónoma de Madrid (UAM), 28040 Madrid, Spain
| | - Pablo Lozano Lominchar
- Surgery Department, Hospital Universitario Gregorio Marañon, Complutense University, 28040 Madrid, Spain
| | | | - Ramiro Méndez
- Radiology Department, Hospital Universitario Clínico San Carlos, 28040 Madrid, Spain
| | - Mercedes Muñoz
- Radiation Oncology Department, Hospital Universitario Gregorio Marañon, Complutense University, 28007 Madrid, Spain
| | | | - Francisco Orbis Castellanos
- Sarcoma Multidisciplinary Unit, Surgery Department, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
| | | | - Miguel Paniagua González
- Radiology Department, Hospital Universitario Gregorio Marañon, Complutense University, 28007 Madrid, Spain
| | - Andres Redondo
- Medical Oncology Department, Hospital Universitario La Paz-IdiPAZ, 28046 Madrid, Spain
| | | | - Jose Manuel Asencio
- Surgery Department, Hospital Universitario Gregorio Marañon, Complutense University, 28040 Madrid, Spain
| |
Collapse
|
27
|
Rupp L, Resag A, Potkrajcic V, Warm V, Wehner R, Jöhrens K, Bösmüller H, Eckert F, Schmitz M. Prognostic impact of the post-treatment T cell composition and spatial organization in soft tissue sarcoma patients treated with neoadjuvant hyperthermic radio(chemo)therapy. Front Immunol 2023; 14:1185197. [PMID: 37261361 PMCID: PMC10228739 DOI: 10.3389/fimmu.2023.1185197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 05/03/2023] [Indexed: 06/02/2023] Open
Abstract
Soft tissue sarcomas (STS) form a heterogeneous group of tumors sharing a mesenchymal origin. Despite good local control of the disease, the occurrence of distant metastases often limits survival of STS patients with localized, high-risk tumors of the extremities. Accumulating evidence suggests a central role for the tumor immune microenvironment in determining the clinical outcome and response to therapy. Thus, it has been reported that STS patients with a high immune signature and especially presence of B cells and tertiary lymphoid structures display improved overall survival and response to checkpoint inhibitor treatment. Here, we explored the effect of curative multimodal therapy on the T cell landscape of STS using multiplex immunohistochemistry. We analyzed the phenotype, frequency, and spatial distribution of STS-infiltrating CD8+ T cells by staining for CD8, 4-1BB, Granzyme B, Ki67, PD-1, and LAG-3 as well as CD3+ T helper cells using a panel consisting of CD3, T-bet, GATA3, RORγT, FoxP3, and Ki67. All patients received neoadjuvant radiotherapy plus locoregional hyperthermia with or without chemotherapy. While the treatment-naïve biopsy sample allows an analysis of baseline T cell infiltration levels, both intra- and peritumoral areas of the matched resected tissue were analyzed to assess composition and spatial distribution of the T cell compartment and its therapeutic modulation. Generally, post-treatment tissues displayed lower frequencies of CD3+ and CD8+ T cells. Association with clinical data revealed that higher post-treatment frequencies of peritumoral and intratumoral CD3+ T cells and intratumoral PD-1+ CD8+ T cells were significantly associated with improved disease-free survival (DFS), while these densities had no prognostic significance in the biopsy. Upon spatial analysis, a high ratio of intratumoral to peritumoral CD8+ T cells emerged as an independent prognostic marker for longer DFS. These results indicate that the STS T cell landscape is altered by multimodal therapy and may influence the clinical outcome of patients. An enhanced understanding of the STS immune architecture and its modulation by neoadjuvant therapy may pave the way towards novel treatment modalities and improve the long-term clinical outcome of STS patients.
Collapse
Affiliation(s)
- Luise Rupp
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, Technische Universität (TU) Dresden, Dresden, Germany
| | - Antonia Resag
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, Technische Universität (TU) Dresden, Dresden, Germany
| | - Vlatko Potkrajcic
- Department of Radiation Oncology, University Hospital Tuebingen, Tuebingen, Germany
| | - Verena Warm
- Institute of Pathology, University Hospital Tuebingen, Tuebingen, Germany
| | - Rebekka Wehner
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, Technische Universität (TU) Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Korinna Jöhrens
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute of Pathology, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Hans Bösmüller
- Institute of Pathology, University Hospital Tuebingen, Tuebingen, Germany
| | - Franziska Eckert
- Department of Radiation Oncology, University Hospital Tuebingen, Tuebingen, Germany
- Department of Radiation Oncology, Medical University of Vienna, Comprehensive Cancer Center Vienna, Vienna, Austria
| | - Marc Schmitz
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, Technische Universität (TU) Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
28
|
Schopow N, Hohenberger P, Gockel I, Osterhoff G. [Multimodal treatment of localized high-grade soft tissue sarcomas of the extremities : Summary and discussion of the current S3 guidelines on adult soft tissue sarcomas and the ESMO guidelines on soft tissue and visceral sarcomas]. CHIRURGIE (HEIDELBERG, GERMANY) 2023; 94:424-431. [PMID: 37041391 DOI: 10.1007/s00104-023-01872-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/24/2023] [Indexed: 04/13/2023]
Abstract
Soft tissue sarcomas are rare, heterogeneous tumors that are frequently in the extremities. Treatment includes surgical resection, combination chemotherapy and/or radiotherapy, as well as supplementary procedures such as isolated limb perfusion and regional deep hyperthermia. The prognosis depends on the tumor stage and the approximately 70 histological subtypes, with specific treatment approaches existing only for some subtypes. This review summarizes the recommendations of the German S3 guideline "Adult Soft Tissue Sarcomas" and the European Society for Medical Oncology (ESMO) guideline "Soft tissue and visceral sarcomas" regarding the diagnostic workup and therapy of soft tissue sarcomas of the extremities.
Collapse
Affiliation(s)
- Nikolas Schopow
- Klinik und Poliklinik für Orthopädie, Unfallchirurgie und Plastische Chirurgie, Universitätsklinikum Leipzig, Liebigstraße 20, 04103, Leipzig, Deutschland
| | - Peter Hohenberger
- Sektion Spezielle chirurgische Onkologie und Thoraxchirurgie, Chirurgische Klinik, Universitätsklinikum Mannheim, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Deutschland
| | - Ines Gockel
- Klinik für Viszeral‑, Transplantations‑, Thorax- und Gefäßchirurgie, Universitätsklinikum Leipzig, Liebigstraße 20, 04103, Leipzig, Deutschland
| | - Georg Osterhoff
- Klinik und Poliklinik für Orthopädie, Unfallchirurgie und Plastische Chirurgie, Universitätsklinikum Leipzig, Liebigstraße 20, 04103, Leipzig, Deutschland.
| |
Collapse
|
29
|
Castro-Torres JL, Méndez J, Torres-Lugo M, Juan E. Development of handheld induction heaters for magnetic fluid hyperthermia applications and in-vitroevaluation on ovarian and prostate cancer cell lines. Biomed Phys Eng Express 2023; 9:035010. [PMID: 36827691 PMCID: PMC9999354 DOI: 10.1088/2057-1976/acbeaf] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 02/24/2023] [Indexed: 02/26/2023]
Abstract
Objective:Magnetic fluid hyperthermia (MFH) is a still experimental technique found to have a potential application in the treatment of cancer. The method aims to reach around 41 °C-47 °C in the tumor site by exciting magnetic nanoparticles with an externally applied alternating magnetic field (AMF), where cell death is expected to occur. Applying AMFs with high spatial resolution is still a challenge. The AMFs from current and prospective MFH applicators cover relatively large areas; being not suitable for patients having metallic implants near the treatment area. Thus, there will be a clinical need for smaller magnetic field applicators. To this end, a laparoscopic induction heater (LIH) and a transrectal induction heater (TRIH) were developed.Methods:Miniature 'pancake' coils were wound and inserted into 3D printed enclosures. Ovarian (SKOV-3, A2780) and prostate (PC-3, LNCaP) cancer cell lines were used to evaluate the instruments' capabilities in killing cancer cellsin vitro, using Synomag®-D nanoparticles as the heat mediators. NIH3T3 normal cell lines were also used with both devices to observe if these cells tolerated the conditions applied.Results:Magnetic field intensities reached by the LIH and TRIH were 42.6 kA m-1at 326 kHz and 26.3 kA m-1at 303 kHz, respectively. Temperatures reached in the samples were 41 °C by the LIH and 43 °C by the TRIH. Both instruments successfully accomplished killing cancer cells, with minimal effects on normal cells.Conclusion:This work presents the first line of handheld medical induction heaters and have the potential to be a complement to existing cancer therapies.Significance:These instruments could enable the development of MFH modalities that will facilitate the clinical translation of this thermal treatment.
Collapse
Affiliation(s)
| | - Janet Méndez
- Chemical Engineering Department, University of Puerto Rico, Mayagüez, Puerto Rico
| | - Madeline Torres-Lugo
- Chemical Engineering Department, University of Puerto Rico, Mayagüez, Puerto Rico
| | - Eduardo Juan
- Electrical and Computer Engineering Department, University of Puerto Rico, Mayagüez, Puerto Rico
| |
Collapse
|
30
|
Issels RD, Boeck S, Pelzer U, Mansmann U, Ghadjar P, Lindner LH, Albertsmeier M, Angele MK, Schmidt M, Xu Y, Bahra M, Pratschke J, Schoenberg M, Thasler WE, Salat C, Stoetzer OJ, Knoefel WT, Graf D, Wessalowski R, Keitel-Anselmino V, Koenigsrainer A, Bitzer M, Zips D, Bamberg M, Fietkau R, Ott O, Kawecki M, Wyrwicz L, Rutkowski P, Rentsch M, Ababei J, Reichardt P, Rigamonti M, Weber B, Abdel-Rahman S, Tschoep-Lechner K, Jauch KW, Bruns CJ, Oettle H, von Bergwelt-Baildon M, Heinemann V, Werner J. Regional hyperthermia with cisplatin added to gemcitabine versus gemcitabine in patients with resected pancreatic ductal adenocarcinoma: The HEAT randomised clinical trial. Eur J Cancer 2023; 181:155-165. [PMID: 36657324 DOI: 10.1016/j.ejca.2022.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/13/2022] [Accepted: 12/13/2022] [Indexed: 12/31/2022]
Abstract
BACKGROUND Regional hyperthermia (RHT) with cisplatin added to gemcitabine showed efficacy in gemcitabine-pre-treated patients with advanced pancreatic ductal adenocarcinoma. We conducted a randomised clinical trial to investigate RHT with cisplatin added to gemcitabine (GPH) compared with gemcitabine (G) in the adjuvant setting of resected pancreatic ductal adenocarcinoma. METHODS This randomised, multicentre, open-label trial randomly assigned patients to either GPH (gemcitabine 1000 mg/m2 on day 1, 15 and cisplatin 25 mg/m2 with RHT on day 2, 3 and 15,16) or to G (gemcitabine 1000 mg/m2 on day 1,8,15), four-weekly over six cycles. Disease-free survival (DFS) was the primary end-point. Secondary end-points included overall survival (OS) and safety. RESULTS A total of 117 eligible patients (median age, 63 years) were randomly allocated to treatment (57 GPH; 60 G). With a follow-up time of 56.6 months, the median DFS was 12.7 compared to 11.2 months for GPH and G, respectively (p = 0.394). Median post-recurrence survival was significantly prolonged in the GPH-group (15.3 versus 9.8 months; p = 0.031). Median OS reached 33.2 versus 25.2 months (p = 0.099) with 5-year survival rates of 28.4% versus 18.7%. Excluding eight patients who received additional capecitabine in the G-arm (investigators choice), median OS favoured GPH (p = 0.052). Adverse events CTCAE (Common Terminology Criteria for Adverse Events) grade ≥3 occurred in 61.5% (GPH) versus 63.6% (G) of patients. Two patients in the G-group died because of treatment-related toxic effects. CONCLUSIONS The randomised controlled Hyperthermia European Adjuvant Trial study failed to demonstrate a significant difference in DFS. However, it suggests a difference in post-recurrence survival and a trend for improved OS. CLINICALTRIALS gov, number NCT01077427.
Collapse
Affiliation(s)
- Rolf D Issels
- Ludwig Maximilians University LMU University Hospital Munich, Germany.
| | - Stefan Boeck
- Ludwig Maximilians University LMU University Hospital Munich, Germany
| | - Uwe Pelzer
- Charite University Hospital Berlin, Germany
| | - Ulrich Mansmann
- Ludwig Maximilian University Munich Institute of Medical Data Processing Biometrics and Epidemiology, Germany
| | | | - Lars H Lindner
- Ludwig Maximilians University LMU University Hospital Munich, Germany
| | | | - Martin K Angele
- Ludwig Maximilians University LMU University Hospital Munich, Germany
| | - Michael Schmidt
- Ludwig Maximilian University Munich Institute of Medical Data Processing Biometrics and Epidemiology, Germany
| | - Yujun Xu
- Ludwig Maximilian University Munich Institute of Medical Data Processing Biometrics and Epidemiology, Germany
| | - Marcus Bahra
- Academic Hospital Waldfriede of the Charité, Berlin, Germany
| | | | | | | | - Christoph Salat
- Medical Center for Hematology and Oncology München GmbH, Germany
| | | | | | - Dirk Graf
- Rheinland Hospital Group Grevenbroich St Elizabeth Hospital, Grevenbroich, Germany
| | | | | | | | | | | | | | | | | | - Maciej Kawecki
- Maria Skłodowska Curie Memorial Cancer Centre, Warsaw, Poland
| | - Lucjan Wyrwicz
- Maria Skłodowska Curie Memorial Cancer Centre, Warsaw, Poland
| | - Piotr Rutkowski
- Maria Skłodowska Curie Memorial Cancer Centre, Warsaw, Poland
| | | | | | | | | | | | | | | | - Karl-Walter Jauch
- Ludwig Maximilians University LMU University Hospital Munich, Germany
| | | | | | | | - Volker Heinemann
- Ludwig Maximilians University LMU University Hospital Munich, Germany
| | - Jens Werner
- Ludwig Maximilians University LMU University Hospital Munich, Germany
| | | |
Collapse
|
31
|
Huffman OG, Chau DB, Dinicu AI, DeBernardo R, Reizes O. Mechanistic Insights on Hyperthermic Intraperitoneal Chemotherapy in Ovarian Cancer. Cancers (Basel) 2023; 15:cancers15051402. [PMID: 36900195 PMCID: PMC10000881 DOI: 10.3390/cancers15051402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 02/10/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
Epithelial ovarian cancer is an aggressive disease of the female reproductive system and a leading cause of cancer death in women. Standard of care includes surgery and platinum-based chemotherapy, yet patients continue to experience a high rate of recurrence and metastasis. Hyperthermic intraperitoneal chemotherapy (HIPEC) treatment in highly selective patients extends overall survival by nearly 12 months. The clinical studies are highly supportive of the use of HIPEC in the treatment of ovarian cancer, though the therapeutic approach is limited to academic medical centers. The mechanism underlying HIPEC benefit remains unknown. The efficacy of HIPEC therapy is impacted by several procedural and patient/tumor factors including the timing of surgery, platinum sensitivity, and molecular profiling such as homologous recombination deficiency. The present review aims to provide insight into the mechanistic benefit of HIPEC treatment with a focus on how hyperthermia activates the immune response, induces DNA damage, impairs DNA damage repair pathways, and has a synergistic effect with chemotherapy, with the ultimate outcome of increasing chemosensitivity. Identifying the points of fragility unmasked by HIPEC may provide the key pathways that could be the basis of new therapeutic strategies for ovarian cancer patients.
Collapse
Affiliation(s)
- Olivia G. Huffman
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Danielle B. Chau
- Division of Gynecologic Oncology, Obstetrics, Gynecology and Women’s Health Institute, Cleveland Clinic, Cleveland, OH 44124, USA
| | - Andreea I. Dinicu
- Division of Gynecologic Oncology, Obstetrics, Gynecology and Women’s Health Institute, Cleveland Clinic, Cleveland, OH 44124, USA
| | - Robert DeBernardo
- Division of Gynecologic Oncology, Obstetrics, Gynecology and Women’s Health Institute, Cleveland Clinic, Cleveland, OH 44124, USA
- Case Comprehensive Cancer Center, Cleveland, OH 44106, USA
| | - Ofer Reizes
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA
- Case Comprehensive Cancer Center, Cleveland, OH 44106, USA
- Correspondence: ; Tel.: +1-216-445-0880
| |
Collapse
|
32
|
Brummelhuis ISG, Crezee J, Witjes JA. Evaluation of thermal dose effect in radiofrequency-induced hyperthermia with intravesical chemotherapy for nonmuscle invasive bladder cancer. Int J Hyperthermia 2023; 40:2157498. [PMID: 36755433 DOI: 10.1080/02656736.2022.2157498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
Abstract
PURPOSE In nonmuscle invasive bladder cancer (NMIBC) patients who fail standard intravesical treatment and are unfit or unwilling to undergo a radical cystectomy, radiofrequency (RF)-induced hyperthermia combined with intravesical chemotherapy (RF-CHT) has shown promising results. We studied whether higher thermal dose improves clinical NMIBC outcome. METHODS AND MATERIALS The cohort comprised 108 patients who started with RF-CHT between November 2013 and December 2019. Patients received intravesical mitomycin-C or epirubicin. Bladder hyperthermia was accomplished with an intravesical 915 MHz RF device guided by intravesical thermometry. We assessed the association between thermal dose parameters (including median temperature and Cumulative Equivalent Minutes of T50 at 43 °C [CEM43T50]) and complete response (CR) at six months for patients with (concomitant) carcinoma in situ (CIS), and recurrence-free survival (RFS) for patients with papillary disease. RESULTS Median temperature and CEM43T50 per treatment were 40.9 (IQR 40.8-41.1) °C and 3.1 (IQR 0.9-2.4) minutes, respectively. Analyses showed no association between any thermal dose parameter and CR or RFS (p > 0.05). Less bladder spasms during treatment sessions was associated with increased median temperature and CEM43T50 (adjusted OR 0.01 and 0.34, both p < 0.001). CONCLUSIONS No significant association between thermal dose and NMIBC outcome was found. Possibly thermal dose effect in patients of the current cohort exceeds a certain threshold value. On the other hand, occurrence of bladder spasms had a thermal dose limiting effect. We advise to treat patients with temperatures >40.5 °C for at least 45 min while respecting individual tolerability, including occurrence of bladder spasms.
Collapse
Affiliation(s)
- Iris S G Brummelhuis
- Department of Urology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Johannes Crezee
- Department of Radiotherapy, Amsterdam University Medical Centers, Cancer Center Amsterdam, University of Amsterdam, Amsterdam, The Netherlands
| | - J Alfred Witjes
- Department of Urology, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
33
|
Fausti V, De Vita A, Vanni S, Ghini V, Gurrieri L, Riva N, Casadei R, Maraldi M, Ercolani G, Cavaliere D, Pacilio CA, Pieri F, Foca F, Bongiovanni A, Ranallo N, Calpona S, Frassineti GL, Ibrahim T, Mercatali L. Systemic Inflammatory Indices in Second-Line Soft Tissue Sarcoma Patients: Focus on Lymphocyte/Monocyte Ratio and Trabectedin. Cancers (Basel) 2023; 15:cancers15041080. [PMID: 36831421 PMCID: PMC9954182 DOI: 10.3390/cancers15041080] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 01/29/2023] [Accepted: 01/31/2023] [Indexed: 02/11/2023] Open
Abstract
A second-line standard of treatment has not yet been identified in patients with soft tissue sarcomas (STS), so identifying predictive markers could be a valuable tool. Recent studies have shown that the intratumoral and inflammatory systems significantly influence tumor aggressiveness. We aimed to investigate prognostic values of pre-therapy neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), lymphocyte-to-monocyte ratio (LMR), systemic inflammatory index (SII), progression-free survival (PFS), and overall survival (OS) of STS patients receiving second-line treatment. In this single-center retrospective analysis, ninety-nine patients with STS were enrolled. All patients received second-line treatment after progressing to anthracycline. PFS and OS curves were calculated using the Kaplan-Meier method of RNA sequencing, and CIBERSORT analysis was performed on six surgical specimens of liposarcoma patients. A high NLR, PLR, and SII were significantly associated with worse PFS (p = 0.019; p = 0.004; p = 0.006). Low LMR was significantly associated with worse OS (p = 0.006). Patients treated with Trabectedin showed a better PFS when the LMR was low, while patients treated with other regimens showed a worse PFS when the LMR was low (p = 0.0154). The intratumoral immune infiltrates analysis seems to show a correlation between intratumoral macrophages and LMR. PS ECOG. The metastatic onset and tumor burden showed prognostic significance for PFS (p = 0.004; p = 0.041; p = 0.0086). According to the histologies, PFS was: 5.7 mo in liposarcoma patients vs. 3.8 mo in leiomyosarcoma patients vs. 3.1 months in patients with other histologies (p = 0.053). Our results confirm the prognostic role of systemic inflammatory markers in patients with STS. Moreover, we demonstrated that LMR is a specific predictor of Trabectedin efficacy and could be useful in daily clinical practice. We also highlighted a possible correlation between LMR levels and the percentage of intratumoral macrophages.
Collapse
Affiliation(s)
- Valentina Fausti
- Clinical and Experimental Oncology, Immunotherapy, Rare Cancers and Biological Resource Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
| | - Alessandro De Vita
- Preclinic and Osteoncology Unit, Bioscience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
- Correspondence: ; Tel.: +39-0543-739239; Fax: +39-0543-739221
| | - Silvia Vanni
- Preclinic and Osteoncology Unit, Bioscience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
| | - Virginia Ghini
- Clinical and Experimental Oncology, Immunotherapy, Rare Cancers and Biological Resource Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
| | - Lorena Gurrieri
- Clinical and Experimental Oncology, Immunotherapy, Rare Cancers and Biological Resource Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
| | - Nada Riva
- Clinical and Experimental Oncology, Immunotherapy, Rare Cancers and Biological Resource Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
| | - Roberto Casadei
- Orthopedic Unit, “Morgagni-Pierantoni” Hospital, 47121 Forlì, Italy
| | - Marco Maraldi
- Orthopedic Unit, “Morgagni-Pierantoni” Hospital, 47121 Forlì, Italy
| | - Giorgio Ercolani
- General and Oncologic Surgery, “Morgagni-Pierantoni” Hospital, 47121 Forlì, Italy
| | - Davide Cavaliere
- General and Oncologic Surgery, “Morgagni-Pierantoni” Hospital, 47121 Forlì, Italy
| | | | - Federica Pieri
- Pathology Unit, “Morgagni-Pierantoni” Hospital, 47121 Forlì, Italy
| | - Flavia Foca
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
| | - Alberto Bongiovanni
- Clinical and Experimental Oncology, Immunotherapy, Rare Cancers and Biological Resource Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
| | - Nicoletta Ranallo
- Clinical and Experimental Oncology, Immunotherapy, Rare Cancers and Biological Resource Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
| | - Sebastiano Calpona
- Clinical and Experimental Oncology, Immunotherapy, Rare Cancers and Biological Resource Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
| | - Giovanni Luca Frassineti
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
| | - Toni Ibrahim
- Osteoncologia, Sarcomi dell’osso e dei tessuti molli, e Terapie Innovative, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Laura Mercatali
- Preclinic and Osteoncology Unit, Bioscience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
| |
Collapse
|
34
|
Abstract
Immunotherapy has revolutionized the treatment of patients with cancer. However, promoting antitumour immunity in patients with tumours that are resistant to these therapies remains a challenge. Thermal therapies provide a promising immune-adjuvant strategy for use with immunotherapy, mostly owing to the capacity to reprogramme the tumour microenvironment through induction of immunogenic cell death, which also promotes the recruitment of endogenous immune cells. Thus, thermal immunotherapeutic strategies for various cancers are an area of considerable research interest. In this Review, we describe the role of the various thermal therapies and provide an update on attempts to combine these with immunotherapies in clinical trials. We also provide an overview of the preclinical development of various thermal immuno-nanomedicines, which are capable of combining thermal therapies with various immunotherapy strategies in a single therapeutic platform. Finally, we discuss the challenges associated with the clinical translation of thermal immuno-nanomedicines and emphasize the importance of multidisciplinary and inter-professional collaboration to facilitate the optimal translation of this technology from bench to bedside.
Collapse
|
35
|
Rothermundt C, Andreou D, Blay JY, Brodowicz T, Desar IME, Dileo P, Gelderblom H, Haas R, Jakob J, Jones RL, Judson I, Kunz WG, Liegl-Atzwanger B, Lindner LH, Messiou C, Miah AB, Reichardt P, Szkandera J, van der Graaf WTA, van Houdt WJ, Wardelmann E, Hofer S. Controversies in the management of patients with soft tissue sarcoma: Recommendations of the Conference on State of Science in Sarcoma 2022. Eur J Cancer 2023; 180:158-179. [PMID: 36599184 DOI: 10.1016/j.ejca.2022.11.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 11/04/2022] [Indexed: 11/17/2022]
Abstract
BACKGROUND Owing to the rarity and heterogeneity in biology and presentation, there are multiple areas in the diagnosis, treatment and follow-up of soft tissue sarcoma (STS), with no, low-level or conflicting evidence. METHODS During the first Consensus Conference on the State of Science in Sarcoma (CSSS), we used a modified Delphi process to identify areas of controversy in the field of sarcoma, to name topics with limited evidence-based data in which a scientific and knowledge gap may remain and a consensus statement will help to guide patient management. We determined scientific questions which need to be addressed in the future in order to generate evidence and to inform physicians and caregivers in daily clinical practice in order to improve the outcomes of patients with sarcoma. We conducted a vote on STS key questions and controversies prior to the CSSS meeting, which took place in May 2022. RESULTS Sixty-two European sarcoma experts participated in the survey. Sixteen strong consensus (≥95%) items were identified by the experts, as well as 30 items with a ≥75% consensus on diagnostic and therapeutic questions. Ultimately, many controversy topics remained without consensus. CONCLUSIONS In this manuscript, we summarise the voting results and the discussion during the CSSS meeting. Future scientific questions, priorities for clinical trials, registries, quality assurance, and action by stakeholders are proposed. Platforms and partnerships can support innovative approaches to improve management and clinical research in STS.
Collapse
Affiliation(s)
- Christian Rothermundt
- Department of Medical Oncology and Haematology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland.
| | - Dimosthenis Andreou
- Department of Orthopedics and Trauma, Medical University of Graz, Graz, Austria
| | - Jean-Yves Blay
- Department of Medicine, Léon Bérard Center, Lyon, France
| | - Thomas Brodowicz
- Department of Medical Oncology, General Hospital - Medical University of Vienna, Vienna, Austria
| | - Ingrid M E Desar
- Department of Medical Oncology, Radboud University Medical Center Nijmegen, the Netherlands
| | - Palma Dileo
- London Sarcoma Service, Department of Oncology, University College Hospital London, United Kingdom
| | - Hans Gelderblom
- Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Rick Haas
- Department of Radiotherapy, The Netherlands Cancer Institute, Amsterdam and the Leiden University Medical Center, Leiden, the Netherlands
| | - Jens Jakob
- Sarcoma Unit, Department of Surgery, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany
| | - Robin L Jones
- Royal Marsden Hospital and Institute of Cancer Research, London, United Kingdom
| | - Ian Judson
- The Institute of Cancer Research, London, United Kingdom
| | - Wolfgang G Kunz
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | | | - Lars H Lindner
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
| | - Christina Messiou
- Department of Radiology, The Royal Marsden NHS Foundation Trust and The Institute of Cancer Research, London, United Kingdom
| | - Aisha B Miah
- Department of Radiotherapy, The Royal Marsden NHS Foundation Trust and The Institute of Cancer Research, London, United Kingdom
| | - Peter Reichardt
- Department of Oncology and Palliative Care, Helios Klinikum Berlin-Buch, Berlin, Germany
| | - Joanna Szkandera
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | | | - Winan J van Houdt
- Department of Surgical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Eva Wardelmann
- Gerhard-Domagk-Institute of Pathology, University Hospital Muenster, Muenster, Germany
| | - Silvia Hofer
- Department of Neurology, University Hospital Zurich, Zurich, Switzerland
| | | |
Collapse
|
36
|
Chia BSH, Ho SZ, Tan HQ, Chua MLK, Tuan JKL. A Review of the Current Clinical Evidence for Loco-Regional Moderate Hyperthermia in the Adjunct Management of Cancers. Cancers (Basel) 2023; 15:cancers15020346. [PMID: 36672300 PMCID: PMC9856725 DOI: 10.3390/cancers15020346] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/23/2022] [Accepted: 12/27/2022] [Indexed: 01/06/2023] Open
Abstract
Regional hyperthermia therapy (RHT) is a treatment that applies moderate heat to tumours in an attempt to potentiate the effects of oncological treatments and improve responses. Although it has been used for many years, the mechanisms of action are not fully understood. Heterogenous practices, poor quality assurance, conflicting clinical evidence and lack of familiarity have hindered its use. Despite this, several centres recognise its potential and have adopted it in their standard treatment protocols. In recent times, significant technical improvements have been made and there is an increasing pool of evidence that could revolutionise its use. Our narrative review aims to summarise the recently published prospective trial evidence and present the clinical effects of RHT when added to standard cancer treatments. In total, 31 studies with higher-quality evidence across various subsites are discussed herein. Although not all of these studies are level 1 evidence, benefits of moderate RHT in improving local tumour control, survival outcomes and quality of life scores were observed across the different cancer subsites with minimal increase in toxicities. This paper may serve as a reference when considering this technique for specific indications.
Collapse
Affiliation(s)
- Brendan Seng Hup Chia
- Division of Radiation Oncology, National Cancer Centre Singapore, 11 Hospital Drive, Singapore 169610, Singapore
- Correspondence:
| | - Shaun Zhirui Ho
- Department of Radiation Oncology, 585 North Bridge Rd, Level 10 Raffles Specialist Centre, Singapore 188770, Singapore
| | - Hong Qi Tan
- Division of Radiation Oncology, National Cancer Centre Singapore, 11 Hospital Drive, Singapore 169610, Singapore
| | - Melvin Lee Kiang Chua
- Division of Radiation Oncology, National Cancer Centre Singapore, 11 Hospital Drive, Singapore 169610, Singapore
| | - Jeffrey Kit Loong Tuan
- Division of Radiation Oncology, National Cancer Centre Singapore, 11 Hospital Drive, Singapore 169610, Singapore
| |
Collapse
|
37
|
Ott OJ, Gaipl US, Lamrani A, Fietkau R. The Emerging Evidence Supporting Integration of Deep Regional Hyperthermia With Chemoradiation in Bladder Cancer. Semin Radiat Oncol 2023; 33:82-90. [PMID: 36517198 DOI: 10.1016/j.semradonc.2022.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
For decades, the antineoplastic potential of hyperthermia alone or in combination with radiotherapy and/or chemotherapy has been subject of intensive preclinical and clinical research in various tumor entities. The clinical evidence on the beneficial effects of additional hyperthermia in combination with intravesical Mitomycin C for superficial non-muscle-invasive bladder cancer as well as for deep regional microwave hyperthermia techniques applied during an external beam radiotherapy or chemoradiation treatment for more advanced tumors are summarized. In some series, deep regional hyperthermia in combination with an initial transurethral resection and Cisplatin-based chemoradiation increased the 5-year overall survival rates up to 20%. The presented data justifies a fresh irrespective chance for mild regional hyperthermia in the context of new progressive prospective trials on multimodality treatment for bladder preservation.
Collapse
Affiliation(s)
- Oliver J Ott
- Universitätsklinikum Erlangen, Department of Radiation Oncology, Erlangen, Germany; Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany.
| | - Udo S Gaipl
- Universitätsklinikum Erlangen, Department of Radiation Oncology, Erlangen, Germany; Universitätsklinikum Erlangen, Department of Radiation Oncology, Translational Radiobiology, Erlangen, Germany
| | - Allison Lamrani
- Universitätsklinikum Erlangen, Department of Radiation Oncology, Erlangen, Germany
| | - Rainer Fietkau
- Universitätsklinikum Erlangen, Department of Radiation Oncology, Erlangen, Germany; Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| |
Collapse
|
38
|
Veltsista PD, Oberacker E, Ademaj A, Corradini S, Eckert F, Flörcken A, Kaul D, Lindner LH, Issels R, Ott OJ, Pink D, Potkrajcic V, Reichardt P, Roohani S, Spalek MJ, Riesterer O, Zips D, Ghadjar P. Hyperthermia in the treatment of high-risk soft tissue sarcomas: a systematic review. Int J Hyperthermia 2023; 40:2236337. [PMID: 37468132 DOI: 10.1080/02656736.2023.2236337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/07/2023] [Accepted: 07/07/2023] [Indexed: 07/21/2023] Open
Abstract
BACKGROUND The therapy of high-risk soft tissue sarcomas (STS) remains an interdisciplinary challenge. Regional hyperthermia (RHT) sparked interest as it has been shown to improve overall survival when added to perioperative chemotherapy (CTX). However, questions arise on how RHT should be optimally integrated into current multi-modal therapies. MATERIALS AND METHODS We performed a systematic literature review according to Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Studies written in English and focused mainly on radiative RHT and superficial hyperthermia were evaluated and included. Studies including patients below the age of 18, with metastatic disease or review articles, were excluded. RESULTS We identified 15 clinical reports from 1990 until July 2022. Three articles combined RHT + CTX, and twelve focused on combined RHT + radiotherapy (RT) or neoadjuvant chemoradiotherapy (CRT). Most treatments were based on invasive thermometry, and less on magnetic resonance imaging (MRI)-based, noninvasive thermometry for STS of the extremities. Perioperative chemotherapy was used for the combination of RHT and CTX, mostly Ifosfamide-based. The effectiveness of RT appeared to be increased by RHT, especially with two RHT sessions/week. The trimodal simultaneous approach of neoadjuvant RHT and CRT was also feasible. No significant toxicity of RHT was reported. CONCLUSIONS The gathered data strengthen the beneficial role of RHT in the multimodal setting. Further expert consensus and clinical trials are required to determine the optimal integration of RHT in treating STS.
Collapse
Affiliation(s)
| | - Eva Oberacker
- Department of Radiation Oncology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Adela Ademaj
- Kantonsspital Aarau, Centre for Radiation Oncology KSA-KSB, Aarau, Switzerland
- Doctoral Clinical Science Program, University of Zürich, Zürich, Switzerland
| | - Stefanie Corradini
- Department of Radiation Oncology, University Hospital Ludwig Maximilians Universität, München, Germany
| | - Franziska Eckert
- Department of Radiation Oncology, Eberhard Karls University Tuebingen, Tübingen, Germany
- Department of Radiation Oncology, Medical University of Vienna/AKH, Vienna, Austria
| | - Anne Flörcken
- Department of Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - David Kaul
- Department of Radiation Oncology, Charité Universitätsmedizin Berlin, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Berlin, Germany; German Cancer Consortium (DKTK), partner site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lars H Lindner
- Department of Medical Oncology, University Hospital Ludwig Maximilians Universität, München, Germany
| | - Rolf Issels
- Department of Medical Oncology, University Hospital Ludwig Maximilians Universität, München, Germany
| | - Oliver J Ott
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Daniel Pink
- Department of Medical Oncology, Helios Klinikum Bad Saarow, Bad Saarow, Germany
- Clinic for Internal Medicine C - Haematology and Oncology, Stem Cell Transplantation and Palliative Care, University Medicine Greifswald, Greifswald, Germany
| | - Vlatko Potkrajcic
- Department of Radiation Oncology, Eberhard Karls University Tuebingen, Tübingen, Germany
| | - Peter Reichardt
- Department of Medical Oncology, Helios Hospital Berlin-Buch, Berlin, Germany
| | - Siyer Roohani
- Department of Radiation Oncology, Charité Universitätsmedizin Berlin, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Berlin, Germany; German Cancer Consortium (DKTK), partner site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Mateusz Jacek Spalek
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Oliver Riesterer
- Kantonsspital Aarau, Centre for Radiation Oncology KSA-KSB, Aarau, Switzerland
| | - Daniel Zips
- Department of Radiation Oncology, Charité Universitätsmedizin Berlin, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Berlin, Germany; German Cancer Consortium (DKTK), partner site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Pirus Ghadjar
- Department of Radiation Oncology, Charité Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
39
|
Willner A, Agaimy A, Fechner K, Ott O, Denz A, Weissmann T, Meidenbauer N, Höfler D, Gaipl U, Frey B, Schmidt M, Haller F, Horch R, Hartmann A, Grützmann R, Fietkau R, Semrau S. Chemoradiotherapy plus hyperthermia (CRTH) versus chemoradiotherapy (CRT) alone in neoadjuvant treatment of soft tissue sarcoma: tumor response, treatment toxicity and disease control. Int J Hyperthermia 2023; 40:2248424. [PMID: 37611915 DOI: 10.1080/02656736.2023.2248424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 08/01/2023] [Accepted: 08/09/2023] [Indexed: 08/25/2023] Open
Abstract
INTRODUCTION Neoadjuvant chemotherapy and radiotherapy for the management of soft tissue sarcomas (STS) are still preferably delivered sequentially, with or without concurrent hyperthermia. Concurrent delivery of chemo-, radio- and thermotherapy may produce synergistic effects and reduce chemotherapy-free intervals. The few available studies suggest that concurrent chemoradiation (CRT) has a greater local effect. Data on the efficacy and toxicity of adding hyperthermia to CRT (CRTH) are sparse. MATERIALS AND METHODS A cohort of 101 patients with STS of the extremities and trunk who received CRT (n = 33) or CRTH (n = 68) before resection of macroscopic tumor (CRT: n = 19, CRTH: n = 49) or re-resection following a non-oncological resection, so called 'whoops procedure', (CRT: n = 14, CRTH: n = 19) were included in this retrospective study. CRT consisted of two cycles of doxorubicine (50 mg/m2 on d2) plus ifosfamide (1500 mg/m2 on d1-5, q28) plus radiation doses of up to 60 Gy. Hyperthermia was delivered in two sessions per week. RESULTS All patients received the minimum dose of 50 Gy. Median doses of ifosfamide and doxorubicin were comparable between CRT (75%/95%) and CRTH (78%/97%). The median number of hyperthermia sessions was seven. There were no differences in acute toxicities. Major wound complications occurred in 15% (CRT) vs. 25% (CRTH) (p = 0.19). In patients with macroscopic disease, the addition of hyperthermia resulted in a tendency toward improved remission: regression ≥90% occurred in 21/48 (CRTH) vs. 4/18 (CRT) patients (p = 0.197). With a median postoperative follow-up of 72 months, 6-year local control and overall survival rates for CRTH vs. CRT alone were 85 vs. 78% (p = 0.938) and 79 vs. 71% (p = 0.215). CONCLUSIONS Both CRT and CRTH are well tolerated with an expected rate of wound complications. The results suggest that adding hyperthermia may improve tumor response.
Collapse
Affiliation(s)
- Alexander Willner
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Friedrich-Alexander-University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Abbas Agaimy
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Katja Fechner
- Department of Haematology and Oncology, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Oliver Ott
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Axel Denz
- Department of Haematology and Oncology, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Thomas Weissmann
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Norbert Meidenbauer
- Department of Surgery, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Daniel Höfler
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Udo Gaipl
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Benjamin Frey
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Manfred Schmidt
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Florian Haller
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Raymund Horch
- Department of Hand- and Plastic Surgery, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Arndt Hartmann
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Robert Grützmann
- Department of Haematology and Oncology, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Rainer Fietkau
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Sabine Semrau
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
40
|
Zanoli M, Dobšíček Trefná H. The hot-to-cold spot quotient for SAR-based treatment planning in deep microwave hyperthermia. Int J Hyperthermia 2022; 39:1421-1439. [DOI: 10.1080/02656736.2022.2136411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Affiliation(s)
- Massimiliano Zanoli
- Department of Electrical Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Hana Dobšíček Trefná
- Department of Electrical Engineering, Chalmers University of Technology, Gothenburg, Sweden
| |
Collapse
|
41
|
Bakker A, Zweije R, Kok HP, Stalpers LJA, Westerveld GH, Hinnen KA, van Tienhoven G, Kolff MW, Crezee H. Comparison of the clinical performance of a hybrid Alba 4D and the AMC-4 locoregional hyperthermia systems. Int J Hyperthermia 2022; 39:1408-1414. [DOI: 10.1080/02656736.2022.2140841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Affiliation(s)
- Akke Bakker
- Department of Radiation Oncology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, University of Amsterdam, Amsterdam, The Netherlands
| | - Remko Zweije
- Department of Radiation Oncology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, University of Amsterdam, Amsterdam, The Netherlands
| | - H. Petra Kok
- Department of Radiation Oncology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, University of Amsterdam, Amsterdam, The Netherlands
| | - Lukas J. A. Stalpers
- Department of Radiation Oncology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, University of Amsterdam, Amsterdam, The Netherlands
| | - G. Henrike Westerveld
- Department of Radiation Oncology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, University of Amsterdam, Amsterdam, The Netherlands
- Department of Radiation Oncology, Erasmus MC, Rotterdam, The Netherlands
| | - Karel A. Hinnen
- Department of Radiation Oncology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, University of Amsterdam, Amsterdam, The Netherlands
| | - Geertjan van Tienhoven
- Department of Radiation Oncology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, University of Amsterdam, Amsterdam, The Netherlands
| | - M. Willemijn Kolff
- Department of Radiation Oncology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, University of Amsterdam, Amsterdam, The Netherlands
| | - Hans Crezee
- Department of Radiation Oncology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
42
|
Du Z, Yin H, Zhao S, Ma Y, Sun Z, Dong B, Zhu M, Zhu C, Peng J, Yang T. Case report: Gemcitabine intravesical hyperthermic infusion combined with tislelizumab in muscle invasive bladder urothelium carcinoma. Front Oncol 2022; 12:1062655. [PMID: 36620538 PMCID: PMC9816863 DOI: 10.3389/fonc.2022.1062655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 12/12/2022] [Indexed: 12/25/2022] Open
Abstract
Background Muscle invasive bladder urothelium carcinoma is a common urinary tract tumor. With the deepening of research, more and more treatment methods are applied in clinical practice, extending the life of patients. Among them, the clinical application of chemotherapeutic intravesical hyperthermia and tumor immunotherapy provides new ideas for our treatment. Case report An 81-year-old female patient was diagnosed with stage T2N0M0 bladder cancer in our hospital. Because the patient and her family were keen to preserve her bladder, they declined surgery and opted for combined chemotherapy. After informed consent from the patient and her family, she received cisplatin combined with gemcitabine intravesical hyperthermic infusion. But the side effects of cisplatin made her intolerable to chemotherapy. With their informed consent we changed her to intravenous tislelizumab in combination with gemcitabine intravesical hyperthermic infusion to continue her treatment. During the subsequent follow-up visits, we found a surprising effect of the treatment. Conclusion Gemcitabine intravesical hyperthermia therapy combined with intravenous tislelizumab in the treatment of muscle invasive bladder urothelium carcinoma may provide a new possible therapeutic strategy of some patients who are inoperable or refuse surgery.
Collapse
|
43
|
Khurshed M, Prades-Sagarra E, Saleh S, Sminia P, Wilmink JW, Molenaar RJ, Crezee H, van Noorden CJF. Hyperthermia as a Potential Cornerstone of Effective Multimodality Treatment with Radiotherapy, Cisplatin and PARP Inhibitor in IDH1-Mutated Cancer Cells. Cancers (Basel) 2022; 14:cancers14246228. [PMID: 36551714 PMCID: PMC9777513 DOI: 10.3390/cancers14246228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/02/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022] Open
Abstract
Mutations in the isocitrate dehydrogenase 1 (IDH1MUT) gene occur in various types of malignancies, including ~60% of chondrosarcomas, ~30% of intrahepatic cholangiocarcinomas and >80% of low-grade gliomas. IDH1MUT are causal in the development and progression of these types of cancer due to neomorphic production of the oncometabolite D-2-hydroxyglutarate (D-2HG). Intracellular accumulation of D-2HG has been implicated in suppressing homologous recombination and renders IDH1MUT cancer cells sensitive to DNA-repair-inhibiting agents, such as poly-(adenosine 5′-diphosphate−ribose) polymerase inhibitors (PARPi). Hyperthermia increases the efficacy of DNA-damaging therapies such as radiotherapy and platinum-based chemotherapy, mainly by inhibition of DNA repair. In the current study, we investigated the additional effects of hyperthermia (42 °C for 1 h) in the treatment of IDH1MUT HCT116 colon cancer cells and hyperthermia1080 chondrosarcoma cancer cells in combination with radiation, cisplatin and/or a PARPi on clonogenic cell survival, cell cycle distribution and the induction and repair of DNA double-strand breaks. We found that hyperthermia in combination with radiation or cisplatin induces an increase in double-strand breaks and cell death, up to 10-fold in IDH1MUT cancer cells compared to IDH1 wild-type cells. This vulnerability was abolished by the IDH1MUT inhibitor AGI-5198 and was further increased by the PARPi. In conclusion, our study shows that IDH1MUT cancer cells are sensitized to hyperthermia in combination with irradiation or cisplatin and a PARPi. Therefore, hyperthermia may be an efficacious sensitizer to cytotoxic therapies in tumors where the clinical application of hyperthermia is feasible, such as IDH1MUT chondrosarcoma of the extremities.
Collapse
Affiliation(s)
- Mohammed Khurshed
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands
- Correspondence:
| | - Elia Prades-Sagarra
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Sarah Saleh
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Peter Sminia
- Department of Radiation Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands
| | - Johanna W. Wilmink
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands
| | - Remco J. Molenaar
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands
| | - Hans Crezee
- Department of Radiation Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Cornelis J. F. van Noorden
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, 1000 Ljubljana, Slovenia
| |
Collapse
|
44
|
Clinical-Radiomics Nomogram from T1W, T1CE, and T2FS MRI for Improving Diagnosis of Soft-Tissue Sarcoma. Mol Imaging Biol 2022; 24:995-1006. [PMID: 35799035 DOI: 10.1007/s11307-022-01751-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 06/02/2022] [Accepted: 06/16/2022] [Indexed: 12/29/2022]
Abstract
PURPOSE To compare values of multiparametric magnetic resonance imaging (MRI) sequences and propose clinical-radiomics nomogram for diagnosis of soft-tissue sarcoma (STS). PROCEDURES This study enrolled 148 patients from Dec. 2017 to Feb. 2021. All patients underwent T1-weighted (T1W), contrast-enhanced T1-weighted (T1CE), and T2-weighted fat-suppressed (T2FS) MRI scans. A total of 1967 radiomic features were extracted from the segmented regions of interest (ROIs) in each MRI sequence. Highly diagnostic radiomic features were selected with Mann-Whitney U test, elastic net, and Akaike's information criterion (AIC) based on MRI images. Logistical regression was used to build Rad scores. Clinical factors were analyzed using the chi-square test or Mann-Whitney U test. The performance of the Rad scores was judged using the area under the receiver operating characteristic area under the curve (ROC AUC), sensitivity, specificity, and accuracy. The nomogram was developed by integrating the Rad score and the most important clinical factor. RESULTS By combining the three MRI sequences, the Rad-Com was developed consisting of twelve features selected by with Mann-Whitney U test, elastic net, and AIC: four from T1W, three from TICE, and five from T2FS MRI. The margin (P < 0.05) demonstrated a statistically significant difference between patients with benign and malignant soft-tissue tumors (STT). The nomogram was constructed by integrating the Rad-Com and margin, which yielded favorable diagnostic AUCs of 0.919 (sensitivity (Sen) = 0.784, specificity (Spe) = 0.936) and 0.913 (Sen = 0.923, Spe = 0.792) in the training and validation cohort. CONCLUSION The proposed nomogram may have potential as a noninvasive marker for STS diagnosis.
Collapse
|
45
|
Radiofrequency Electromagnetic Fields Cause Non-Temperature-Induced Physical and Biological Effects in Cancer Cells. Cancers (Basel) 2022; 14:cancers14215349. [PMID: 36358768 PMCID: PMC9655505 DOI: 10.3390/cancers14215349] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 10/12/2022] [Accepted: 10/21/2022] [Indexed: 11/30/2022] Open
Abstract
Simple Summary Radiofrequency electromagnetic fields are used for tumor heating as adjunct therapy, but it appears that sufficient temperatures can sometimes not be reached. We therefore aimed to study potential non-temperature-induced anticancer effects when adding amplitude modulation to the radiofrequency waves. We could demonstrate in a colorectal cancer model that radiofrequency electromagnetic fields do have anticancer effects when not being induced by increased temperature that can be further increased by amplitude modulation. Therefore, this treatment could potentially serve as a more effective tumor therapy. Abstract Non-temperature-induced effects of radiofrequency electromagnetic fields (RF) have been controversial for decades. Here, we established measurement techniques to prove their existence by investigating energy deposition in tumor cells under RF exposure and upon adding amplitude modulation (AM) (AMRF). Using a preclinical device LabEHY-200 with a novel in vitro applicator, we analyzed the power deposition and system parameters for five human colorectal cancer cell lines and measured the apoptosis rates in vitro and tumor growth inhibition in vivo in comparison to water bath heating. We showed enhanced anticancer effects of RF and AMRF in vitro and in vivo and verified the non-temperature-induced origin of the effects. Furthermore, apoptotic enhancement by AM was correlated with cell membrane stiffness. Our findings not only provide a strategy to significantly enhance non-temperature-induced anticancer cell effects in vitro and in vivo but also provide a perspective for a potentially more effective tumor therapy.
Collapse
|
46
|
Aram E, Moeni M, Abedizadeh R, Sabour D, Sadeghi-Abandansari H, Gardy J, Hassanpour A. Smart and Multi-Functional Magnetic Nanoparticles for Cancer Treatment Applications: Clinical Challenges and Future Prospects. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:nano12203567. [PMID: 36296756 PMCID: PMC9611246 DOI: 10.3390/nano12203567] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/16/2022] [Accepted: 09/27/2022] [Indexed: 05/14/2023]
Abstract
Iron oxide nanoparticle (IONPs) have become a subject of interest in various biomedical fields due to their magnetism and biocompatibility. They can be utilized as heat mediators in magnetic hyperthermia (MHT) or as contrast media in magnetic resonance imaging (MRI), and ultrasound (US). In addition, their high drug-loading capacity enabled them to be therapeutic agent transporters for malignancy treatment. Hence, smartening them allows for an intelligent controlled drug release (CDR) and targeted drug delivery (TDD). Smart magnetic nanoparticles (SMNPs) can overcome the impediments faced by classical chemo-treatment strategies, since they can be navigated and release drug via external or internal stimuli. Recently, they have been synchronized with other modalities, e.g., MRI, MHT, US, and for dual/multimodal theranostic applications in a single platform. Herein, we provide an overview of the attributes of MNPs for cancer theranostic application, fabrication procedures, surface coatings, targeting approaches, and recent advancement of SMNPs. Even though MNPs feature numerous privileges over chemotherapy agents, obstacles remain in clinical usage. This review in particular covers the clinical predicaments faced by SMNPs and future research scopes in the field of SMNPs for cancer theranostics.
Collapse
Affiliation(s)
- Elham Aram
- Department of Cancer Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Babol 47138-18981, Iran
- Department of Polymer Engineering, Faculty of Engineering, Golestan University, Gorgan 49188-88369, Iran
| | - Masome Moeni
- School of Chemical and Process Engineering, University of Leeds, Leeds LS2 9JT, UK
| | - Roya Abedizadeh
- Department of Cancer Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Babol 47138-18981, Iran
| | - Davood Sabour
- Department of Cancer Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Babol 47138-18981, Iran
| | - Hamid Sadeghi-Abandansari
- Department of Cancer Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Babol 47138-18981, Iran
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 16635-148, Iran
| | - Jabbar Gardy
- School of Chemical and Process Engineering, University of Leeds, Leeds LS2 9JT, UK
- Correspondence: (J.G.); (A.H.)
| | - Ali Hassanpour
- School of Chemical and Process Engineering, University of Leeds, Leeds LS2 9JT, UK
- Correspondence: (J.G.); (A.H.)
| |
Collapse
|
47
|
Amin M, Lammers T, Ten Hagen TLM. Temperature-sensitive polymers to promote heat-triggered drug release from liposomes: Towards bypassing EPR. Adv Drug Deliv Rev 2022; 189:114503. [PMID: 35998827 DOI: 10.1016/j.addr.2022.114503] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 06/14/2022] [Accepted: 08/17/2022] [Indexed: 01/24/2023]
Abstract
Heat-triggered drug release from temperature-sensitive nanocarriers upon the application of mild hyperthermia is a promising approach to achieve site-specific delivery of drugs. The combination of mild hyperthermia (41-42 °C) and temperature-sensitive liposomes (TSL) that undergo lipid phase-transition and drug release has been studied extensively and has shown promising therapeutic outcome in a variety of animal tumor models as well as initial indications of success in humans. Sensitization of liposomes to mild hyperthermia by means of exploiting the thermal behavior of temperature-sensitive polymers (TSP) provides novel opportunities. Recently, TSP-modified liposomes (TSPL) have shown potential for enhancing tumor-directed drug delivery, either by triggered drug release or by triggered cell interactions in response to heat. In this review, we describe different classes of TSPL, and analyze and discuss the mechanisms and kinetics of content release from TSPL in response to local heating. In addition, the impact of lipid composition, polymer and copolymer characteristics, serum components and PEGylation on the mechanism of content release and TSPL performance is addressed. This is done from the perspective of rationally designing TSPL, with the overall goal of conceiving efficient strategies to increase the efficacy of TSPL plus hyperthermia to improve the outcome of targeted anticancer therapy.
Collapse
Affiliation(s)
- Mohamadreza Amin
- Laboratory of Experimental Oncology (LEO), Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands; Nanomedicine Innovation Center Erasmus (NICE), Erasmus Medical Center, Rotterdam, The Netherlands.
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Center for Biohybrid Medical Systems, Aachen, Germany.
| | - Timo L M Ten Hagen
- Laboratory of Experimental Oncology (LEO), Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands; Nanomedicine Innovation Center Erasmus (NICE), Erasmus Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
48
|
Inflammatory Surrogate Parameters for Predicting Ifosfamide-Induced Neurotoxicity in Sarcoma Patients. J Clin Med 2022; 11:jcm11195798. [PMID: 36233666 PMCID: PMC9572151 DOI: 10.3390/jcm11195798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/24/2022] [Accepted: 09/27/2022] [Indexed: 11/25/2022] Open
Abstract
Sarcomas compromise a heterogenous group of tumors of a mesenchymal origin. Although treatment options in many solid tumors have evolved over the past decades, the treatment of advanced sarcoma is still based on conventional chemotherapeutic agents. Beside anthracyclines, alkylating agents such as ifosfamide are frequently used in sarcoma treatment. However, treatment with ifosfamide can cause severe dose- and treatment-limiting side effects, such as ifosfamide-induced neurotoxicity (IIN). Especially in sarcoma, consecutive risk assessment analyses investigating the individual factors associated with the increased incidence in IIN, remain insufficient so far. In this retrospective analysis, we investigated 172 sarcoma patients treated with ifosfamide. Out of 172 patients, 49 patients (28.5%) developed IIN. While gender, age, histologic origin, and tumor stage were not associated with the occurrence of IIN, infusion times, simultaneous radiotherapy, and concomitant use of opioids or anticonvulsants affected the risk of developing IIN. Sarcoma patients with IIN showed an alteration in several inflammatory markers, including a lower lymphocyte count, hemoglobin levels, and calcium levels, as well as elevated GGT, sodium, and CRP levels. Remarkably, the occurrence of IIN was associated with a worse prognosis regarding progression free and overall survival. In addition, high CTCAE grades were negatively associated with overall survival in sarcoma. The observation that an inflammatory state is associated with an increased risk of IIN in sarcoma patients can be used prospectively to further investigate the relationship of inflammation and IIN. In addition, the easily accessible blood markers used in our study to predict IIN can be incorporated into clinical decision making.
Collapse
|
49
|
Resag A, Toffanin G, Benešová I, Müller L, Potkrajcic V, Ozaniak A, Lischke R, Bartunkova J, Rosato A, Jöhrens K, Eckert F, Strizova Z, Schmitz M. The Immune Contexture of Liposarcoma and Its Clinical Implications. Cancers (Basel) 2022; 14:cancers14194578. [PMID: 36230502 PMCID: PMC9559230 DOI: 10.3390/cancers14194578] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/14/2022] [Accepted: 09/16/2022] [Indexed: 11/16/2022] Open
Abstract
Liposarcomas (LPS) are the most frequent malignancies in the soft tissue sarcoma family and consist of five distinctive histological subtypes, termed well-differentiated LPS, dedifferentiated LPS (DDLPS), myxoid LPS (MLPS), pleomorphic LPS, and myxoid pleomorphic LPS. They display variations in genetic alterations, clinical behavior, and prognostic course. While accumulating evidence implicates a crucial role of the tumor immune contexture in shaping the response to anticancer treatments, the immunological landscape of LPS is highly variable across different subtypes. Thus, DDLPS is characterized by a higher abundance of infiltrating T cells, yet the opposite was reported for MLPS. Interestingly, a recent study indicated that the frequency of pre-existing T cells in soft tissue sarcomas has a predictive value for immune checkpoint inhibitor (CPI) therapy. Additionally, B cells and tertiary lymphoid structures were identified as potential biomarkers for the clinical outcome of LPS patients and response to CPI therapy. Furthermore, it was demonstrated that macrophages, predominantly of M2 polarization, are frequently associated with poor prognosis. An improved understanding of the complex LPS immune contexture enables the design and refinement of novel immunotherapeutic approaches. Here, we summarize recent studies focusing on the clinicopathological, genetic, and immunological determinants of LPS.
Collapse
Affiliation(s)
- Antonia Resag
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Fetscherstraße 74, 01307 Dresden, Germany
| | - Giulia Toffanin
- Department of Surgery Oncology and Gastroenterology, University of Padova, Via Gattamelata 64, 35128 Padova, Italy
| | - Iva Benešová
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Fetscherstraße 74, 01307 Dresden, Germany
- Department of Immunology, Second Faculty of Medicine, Charles University, University Hospital Motol, V Úvalu 84, 150 06 Prague, Czech Republic
| | - Luise Müller
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Fetscherstraße 74, 01307 Dresden, Germany
| | - Vlatko Potkrajcic
- Department of Radiation Oncology, Eberhard-Karls-University Tuebingen, Hoppe-Seyler-Straße 3, 72076 Tuebingen, Germany
| | - Andrej Ozaniak
- Third Department of Surgery, First Faculty of Medicine, Charles University, University Hospital Motol, V Úvalu 84, 150 06 Prague, Czech Republic
| | - Robert Lischke
- Third Department of Surgery, First Faculty of Medicine, Charles University, University Hospital Motol, V Úvalu 84, 150 06 Prague, Czech Republic
| | - Jirina Bartunkova
- Department of Immunology, Second Faculty of Medicine, Charles University, University Hospital Motol, V Úvalu 84, 150 06 Prague, Czech Republic
| | - Antonio Rosato
- Department of Surgery Oncology and Gastroenterology, University of Padova, Via Gattamelata 64, 35128 Padova, Italy
- Veneto Institute of Oncology IOV-IRCCS, Via Gattamelata 64, 35128 Padova, Italy
| | - Korinna Jöhrens
- Institute of Pathology, University Hospital Carl Gustav Carus, Fetscherstraße 74, 01307 Dresden, Germany
- National Center for Tumor Diseases (NCT), University Hospital Carl Gustav Carus, TU Dresden, Fetscherstraße 74, 01307 Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Franziska Eckert
- Department of Radiation Oncology, Eberhard-Karls-University Tuebingen, Hoppe-Seyler-Straße 3, 72076 Tuebingen, Germany
- Department of Radiation Oncology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Zuzana Strizova
- Department of Immunology, Second Faculty of Medicine, Charles University, University Hospital Motol, V Úvalu 84, 150 06 Prague, Czech Republic
- Correspondence: (Z.S.); (M.S.); Tel.: +420-604712471 (Z.S.); +49-351-458-6501 (M.S.)
| | - Marc Schmitz
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Fetscherstraße 74, 01307 Dresden, Germany
- National Center for Tumor Diseases (NCT), University Hospital Carl Gustav Carus, TU Dresden, Fetscherstraße 74, 01307 Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- Correspondence: (Z.S.); (M.S.); Tel.: +420-604712471 (Z.S.); +49-351-458-6501 (M.S.)
| |
Collapse
|
50
|
Roohani S, Ehret F, Kobus M, Flörcken A, Märdian S, Striefler JK, Rau D, Öllinger R, Jarosch A, Budach V, Kaul D. Preoperative hypofractionated radiotherapy for soft tissue sarcomas: a systematic review. Radiat Oncol 2022; 17:159. [PMID: 36104789 PMCID: PMC9472188 DOI: 10.1186/s13014-022-02072-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 05/24/2022] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND Soft tissue sarcomas (STS) represent a diverse group of rare malignant tumors. Currently, five to six weeks of preoperative radiotherapy (RT) combined with surgery constitute the mainstay of therapy for localized high-grade sarcomas (G2-G3). Growing evidence suggests that shortening preoperative RT courses by hypofractionation neither increases toxicity rates nor impairs oncological outcomes. Instead, shortening RT courses may improve therapy adherence, raise cost-effectiveness, and provide more treatment opportunities for a wider range of patients. Presumed higher rates of adverse effects and worse outcomes are concerns about hypofractionated RT (HFRT) for STS. This systematic review summarizes the current evidence on preoperative HFRT for the treatment of STS and discusses toxicity and oncological outcomes compared to normofractionated RT. METHODS We conducted a systematic review of clinical trials describing outcomes for preoperative HFRT in the management of STS using PubMed, the Cochrane library, the Cochrane Central Register of Controlled Trials, ClinicalTrials.gov, Embase, and Ovid Medline. We followed the 2020 Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Trials on retroperitoneal sarcomas, postoperative RT, and hyperthermia were excluded. Articles published until November 30th, 2021, were included. RESULTS Initial search yielded 94 articles. After removal of duplicate and ineligible articles, 13 articles qualified for analysis. Eight phase II trials and five retrospective analyses were reviewed. Most trials applied 5 × 5 Gy preoperatively in patients with high-grade STS. HFRT courses did not show increased rates of adverse events compared to historical trials of normofractionated RT. Toxicity rates were mostly comparable or lower than in trials of normofractionated RT. Moreover, HFRT achieved comparable local control rates with shorter duration of therapy. Currently, more than 15 prospective studies on HFRT + / - chemotherapy are ongoing. CONCLUSIONS Retrospective data and phase II trials suggest preoperative HFRT to be a reasonable treatment modality for STS. Oncological outcomes and toxicity profiles were favorable. To date, our knowledge is mostly derived from phase II data. No randomized phase III trial comparing normofractionated and HFRT in STS has been published yet. Multiple ongoing phase II trials applying HFRT to investigate acute and late toxicity will hopefully bring forth valuable findings.
Collapse
Affiliation(s)
- Siyer Roohani
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.
| | - Felix Ehret
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Marta Kobus
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Anne Flörcken
- Department of Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Berlin, Germany; German Cancer Consortium (DKTK), partner site Berlin, and German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Sven Märdian
- Centre for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Jana Käthe Striefler
- Department of Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Daniel Rau
- Centre for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Robert Öllinger
- Department of Surgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Armin Jarosch
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Volker Budach
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - David Kaul
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Berlin, Germany; German Cancer Consortium (DKTK), partner site Berlin, and German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| |
Collapse
|