1
|
Ma L, Li Y, Sakamoto Y, Xie L, Suzuki S, Yoshida Y, Sui L, Guo G, Wen J, Ren W, Kakimi K, Osada K, Takahashi A, Shimokawa T. Optimal radiation dose to induce an abscopal effect by combining carbon-ion radiotherapy and anti-CTLA4 antibody. Neoplasia 2024; 60:101099. [PMID: 39674115 PMCID: PMC11699741 DOI: 10.1016/j.neo.2024.101099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 11/19/2024] [Accepted: 11/27/2024] [Indexed: 12/16/2024]
Abstract
BACKGROUND AND PURPOSE Although carbon-ion radiotherapy (CIRT) has led to good outcomes, controlling metastasis is still crucial for improving overall survival. This study aimed to evaluate the effectiveness of by two combinations, one of CIRT and anti-CTLA4 antibody, the other of CIRT and anti-PD-1 antibody, applied at different radiation doses for distal tumour and metastasis suppression. MATERIALS AND METHODS Murine cancer cells (colon carcinoma Colon-26 cells for experiments and osteosarcoma LM8 cells for verification) were grafted into both sides of the hind legs of syngeneic mice. Right-side tumours were irradiated with 3 Gy or 10 Gy CIRT while the left-side tumours were not irradiated, followed by the administration of the anti-CTLA4 antibody or anti-PD-1 antibody. The diameter of the tumours in both legs was measured 3 times per week after irradiation. The number of pulmonary metastases was evaluated within 3 weeks after irradiation. RESULTS Compared with the control group, the high-dose group showed promising anti-cancer benefits in terms of both irradiated tumours and lung metastasis, but neither 10 Gy CIRT combined with the anti-CTLA4 antibody nor 10 Gy CIRT combined with the anti-PD-1 antibody suppressed the growth of distant unirradiated tumours. In the low-dose group, the effect on primary tumour control was slightly weaker than that in the high-dose treatment group, but significant suppressive effects on both distant unirradiated tumours and metastases were observed following 3 Gy CIRT combined with anti-CTLA4 antibody treatment. Specifically, the volume of distant unirradiated tumours decreased by 40 % compared with that of the control group, and no lung metastasis was observed. CONCLUSION Our findings suggest that there is an optimal dose range for the abscopal effect generated with the CIRT combined with anti-CTLA4 antibody, and it highlights a new opportunity for increased induction efficiency of the abscopal effect of combination therapy.
Collapse
Affiliation(s)
- Liqiu Ma
- Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba 263-8555, Japan; Gunma University Heavy Ion Medical Center, Gunma 371-8511, Japan; Department of Nuclear Physics, China Institute of Atomic Energy, Beijing 102413, China.
| | - Yang Li
- Gunma University Heavy Ion Medical Center, Gunma 371-8511, Japan
| | - Yoshimitsu Sakamoto
- Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba 263-8555, Japan
| | - Lin Xie
- Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba 263-8555, Japan
| | - Saaya Suzuki
- Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba 263-8555, Japan
| | - Yukari Yoshida
- Gunma University Heavy Ion Medical Center, Gunma 371-8511, Japan
| | - Li Sui
- Department of Nuclear Physics, China Institute of Atomic Energy, Beijing 102413, China
| | - Gang Guo
- Department of Nuclear Physics, China Institute of Atomic Energy, Beijing 102413, China
| | - Jialing Wen
- Department of Nuclear Physics, China Institute of Atomic Energy, Beijing 102413, China
| | - Wangcai Ren
- Department of Nuclear Physics, China Institute of Atomic Energy, Beijing 102413, China
| | - Kazuhiro Kakimi
- Department of Immunology, Kindai University Faculty of Medicine, Osaka 589-0014, Japan
| | - Kensuke Osada
- Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba 263-8555, Japan
| | | | - Takashi Shimokawa
- Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba 263-8555, Japan.
| |
Collapse
|
2
|
Teodoro L, Carreira ACO, Sogayar MC. Exploring the Complexity of Pan-Cancer: Gene Convergences and in silico Analyses. BREAST CANCER (DOVE MEDICAL PRESS) 2024; 16:913-934. [PMID: 39691553 PMCID: PMC11651076 DOI: 10.2147/bctt.s489246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 11/06/2024] [Indexed: 12/19/2024]
Abstract
Cancer is a complex and multifaceted group of diseases characterized by highly intricate mechanisms of tumorigenesis and tumor progression, which complicates diagnosis, prognosis, and treatment. In recent years, targeted therapies have gained prominence by focusing on specific mutations and molecular features unique to each tumor type, offering more effective and personalized treatment options. However, it is equally critical to explore the genetic commonalities across different types of cancer, which has led to the rise of pan-cancer studies. These approaches help identify shared therapeutic targets across various tumor types, enabling the development of broader and potentially more widely applicable treatment strategies. This review aims to provide a comprehensive overview of key concepts related to tumors, including tumorigenesis processes, the tumor microenvironment, and the role of extracellular vesicles in tumor biology. Additionally, we explore the molecular interactions and mechanisms driving tumor progression, with a particular focus on the pan-cancer perspective. To achieve this, we conducted an in silico analysis using publicly available datasets, which facilitated the identification of both common and divergent genetic and molecular patterns across different tumor types. By integrating these diverse areas, this review offers a clearer and deeper understanding of the factors influencing tumorigenesis and highlights potential therapeutic targets.
Collapse
Affiliation(s)
- Leandro Teodoro
- Cell and Molecular Therapy NUCEL Group, School of Medicine, University of São Paulo, São Paulo, São Paulo, 01246-903, Brazil
- Biochemistry Department, Chemistry Institute, University of São Paulo, São Paulo, São Paulo, 05508-900, Brazil
| | - Ana Claudia O Carreira
- Cell and Molecular Therapy NUCEL Group, School of Medicine, University of São Paulo, São Paulo, São Paulo, 01246-903, Brazil
- Center of Human and Natural Sciences, Federal University of ABC, Santo André, São Paulo, 09280-560, Brazil
| | - Mari C Sogayar
- Cell and Molecular Therapy NUCEL Group, School of Medicine, University of São Paulo, São Paulo, São Paulo, 01246-903, Brazil
- Biochemistry Department, Chemistry Institute, University of São Paulo, São Paulo, São Paulo, 05508-900, Brazil
| |
Collapse
|
3
|
Yang G, Koom WS, Lee BM, Isozaki T, Shinoto M, Yamada S, Seong J. Reduced Risk of Severe Radiation-Induced Lymphopenia in Carbon Ion Radiation Therapy for Locally Advanced Pancreatic Cancer: A Comparative Analysis of Carbon Versus Photon Therapy. Int J Radiat Oncol Biol Phys 2024; 120:544-554. [PMID: 38713122 DOI: 10.1016/j.ijrobp.2024.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 03/15/2024] [Accepted: 04/03/2024] [Indexed: 05/08/2024]
Abstract
PURPOSE Radiation-induced lymphopenia (RIL) is associated with poor prognosis in patients with locally advanced pancreatic cancers. However, there are no reports comparing the effects of carbon ion radiation therapy (CIRT) and photon beam radiation therapy (RT) on the development of RIL. Differences in RIL after CIRT or photon beam RT and predictive factors for RIL in patients with locally advanced pancreatic cancer were investigated. MATERIALS AND METHODS This retrospective study cohort included 834 patients who received concurrent chemoradiotherapy (CCRT) in 2 separate institutions: 337 and 497 in the CIRT and photon beam RT groups, respectively. Severe RIL was defined as an absolute lymphocyte count (ALC) <0.5 × 109 cells/L. A 1:1 propensity score-matching analysis was performed between the CIRT and photon beam RT groups. Patients were categorized into 3 groups according to the development of recovery from severe RIL: no severe RIL (Group A), recovery from severe RIL (Group B), and no recovery from severe RIL (Group C). Logistic regression analysis was performed to identify the predictive value of severe RIL. The prognostic factors of overall survival (OS) were determined using Cox regression analysis. RESULTS After propensity score matching, the baseline ALC and planning target volume of the CIRT and photon beam RT groups were comparable. During CCRT, the ALC of the entire cohort decreased and was significantly lower in the photon beam RT group than in the CIRT group (P < .001). Multivariate logistic regression analysis showed that CIRT reduced severe RIL more than photon beam RT. After adjusting for other factors, the RT modality and RIL were significantly associated with OS. Photon beam RT showed a significantly worse OS than CIRT, and Group C showed a significantly worse OS than Group A. CONCLUSIONS CIRT seems to reduce the development of severe RIL. The RT modality and development/recovery from severe RIL were associated with OS in patients who received CCRT for locally advanced pancreatic cancer. The reduction of severe RIL through optimized RT may be essential for improving treatment outcomes.
Collapse
Affiliation(s)
- Gowoon Yang
- Department of Radiation Oncology, Yonsei Cancer Center, Heavy Ion Therapy Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Woong Sub Koom
- Department of Radiation Oncology, Yonsei Cancer Center, Heavy Ion Therapy Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Byung Min Lee
- Department of Radiation Oncology, Yonsei Cancer Center, Heavy Ion Therapy Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Tetsuro Isozaki
- QST Hospital, National Institutes for Quantum Science and Technology, Inageku, Chiba, Japan
| | - Makoto Shinoto
- QST Hospital, National Institutes for Quantum Science and Technology, Inageku, Chiba, Japan
| | - Shigeru Yamada
- QST Hospital, National Institutes for Quantum Science and Technology, Inageku, Chiba, Japan.
| | - Jinsil Seong
- Department of Radiation Oncology, Yonsei Cancer Center, Heavy Ion Therapy Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
4
|
Paganetti H. A review on lymphocyte radiosensitivity and its impact on radiotherapy. Front Oncol 2023; 13:1201500. [PMID: 37601664 PMCID: PMC10435323 DOI: 10.3389/fonc.2023.1201500] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 07/17/2023] [Indexed: 08/22/2023] Open
Abstract
It is well known that radiation therapy causes lymphopenia in patients and that this is correlated with a negative outcome. The mechanism is not well understood because radiation can have both immunostimulatory and immunosuppressive effects. How tumor dose conformation, dose fractionation, and selective lymph node irradiation in radiation therapy does affect lymphopenia and immune response is an active area of research. In addition, understanding the impact of radiation on the immune system is important for the design and interpretation of clinical trials combining radiation with immune checkpoint inhibitors, both in terms of radiation dose and treatment schedules. Although only a few percent of the total lymphocyte population are circulating, it has been speculated that their increased radiosensitivity may contribute to, or even be the primary cause of, lymphopenia. This review summarizes published data on lymphocyte radiosensitivity based on human, small animal, and in vitro studies. The data indicate differences in radiosensitivity among lymphocyte subpopulations that affect their relative contribution and thus the dynamics of the immune response. In general, B cells appear to be more radiosensitive than T cells and NK cells appear to be the most resistant. However, the reported dose-response data suggest that in the context of lymphopenia in patients, aspects other than cell death must also be considered. Not only absolute lymphocyte counts, but also lymphocyte diversity and activity are likely to be affected by radiation. Taken together, the reviewed data suggest that it is unlikely that radiation-induced cell death in lymphocytes is the sole factor in radiation-induced lymphopenia.
Collapse
Affiliation(s)
- Harald Paganetti
- Department of Radiation Oncology, Massachusetts General Hospital, Boston MA, United States
- Harvard Medical School, Boston MA, United States
| |
Collapse
|
5
|
Galluzzi L, Aryankalayil MJ, Coleman CN, Formenti SC. Emerging evidence for adapting radiotherapy to immunotherapy. Nat Rev Clin Oncol 2023:10.1038/s41571-023-00782-x. [PMID: 37280366 DOI: 10.1038/s41571-023-00782-x] [Citation(s) in RCA: 107] [Impact Index Per Article: 53.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/09/2023] [Indexed: 06/08/2023]
Abstract
Immunotherapy has revolutionized the clinical management of many malignancies but is infrequently associated with durable objective responses when used as a standalone treatment approach, calling for the development of combinatorial regimens with superior efficacy and acceptable toxicity. Radiotherapy, the most commonly used oncological treatment, has attracted considerable attention as a combination partner for immunotherapy owing to its well-known and predictable safety profile, widespread clinical availability, and potential for immunostimulatory effects. However, numerous randomized clinical trials investigating radiotherapy-immunotherapy combinations have failed to demonstrate a therapeutic benefit compared with either modality alone. Such a lack of interaction might reflect suboptimal study design, choice of end points and/or administration of radiotherapy according to standard schedules and target volumes. Indeed, radiotherapy has empirically evolved towards radiation doses and fields that enable maximal cancer cell killing with manageable toxicity to healthy tissues, without much consideration of potential radiation-induced immunostimulatory effects. Herein, we propose the concept that successful radiotherapy-immunotherapy combinations might require modifications of standard radiotherapy regimens and target volumes to optimally sustain immune fitness and enhance the antitumour immune response in support of meaningful clinical benefits.
Collapse
Affiliation(s)
- Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA
| | - Molykutty J Aryankalayil
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - C Norman Coleman
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Silvia C Formenti
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.
- Sandra and Edward Meyer Cancer Center, New York, NY, USA.
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
6
|
Seneviratne DS, Saifi O, Mackeyev Y, Malouff T, Krishnan S. Next-Generation Boron Drugs and Rational Translational Studies Driving the Revival of BNCT. Cells 2023; 12:1398. [PMID: 37408232 DOI: 10.3390/cells12101398] [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: 03/29/2023] [Revised: 04/26/2023] [Accepted: 05/04/2023] [Indexed: 07/07/2023] Open
Abstract
BNCT is a high-linear-energy transfer therapy that facilitates tumor-directed radiation delivery while largely sparing adjacent normal tissues through the biological targeting of boron compounds to tumor cells. Tumor-specific accumulation of boron with limited accretion in normal cells is the crux of successful BNCT delivery. Given this, developing novel boronated compounds with high selectivity, ease of delivery, and large boron payloads remains an area of active investigation. Furthermore, there is growing interest in exploring the immunogenic potential of BNCT. In this review, we discuss the basic radiobiological and physical aspects of BNCT, traditional and next-generation boron compounds, as well as translational studies exploring the clinical applicability of BNCT. Additionally, we delve into the immunomodulatory potential of BNCT in the era of novel boron agents and examine innovative avenues for exploiting the immunogenicity of BNCT to improve outcomes in difficult-to-treat malignancies.
Collapse
Affiliation(s)
| | - Omran Saifi
- Department of Radiation Oncology, Mayo Clinic Florida, Jacksonville, FL 32224, USA
| | - Yuri Mackeyev
- Department of Neurosurgery, UTHealth, Houston, TX 77030, USA
| | - Timothy Malouff
- Department of Radiation Oncology, University of Oklahoma, Oklahoma City, OK 73019, USA
| | - Sunil Krishnan
- Department of Neurosurgery, UTHealth, Houston, TX 77030, USA
| |
Collapse
|
7
|
Helm A, Totis C, Durante M, Fournier C. Are charged particles a good match for combination with immunotherapy? Current knowledge and perspectives. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 376:1-36. [PMID: 36997266 DOI: 10.1016/bs.ircmb.2023.01.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
Charged particle radiotherapy, mainly using protons and carbon ions, provides physical characteristics allowing for a volume conformal irradiation and a reduction of the integral dose to normal tissue. Carbon ion therapy additionally features an increased biological effectiveness resulting in peculiar molecular effects. Immunotherapy, mostly performed with immune checkpoint inhibitors, is nowadays considered a pillar in cancer therapy. Based on the advantageous features of charged particle radiotherapy, we review pre-clinical evidence revealing a strong potential of its combination with immunotherapy. We argue that the combination therapy deserves further investigation with the aim of translation in clinics, where a few studies have been set up already.
Collapse
Affiliation(s)
- A Helm
- Biophysics Department, GSI, Darmstadt, Germany
| | - C Totis
- Biophysics Department, GSI, Darmstadt, Germany
| | - M Durante
- Biophysics Department, GSI, Darmstadt, Germany.
| | - C Fournier
- Biophysics Department, GSI, Darmstadt, Germany
| |
Collapse
|
8
|
Photon- and Proton-Mediated Biological Effects: What Has Been Learned? LIFE (BASEL, SWITZERLAND) 2022; 13:life13010030. [PMID: 36675979 PMCID: PMC9866122 DOI: 10.3390/life13010030] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/14/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022]
Abstract
The current understanding of the effects of radiation is gradually becoming broader. However, it still remains unclear why some patients respond to radiation with a pronounced positive response, while in some cases the disease progresses. This is the motivation for studying the effects of radiation therapy not only on tumor cells, but also on the tumor microenvironment, as well as studying the systemic effects of radiation. In this framework, we review the biological effects of two types of radiotherapy: photon and proton irradiations. Photon therapy is a commonly used type of radiation therapy due to its wide availability and long-term history, with understandable and predictable outcomes. Proton therapy is an emerging technology, already regarded as the method of choice for many cancers in adults and children, both dosimetrically and biologically. This review, written after the analysis of more than 100 relevant literary sources, describes the local effects of photon and proton therapy and shows the mechanisms of tumor cell damage, interaction with tumor microenvironment cells and effects on angiogenesis. After systematic analysis of the literature, we can conclude that proton therapy has potentially favorable toxicological profiles compared to photon irradiation, explained mainly by physical but also biological properties of protons. Despite the fact that radiobiological effects of protons and photons are generally similar, protons inflict reduced damage to healthy tissues surrounding the tumor and hence promote fewer adverse events, not only local, but also systemic.
Collapse
|
9
|
Pompos A, Foote RL, Koong AC, Le QT, Mohan R, Paganetti H, Choy H. National Effort to Re-Establish Heavy Ion Cancer Therapy in the United States. Front Oncol 2022; 12:880712. [PMID: 35774126 PMCID: PMC9238353 DOI: 10.3389/fonc.2022.880712] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 04/27/2022] [Indexed: 11/13/2022] Open
Abstract
In this review, we attempt to make a case for the establishment of a limited number of heavy ion cancer research and treatment facilities in the United States. Based on the basic physics and biology research, conducted largely in Japan and Germany, and early phase clinical trials involving a relatively small number of patients, we believe that heavy ions have a considerably greater potential to enhance the therapeutic ratio for many cancer types compared to conventional X-ray and proton radiotherapy. Moreover, with ongoing technological developments and with research in physical, biological, immunological, and clinical aspects, it is quite plausible that cost effectiveness of radiotherapy with heavier ions can be substantially improved.
Collapse
Affiliation(s)
- Arnold Pompos
- Department of Radiation Oncology, University of Texas (UT) Southwestern Medical Center, Dallas, TX, United States
| | - Robert L. Foote
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, United States
- *Correspondence: Robert L. Foote,
| | - Albert C. Koong
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Quynh Thu Le
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, United States
| | - Radhe Mohan
- Department of Radiation Physics, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Harald Paganetti
- Department of Radiation Oncology, Harvard Medical School and Massachusetts General Hospital, Boston, MA, United States
| | - Hak Choy
- Department of Radiation Oncology, University of Texas (UT) Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
10
|
Riva G, Vischioni B, Gandini S, Cavalieri S, Ronchi S, Barcellini A, Bonora M, Chalaszczyk A, Ingargiola R, Vitolo V, Fiore MR, Iannalfi A, Orlandi E. Particle Beam Therapy Tolerance and Outcome on Patients with Autoimmune Diseases: A Single Institution Matched Case-Control Study. Cancers (Basel) 2021; 13:cancers13205183. [PMID: 34680331 PMCID: PMC8534022 DOI: 10.3390/cancers13205183] [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: 08/15/2021] [Revised: 10/08/2021] [Accepted: 10/12/2021] [Indexed: 11/16/2022] Open
Abstract
It is unclear whether autoimmune diseases (ADs) may predispose patients to higher radiation-induced toxicity, and no data are available regarding particle therapy. Our objective was to determine if cancer patients with ADs have a higher incidence of complications after protons (PT) or carbon ion (CIRT) therapy. METHODS In our retrospective monocentric study, 38 patients with ADs over 1829 patients were treated with particle therapy between 2011 and 2020. Thirteen patients had collagen vascular disease (CVD), five an inflammatory bowel disease (IBD) and twenty patients an organ-specific AD. Each patient was matched with two control patients without ADs on the basis of type/site of cancer, type of particle treatment, age, sex, hypertension and/or diabetes and previous surgery. RESULTS No G4-5 complications were reported. In the AD group, the frequency of acute grade 3 (G3) toxicity was higher than in the control group (15.8% vs. 2.6%, p = 0.016). Compared to their matched controls, CVD-IBD patients had a higher frequency of G3 acute complications (27.7 vs. 2.6%, p = 0.002). There was no difference between AD patients (7.9%) and controls (2.6%) experiencing late G3 toxicity (p = 0.33). The 2 years disease-free survival was lower in AD patients than in controls (74% vs. 91%, p = 0.01), although the differences in terms of survival were not significant. CONCLUSIONS G3 acute toxicity was more frequently reported in AD patients after PT or CIRT. Since no severe G4-G5 events were reported and in consideration of the benefit of particle therapy for selected cancers, we conclude that particle therapy should be not discouraged for patients with ADs. Further prospective studies are warranted to gain insight into toxicity in cancer patients with ADs enrolled for particle therapy.
Collapse
Affiliation(s)
- Giulia Riva
- Clinical Department, National Center for Oncological Hadrontherapy (CNAO), 27100 Pavia, Italy; (B.V.); (S.R.); (A.B.); (M.B.); (A.C.); (R.I.); (V.V.); (M.R.F.); (A.I.); (E.O.)
- Correspondence: ; Tel.: +39-0382-078-501
| | - Barbara Vischioni
- Clinical Department, National Center for Oncological Hadrontherapy (CNAO), 27100 Pavia, Italy; (B.V.); (S.R.); (A.B.); (M.B.); (A.C.); (R.I.); (V.V.); (M.R.F.); (A.I.); (E.O.)
| | - Sara Gandini
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, 20139 Milan, Italy;
| | - Stefano Cavalieri
- Head and Neck Cancer Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy;
| | - Sara Ronchi
- Clinical Department, National Center for Oncological Hadrontherapy (CNAO), 27100 Pavia, Italy; (B.V.); (S.R.); (A.B.); (M.B.); (A.C.); (R.I.); (V.V.); (M.R.F.); (A.I.); (E.O.)
| | - Amelia Barcellini
- Clinical Department, National Center for Oncological Hadrontherapy (CNAO), 27100 Pavia, Italy; (B.V.); (S.R.); (A.B.); (M.B.); (A.C.); (R.I.); (V.V.); (M.R.F.); (A.I.); (E.O.)
| | - Maria Bonora
- Clinical Department, National Center for Oncological Hadrontherapy (CNAO), 27100 Pavia, Italy; (B.V.); (S.R.); (A.B.); (M.B.); (A.C.); (R.I.); (V.V.); (M.R.F.); (A.I.); (E.O.)
| | - Agnieszka Chalaszczyk
- Clinical Department, National Center for Oncological Hadrontherapy (CNAO), 27100 Pavia, Italy; (B.V.); (S.R.); (A.B.); (M.B.); (A.C.); (R.I.); (V.V.); (M.R.F.); (A.I.); (E.O.)
| | - Rossana Ingargiola
- Clinical Department, National Center for Oncological Hadrontherapy (CNAO), 27100 Pavia, Italy; (B.V.); (S.R.); (A.B.); (M.B.); (A.C.); (R.I.); (V.V.); (M.R.F.); (A.I.); (E.O.)
| | - Viviana Vitolo
- Clinical Department, National Center for Oncological Hadrontherapy (CNAO), 27100 Pavia, Italy; (B.V.); (S.R.); (A.B.); (M.B.); (A.C.); (R.I.); (V.V.); (M.R.F.); (A.I.); (E.O.)
| | - Maria Rosaria Fiore
- Clinical Department, National Center for Oncological Hadrontherapy (CNAO), 27100 Pavia, Italy; (B.V.); (S.R.); (A.B.); (M.B.); (A.C.); (R.I.); (V.V.); (M.R.F.); (A.I.); (E.O.)
| | - Alberto Iannalfi
- Clinical Department, National Center for Oncological Hadrontherapy (CNAO), 27100 Pavia, Italy; (B.V.); (S.R.); (A.B.); (M.B.); (A.C.); (R.I.); (V.V.); (M.R.F.); (A.I.); (E.O.)
| | - Ester Orlandi
- Clinical Department, National Center for Oncological Hadrontherapy (CNAO), 27100 Pavia, Italy; (B.V.); (S.R.); (A.B.); (M.B.); (A.C.); (R.I.); (V.V.); (M.R.F.); (A.I.); (E.O.)
| |
Collapse
|
11
|
Raghavapudi H, Singroul P, Kohila V. Brain Tumor Causes, Symptoms, Diagnosis and Radiotherapy Treatment. Curr Med Imaging 2021; 17:931-942. [PMID: 33573575 DOI: 10.2174/1573405617666210126160206] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 12/15/2020] [Accepted: 12/17/2020] [Indexed: 11/22/2022]
Abstract
The strategy used for the treatment of given brain cancer is critical in determining the post effects and survival. An oncological diagnosis of tumor evaluates a range of parameters such as shape, size, volume, location and neurological complexity that define the symptomatic severity. The evaluation determines a suitable treatment approach chosen from a range of options such as surgery, chemotherapy, hormone therapy, radiation therapy and other targeted therapies. Often, a combination of such therapies is applied to achieve superior results. Radiotherapy serves as a better treatment strategy because of a higher survival rate. It offers the flexibility of synergy with other treatment strategies and fewer side effects on organs at risk. This review presents a radiobiological perspective in the treatment of brain tumor. The cause, symptoms, diagnosis, treatment, post-treatment effects and the framework involved in its elimination are summarized.
Collapse
Affiliation(s)
- Haarika Raghavapudi
- Department of Biotechnology, National Institute of Technology Warangal, Warangal -506004, Telangana, India
| | - Pankaj Singroul
- Department of Biotechnology, National Institute of Technology Warangal, Warangal -506004, Telangana, India
| | - V Kohila
- Department of Biotechnology, National Institute of Technology Warangal, Warangal -506004, Telangana, India
| |
Collapse
|
12
|
Abstract
Radiotherapy delivered using photons induces an immune response that leads to modulation of the tumor microenvironment. Clinical studies are ongoing to evaluate immune checkpoint inhibitors in association with photon radiotherapy. At present, there is no publication on the radio-induced immune response after proton therapy. Balb/c mice bearing subcutaneous CT26 colon tumors were irradiated by a single fraction of 16.4 Gy using a proton beam extracted from a TR24 cyclotron. RNA sequencing analysis was assessed at 3 days post-treatment. Proton therapy immune response was monitored by flow cytometry using several panels (lymphoid, myeloid cells, lymphoid cytokines) at 7 and 14 days post-irradiation. RNA-Seq functional profiling identified a large number of GO categories linked to “immune response” and “interferon signaling”. Immunomonitoring evaluation showed induced tumor infiltration by immune cells. This is the first study showing the effect of proton therapy on immune response. These interesting results provide a sound basis to assess the efficacy of a combination of proton therapy and immune checkpoint inhibitors.
Collapse
|
13
|
Hoshino H, Aokage K, Miyoshi T, Tane K, Kojima M, Sugano M, Kuwata T, Ochiai A, Suzuki K, Tsuboi M, Ishii G. Correlation between the number of viable tumor cells and immune cells in the tumor microenvironment in non-small cell lung cancer after induction therapy. Pathol Int 2021; 71:512-520. [PMID: 34115921 DOI: 10.1111/pin.13110] [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: 01/14/2021] [Accepted: 04/26/2021] [Indexed: 12/24/2022]
Abstract
This study aims to determine the correlation between the percent viable tumor cells (%VTC) and the tumor microenvironment in resected non-small cell lung cancer after induction therapy. We enrolled 72 patients with non-small cell lung cancer (NSCLC) who received chemoradiotherapy (CRT) or chemotherapy (CT) prior to surgery. The ratio of the area of viable tumor cells to the total tumor area was calculated to obtain the %VTC. We also examined the number of CD4 (+), CD8 (+), CD20 (+) and FOXP3 (+) tumor-infiltrating lymphocytes (TILs), podoplanin (PDPN) (+) cancer-associated fibroblasts (CAFs), and CD204 (+) tumor-associated macrophages (TAMs) by immunohistochemistry (IHC). In the CRT group (n = 37), the tumors had significantly lower %VTC than the CT group (n = 35) (P < 0.001). In both of the CT group and CRT group, the %VTC showed a significant positive correlation with the number of CD204 (+)-TAMs (P = 0.014 and 0.005, respectively). Only in the CRT group, a higher number of CD204 (+) TAMs was associated with a shorter overall survival (OS) (P = 0.007) and recurrence-free survival (RFS) (P = 0.015). In the CRT group, the number of CD204 (+) TAMs is associated with %VTC and prognosis, suggesting that these cells may have tumor-promoting effects on the residual lung cancer in specific microenvironments after CRT.
Collapse
Affiliation(s)
- Hironobu Hoshino
- Division of Pathology, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Chiba, Japan.,Department of Thoracic Surgery, National Cancer Center Hospital East, Kashiwa, Chiba, Japan.,Department of General Thoracic Surgery, Juntendo University School of Medicine, Tokyo, Japan.,Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - Keiju Aokage
- Department of Thoracic Surgery, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - Tomohiro Miyoshi
- Department of Thoracic Surgery, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - Kenta Tane
- Department of Thoracic Surgery, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - Motohiro Kojima
- Division of Pathology, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Chiba, Japan.,Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - Masato Sugano
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - Takeshi Kuwata
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - Atsushi Ochiai
- Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Chiba, Japan
| | - Kenji Suzuki
- Department of General Thoracic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Masahiro Tsuboi
- Department of Thoracic Surgery, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - Genichiro Ishii
- Division of Pathology, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Chiba, Japan.,Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| |
Collapse
|
14
|
Durante M. Failla Memorial Lecture: The Many Facets of Heavy-Ion Science. Radiat Res 2021; 195:403-411. [PMID: 33979440 DOI: 10.1667/rade-21-00029.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 02/22/2021] [Indexed: 11/03/2022]
Abstract
Heavy ions are riveting in radiation biophysics, particularly in the areas of radiotherapy and space radiation protection. Accelerated charged particles can indeed penetrate deeply in the human body to sterilize tumors, exploiting the favorable depth-dose distribution of ions compared to conventional X rays. Conversely, the high biological effectiveness in inducing late effects presents a hazard for manned space exploration. Even after half a century of accelerator-based experiments, clinical applications and flight research, these two topics remain both fascinating and baffling. Heavy-ion therapy is very expensive, and despite the clinical success it remains controversial. Research on late radiation morbidity in spaceflight led to a reduction in uncertainty, but also pointed to new risks previously underestimated, such as possible damage to the central nervous system. Recently, heavy ions have also been used in other, unanticipated biomedical fields, such as treatment of heart arrhythmia or inactivation of viruses for vaccine development. Heavy-ion science nicely merges physics and biology and remains an extraordinary research field for the 21st century.
Collapse
Affiliation(s)
- Marco Durante
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung, 64291 Darmstadt, Germany; and Technische Universität Darmstadt, Institute of Condensed Matter Physics, 64289 Darmstadt, Germany
| |
Collapse
|
15
|
Friedrich T, Henthorn N, Durante M. Modeling Radioimmune Response-Current Status and Perspectives. Front Oncol 2021; 11:647272. [PMID: 33796470 PMCID: PMC8008061 DOI: 10.3389/fonc.2021.647272] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 02/25/2021] [Indexed: 12/13/2022] Open
Abstract
The combination of immune therapy with radiation offers an exciting and promising treatment modality in cancer therapy. It has been hypothesized that radiation induces damage signals within the tumor, making it more detectable for the immune system. In combination with inhibiting immune checkpoints an effective anti-tumor immune response may be established. This inversion from tumor immune evasion raises numerous questions to be solved to support an effective clinical implementation: These include the optimum immune drug and radiation dose time courses, the amount of damage and associated doses required to stimulate an immune response, and the impact of lymphocyte status and dynamics. Biophysical modeling can offer unique insights, providing quantitative information addressing these factors and highlighting mechanisms of action. In this work we review the existing modeling approaches of combined ‘radioimmune’ response, as well as associated fields of study. We propose modeling attempts that appear relevant for an effective and predictive model. We emphasize the importance of the time course of drug and dose delivery in view to the time course of the triggered biological processes. Special attention is also paid to the dose distribution to circulating blood lymphocytes and the effect this has on immune competence.
Collapse
Affiliation(s)
- Thomas Friedrich
- Biophysics Department, GSI Helmholtz Center for Heavy Ion Research, Darmstadt, Germany
| | - Nicholas Henthorn
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom.,The Christie NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Marco Durante
- Biophysics Department, GSI Helmholtz Center for Heavy Ion Research, Darmstadt, Germany.,Institute for Solid State Physics, Technical University Darmstadt, Darmstadt, Germany
| |
Collapse
|
16
|
Paganetti H, Beltran C, Both S, Dong L, Flanz J, Furutani K, Grassberger C, Grosshans DR, Knopf AC, Langendijk JA, Nystrom H, Parodi K, Raaymakers BW, Richter C, Sawakuchi GO, Schippers M, Shaitelman SF, Teo BKK, Unkelbach J, Wohlfahrt P, Lomax T. Roadmap: proton therapy physics and biology. Phys Med Biol 2021; 66. [DOI: 10.1088/1361-6560/abcd16] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 11/23/2020] [Indexed: 12/12/2022]
|
17
|
Senchukova MA, Makarova EV, Kalinin EA, Tkachev VV, Zubareva EY. Modern concepts on the role of hypoxia in the development of tumor radioresistance. SIBERIAN JOURNAL OF ONCOLOGY 2020. [DOI: 10.21294/1814-4861-2020-19-6-141-147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The purpose of the study was to systematize and summarize modern ideas about the role of hypoxia in the development of tumor radioresistance.Material and Methods. PubMed, eLibrary and Springer databases were used to identify reviews published from 1953 to 2020, of which 57 were selected to write our review.Results. Radiation therapy is one of the most important components in cancer treatment. The major drawback of radiation therapy is the development radiation resistance in cancerous cells and secondary malignancies. The mechanisms of cancer radioresistance are very complicated and affected by many factors, of which hypoxia is the most important. Hypoxia is able to activate the mechanisms of angiogenesis, epithelial-mesenchymal transformation and contribute to the formation of the pool of cancer stem cell, which are characterized by chemo- and radioresistance. In turn, the severity of hypoxia largely dependent on tumor blood flow. Moreover, not only the quantitative but also the qualitative characteristics of blood vessels can affect the development of tissue hypoxia in the tumor.Conclusion. A comprehensive assessment of the severity of hypoxia, as well as characteristics of angiogenesis and EMT can contribute to a better understanding of the mechanisms of development of cancer radioresistance.
Collapse
Affiliation(s)
- M. A. Senchukova
- Orenburg State Medical University;
Orenburg Regional Oncology Clinic
| | - E. V. Makarova
- Orenburg State Medical University;
Orenburg Regional Oncology Clinic
| | | | | | - E. Y. Zubareva
- Orenburg State Medical University;
Orenburg Regional Oncology Clinic
| |
Collapse
|
18
|
Tinganelli W, Durante M. Tumor Hypoxia and Circulating Tumor Cells. Int J Mol Sci 2020; 21:ijms21249592. [PMID: 33339353 PMCID: PMC7766826 DOI: 10.3390/ijms21249592] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 12/12/2022] Open
Abstract
Circulating tumor cells (CTCs) are a rare tumor cell subpopulation induced and selected by the tumor microenvironment's extreme conditions. Under hypoxia and starvation, these aggressive and invasive cells are able to invade the lymphatic and circulatory systems. Escaping from the primary tumor, CTCs enter into the bloodstream to form metastatic deposits or re-establish themselves in cancer's primary site. Although radiotherapy is widely used to cure solid malignancies, it can promote metastasis. Radiation can disrupt the primary tumor vasculature, increasing the dissemination of CTCs. Radiation also induces epithelial-mesenchymal transition (EMT) and eliminates suppressive signaling, causing the proliferation of existent, but previously dormant, disseminated tumor cells (DTCs). In this review, we collect the results and evidence underlying the molecular mechanisms of CTCs and DTCs and the effects of radiation and hypoxia in developing these cells.
Collapse
Affiliation(s)
- Walter Tinganelli
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung GmbH, 64291 Darmstadt, Germany;
| | - Marco Durante
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung GmbH, 64291 Darmstadt, Germany;
- Institut für Festkörperphysik, Technische Universität Darmstadt, 64291 Darmstadt, Germany
- Correspondence:
| |
Collapse
|
19
|
Hartmann L, Schröter P, Osen W, Baumann D, Offringa R, Moustafa M, Will R, Debus J, Brons S, Rieken S, Eichmüller SB. Photon versus carbon ion irradiation: immunomodulatory effects exerted on murine tumor cell lines. Sci Rep 2020; 10:21517. [PMID: 33299018 PMCID: PMC7726046 DOI: 10.1038/s41598-020-78577-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 11/23/2020] [Indexed: 12/18/2022] Open
Abstract
While for photon radiation hypofractionation has been reported to induce enhanced immunomodulatory effects, little is known about the immunomodulatory potential of carbon ion radiotherapy (CIRT). We thus compared the radio-immunogenic effects of photon and carbon ion irradiation on two murine cancer cell lines of different tumor entities. We first calculated the biological equivalent doses of carbon ions corresponding to photon doses of 1, 3, 5, and 10 Gy of the murine breast cancer cell line EO771 and the OVA-expressing pancreatic cancer cell line PDA30364/OVA by clonogenic survival assays. We compared the potential of photon and carbon ion radiation to induce cell cycle arrest, altered surface expression of immunomodulatory molecules and changes in the susceptibility of cancer cells to cytotoxic T cell (CTL) mediated killing. Irradiation induced a dose-dependent G2/M arrest in both cell lines irrespective from the irradiation source applied. Likewise, surface expression of the immunomodulatory molecules PD-L1, CD73, H2-Db and H2-Kb was increased in a dose-dependent manner. Both radiation modalities enhanced the susceptibility of tumor cells to CTL lysis, which was more pronounced in EO771/Luci/OVA cells than in PDA30364/OVA cells. Overall, compared to photon radiation, the effects of carbon ion radiation appeared to be enhanced at higher dose range for EO771 cells and extenuated at lower dose range for PDA30364/OVA cells. Our data show for the first time that equivalent doses of carbon ion and photon irradiation exert similar immunomodulating effects on the cell lines of both tumor entities, highlighted by an enhanced susceptibility to CTL mediated cytolysis in vitro.
Collapse
Affiliation(s)
- Laura Hartmann
- German Cancer Research Center (DKFZ), Research Group GMP & T Cell Therapy, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Philipp Schröter
- German Cancer Research Center (DKFZ), Research Group GMP & T Cell Therapy, Heidelberg, Germany
- Department of Radiation Oncology, Heidelberg University Hospital (UKHD), Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany
- Department of Radiation Oncology, Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg University Hospital (UKHD), Heidelberg, Germany
| | - Wolfram Osen
- German Cancer Research Center (DKFZ), Research Group GMP & T Cell Therapy, Heidelberg, Germany
| | - Daniel Baumann
- German Cancer Research Center (DKFZ), Molecular Oncology of Gastrointestinal Tumors, Heidelberg, Germany
- Department of Surgery, Heidelberg University Hospital (UKHD), Heidelberg, Germany
| | - Rienk Offringa
- German Cancer Research Center (DKFZ), Molecular Oncology of Gastrointestinal Tumors, Heidelberg, Germany
- Department of Surgery, Heidelberg University Hospital (UKHD), Heidelberg, Germany
| | - Mahmoud Moustafa
- Department of Radiation Oncology, Heidelberg University Hospital (UKHD), Heidelberg, Germany
- Faculty of Medicine Heidelberg (MFHD), Division of Molecular and Translational Radiation Oncology, Heidelberg, Germany
- German Cancer Consortium (DKTK) Core-Center Heidelberg, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Clinical Pathology, Suez Canal University, Ismailia, Egypt
| | - Rainer Will
- German Cancer Research Center (DKFZ), Genomics and Proteomics Core Facility, Heidelberg, Germany
| | - Jürgen Debus
- Department of Radiation Oncology, Heidelberg University Hospital (UKHD), Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany
- Department of Radiation Oncology, Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg University Hospital (UKHD), Heidelberg, Germany
- Faculty of Medicine Heidelberg (MFHD), Division of Molecular and Translational Radiation Oncology, Heidelberg, Germany
- German Cancer Consortium (DKTK) Core-Center Heidelberg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stephan Brons
- Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany
- Department of Radiation Oncology, Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg University Hospital (UKHD), Heidelberg, Germany
| | - Stefan Rieken
- Department of Radiation Oncology, Heidelberg University Hospital (UKHD), Heidelberg, Germany.
- Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany.
- Department of Radiation Oncology, Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg University Hospital (UKHD), Heidelberg, Germany.
- Department of Radiation Oncology, University Medical Center Göttingen, Göttingen, Germany.
| | - Stefan B Eichmüller
- German Cancer Research Center (DKFZ), Research Group GMP & T Cell Therapy, Heidelberg, Germany.
| |
Collapse
|
20
|
Guo T, Zou L, Ni J, Chu X, Zhu Z. Radiotherapy for unresectable locally advanced non-small cell lung cancer: a narrative review of the current landscape and future prospects in the era of immunotherapy. Transl Lung Cancer Res 2020; 9:2097-2112. [PMID: 33209629 PMCID: PMC7653144 DOI: 10.21037/tlcr-20-511] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Significant recent advances have occurred in the use of radiation therapy for locally advanced non-small cell lung cancer (LA-NSCLC). In fact, the past few decades have seen both therapeutic gains and setbacks in the evolution of radiotherapy for LA-NSCLC. The PACIFIC trial has heralded a new era of immunotherapy and has raised important questions for future study, such as the future directions of radiation therapy for LA-NSCLC in the era of immunotherapy. Modern radiotherapy techniques such as three-dimensional (3D) conformal radiotherapy and intensity-modulated radiotherapy (IMRT) provide opportunities for improved target conformity and reduced normal-tissue exposure. However, the low-dose radiation volume brought by IMRT and its effects on the immune system deserve particular attention when combing radiotherapy and immunotherapy. Particle radiotherapy offers dosimetric advantages and exhibits great immunoregulatory potential. With the ongoing improvement in particle radiotherapy techniques and knowledge, the combination of immunotherapy and particle radiotherapy has tremendous potential to improve treatment outcomes. Of particular importance are questions on the optimal radiation schedule in the settings of radio-immunotherapy. Strategies for the reduction of the irradiated field such as involved-field irradiation (IFI) and omission of clinical target volume (CTV) hold promise for better preservation of immune function while not compromising locoregional and distant control. In addition, different dose-fractionation regimens can have diverse effects on the immune system. Thus, prospective trials are urgently needed to establish the optimal dose fractionation regimen. Moreover, personalized radiotherapy which allows the tailoring of radiation dose to each individual's genetic background and immune state is of critical importance in maximizing the benefit of radiation to patients with LA-NSCLC.
Collapse
Affiliation(s)
- Tiantian Guo
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College
| | - Liqing Zou
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College
| | - Jianjiao Ni
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College
| | - Xiao Chu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College
| | - Zhengfei Zhu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College.,Institute of Thoracic Oncology, Fudan University, Shanghai, China
| |
Collapse
|
21
|
Helm A, Tinganelli W, Simoniello P, Kurosawa F, Fournier C, Shimokawa T, Durante M. Reduction of Lung Metastases in a Mouse Osteosarcoma Model Treated With Carbon Ions and Immune Checkpoint Inhibitors. Int J Radiat Oncol Biol Phys 2020; 109:594-602. [PMID: 32980497 DOI: 10.1016/j.ijrobp.2020.09.041] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 08/12/2020] [Accepted: 09/21/2020] [Indexed: 12/11/2022]
Abstract
PURPOSE The combination of radiation therapy and immunotherapy is recognized as a very promising strategy for metastatic cancer treatment. The purpose of this work is to compare the effectiveness of x-ray and high-energy carbon ion therapy in combination with checkpoint inhibitors in a murine model. METHODS AND MATERIALS We used an osteosarcoma mouse model irradiated with either carbon ions or x-rays in combination with 2 immune checkpoint inhibitors (anti-PD-1 and anti-CTLA-4). LM8 osteosarcoma cells were injected in both hind limbs of female C3H/He mice 7 days before exposure to carbon ions or x-rays. In experimental groups receiving irradiation, only the tumor on the left limb was exposed, whereas the tumor on the right limb served as an abscopal mimic. Checkpoint inhibitors were injected intraperitoneally 1 day before exposure as well as concomitant to and after exposure. Tumor growth was measured regularly up to day 21 after exposure, when mice were sacrificed. Both tumors as well as lungs were extracted. RESULTS A reduced growth of the abscopal tumor was most pronounced after the combined protocol of carbon ions and the immune checkpoint inhibitors administered sequentially. Radiation or checkpoint inhibitors alone were not sufficient to reduce the growth of the abscopal tumors. Carbon ions alone reduced the number of lung metastases more efficiently than x-rays, and in combination with immunotherapy both radiation types essentially suppressed the metastasis, with carbon ions being again more efficient. Investigation of the infiltration of immune cells in the abscopal tumors of animals treated with combination revealed an increase in CD8+ cells. CONCLUSIONS Combination of checkpoint inhibitors with high-energy carbon ion radiation therapy can be an effective strategy for the treatment of advanced tumors.
Collapse
Affiliation(s)
- Alexander Helm
- GSI Helmholtzzentrum für Schwerionenforschung GmbH, Darmstadt, Germany
| | - Walter Tinganelli
- GSI Helmholtzzentrum für Schwerionenforschung GmbH, Darmstadt, Germany
| | - Palma Simoniello
- Department of Science and Technology, Parthenope University of Naples, Naples, Italy; Istituto Nazionale di Fisica Nucleare, Sezione di Napoli, Naples, Italy
| | - Fuki Kurosawa
- National Institute of Radiological Sciences, National Institutes of Quantum and Radiological Science and Technology, Chiba, Japan
| | - Claudia Fournier
- GSI Helmholtzzentrum für Schwerionenforschung GmbH, Darmstadt, Germany
| | - Takashi Shimokawa
- National Institute of Radiological Sciences, National Institutes of Quantum and Radiological Science and Technology, Chiba, Japan
| | - Marco Durante
- GSI Helmholtzzentrum für Schwerionenforschung GmbH, Darmstadt, Germany; Technische Universität Darmstadt, Institut für Festkörperphysik, Darmstadt, Germany.
| |
Collapse
|
22
|
Nielsen S, Bassler N, Grzanka L, Swakon J, Olko P, Horsman MR, Sørensen BS. Proton scanning and X-ray beam irradiation induce distinct regulation of inflammatory cytokines in a preclinical mouse model. Int J Radiat Biol 2020; 96:1238-1244. [PMID: 32780616 DOI: 10.1080/09553002.2020.1807644] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
PURPOSE Conventional X-ray radiotherapy induces a pro-inflammatory response mediated by altered expression of inflammation-regulating cytokines. Proton scanning and X-ray irradiation produce distinct changes to cytokine gene expression in vitro suggesting that proton beam therapy may induce an inflammatory response dissimilar to that of X-ray radiation. The purpose of the present study was to determine whether proton scanning beam radiation and conventional X-ray photon radiation would induce differential regulation of circulating cytokines in vivo. MATERIALS AND METHODS Female CDF1 mice were irradiated locally at the right hind leg using proton pencil beam scanning or X-ray photons. Blood samples were obtained from two separate mice groups. Samples from one group were drawn by retro-orbital puncture 16 months post irradiation, while samples from the other group were drawn 5 and 30 days post irradiation. Concentration of the cytokines IL-6, IL-1β, IL-10, IL-17A, IFN-γ, and TNFα was measured in plasma using bead-based immunoassays. RESULTS The cytokines IL-6, IL-1β, IL-10, IFN-γ, and TNFα were expressed at lower levels in plasma samples from proton-irradiated mice compared with X-ray-irradiated mice 16 months post irradiation. The same cytokines were downregulated in proton-irradiated mice 5 days post irradiation when compared to controls, while at day 30 expression had increased to the same level or higher. X-ray radiation did not markedly change expression levels at days 5 and 30. CONCLUSIONS The inflammatory response to proton and X-ray irradiation seem to be distinct as the principal pro-inflammatory cytokines are differentially regulated short- and long-term following irradiation. Both the development of normal tissue damage and efficacy of immunotherapy could be influenced by an altered inflammatory response to irradiation.
Collapse
Affiliation(s)
- Steffen Nielsen
- Experimental Clinical Oncology, Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Niels Bassler
- Medical Radiation Physics, Department of Physics, Stockholm University, Stockholm, Sweden
| | - Leszek Grzanka
- Proton Radiotherapy Group, Institute of Nuclear Physics, Polish Academy of Sciences, Krakow, Poland
| | - Jan Swakon
- Proton Radiotherapy Group, Institute of Nuclear Physics, Polish Academy of Sciences, Krakow, Poland
| | - Pawel Olko
- Proton Radiotherapy Group, Institute of Nuclear Physics, Polish Academy of Sciences, Krakow, Poland
| | - Michael R Horsman
- Experimental Clinical Oncology, Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Brita Singers Sørensen
- Experimental Clinical Oncology, Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
23
|
Ho AY, Wright JL, Blitzblau RC, Mutter RW, Duda DG, Norton L, Bardia A, Spring L, Isakoff SJ, Chen JH, Grassberger C, Bellon JR, Beriwal S, Khan AJ, Speers C, Dunn SA, Thompson A, Santa-Maria CA, Krop IE, Mittendorf E, King TA, Gupta GP. Optimizing Radiation Therapy to Boost Systemic Immune Responses in Breast Cancer: A Critical Review for Breast Radiation Oncologists. Int J Radiat Oncol Biol Phys 2020; 108:227-241. [PMID: 32417409 PMCID: PMC7646202 DOI: 10.1016/j.ijrobp.2020.05.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 04/24/2020] [Accepted: 05/07/2020] [Indexed: 12/13/2022]
Abstract
Immunotherapy using immune checkpoint blockade has revolutionized the treatment of many types of cancer. Radiation therapy (RT)-particularly when delivered at high doses using newer techniques-may be capable of generating systemic antitumor effects when combined with immunotherapy in breast cancer. These systemic effects might be due to the local immune-priming effects of RT resulting in the expansion and circulation of effector immune cells to distant sites. Although this concept merits further exploration, several challenges need to be overcome. One is an understanding of how the heterogeneity of breast cancers may relate to tumor immunogenicity. Another concerns the need to develop knowledge and expertise in delivery, sequencing, and timing of RT with immunotherapy. Clinical trials addressing these issues are under way. We here review and discuss the particular opportunities and issues regarding this topic, including the design of informative clinical and translational studies.
Collapse
Affiliation(s)
- Alice Y Ho
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts.
| | - Jean L Wright
- Department of Radiation Oncology, Johns Hopkins Cancer Center, Brooklandville, Maryland
| | - Rachel C Blitzblau
- Department of Radiation Oncology, Duke Cancer Center, Durham, North Carolina
| | - Robert W Mutter
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | - Dan G Duda
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | - Larry Norton
- Department of Medical Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Aditya Bardia
- Department of Medical Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | - Laura Spring
- Department of Medical Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | - Steven J Isakoff
- Department of Medical Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | - Jonathan H Chen
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | - Clemens Grassberger
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | - Jennifer R Bellon
- Department of Radiation Oncology, Dana Farber Cancer Institute, Boston, Massachusetts
| | - Sushil Beriwal
- Department of Radiation Oncology, University of Pittsburgh Cancer Center, Pittsburgh, Pennslyvania
| | - Atif J Khan
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Corey Speers
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Samantha A Dunn
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | - Alastair Thompson
- Department of Surgical Oncology, Baylor College of Medicine Medical Center, Houston, Texas
| | - Cesar A Santa-Maria
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ian E Krop
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts
| | - Elizabeth Mittendorf
- Department of Surgical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts
| | - Tari A King
- Department of Surgical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts
| | - Gaorav P Gupta
- Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
24
|
Overall Survival After Treatment of Localized Prostate Cancer With Proton Beam Therapy, External-Beam Photon Therapy, or Brachytherapy. Clin Genitourin Cancer 2020; 19:255-266.e7. [PMID: 32972877 PMCID: PMC7914293 DOI: 10.1016/j.clgc.2020.08.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 08/19/2020] [Accepted: 08/22/2020] [Indexed: 01/22/2023]
Abstract
With limited high-level evidence, we carried out a comparative effectiveness study for the effect of proton beam therapy (PBT) on overall survival compared to external-beam radiotherapy (EBRT) and brachytherapy (BT) among patients with localized prostate cancer using a national database. PBT was associated with a significant overall survival benefit compared to EBRT and had a similar performance as BT.
Collapse
|
25
|
Lu G, Luo H, Zhu X. Targeting the GRP78 Pathway for Cancer Therapy. Front Med (Lausanne) 2020; 7:351. [PMID: 32850882 PMCID: PMC7409388 DOI: 10.3389/fmed.2020.00351] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 06/11/2020] [Indexed: 12/24/2022] Open
Abstract
The 78-kDa glucose-regulated protein (GRP78) plays an important part in maintaining protein stability, regulating protein folding, and inducing apoptosis autophagy, which is considered as a powerful protein. Meanwhile, it also plays a role in ensuring the normal function of organs. In recent years, more and more researches have been carried out on the targeted therapy of GRP78, mainly focusing on its relevant role in tumor and its role as a major modulator and modulator of subordinate pathways. The ability of GRP78 to respond to endoplasmic reticulum stress (ERS) determines whether tumor cells survive and whether the changes in expression level of GRP78 regulated by endoplasmic reticulum (ER) caused by various factors will directly or indirectly affect cell proliferation, apoptosis, and injury, or reduce the body's defense ability, or have protective effects on various organs.
Collapse
Affiliation(s)
- Guanhua Lu
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China
| | - Hui Luo
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China.,The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China.,Southern Marine Science and Engineering Guangdong Laboratory, Zhanjiang, China.,The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China
| | - Xiao Zhu
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China.,The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China.,Southern Marine Science and Engineering Guangdong Laboratory, Zhanjiang, China.,The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
26
|
Galalae R, Hannoun-Lévi JM. Accelerated partial breast irradiation by brachytherapy: present evidence and future developments. Jpn J Clin Oncol 2020; 50:743-752. [PMID: 32444872 DOI: 10.1093/jjco/hyaa064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 04/16/2020] [Accepted: 04/20/2020] [Indexed: 11/14/2022] Open
Abstract
Accelerated partial breast irradiation (APBI) delivers a short course of adjuvant RT after breast conserving surgery to only a limited part of the breast where the tumor was located. This procedure requires expertise, good communication, and close collaboration between specialized surgeons and attending radiation oncologists with adequate intraoperative tumor bed clip marking. However, APBI offers several intrinsic benefits when compared with whole breast irradiation (WBIR) including reduced treatment time (1 versus 4-6 weeks) and better sparing of surrounding healthy tissues. The present publication reviews the APBI level 1-evidence provided with various radiation techniques supplemented by long-term experience obtained from large multi-institutional phase II studies. Additionally, it offers an outlook on recent research with ultra-short or single-fraction APBI courses and new brachytherapy sources. Mature data from three randomized controlled trials (RCTs) clearly prove the noninferiority of APBI with 'only two techniques-1/MIBT (multicatheter interstitial brachytherapy) (two trials) and 2/intensity modulated radiotherapy (one trial)'-in terms of equivalent local control/overall survival to the previous standard 'conventionally fractionated WBIR'. However, MIBT-APBI techniques were superior in both toxicity and patient-reported outcomes (PROs) versus WBIR at long-term follow-up. Currently, in RCT-setting, alternative APBI techniques such as intraoperative electrons, 50-kV x-rays and three-dimensional conformal external beam radiotherapy (3D-CRT) failed to demonstrate noninferiority to conventionally fractionated WBIR. However, 3D-CRT-APBI compared noninferior to hypo-fractionated WBIR in preventing ipsilateral breast tumor recurrence (randomized RAPID-trial) but was associated with a higher rate of late radiation toxicity. Ultimately, MIBT remains the only APBI modality with noninferior survival/superior toxicity/PROs at 10-years and therefore should be prioritized over alternative methods in patients with breast cancer considered at low-risk for local recurrence according to recent international guidelines.
Collapse
Affiliation(s)
- Razvan Galalae
- MedAustron, Center for Ion Therapy and Research, Wiener Neustadt, Austria
- Department of Radiation Oncology, Medical Faculty, Christian-Albrechts-University, Kiel, Germany
| | - Jean-Michel Hannoun-Lévi
- Department of Radiation Oncology, Centre Antoine Lacassagne, University of Cote d'Azur, Nice, France
| |
Collapse
|
27
|
Wang L, Yang L, Han S, Zhu J, Li Y, Wang Z, Fan YH, Lin E, Zhang R, Sahoo N, Li Y, Zhang X, Wang X, Li T, Zhu XR, Zhu H, Heymach JV, Myers JN, Frank SJ. Patterns of protein expression in human head and neck cancer cell lines differ after proton vs photon radiotherapy. Head Neck 2020; 42:289-301. [PMID: 31710172 DOI: 10.1002/hed.26005] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 09/26/2019] [Accepted: 10/18/2019] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Proton radiotherapy (PRT) may be a less toxic alternative to photon radiotherapy (XRT) for patients with head and neck squamous cell carcinoma (HNSCC). However, the molecular responses of HNSCC cells to PRT vs XRT are unclear. METHODS Proteomics analyses of protein expression profiles by reverse-phase protein arrays were done for two human papillomavirus [HPV]-negative and two HPV+ cell lines. Expression patterns of 175 proteins involved in several signaling pathways were tested. RESULTS Compared with PRT, XRT tended to induce lower expression of DNA damage repair-and cell cycle arrest-related proteins and higher expression of cell survival- and proliferation-related proteins. CONCLUSIONS Under these experimental conditions, PRT and XRT induced different protein expression and activation profiles. Further preclinical verification is needed, as are studies of tumor pathway mutations as biomarkers for choice of treatment or as radiosensitization targets to improve the response of HNSCC to PRT or XRT.
Collapse
Affiliation(s)
- Li Wang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Liuqing Yang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shichao Han
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jinming Zhu
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yuting Li
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Zeming Wang
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - You-Hong Fan
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Eric Lin
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ruiping Zhang
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Narayan Sahoo
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yupeng Li
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xiaodong Zhang
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xiaochun Wang
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Tengfei Li
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xiaorong R Zhu
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hongtu Zhu
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - John V Heymach
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jeffrey N Myers
- Department of Head & Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Steven J Frank
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
28
|
Clinical evidence of abscopal effect in cutaneous squamous cell carcinoma treated with diffusing alpha emitters radiation therapy: a case report. J Contemp Brachytherapy 2019; 11:449-457. [PMID: 31749854 PMCID: PMC6854861 DOI: 10.5114/jcb.2019.88138] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 07/25/2019] [Indexed: 12/19/2022] Open
Abstract
Purpose Alpha particle treatments could enhance the probability of an immune response, which can lead to abscopal effects (AE). We report a case of a patient affected by multiple cutaneous squamous cell carcinoma (cSCC). After the treatment with diffusing alpha emitters radiation therapy (DaRT) of one lesion, an AE was observed on at least two distant ones. Material and methods We investigated a case of a 65-year-old female patient with multiple synchronous lesions of the skin of lower limbs confirmed by a biopsy. Patient was enrolled in a clinical trial N.CTP-SCC-00 (NCT03015883), with the objective to assess effectiveness of DaRT technique. DaRT is based on the insertion of locally 224Ra-loaded seeds in a clinical target volume (CTV). Treatment plan with positron emission tomography/computed tomography (PET/CT) was used to entirely cover the CTV. Follow-up and biopsy evaluations were employed to outline the patient outcome. Results We performed seeds implantation according to the Paris system. At 28th day, an evident lesion shrinkage with a persistent minimal area of hyperkeratosis was noted. 76 days after implantation, a complete remission of the treated lesion was observed and an evident reduction of the area with two more distant lesion, which could be associated to an immune-mediated response. One year after the treatment, a complete remission of treated lesion was observed as well as spontaneous regression of untreated distant ones. Conclusions In this study, we reported evidences of an AE in cSCC stimulated by radiation and possibly mediated by immune system. In the next DaRT treatments, our intent is to monitor T-lymphocytes variations in peripheral blood in order to demonstrate indirect activation of the immune system mediated by radiation also in patients with solitary lesions, in which, by definition, an AE cannot be observed.
Collapse
|
29
|
Bachi N, Otranto S, Otero GS, Olson RE. The role of multiple ionization of H 2O in heavy ion collisions. Phys Med Biol 2019; 64:205020. [PMID: 31487696 DOI: 10.1088/1361-6560/ab41db] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Collisional ionization processes involving H2O molecules and C6+, O8+, Si13+ ions are studied by means of the classical trajectory Monte Carlo method using molecular orbital calculations to define the ionization stages of the water molecule. Net total and single-differential cross sections in energy and angle are obtained by using a newly developed model that goes beyond the commonly applied one-active electron approximation. This model allows us to access the fraction of electron emission arising from single and multiple electron ionization. Calculated cross sections are contrasted and benchmarked against available experimental data at impact energies in the MeV/u range. The present results highlight the important role of multiple ionization in the emission of electrons where we find the majority of electrons emitted with energies greater than ~50 eV arise from multiple ionization collisions.
Collapse
Affiliation(s)
- N Bachi
- Instituto de Física del Sur (IFISUR), Departamento de Física, Universidad Nacional del Sur (UNS), CONICET, Av. L. N. Alem 1253, B8000CPB-Bahía Blanca, Argentina. Author to whom any correspondence should be addressed
| | | | | | | |
Collapse
|
30
|
Andersson P, Ostheimer C. Editorial: Combinatorial Approaches to Enhance Anti-tumor Immunity: Focus on Immune Checkpoint Blockade Therapy. Front Immunol 2019; 10:2083. [PMID: 31555290 PMCID: PMC6742972 DOI: 10.3389/fimmu.2019.02083] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 08/16/2019] [Indexed: 12/12/2022] Open
Affiliation(s)
- Patrik Andersson
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, United States.,Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Christian Ostheimer
- Medical Science Division, Department of Oncology, Oxford Institute for Radiation Oncology, University of Oxford, Oxford, United Kingdom.,Department of Radiation Oncology, Martin Luther University Halle-Wittenberg, Halle, Germany
| |
Collapse
|
31
|
Durante M, Formenti S. Harnessing radiation to improve immunotherapy: better with particles? Br J Radiol 2019; 93:20190224. [PMID: 31317768 DOI: 10.1259/bjr.20190224] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The combination of radiotherapy and immunotherapy is one of the most promising strategies for cancer treatment. Recent clinical results support the pre-clinical experiments pointing to a benefit for the combined treatment in metastatic patients. Charged particle therapy (using protons or heavier ions) is considered one of the most advanced radiotherapy techniques, but its cost remains higher than conventional X-ray therapy. The most important question to be addressed to justify a more widespread use of particle therapy is whether they can be more effective than X-rays in combination with immunotherapy. Protons and heavy ions have physical advantages compared to X-rays that lead to a reduced damage to the immune cells, that are required for an effective immune response. Moreover, densely ionizing radiation may have biological advantages, due to different cell death pathways and release of cytokine mediators of inflammation. We will discuss results in esophageal cancer patients showing that charged particles can reduce the damage to blood lymphocytes compared to X-rays, and preliminary in vitro studies pointing to an increased release of immune-stimulating cytokines after heavy ion exposure. Pre-clinical and clinical studies are ongoing to test these hypotheses.
Collapse
Affiliation(s)
- Marco Durante
- GSI Helmholtzzentrum für Schwerionenforschung, Biophysics Department, Darmstadt, Germany.,Technische Universität Darmstadt, Institut für Festkörperphysik, Darmstadt, Germany
| | - Silvia Formenti
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
32
|
Tumor Microenvironment as A "Game Changer" in Cancer Radiotherapy. Int J Mol Sci 2019; 20:ijms20133212. [PMID: 31261963 PMCID: PMC6650939 DOI: 10.3390/ijms20133212] [Citation(s) in RCA: 300] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 06/19/2019] [Accepted: 06/25/2019] [Indexed: 12/24/2022] Open
Abstract
Radiotherapy (RT), besides cancer cells, also affects the tumor microenvironment (TME): tumor blood vessels and cells of the immune system. It damages endothelial cells and causes radiation-induced inflammation. Damaged vessels inhibit the infiltration of CD8+ T lymphocytes into tumors, and immunosuppressive pathways are activated. They lead to the accumulation of radioresistant suppressor cells, including tumor-associated macrophages (TAMs) with the M2 phenotype, myeloid-derived suppressor cells (MDSCs), and regulatory T cells (Tregs). The area of tumor hypoxia increases. Hypoxia reduces oxygen-dependent DNA damage and weakens the anti-cancer RT effect. It activates the formation of new blood vessels and leads to cancer relapse after irradiation. Irradiation may also activate the immune response through immunogenic cell death induction. This leads to the "in situ" vaccination effect. In this article, we review how changes in the TME affect radiation-induced anticancer efficacy. There is a very delicate balance between the activation of the immune system and the immunosuppression induced by RT. The effects of RT doses on immune system reactions and also on tumor vascularization remain unclear. A better understanding of these interactions will contribute to the optimization of RT treatment, which may prevent the recurrence of cancer.
Collapse
|
33
|
Zhu C, Lin S, Liang J, Zhu Y. PD-1 blockade enhances the anti-tumor immune response induced by cryoablation in a murine model of renal cell carcinoma. Cryobiology 2019; 87:86-90. [DOI: 10.1016/j.cryobiol.2019.01.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 01/29/2019] [Accepted: 01/29/2019] [Indexed: 12/24/2022]
|
34
|
|
35
|
Helm A, Ebner DK, Tinganelli W, Simoniello P, Bisio A, Marchesano V, Durante M, Yamada S, Shimokawa T. Combining Heavy-Ion Therapy with Immunotherapy: An Update on Recent Developments. Int J Part Ther 2018; 5:84-93. [PMID: 31773022 PMCID: PMC6871592 DOI: 10.14338/ijpt-18-00024.1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 07/05/2018] [Indexed: 12/18/2022] Open
Abstract
Clinical trials and case reports of cancer therapies combining radiation therapy with immunotherapy have at times demonstrated total reduction or elimination of metastatic disease. While virtually all trials focus on the use of immunotherapy combined with conventional photon irradiation, the dose-distributive benefits of particles, in particular the distinct biological effects of heavy ions, have unknown potential vis-a-vis systemic disease response. Here, we review recent developments and evidence with a focus on the potential for heavy-ion combination therapy.
Collapse
Affiliation(s)
- Alexander Helm
- Trento Institute for Fundamental Physics and Applications-National Institute for Nuclear Physics, Trento, Italy
| | - Daniel K. Ebner
- Brown University Alpert Medical School, Providence, RI, USA
- Hospital of the National Institute of Radiological Sciences, National Institutes of Quantum and Radiological Science and Technology, Chiba, Japan
| | - Walter Tinganelli
- Trento Institute for Fundamental Physics and Applications-National Institute for Nuclear Physics, Trento, Italy
| | - Palma Simoniello
- Department of Science and Technology, Parthenope University of Naples, Naples, Italy
| | - Alessandra Bisio
- Center for Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Valentina Marchesano
- Trento Institute for Fundamental Physics and Applications-National Institute for Nuclear Physics, Trento, Italy
- Center for Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Marco Durante
- Trento Institute for Fundamental Physics and Applications-National Institute for Nuclear Physics, Trento, Italy
| | - Shigeru Yamada
- Hospital of the National Institute of Radiological Sciences, National Institutes of Quantum and Radiological Science and Technology, Chiba, Japan
| | - Takashi Shimokawa
- National Institute of Radiological Sciences, National Institutes of Quantum and Radiological Science and Technology, Chiba, Japan
| |
Collapse
|
36
|
Tsuboi K. Advantages and Limitations in the Use of Combination Therapies with Charged Particle Radiation Therapy. Int J Part Ther 2018; 5:122-132. [PMID: 31773024 DOI: 10.14338/ijpt-18-00019.1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 06/21/2018] [Indexed: 12/15/2022] Open
Abstract
Purpose Studies are currently underway to help provide basic and clinical evidence for combination particle beam radiation therapy, on which there are few published reports. The purpose of this article is to summarize the current status in the use of particle beams combined with other modalities. Results Following from experiences in x-ray radiation therapy, combination therapy with proton beams (PBT) has been attempted, and several clinical studies have reported improved survival rates for patients with non-small cell lung cancer, pancreatic cancers, esophageal cancers, and glioblastomas. Recently, basic studies combining PBT with PARP inhibitors and histone deacetylase inhibitors have also reported promising results. In the area of carbon ion therapy (CIT), there are few clinical reports on combination therapy; however, the number of basic research reports exceeds that for PBT. So far, the combined use of cytotoxic drugs with CIT yields independent additive effects. In addition, it is notable that combination therapy with CIT is effective against radioresistant cancer stem-like cells. Recent evidence also suggests that local radiation therapy can induce an effective antitumor immune response. There has been an increased use of combination immune-modulating agents and cytokines with particle beams, especially CIT. The field of radiation therapy is evolving from a strong reliance on local-regional treatment to a growing reliance on systemic immunotherapy. Conclusions The combined use of anticancer agents with particle radiation therapy has a considerable potential effect. Future research in molecular targeting therapy and immunotherapy may help identify the most efficacious approach for combination therapy with protons and carbon ions.
Collapse
Affiliation(s)
- Koji Tsuboi
- Proton Medical Research Center, Faculty of Medicine, University of Tsukuba, Japan
| |
Collapse
|
37
|
Lühr A, von Neubeck C, Pawelke J, Seidlitz A, Peitzsch C, Bentzen SM, Bortfeld T, Debus J, Deutsch E, Langendijk JA, Loeffler JS, Mohan R, Scholz M, Sørensen BS, Weber DC, Baumann M, Krause M. "Radiobiology of Proton Therapy": Results of an international expert workshop. Radiother Oncol 2018; 128:56-67. [PMID: 29861141 DOI: 10.1016/j.radonc.2018.05.018] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 05/17/2018] [Accepted: 05/17/2018] [Indexed: 12/25/2022]
Abstract
The physical properties of proton beams offer the potential to reduce toxicity in tumor-adjacent normal tissues. Toward this end, the number of proton radiotherapy facilities has steeply increased over the last 10-15 years to currently around 70 operational centers worldwide. However, taking full advantage of the opportunities offered by proton radiation for clinical radiotherapy requires a better understanding of the radiobiological effects of protons alone or combined with drugs or immunotherapy on normal tissues and tumors. This report summarizes the main results of the international expert workshop "Radiobiology of Proton Therapy" that was held in November 2016 in Dresden. It addresses the major topics (1) relative biological effectiveness (RBE) in proton beam therapy, (2) interaction of proton radiobiology with radiation physics in current treatment planning, (3) biological effects in proton therapy combined with systemic treatments, and (4) testing biological effects of protons in clinical trials. Finally, important research avenues for improvement of proton radiotherapy based on radiobiological knowledge are identified. The clinical distribution of radiobiological effectiveness of protons alone or in combination with systemic chemo- or immunotherapies as well as patient stratification based on biomarker expressions are key to reach the full potential of proton beam therapy. Dedicated preclinical experiments, innovative clinical trial designs, and large high-quality data repositories will be most important to achieve this goal.
Collapse
Affiliation(s)
- Armin Lühr
- Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Germany; OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany; German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Cläre von Neubeck
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany; German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jörg Pawelke
- Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Germany; OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany
| | - Annekatrin Seidlitz
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany; German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany
| | - Claudia Peitzsch
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany; German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany; National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany, and; Helmholtz Association/Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Germany
| | - Søren M Bentzen
- Division of Biostatistics and Bioinformatics, Department of Epidemiology and Public Health and the Maryland Proton Therapy Center, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, USA
| | - Thomas Bortfeld
- Physics Division, Department of Radiation Oncology, Massachusetts General Hospital, Boston, USA
| | - Jürgen Debus
- German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Radiation Oncology, University Heidelberg German Consortium for Translational Oncology (DKTK), Germany; National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Eric Deutsch
- Department of Radiation Oncology Gustave Roussy Cancer Campus, INSERM, 1030 Villejuif, France; Université Paris-Sud, Faculté de Medecine du Kremlin Bicetre Paris Sud, Le Kremlin-Bicêtre, France
| | - Johannes A Langendijk
- Department of Radiation Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jay S Loeffler
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, USA; Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, USA
| | - Radhe Mohan
- Department of Radiation Physics, UT MD Anderson Cancer Center, Houston, USA
| | - Michael Scholz
- GSI Helmholtz Center for Heavy Ion Research, Department of Biophysics, Darmstadt, Germany
| | - Brita S Sørensen
- Dept. Experimental Clinical Oncology, Aarhus University Hospital, Denmark
| | - Damien C Weber
- Center for Proton Therapy, Paul Scherrer Institute, ETH Domain, Villigen, Switzerland
| | - Michael Baumann
- Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Germany; OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany; German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany, and; Helmholtz Association/Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Mechthild Krause
- Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Germany; OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany; German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany, and; Helmholtz Association/Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Germany
| | | |
Collapse
|
38
|
Frey B, Rückert M, Deloch L, Rühle PF, Derer A, Fietkau R, Gaipl US. Immunomodulation by ionizing radiation-impact for design of radio-immunotherapies and for treatment of inflammatory diseases. Immunol Rev 2018; 280:231-248. [PMID: 29027224 DOI: 10.1111/imr.12572] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ionizing radiation is often regarded as an element of danger. But, danger responses on the cellular and molecular level are often beneficial with regard to the induction of anti-tumor immunity and for amelioration of inflammation. We outline how in dependence of radiation dose and fraction, radiation itself-and especially in combination with immune modulators-impacts on the innate and adaptive immune system. Focus is set on radiation-induced changes of the tumor cell phenotype and the cellular microenvironment including immunogenic cancer cell death. Mechanisms how anti-tumor immune responses are triggered by radiotherapy in combination with hyperthermia, inhibition of apoptosis, the adjuvant AnnexinA5, or vaccination with high hydrostatic pressure-killed autologous tumor cells are discussed. Building on this, feasible multimodal radio-immunotherapy concepts are reviewed including overcoming immune suppression by immune checkpoint inhibitors and by targeting TGF-β. Since radiation-induced tissue damage, inflammation, and anti-tumor immune responses are interconnected, the impact of lower doses of radiation on amelioration of inflammation is outlined. Closely meshed immune monitoring concepts based on the liquid biopsy blood are suggested for prognosis and prediction of cancer and non-cancer inflammatory diseases. Finally, challenges and visions for the design of cancer radio-immunotherapies and for treatment of benign inflammatory diseases are given.
Collapse
Affiliation(s)
- Benjamin Frey
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Michael Rückert
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Lisa Deloch
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Paul F Rühle
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Anja Derer
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Rainer Fietkau
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Udo S Gaipl
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
39
|
Durante M, Formenti SC. Radiation-Induced Chromosomal Aberrations and Immunotherapy: Micronuclei, Cytosolic DNA, and Interferon-Production Pathway. Front Oncol 2018; 8:192. [PMID: 29911071 PMCID: PMC5992419 DOI: 10.3389/fonc.2018.00192] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 05/14/2018] [Indexed: 11/13/2022] Open
Abstract
Radiation-induced chromosomal aberrations represent an early marker of late effects, including cell killing and transformation. The measurement of cytogenetic damage in tissues, generally in blood lymphocytes, from patients treated with radiotherapy has been studied for many years to predict individual sensitivity and late morbidity. Acentric fragments are lost during mitosis and create micronuclei (MN), which are well correlated to cell killing. Immunotherapy is rapidly becoming a most promising new strategy for metastatic tumors, and combination with radiotherapy is explored in several pre-clinical studies and clinical trials. Recent evidence has shown that the presence of cytosolic DNA activates immune response via the cyclic GMP-AMP synthase/stimulator of interferon genes pathway, which induces type I interferon transcription. Cytosolic DNA can be found after exposure to ionizing radiation either as MN or as small fragments leaking through nuclear envelope ruptures. The study of the dependence of cytosolic DNA and MN on dose and radiation quality can guide the optimal combination of radiotherapy and immunotherapy. The role of densely ionizing charged particles is under active investigation to define their impact on the activation of the interferon pathway.
Collapse
Affiliation(s)
- Marco Durante
- Trento Institute for Fundamental and Applied Physics (TIFPA), National Institute for Nuclear Physics (INFN), University of Trento, Trento, Italy
| | - Silvia C. Formenti
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States
| |
Collapse
|
40
|
Targeted alpha therapy using Radium-223: From physics to biological effects. Cancer Treat Rev 2018; 68:47-54. [PMID: 29859504 DOI: 10.1016/j.ctrv.2018.05.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 05/22/2018] [Accepted: 05/23/2018] [Indexed: 12/13/2022]
Abstract
With the advance of the use of ionizing radiation in therapy, targeted alpha therapy (TAT) has assumed an important role around the world. This kind of therapy can potentially reduce side effects caused by radiation in normal tissues and increased destructive radiobiological effects in tumor cells. However, in many countries, the use of this therapy is still in a pioneering phase. Radium-223 (223Ra), an alpha-emitting radionuclide, has been the first of its kind to be approved for the treatment of bone metastasis in metastatic castration-resistant prostate cancer. Nevertheless, the interaction mechanism and the direct effects of this radiopharmaceutical in tumor cells are not fully understood neither characterized at a molecular level. In fact, the ways how TAT is linked to radiobiological effects in cancer is not yet revised. Therefore, this review introduces some physical properties of TAT that leads to biological effects and links this information to the hallmarks of cancer. The authors also collected the studies developed with 223Ra to correlate with the three categories reviewed - properties of TAT, 5 R's of radiobiology and hallmarks of cancer- and with the promising future to this radiopharmaceutical.
Collapse
|
41
|
Cushman TR, Verma V, Rwigema JCM. Comparison of proton therapy and intensity modulated photon radiotherapy for locally advanced non-small cell lung cancer: considerations for optimal trial design. J Thorac Dis 2018; 10:S988-S990. [PMID: 29849214 DOI: 10.21037/jtd.2018.04.59] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Taylor R Cushman
- The University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Vivek Verma
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE, USA
| | | |
Collapse
|
42
|
Heavy Charged Particles: Does Improved Precision and Higher Biological Effectiveness Translate to Better Outcome in Patients? Semin Radiat Oncol 2018. [DOI: 10.1016/j.semradonc.2017.11.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
43
|
Abdo J, Cornell DL, Mittal SK, Agrawal DK. Immunotherapy Plus Cryotherapy: Potential Augmented Abscopal Effect for Advanced Cancers. Front Oncol 2018; 8:85. [PMID: 29644213 PMCID: PMC5882833 DOI: 10.3389/fonc.2018.00085] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 03/12/2018] [Indexed: 01/10/2023] Open
Abstract
Since the 1920s the gold standard for treating cancer has been surgery, which is typically preceded or followed with chemotherapy and/or radiation, a process that perhaps contributes to the destruction of a patient’s immune defense system. Cryosurgery ablation of a solid tumor is mechanistically similar to a vaccination where hundreds of unique antigens from a heterogeneous population of tumor cells derived from the invading cancer are released. However, releasing tumor-derived self-antigens into circulation may not be sufficient enough to overcome the checkpoint escape mechanisms some cancers have evolved to avoid immune responses. The potentiated immune response caused by blocking tumor checkpoints designed to prevent programmed cell death may be the optimal treatment method for the immune system to recognize these new circulating cryoablated self-antigens. Preclinical and clinical evidence exists for the complementary roles for Cytotoxic T-lymphocyte-associated protein (CTLA-4) and PD-1 antagonists in regulating adaptive immunity, demonstrating that combination immunotherapy followed by cryosurgery provides a more targeted immune response to distant lesions, a phenomenon known as the abscopal effect. We propose that when the host’s immune system has been “primed” with combined anti-CTLA-4 and anti-PD-1 adjuvants prior to cryosurgery, the preserved cryoablated tumor antigens will be presented and processed by the host’s immune system resulting in a robust cytotoxic CD8+ T-cell response. Based on recent investigations and well-described biochemical mechanisms presented herein, a polyvalent autoinoculation of many tumor-specific antigens, derived from a heterogeneous population of tumor cancer cells, would present to an unhindered yet pre-sensitized immune system yielding a superior advantage in locating, recognizing, and destroying tumor cells throughout the body.
Collapse
Affiliation(s)
- Joe Abdo
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, United States
| | - David L Cornell
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, United States.,Department of Surgery, CHI Health Creighton University Medical Center, Omaha, NE, United States
| | - Sumeet K Mittal
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, United States.,Dignity Health, Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, United States
| | - Devendra K Agrawal
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, United States
| |
Collapse
|
44
|
Xie YQ, Wei L, Tang L. Immunoengineering with biomaterials for enhanced cancer immunotherapy. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2018; 10:e1506. [PMID: 29333729 DOI: 10.1002/wnan.1506] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Revised: 11/07/2017] [Accepted: 11/22/2017] [Indexed: 12/17/2022]
Abstract
Cancer immunotherapy has recently shown dramatic clinical success inducing durable response in patients of a wide variety of malignancies. Further improvement of the clinical outcome with immune related cancer treatment requests more exquisite manipulation of a patient's immune system with increased immunity against diseases while mitigating the toxicities. To meet this challenge, biomaterials applied to immunoengineering are being developed to achieve tissue- and/or cell-specific immunomodulation and thus could potentially enhance both the efficacy and safety of current cancer immunotherapies. Here, we review the recent advancement in the field of immunoengineering using biomaterials and their applications in promoting different modalities of cancer immunotherapies, with focus on cell-, antibody-, immunomodulator-, and gene-based immune related treatments and their combinations with conventional therapies. Challenges and opportunities are discussed in applying biomaterials engineering strategies in the development of future cancer immunotherapies. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Therapeutic Approaches and Drug Discovery > Emerging Technologies Implantable Materials and Surgical Technologies > Nanomaterials and Implants.
Collapse
Affiliation(s)
- Yu-Qing Xie
- Institute of Bioengineering, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Lixia Wei
- Institute of Materials Science & Engineering, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Li Tang
- Institute of Bioengineering, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland.,Institute of Materials Science & Engineering, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
45
|
Lacombe S, Porcel E, Scifoni E. Particle therapy and nanomedicine: state of art and research perspectives. Cancer Nanotechnol 2017; 8:9. [PMID: 29213338 PMCID: PMC5698390 DOI: 10.1186/s12645-017-0029-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 09/08/2017] [Indexed: 12/31/2022] Open
Abstract
Cancer radiation therapy with charged particle beams, called particle therapy, is a new therapeutic treatment presenting major advantages when compared to conventional radiotherapy. Because ions have specific ballistic properties and a higher biological effectiveness, they are superior to x-rays. Numerous medical centres are starting in the world using mostly protons but also carbon ions as medical beams. Several investigations are attempting to reduce the cost/benefit ratio and enlarge the range of therapeutic indications. A major limitation of particle therapy is the presence of low but significant damage induced in healthy tissues located at the entrance of the ion track prior to reaching the tumour. It is thus a major challenge to improve the targeting of the tumours, concentrating radiation effects in the malignance. A novel strategy, based on the addition of nanoparticles targeting the tumour, was suggested over a decade ago to improve the performance of conventional photon therapy. Recently, similar developments have emerged for particle therapy and the amount of research is now exploding. In this paper, we review the experimental results, as well as theoretical and simulation studies that shed light in the promising outcomes of this strategy and in the underpinning mechanisms. Several experiments provide consistent evidence of significant enhancement of ion radiation effects in the presence of nanoparticles. In view of implementing this strategy for cancer treatment, simulation studies have begun to establish the rationale and the specificity of this effect. In addition, these studies will help to outline a list of possible mechanisms and to predict the impact of ion beams and nanoparticle characteristics. Many questions remain unsolved, but the findings of these first studies are encouraging and open new challenges. After summarizing the main results in the field, we propose a roadmap to pursue future research with the aim to strengthen the potential interplay between particle therapy and nanomedicine.
Collapse
Affiliation(s)
- Sandrine Lacombe
- Institut des Sciences Moléculaires d'Orsay (UMR 8214) Bât 351, University Paris Saclay, University of Paris Sud, CNRS, 91405 Orsay Cedex, France
| | - Erika Porcel
- Institut des Sciences Moléculaires d'Orsay (UMR 8214) Bât 351, University Paris Saclay, University of Paris Sud, CNRS, 91405 Orsay Cedex, France
| | - Emanuele Scifoni
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung, 64291 Darmstadt, Germany.,TIFPA-INFN, Trento Institute for Fundamental Physics and Applications, University of Trento, 38121 Trento, Italy
| |
Collapse
|
46
|
Abstract
OBJECTIVE This article aims to address the need to rethink the classification of mental illness and to draw attention to the current use of quantum biology and its likely future use in understanding the nature of mental illness. CONCLUSION It is desirable to separate out neuroscience and clinical research and to become better acquainted with the concepts of quantum mechanical/quantum biological theory and its contribution to medicine.
Collapse
|
47
|
Ebner DK, Kamada T, Yamada S. Abscopal effect in recurrent colorectal cancer treated with carbon-ion radiation therapy: 2 case reports. Adv Radiat Oncol 2017; 2:333-338. [PMID: 29114600 PMCID: PMC5605310 DOI: 10.1016/j.adro.2017.06.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 05/31/2017] [Accepted: 06/01/2017] [Indexed: 12/27/2022] Open
Affiliation(s)
- Daniel K. Ebner
- Hospital of the National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Sciences and Technology, Chiba, Japan
- Brown University Alpert Medical School, Providence, Rhode Island
| | - Tadashi Kamada
- Hospital of the National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Sciences and Technology, Chiba, Japan
| | - Shigeru Yamada
- Hospital of the National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Sciences and Technology, Chiba, Japan
| |
Collapse
|