1
|
Najafi M, Jahanbakhshi A, Gomar M, Iotti C, Giaccherini L, Rezaie O, Cavallieri F, Deantonio L, Bardoscia L, Botti A, Sardaro A, Cozzi S, Ciammella P. State of the Art in Combination Immuno/Radiotherapy for Brain Metastases: Systematic Review and Meta-Analysis. Curr Oncol 2022; 29:2995-3012. [PMID: 35621634 PMCID: PMC9139474 DOI: 10.3390/curroncol29050244] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/03/2022] [Accepted: 04/17/2022] [Indexed: 01/10/2023] Open
Abstract
OBJECTIVES Common origins for brain metastases (BMs) are melanoma, lung, breast, and renal cell cancers. BMs account for a large share of morbidity and mortality caused by these cancers. The advent of new immunotherapeutic treatments has made a revolution in the treatment of cancer patients and particularly, as a new concept, if it is combined with radiotherapy, may lead to considerably longer survival. This systematic review and meta-analysis aimed to evaluate the survival rate and toxicities of such a combination in brain metastases. METHODS To perform a systematic review of the literature until January 2021 using electronic databases such as PubMed, Cochrane Library, and Embase; the Newcastle-Ottawa Scale was used to evaluate the quality of cohort studies. For data extraction, two reviewers extracted the data blindly and independently. Hazard ratio with 95% confidence interval (CI), fixed-effect model, and inverse-variance method was calculated. The meta-analysis has been evaluated with the statistical software Stata/MP v.16 (The fastest version of Stata). RESULTS In the first step, 494 studies were selected to review the abstracts, in the second step, the full texts of 86 studies were reviewed. Finally, 28 studies were selected consisting of 1465 patients. The addition of IT to RT in the treatment of brain metastasis from melanoma and non-small-cell lung carcinoma was associated with a 39% reduction in mortality rate and has prolonged overall survival, with an acceptable toxicity profile. The addition of IT to RT compared with RT alone has a hazard ratio of 0.39(95% CI 0.34-0.44). CONCLUSIONS A combination of immuno/radiotherapy (IR) for the treatment of patients with BMs from melanoma and non-small-cell lung carcinoma has prolonged overall survival and reduced mortality rate, with acceptable toxicity. In terms of timing, RT seems to have the best effect on the result when performed before or simultaneously with immunotherapy.
Collapse
Affiliation(s)
- Masoumeh Najafi
- Skull Base Research Center, Iran University of Medical Sciences, Tehran 1997667665, Iran;
| | - Amin Jahanbakhshi
- Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran 1997667665, Iran;
| | - Marzieh Gomar
- Radiation Oncology Research Center, Iran Cancer Institute, Tehran University of Medical Sciences, Tehran 1416753955, Iran;
| | - Cinzia Iotti
- Radiation Therapy Unit, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy; (C.I.); (L.G.); (P.C.)
| | - Lucia Giaccherini
- Radiation Therapy Unit, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy; (C.I.); (L.G.); (P.C.)
| | - Omid Rezaie
- Hematology-Oncology Department, Jam Hospital, Tehran 1997667665, Iran;
| | - Francesco Cavallieri
- Neurology Unit, Neuromotor & Rehabilitation Department, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy;
| | - Letizia Deantonio
- Radiation Oncology Clinic, Oncology Institute of Southern Switzerland (IOSI), 6500 Bellinzona, Switzerland;
| | - Lilia Bardoscia
- Radiation Oncology Unit, S. Luca Hospital, Healthcare Company Tuscany Nord Ovest, 55100 Lucca, Italy;
| | - Andrea Botti
- Medical Physics Unit, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy;
| | - Angela Sardaro
- Section of Radiology and Radiation Oncology, Interdisciplinary Department of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy;
| | - Salvatore Cozzi
- Radiation Therapy Unit, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy; (C.I.); (L.G.); (P.C.)
| | - Patrizia Ciammella
- Radiation Therapy Unit, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy; (C.I.); (L.G.); (P.C.)
| |
Collapse
|
2
|
Zeng W, Fu L, Xu H. MicroRNA-206 relieves irradiation-induced neuroinflammation by regulating connexin 43. Exp Ther Med 2021; 22:1186. [PMID: 34475976 PMCID: PMC8406811 DOI: 10.3892/etm.2021.10620] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 07/26/2021] [Indexed: 01/17/2023] Open
Abstract
Radiation therapy has been widely used for the treatment of various types of cancer; however, it may cause neuroinflammation during the pathological process of the disease. Astrocytes, the most abundant cell type in the central nervous system, have been confirmed to play vital roles in various diseases. Connexin (Cx)43, the main Cx type in astrocytes, which has been identified as a direct target gene of microRNA (miR)-206, was found to be involved in diseases pathologies in regions with astrocytes. The aim of the present study was to investigate the mechanism through which γ-radiation may cause astrocyte neuroinflammation and determine the specific mechanism underlying the effects of miR-206 in irradiation-induced HA-1800 cells. A dual-luciferase reporter system was used to predict and verify the target binding site between Cx43 and miR-206. HA-1800 cell viability and apoptosis were determined using a MTT assay and flow cytometry, respectively. In addition, the HA-1800 cells were induced by γ-radiation, then the protein and mRNA expression levels of Cx43, miR-206 and cleaved-caspase-3 were determined using western blot and reverse transcription-quantitative PCR analyses, respectively. ELISA was also performed to evaluate the concentrations of different inflammatory cytokines (TNF-α, IL-β, IL-6 and IFN-γ). The dual-luciferase reporter system indicated that Cx43 was a direct target of miR-206. miR-206 mimics increased the expression level of miR-206 in the astrocytes. Irradiation suppressed cell proliferation, increased apoptotic cells and enhanced cleaved-caspase-3 expression and inflammatory cytokines secretion in astrocytes. Furthermore, miR-206 was found to be downregulated and its expression was inversely associated with that of Cx43 in γ-radiation-induced astrocytes. Overexpression of miR-206 enhanced miR-206 and suppressed Cx43 expression, while Cx43 was upregulated in HA-1800 cells transfected with miR-206 mimic + Cx43-plasmid. However, the expression level of miR-206 was not significantly different in the Cx43-plasmid transfected group. In addition, it was found that miR-206 mimics relieved irradiation-induced neuroinflammation, which was confirmed by increased cell viability, and reduced cell apoptosis and cleaved caspase-3 protein expression, as well as decreased inflammatory cytokine secretion. Furthermore, all the effects of miR-206 mimics on γ-radiation-induced astrocytes were reversed by Cx43-plasmid. In summary, the results of the present study indicated that miR-206 may relieve irradiation-induced neural damage by regulating Cx43, which may provide a novel research direction and a potential therapeutic target for the clinical treatment of inflammation-associated neuronal injury following irradiation.
Collapse
Affiliation(s)
- Wei Zeng
- Department of Radiology, Affiliated Hospital of Jianghan University, The Sixth Hospital of Wuhan City, Wuhan, Hubei 430019, P.R. China
| | - Li Fu
- Department of Radiology, Affiliated Hospital of Jianghan University, The Sixth Hospital of Wuhan City, Wuhan, Hubei 430019, P.R. China
| | - Hongfang Xu
- Department of Radiology, Affiliated Hospital of Jianghan University, The Sixth Hospital of Wuhan City, Wuhan, Hubei 430019, P.R. China
| |
Collapse
|
3
|
Jacob JB, Jacob MK, Parajuli P. Review of immune checkpoint inhibitors in immuno-oncology. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2021; 91:111-139. [PMID: 34099106 DOI: 10.1016/bs.apha.2021.01.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Tumor cells predominantly express self-antigens and overcoming self-tolerance is the primary challenge to effective immunotherapy. Tumors also express ligands for co-inhibitory molecules on immune cells, in order to suppress anti-tumor immunity. Over a decade ago, the first antibodies generated to block the co-inhibitory molecule CTLA-4 was tested in patients with metastatic melanoma. Results from this landmark trial have informed not only the current landscape of checkpoint blockade but also the way in which immunotherapy trial outcomes are determined. Antibodies targeting PD-1 and its ligand, PD-L1, soon followed and use of these checkpoint inhibitors (ICIs) have expanded exponentially. ICI treatment has shown long-lasting clinical benefit in several tumor types and patients refractory to other treatments can often respond to ICI therapy. On the other hand, in some tumor types, the response to ICI is short-lived and tumors eventually recur. Current clinical trials are focused on enhancing anti-tumor effects through combinations of multiple ICIs with agents which cause tumor death, particularly in solid tumors, in order to enhance antigen presentation. It is also important to define which patients will respond to therapy with ICIs as over half of all patients suffer from immune-related adverse events (irAE), some of which are severe and long-lasting.
Collapse
Affiliation(s)
- Jennifer B Jacob
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States.
| | - Mark K Jacob
- Michigan State University School of Human Medicine, East Lansing, MI, United States
| | - Prahlad Parajuli
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
| |
Collapse
|
4
|
Kataoka H, Nishie H, Tanaka M, Sasaki M, Nomoto A, Osaki T, Okamoto Y, Yano S. Potential of Photodynamic Therapy Based on Sugar-Conjugated Photosensitizers. J Clin Med 2021; 10:jcm10040841. [PMID: 33670714 PMCID: PMC7922816 DOI: 10.3390/jcm10040841] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/08/2021] [Accepted: 02/12/2021] [Indexed: 02/06/2023] Open
Abstract
In 2015, the Japanese health insurance approved the use of a second-generation photodynamic therapy (PDT) using talaporfin sodium (TS); however, its cancer cell selectivity and antitumor effects of TS PDT are not comprehensive. The Warburg effect describes the elevated rate of glycolysis in cancer cells, despite the presence of sufficient oxygen. Because cancer cells absorb considerable amounts of glucose, they are visible using positron emission tomography (PET). We developed a third-generation PDT based on the Warburg effect by synthesizing novel photosensitizers (PSs) in the form of sugar-conjugated chlorins. Glucose-conjugated (tetrafluorophenyl) chlorin (G-chlorin) PDT revealed significantly stronger antitumor effects than TS PDT and induced immunogenic cell death (ICD). ICD induced by PDT enhances cancer immunity, and a combination therapy of PDT and immune checkpoint blockers is expected to synergize antitumor effects. Mannose-conjugated (tetrafluorophenyl) chlorin (M-chlorin) PDT, which targets cancer cells and tumor-associated macrophages (TAMs), also shows strong antitumor effects. Finally, we synthesized a glucose-conjugated chlorin e6 (SC-N003HP) that showed 10,000-50,000 times stronger antitumor effects than TS (IC50) in vitro, and it was rapidly metabolized and excreted. In this review, we discuss the potential and the future of next-generation cancer cell-selective PDT and describe three types of sugar-conjugated PSs expected to be clinically developed in the future.
Collapse
Affiliation(s)
- Hiromi Kataoka
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan; (H.N.); (M.T.); (M.S.)
- Correspondence:
| | - Hirotada Nishie
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan; (H.N.); (M.T.); (M.S.)
| | - Mamoru Tanaka
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan; (H.N.); (M.T.); (M.S.)
| | - Makiko Sasaki
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan; (H.N.); (M.T.); (M.S.)
| | - Akihiro Nomoto
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, Osaka 599-8531, Japan;
| | - Tomohiro Osaki
- Joint Department of Veterinary Clinical Medicine, Faculty of Agriculture, Tottori University, Tottori 680-8553, Japan; (T.O.); (Y.O.)
| | - Yoshiharu Okamoto
- Joint Department of Veterinary Clinical Medicine, Faculty of Agriculture, Tottori University, Tottori 680-8553, Japan; (T.O.); (Y.O.)
| | - Shigenobu Yano
- KYOUSEI Science Center for Life and Nature, Nara Women’s University, Kitauoyahigashi-machi, Nara 630-8506, Japan;
| |
Collapse
|
5
|
Mapanao AK, Che PP, Sarogni P, Sminia P, Giovannetti E, Voliani V. Tumor grafted - chick chorioallantoic membrane as an alternative model for biological cancer research and conventional/nanomaterial-based theranostics evaluation. Expert Opin Drug Metab Toxicol 2021; 17:947-968. [PMID: 33565346 DOI: 10.1080/17425255.2021.1879047] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Introduction: Advancements in cancer management and treatment are associated with strong preclinical research data, in which reliable cancer models are demanded. Indeed, inconsistent preclinical findings and stringent regulations following the 3Rs principle of reduction, refinement, and replacement of conventional animal models currently pose challenges in the development and translation of efficient technologies. The chick embryo chorioallantoic membrane (CAM) is a system for the evaluation of treatment effects on the vasculature, therefore suitable for studies on angiogenesis. Apart from vascular effects, the model is now increasingly employed as a preclinical cancer model following tumor-grafting procedures.Areas covered: The broad application of CAM tumor model is highlighted along with the methods for analyzing the neoplasm and vascular system. The presented and cited investigations focus on cancer biology and treatment, encompassing both conventional and emerging nanomaterial-based modalities.Expert opinion: The CAM tumor model finds increased significance given the influences of angiogenesis and the tumor microenvironment in cancer behavior, then providing a qualified miniature system for oncological research. Ultimately, the establishment and increased employment of such a model may resolve some of the limitations present in the standard preclinical tumor models, thereby redefining the preclinical research workflow.
Collapse
Affiliation(s)
- Ana Katrina Mapanao
- Center for Nanotechnology Innovation@NEST, Istituto Italiano Di Tecnologia, Pisa, Italy.,NEST-Scuola Normale Superiore, Pisa, Italy
| | - Pei Pei Che
- Department of Radiation Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center, Amsterdam, The Netherlands.,Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, The Netherlands
| | - Patrizia Sarogni
- Center for Nanotechnology Innovation@NEST, Istituto Italiano Di Tecnologia, Pisa, Italy
| | - Peter Sminia
- Department of Radiation Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center, Amsterdam, The Netherlands
| | - Elisa Giovannetti
- Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, The Netherlands.,Cancer Pharmacology Lab, AIRC Start-Up Unit, Fondazione Pisana per La Scienza, Pisa, Italy
| | - Valerio Voliani
- Center for Nanotechnology Innovation@NEST, Istituto Italiano Di Tecnologia, Pisa, Italy
| |
Collapse
|
6
|
Trommer M, Kinsky J, Adams A, Hellmich M, Schlaak M, von Bergwelt-Baildon M, Celik E, Rosenbrock J, Morgenthaler J, Herter JM, Linde P, Mauch C, Theurich S, Marnitz S, Baues C. Addition of Radiotherapy to Immunotherapy: Effects on Outcome of Different Subgroups Using a Propensity Score Matching. Cancers (Basel) 2020; 12:cancers12092429. [PMID: 32867046 PMCID: PMC7563550 DOI: 10.3390/cancers12092429] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/05/2020] [Accepted: 08/24/2020] [Indexed: 01/05/2023] Open
Abstract
Immune checkpoint inhibition (ICI) has been established as successful modality in cancer treatment. Combination concepts are used to optimize treatment outcome, but may also induce higher toxicity rates than monotherapy. Several rationales support the combination of radiotherapy (RT) with ICI as radioimmunotherapy (RIT), but it is still unknown in which clinical situation RIT would be most beneficial. Therefore, we have conducted a retrospective matched-pair analysis of 201 patients with advanced-stage cancers and formed two groups treated with programmed cell death protein 1 (PD-1) inhibitors only (PD1i) or in combination with local RT (RIT) at our center between 2013 and 2017. We collected baseline characteristics, programmed death ligand 1 (PD-L1) status, mutational status, PD-1 inhibitor and RT treatment details, and side effects according to the Common Terminology Criteria for Adverse Events (CTCAE) v.5.0. Patients received pembrolizumab (n = 93) or nivolumab (n = 108), 153 with additional RT. For overall survival (OS) and progression-free survival (PFS), there was no significant difference between both groups. After propensity score matching (PSM), we analyzed 96 patients, 67 with additional and 29 without RT. We matched for different covariates that could have a possible influence on the treatment outcome. The RIT group displayed a trend towards a longer OS until the PD1i group reached a survival plateau. PD-L1-positive patients, smokers, patients with a BMI ≤ 25, and patients without malignant melanoma showed a longer OS when treated with RIT. Our data show that some subgroups may benefit more from RIT than others. Suitable biomarkers as well as the optimal timing and dosage must be established in order to achieve the best effect on cancer treatment outcome.
Collapse
Affiliation(s)
- Maike Trommer
- Department of Radiation Oncology and Cyberknife Center, University Hospital of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; (J.K.); (E.C.); (J.R.); (J.M.); (J.M.H.); (P.L.); (S.M.); (C.B.)
- Radio Immune-Oncology Consortium (RIO), University Hospital of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; (M.S.); (M.v.B.-B.); (S.T.)
- Center for Integrated Oncology (CIO), University Hospital of Cologne, Kerpener Str. 62, 50937 Cologne, Germany;
- Center for Molecular Medicine Cologne (CMMC), University Hospital of Cologne, Kerpener Str. 62, 50937 Cologne, Germany
- Correspondence: ; Tel.: +49-221-4780; Fax: +49-221-4786648
| | - Jaika Kinsky
- Department of Radiation Oncology and Cyberknife Center, University Hospital of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; (J.K.); (E.C.); (J.R.); (J.M.); (J.M.H.); (P.L.); (S.M.); (C.B.)
| | - Anne Adams
- Institute of Medical Statistics and Computational Biology, University of Cologne, Faculty of Medicine and University Hospital of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; (A.A.); (M.H.)
| | - Martin Hellmich
- Institute of Medical Statistics and Computational Biology, University of Cologne, Faculty of Medicine and University Hospital of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; (A.A.); (M.H.)
| | - Max Schlaak
- Radio Immune-Oncology Consortium (RIO), University Hospital of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; (M.S.); (M.v.B.-B.); (S.T.)
- Department of Dermatology and Allergology, LMU University Hospital, Ludwig-Maximilians University (LMU), Munich, Frauenlobstr. 9-11, 80377 Munich, Germany
| | - Michael von Bergwelt-Baildon
- Radio Immune-Oncology Consortium (RIO), University Hospital of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; (M.S.); (M.v.B.-B.); (S.T.)
- Department III of Internal Medicine, LMU University Hospital, Ludwig-Maximilians University (LMU), Munich, Marchioninistr. 15, 81377 Munich, Germany
| | - Eren Celik
- Department of Radiation Oncology and Cyberknife Center, University Hospital of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; (J.K.); (E.C.); (J.R.); (J.M.); (J.M.H.); (P.L.); (S.M.); (C.B.)
- Center for Integrated Oncology (CIO), University Hospital of Cologne, Kerpener Str. 62, 50937 Cologne, Germany;
| | - Johannes Rosenbrock
- Department of Radiation Oncology and Cyberknife Center, University Hospital of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; (J.K.); (E.C.); (J.R.); (J.M.); (J.M.H.); (P.L.); (S.M.); (C.B.)
- Center for Integrated Oncology (CIO), University Hospital of Cologne, Kerpener Str. 62, 50937 Cologne, Germany;
| | - Janis Morgenthaler
- Department of Radiation Oncology and Cyberknife Center, University Hospital of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; (J.K.); (E.C.); (J.R.); (J.M.); (J.M.H.); (P.L.); (S.M.); (C.B.)
- Center for Integrated Oncology (CIO), University Hospital of Cologne, Kerpener Str. 62, 50937 Cologne, Germany;
| | - Jan M. Herter
- Department of Radiation Oncology and Cyberknife Center, University Hospital of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; (J.K.); (E.C.); (J.R.); (J.M.); (J.M.H.); (P.L.); (S.M.); (C.B.)
- Center for Integrated Oncology (CIO), University Hospital of Cologne, Kerpener Str. 62, 50937 Cologne, Germany;
- Center for Molecular Medicine Cologne (CMMC), University Hospital of Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Philipp Linde
- Department of Radiation Oncology and Cyberknife Center, University Hospital of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; (J.K.); (E.C.); (J.R.); (J.M.); (J.M.H.); (P.L.); (S.M.); (C.B.)
- Center for Integrated Oncology (CIO), University Hospital of Cologne, Kerpener Str. 62, 50937 Cologne, Germany;
| | - Cornelia Mauch
- Center for Integrated Oncology (CIO), University Hospital of Cologne, Kerpener Str. 62, 50937 Cologne, Germany;
- Department of Dermatology and Allergology, University Hospital of Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Sebastian Theurich
- Radio Immune-Oncology Consortium (RIO), University Hospital of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; (M.S.); (M.v.B.-B.); (S.T.)
- Department III of Internal Medicine, LMU University Hospital, Ludwig-Maximilians University (LMU), Munich, Marchioninistr. 15, 81377 Munich, Germany
- Cancer & Immunometabolism Research Group, Gene Center LMU, Ludwig-Maximilians University, Munich, Feodor-Lynen-Str. 25, 81377 Munich, Germany
| | - Simone Marnitz
- Department of Radiation Oncology and Cyberknife Center, University Hospital of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; (J.K.); (E.C.); (J.R.); (J.M.); (J.M.H.); (P.L.); (S.M.); (C.B.)
- Radio Immune-Oncology Consortium (RIO), University Hospital of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; (M.S.); (M.v.B.-B.); (S.T.)
- Center for Integrated Oncology (CIO), University Hospital of Cologne, Kerpener Str. 62, 50937 Cologne, Germany;
| | - Christian Baues
- Department of Radiation Oncology and Cyberknife Center, University Hospital of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; (J.K.); (E.C.); (J.R.); (J.M.); (J.M.H.); (P.L.); (S.M.); (C.B.)
- Radio Immune-Oncology Consortium (RIO), University Hospital of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; (M.S.); (M.v.B.-B.); (S.T.)
- Center for Integrated Oncology (CIO), University Hospital of Cologne, Kerpener Str. 62, 50937 Cologne, Germany;
- Center for Molecular Medicine Cologne (CMMC), University Hospital of Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| |
Collapse
|
7
|
Pass HI. Commentary: "Hot" gene signatures for checkpoint response prediction: A very cool idea. J Thorac Cardiovasc Surg 2020; 159:1611-1612. [PMID: 32037244 DOI: 10.1016/j.jtcvs.2019.12.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 12/07/2019] [Accepted: 12/10/2019] [Indexed: 11/29/2022]
Affiliation(s)
- Harvey I Pass
- Department of Cardiothoracic Surgery, Division of General Thoracic Surgery NYU Langone Medical Center, New York, NY.
| |
Collapse
|
8
|
Dahl O, Brydøy M. The pioneers behind immune checkpoint blockers awarded the Nobel Prize in physiology or medicine 2018. Acta Oncol 2019; 58:1-8. [PMID: 30698061 DOI: 10.1080/0284186x.2018.1555375] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Olav Dahl
- Department of Clinical Science Faculty of Medicine, University of Bergen, Bergen, Norway
- Department of Oncology and Medical Physics, Haukeland University Hospital, Bergen, Norway
| | - Marianne Brydøy
- Department of Oncology and Medical Physics, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|