1
|
Pepper NB, Eich HT, Müther M, Oertel M, Rehn S, Spille DC, Stummer W. ALA-RDT in GBM: protocol of the phase I/II dose escalation trial of radiodynamic therapy with 5-Aminolevulinic acid in patients with recurrent glioblastoma. Radiat Oncol 2024; 19:11. [PMID: 38254201 PMCID: PMC10804590 DOI: 10.1186/s13014-024-02408-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 01/16/2024] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND Despite improvements in surgical as well as adjuvant therapies over the last decades, the prognosis for patients with glioblastoma remains poor. Five-Aminolevulinic acid (5-ALA) induced porphyrins are already used for fluorescence-guided resection and as photosensitizer for photodynamic therapy. New findings reveal their potential use as sensitizing agents in combination with ionizing radiation. METHODS We initiated a phase I/II dose escalation study, treating patients with recurrence of glioblastoma with oral 5-ALA concurrent to radiotherapy (RT). This prospective single-center study based in the University Hospital Münster aims to recruit 30 patients over 18 years of age with histologically verified recurrence of supratentorial glioblastoma in good performance status (KPS ≥ 60). Following a 3 + 3 dose-escalation design, patients having undergone re-resection will receive a 36 Gy RT including radiodynamic therapy fractions (RDT). RDT constitutes of oral administration of 5-ALA before the irradiation session. Two cohorts will additionally receive two fractions of neoadjuvant treatment three and two days before surgery. To determine the maximum tolerated dose of repeated 5-ALA-administration, the number of RDT-fractions will increase, starting with one to a maximum of eight fractions, while closely monitoring for safety and toxicity. Follow-up will be performed at two and five months after treatment. Primary endpoint will be the maximum tolerated dose (MTD) of repeated ALA-administration, secondary endpoints are event-free-, progression-free-, and overall-survival. Additionally, 5-ALA metabolites and radiobiological markers will be analysed throughout the course of therapy and tissue effects after neoadjuvant treatment will be determined in resected tissue. This protocol is in accordance with the SPIRIT guidelines for clinical trial protocols. DISCUSSION This is the protocol of the ALA-RDT in GBM-study, the first-in-man evaluation of repeated administration of 5-ALA as a radiosensitizer for treatment of recurrent glioblastoma. TRIAL REGISTRATION This study was approved by the local ethics committee of the Medical Association of Westphalia-Lippe and the University of Münster on 12.10.2022, the German federal institute for Drugs and medical devices on 13.10.2022 and the federal office for radiation protection on 29.08.2022. This trial was registered on the public European EudraCT database (EudraCT-No.: 2021-004631-92) and is registered under www.cliniclatrials.gov (Identifier: NCT05590689).
Collapse
Affiliation(s)
- Niklas Benedikt Pepper
- Department of Radiation Oncology, University Hospital of Münster, Albert-Schweitzer-Campus 1, Building A1, 48149, Münster, Germany.
| | - Hans Theodor Eich
- Department of Radiation Oncology, University Hospital of Münster, Albert-Schweitzer-Campus 1, Building A1, 48149, Münster, Germany
| | - Michael Müther
- Department of Neurosurgery, University Hospital of Münster, Albert-Schweitzer-Campus 1, Building A1, 48149, Münster, Germany
| | - Michael Oertel
- Department of Radiation Oncology, University Hospital of Münster, Albert-Schweitzer-Campus 1, Building A1, 48149, Münster, Germany
| | - Stephan Rehn
- Department of Radiation Oncology, University Hospital of Münster, Albert-Schweitzer-Campus 1, Building A1, 48149, Münster, Germany
| | - Dorothee Cäcilia Spille
- Department of Neurosurgery, University Hospital of Münster, Albert-Schweitzer-Campus 1, Building A1, 48149, Münster, Germany
| | - Walter Stummer
- Department of Neurosurgery, University Hospital of Münster, Albert-Schweitzer-Campus 1, Building A1, 48149, Münster, Germany
| |
Collapse
|
2
|
Frosina G. Improving control of high‐grade glioma by ultra‐hyper‐fractionated radiotherapy. J Neurosci Res 2022; 100:933-946. [DOI: 10.1002/jnr.25030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 10/05/2021] [Accepted: 12/13/2021] [Indexed: 11/11/2022]
Affiliation(s)
- Guido Frosina
- Mutagenesis & Cancer Prevention Unit IRCCS Ospedale Policlinico San Martino Genova Italy
| |
Collapse
|
3
|
Scirocco E, Cellini F, Zamagni A, Macchia G, Deodato F, Cilla S, Strigari L, Buwenge M, Rizzo S, Cammelli S, Morganti AG. Clinical Studies on Ultrafractionated Chemoradiation: A Systematic Review. Front Oncol 2021; 11:748200. [PMID: 34868948 PMCID: PMC8635188 DOI: 10.3389/fonc.2021.748200] [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: 07/27/2021] [Accepted: 10/25/2021] [Indexed: 01/08/2023] Open
Abstract
Aim The efficacy of low-dose fractionated radiotherapy (LDFRT) and chemotherapy (CHT) combination has large preclinical but little clinical evidence. Therefore, the aim of this review was to collect and analyze the clinical results of LDRT plus concurrent CHT in patients with advanced cancers. Methods A systematic literature search was conducted on PubMed using the PRISMA methodology. Only studies based on the combination of LDFRT (< 1 Gy/fraction) and CHT were included. Endpoints of the analysis were tumor response, toxicity, and overall survival, with particular focus on any differences between LDFRT-CHT and CHT alone. Results Twelve studies (307 patients) fulfilled the selection criteria and were included in this review. Two studies were retrospective, one was a prospective pilot trial, six were phase II studies, two were phase I trials, and one was a phase I/II open label study. No randomized controlled trials were found. Seven out of eight studies comparing clinical response showed higher rates after LDFRT-CHT compared to CHT alone. Three out of four studies comparing survival reported improved results after combined treatment. Three studies compared toxicity of CHT and LDFRT plus CHT, and all of them reported similar adverse events rates. In most cases, toxicity was manageable with only three likely LDFRT-unrelated fatal events (1%), all recorded in the same series on LDFRT plus temozolomide in glioblastoma multiforme patients. Conclusion None of the analyzed studies provided level I evidence on the clinical impact of LDFRT plus CHT. However, it should be noted that, apart from two small series of breast cancers, all studies reported improved therapeutic outcomes and similar tolerability compared to CHT alone. Systematic Review Registration www.crd.york.ac.uk/prospero/, identifier CRD42020206639.
Collapse
Affiliation(s)
- Erica Scirocco
- Radiation Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Experimental, Diagnostic and Specialty Medicine-Alma Mater Studiorum Bologna University, Bologna, Italy
| | - Francesco Cellini
- Università Cattolica del Sacro Cuore, Dipartimento Universitario Diagnostica per immagini, Radioterapia Oncologica ed Ematologia, Roma, Italy.,Fondazione Policlinico Universitario "A. Gemelli" Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Roma, Italy
| | - Alice Zamagni
- Radiation Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Experimental, Diagnostic and Specialty Medicine-Alma Mater Studiorum Bologna University, Bologna, Italy
| | - Gabriella Macchia
- Radiotherapy Unit, Gemelli Molise Hospital, Catholic University of Sacred Heart, Campobasso, Italy
| | - Francesco Deodato
- Radiotherapy Unit, Gemelli Molise Hospital, Catholic University of Sacred Heart, Campobasso, Italy
| | - Savino Cilla
- Medical Physic Unit, Gemelli Molise Hospital, Catholic University of Sacred Heart, Campobasso, Italy
| | - Lidia Strigari
- Medical Physics Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Milly Buwenge
- Department of Experimental, Diagnostic and Specialty Medicine-Alma Mater Studiorum Bologna University, Bologna, Italy
| | - Stefania Rizzo
- Service of Radiology, Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale (EOC), Lugano, Switzerland
| | - Silvia Cammelli
- Radiation Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Experimental, Diagnostic and Specialty Medicine-Alma Mater Studiorum Bologna University, Bologna, Italy
| | - Alessio Giuseppe Morganti
- Radiation Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Experimental, Diagnostic and Specialty Medicine-Alma Mater Studiorum Bologna University, Bologna, Italy
| |
Collapse
|
4
|
Frosina G, Fontana V, Verzola D, Rosa A, Gaggero G, Garibotto G, Vagge S, Pigozzi S, Daga A. Ultra-hyper-fractionated radiotherapy for high-grade gliomas. J Neurosci Res 2021; 99:3182-3203. [PMID: 34747065 DOI: 10.1002/jnr.24929] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
High-grade gliomas (HGGs; WHO grades III and IV) are invariably lethal brain tumors. Low-dose hyper-radiosensitivity (HRS) of HGG is a well-established phenomenon in vitro. However, possibly linked to the unavailability of accurate animal models of the diseases, this therapeutic effect could not be consistently translated to the animal setting, thus impairing its subsequent clinical development. The purpose of this study was to develop radiotherapeutic (RT) schedules permitting to significantly improve the overall survival of faithful animal models of HGG that have been recently made available. We used primary glioma initiating cell (GIC)-driven orthotopic animal models that accurately recapitulate the heterogeneity and growth patterns of the patients' tumors, to investigate the therapeutic effects of low radiation doses toward HGG. With the same total dose, RT fractions ≤0.5 Gy twice per week [ultra-hyper-fractionation (ultra-hyper-FRT)] started at early stages of tumor progression (a condition that in the clinical setting often occurs at the end of the guidelines treatment) improved the effectiveness of RT and the animal survival in comparison to standard fractions. For the same cumulative dose, the use of fractions ≤0.5 Gy may permit to escape one or more tumor resistance mechanisms thus increasing the effectiveness of RT and the overall animal survival. These findings suggest investigating in the clinical setting the therapeutic effect of an ultra-hyper-FRT schedule promptly extending the conventional RT component of the current guideline ("Stupp") therapeutic protocol.
Collapse
Affiliation(s)
- Guido Frosina
- Mutagenesis & Cancer Prevention, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Vincenzo Fontana
- Clinical Epidemiology, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Daniela Verzola
- Department of Internal Medicine and Medical Specialties - Dimi, University of Genova, Genova, Italy
| | - Alessandra Rosa
- Clinical Epidemiology, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Gabriele Gaggero
- Pathological Anatomy and Histology, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Giacomo Garibotto
- Department of Internal Medicine and Medical Specialties - Dimi, University of Genova, Genova, Italy
| | - Stefano Vagge
- Radiation Oncology, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Simona Pigozzi
- Department of Surgical Sciences and Integrated Diagnostics - Disc, University of Genova, Genova, Italy
| | - Antonio Daga
- Cellular Oncology, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| |
Collapse
|
5
|
Straube C, Kessel KA, Zimmer C, Schmidt-Graf F, Schlegel J, Gempt J, Meyer B, Combs SE. A Second Course of Radiotherapy in Patients with Recurrent Malignant Gliomas: Clinical Data on Re-irradiation, Prognostic Factors, and Usefulness of Digital Biomarkers. Curr Treat Options Oncol 2019; 20:71. [PMID: 31324990 DOI: 10.1007/s11864-019-0673-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OPINION STATEMENT The treatment of malignant gliomas has undergone a significant intensification during the past decade, and the interdisciplinary treatment team has learned that all treatment opportunities, including surgery and radiotherapy (RT), also have a central role in recurrent gliomas. Throughout the decades, re-irradiation (re-RT) has achieved a prominent place in the treatment of recurrent gliomas. A solid body of evidence supports the safety and efficacy of re-RT, especially when modern techniques are used, and justifies the early use of this regimen, especially in the case when macroscopic disease is present. Additionally, a second adjuvant re-RT to the resection cavity is currently being investigated by several investigators and seems to offer promising results. Although advanced RT technologies, such as stereotactic radiosurgery (SRS), fractionated stereotactic radiotherapy (FSRT), intensity-modulated radiotherapy (IMRT), and image-guided radiotherapy (IGRT) have become available in many centers, re-RT should continue to be kept in experienced hands so that they can select the optimal regimen, the ideal treatment volume, and the appropriate techniques from their tool-boxes. Concomitant or adjuvant use of systemic treatment options should also strongly be taken into consideration, especially because temozolomide (TMZ), cyclohexyl-nitroso-urea (CCNU), and bevacizumab have shown a good safety profile; they should be considered, if available. Nonetheless, the selection of patients for re-RT remains crucial. Single factors, such as patient age or the progression-free interval (PFI), fall too short. Therefore, powerful prognostic scores have been generated and validated, and these scores should be used for patient selection and counseling.
Collapse
Affiliation(s)
- Christoph Straube
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München (TUM), Ismaninger Straße 22, 81675, Munich, Germany
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partner Site Munich, Munich, Germany
- Institute for Radiation Medicine (IRM), Department of Radiation Sciences (DRS), Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
| | - Kerstin A Kessel
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München (TUM), Ismaninger Straße 22, 81675, Munich, Germany
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partner Site Munich, Munich, Germany
- Institute for Radiation Medicine (IRM), Department of Radiation Sciences (DRS), Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
| | - Claus Zimmer
- Department of Neuroradiology, Klinikum rechts der Isar, Technical University of Munich (TUM), Munich, Germany
| | - Friederike Schmidt-Graf
- Department of Neurology, Klinikum rechts der Isar, Technical University of Munich (TUM), Munich, Germany
| | - Jürgen Schlegel
- Department of Neuroradiology, Klinikum rechts der Isar, Technical University of Munich (TUM), Munich, Germany
| | - Jens Gempt
- Department of Neurosurgery, Klinikum rechts der Isar, Technical University of Munich (TUM), Munich, Germany
| | - Bernhard Meyer
- Department of Neurosurgery, Klinikum rechts der Isar, Technical University of Munich (TUM), Munich, Germany
| | - Stephanie E Combs
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München (TUM), Ismaninger Straße 22, 81675, Munich, Germany.
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partner Site Munich, Munich, Germany.
- Institute for Radiation Medicine (IRM), Department of Radiation Sciences (DRS), Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany.
| |
Collapse
|
6
|
Re-irradiation for recurrent glioblastoma (GBM): a systematic review and meta-analysis. J Neurooncol 2018; 142:79-90. [PMID: 30523605 DOI: 10.1007/s11060-018-03064-0] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 11/24/2018] [Indexed: 12/12/2022]
Abstract
PURPOSE To determine the efficacy and toxicity of re-irradiation for patients with recurrent GBM. MATERIALS AND METHODS We searched various biomedical databases from 1998 to 2018, for eligible studies where patients were treated with re-irradiation for recurrent GBM. Outcomes of interest were 6 and 12-month overall survival (OS-6, OS-12), 6 and 12-month progression free survival (PFS-6, PFS-12) and serious (Grade 3 +) adverse events (AE). We used the random effects model to pool outcomes across studies and compared pre-defined subgroups using interaction test. Methodological quality of each study was assessed using the Newcastle-Ottawa scoring system. RESULTS We found 50 eligible non-comparative studies including 2095 patients. Of these, 42% were of good or fair quality. The pooled results were as follows: OS-6 rate 73% (95% confidence interval (CI) 69-77%), OS-12 rate 36% (95% CI 32-40%), PFS-6 rate 43% (95% CI 35-50%), PFS-12 rate 17% (95% CI 13-20%), and Grade 3 + AE rate 7% (95% CI 4-10%). Subgroup analysis showed that prospective studies reported higher toxicity rates, and studies which utilized brachytherapy to have a longer OS-12. Within the external beam radiotherapy group, there was no dose-response [above or below 36 Gy in 2 Gy equivalent doses (EQD2)]. However, a short fractionation regimen (≤ 5 fractions) seemed to provide superior PFS-6. CONCLUSION The available evidence, albeit mostly level III, suggests that re-irradiation provides encouraging disease control and survival rates. Toxicity was not uniformly reported, but seemed to be low from the included studies. Randomized controlled trials (RCT) are needed to establish the optimal management strategy for recurrent GBM.
Collapse
|
7
|
Fròsina G, Profumo A, Marubbi D, Marcello D, Ravetti JL, Daga A. ATR kinase inhibitors NVP-BEZ235 and AZD6738 effectively penetrate the brain after systemic administration. Radiat Oncol 2018; 13:76. [PMID: 29685176 PMCID: PMC5914052 DOI: 10.1186/s13014-018-1020-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 04/06/2018] [Indexed: 12/27/2022] Open
Abstract
Ataxia Telangiectasia and Rad3 related protein (ATR) is a central mediator of the response to DNA damage that may cause the quiescent resistance of cancer initiating cells to genotoxic radiotherapy. NVP-BEZ235 is a dual PI3K/mTOR inhibitor that also effectively targets ATR with IC50 = 21 × 10- 9 M in cells. AZD6738 does not target significantly PI3K/mTOR-related kinases but specifically inhibits ATR with IC50 = 74 × 10- 9 M in cells. Both drugs have been proposed as radiosensitizers of different tumors including glioblastoma (GB), the most malignant brain tumor. In order to study the radiosensitizing properties of ATR inhibitors NVP-BEZ235 and AZD6738 towards GB, we have preliminarily investigated their capacity to penetrate the brain after systemic administration. Tumor-free CD-1 mice were inoculated i.p. with 25 mg/Kg body weight of NVP-BEZ235 or AZD6738. 1, 2, 6 and 8 h later, blood was collected by retro-orbital bleeding after which the mice were euthanized and the brains explanted. Blood and brain samples were then extracted and NVP-BEZ235 and AZD6738 concentrations determined by High Performance Liquid Chromatography/Mass Spectrometry. We found for NVP-BEZ235 and especially for AZD6738, elevated bioavailability and effective brain penetration after intraperitoneal administration. Albeit low drug and radiation dosages were used, a trend to toxicity of NVP-BEZ235 followed by ionizing radiation (IR) towards mice bearing primary glioma initiating cells (GIC)-driven orthotopic tumors was yet observed, as compared to AZD6738 + IR and vehicle+IR. Survival was never improved with median values of 99, 86 and 101 days for vehicle+IR, NVP-BEZ235 + IR and AZD6738 + IR-treated mice, respectively. Although the present results indicate favorable pharmacokinetics properties of ATR inhibitors NVP-BEZ235 and AZD6738, they do not lend support to their use as radiosensitizers of GB.
Collapse
Affiliation(s)
- Guido Fròsina
- Mutagenesis & Cancer Prevention, Ospedale Policlinico San Martino, Genoa, Italy.
| | - Aldo Profumo
- Biopolymers and Proteomics, Ospedale Policlinico San Martino, Genoa, Italy
| | - Daniela Marubbi
- Department of Experimental Medicine (DIMES), University of Genova, Genoa, Italy
| | - Diana Marcello
- Mutagenesis & Cancer Prevention, Ospedale Policlinico San Martino, Genoa, Italy
| | - Jean Louis Ravetti
- Pathological Anatomy and Histology, Ospedale Policlinico San Martino, Genoa, Italy
| | - Antonio Daga
- Regenerative Medicine, Ospedale Policlinico San Martino, Genoa, Italy
| |
Collapse
|
8
|
Abstract
There is no standard treatment available for recurrent high-grade gliomas. This monoinstitutional retrospective analysis evaluates the differences in overall survival and progression-free survival in patients according to the timing of re-irradiation. Patients suffering from a glioblastoma who received re-irradiation for recurrence were evaluated retrospectively. The median overall survival (OS) and the median progression-free survival were compared with different treatment options and within various time periods. From January 2007 until March 2015, 41 patients suffering from recurrent high-grade gliomas received re-irradiation [median dose of 30.6 Gy (range 20-40 Gy) in median 4 Gy fractions (range 1.8-5 Gy)] in our institution after initial postoperative irradiation or combined radiochemotherapy. The OS in this population was 34 months, and the OS after recurrence (OS-R) was 13 months. After diagnosis of recurrence, patients underwent additional surgical resection after a median of 1.2 months, received a second-line systemic therapy after 2.2 months with or without re-irradiation after 5.7 months. Growth of the tumour was assessed 4.3 months after the start of re-irradiation. The OS after the second surgical resection was 12.2 months, 11.7 months after the start of the second-line systemic therapy, and 6.7 months after the start of re-irradiation. The OS-R was not significantly correlated with the start of re-irradiation after a diagnosis of recurrence or the time period after the previous surgery. At this institution, re-irradiation was performed later compared to other treatment options. However, select patients could benefit from irradiation at an earlier time point. A precise time point should still be evaluated on an individual basis due to the patient's diverse conditions.
Collapse
|
9
|
Jennewein L, Ronellenfitsch MW, Antonietti P, Ilina EI, Jung J, Stadel D, Flohr LM, Zinke J, von Renesse J, Drott U, Baumgarten P, Braczynski AK, Penski C, Burger MC, Theurillat JP, Steinbach JP, Plate KH, Dikic I, Fulda S, Brandts C, Kögel D, Behrends C, Harter PN, Mittelbronn M. Diagnostic and clinical relevance of the autophago-lysosomal network in human gliomas. Oncotarget 2018; 7:20016-32. [PMID: 26956048 PMCID: PMC4991435 DOI: 10.18632/oncotarget.7910] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 02/15/2016] [Indexed: 12/19/2022] Open
Abstract
Recently, the conserved intracellular digestion mechanism ‘autophagy’ has been considered to be involved in early tumorigenesis and its blockade proposed as an alternative treatment approach. However, there is an ongoing debate about whether blocking autophagy has positive or negative effects in tumor cells. Since there is only poor data about the clinico-pathological relevance of autophagy in gliomas in vivo, we first established a cell culture based platform for the in vivo detection of the autophago-lysosomal components. We then investigated key autophagosomal (LC3B, p62, BAG3, Beclin1) and lysosomal (CTSB, LAMP2) molecules in 350 gliomas using immunohistochemistry, immunofluorescence, immunoblotting and qPCR. Autophagy was induced pharmacologically or by altering oxygen and nutrient levels. Our results show that autophagy is enhanced in astrocytomas as compared to normal CNS tissue, but largely independent from the WHO grade and patient survival. A strong upregulation of LC3B, p62, LAMP2 and CTSB was detected in perinecrotic areas in glioblastomas suggesting micro-environmental changes as a driver of autophagy induction in gliomas. Furthermore, glucose restriction induced autophagy in a concentration-dependent manner while hypoxia or amino acid starvation had considerably lesser effects. Apoptosis and autophagy were separately induced in glioma cells both in vitro and in vivo. In conclusion, our findings indicate that autophagy in gliomas is rather driven by micro-environmental changes than by primary glioma-intrinsic features thus challenging the concept of exploitation of the autophago-lysosomal network (ALN) as a treatment approach in gliomas.
Collapse
Affiliation(s)
- Lukas Jennewein
- Neurological Institute (Edinger Institute), Goethe University, Frankfurt am Main, Germany
| | - Michael W Ronellenfitsch
- Senckenberg Institute of Neurooncology, Goethe University, Frankfurt am Main, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Patrick Antonietti
- Experimental Neurosurgery, Department of Neurosurgery, Goethe University, Frankfurt am Main, Germany
| | - Elena I Ilina
- Neurological Institute (Edinger Institute), Goethe University, Frankfurt am Main, Germany
| | - Jennifer Jung
- Institute of Biochemistry II, Goethe University, Frankfurt am Main, Germany
| | - Daniela Stadel
- Institute of Biochemistry II, Goethe University, Frankfurt am Main, Germany
| | - Lisa-Marie Flohr
- Neurological Institute (Edinger Institute), Goethe University, Frankfurt am Main, Germany
| | - Jenny Zinke
- Neurological Institute (Edinger Institute), Goethe University, Frankfurt am Main, Germany
| | - Janusz von Renesse
- Neurological Institute (Edinger Institute), Goethe University, Frankfurt am Main, Germany
| | - Ulrich Drott
- Neurological Institute (Edinger Institute), Goethe University, Frankfurt am Main, Germany
| | - Peter Baumgarten
- Neurological Institute (Edinger Institute), Goethe University, Frankfurt am Main, Germany.,Department of Neurosurgery, Goethe University, Frankfurt am Main, Germany
| | - Anne K Braczynski
- Neurological Institute (Edinger Institute), Goethe University, Frankfurt am Main, Germany
| | - Cornelia Penski
- Neurological Institute (Edinger Institute), Goethe University, Frankfurt am Main, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael C Burger
- Senckenberg Institute of Neurooncology, Goethe University, Frankfurt am Main, Germany
| | | | - Joachim P Steinbach
- Senckenberg Institute of Neurooncology, Goethe University, Frankfurt am Main, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Karl-Heinz Plate
- Neurological Institute (Edinger Institute), Goethe University, Frankfurt am Main, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ivan Dikic
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Institute of Biochemistry II, Goethe University, Frankfurt am Main, Germany
| | - Simone Fulda
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Institute for Experimental Cancer Research in Pediatrics, Goethe University, Frankfurt am Main, Germany
| | - Christian Brandts
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Medicine, Hematology/Oncology, Goethe University, Frankfurt am Main, Germany
| | - Donat Kögel
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Experimental Neurosurgery, Department of Neurosurgery, Goethe University, Frankfurt am Main, Germany
| | - Christian Behrends
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Institute of Biochemistry II, Goethe University, Frankfurt am Main, Germany
| | - Patrick N Harter
- Neurological Institute (Edinger Institute), Goethe University, Frankfurt am Main, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michel Mittelbronn
- Neurological Institute (Edinger Institute), Goethe University, Frankfurt am Main, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
10
|
Re-irradiation after gross total resection of recurrent glioblastoma : Spatial pattern of recurrence and a review of the literature as a basis for target volume definition. Strahlenther Onkol 2017; 193:897-909. [PMID: 28616821 DOI: 10.1007/s00066-017-1161-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 05/23/2017] [Indexed: 12/20/2022]
Abstract
BACKGROUND Currently, patients with gross total resection (GTR) of recurrent glioblastoma (rGBM) undergo adjuvant chemotherapy or are followed up until progression. Re-irradiation, as one of the most effective treatments in macroscopic rGBM, is withheld in this situation, as uncertainties about the pattern of re-recurrence, the target volume, and also the efficacy of early re-irradiation after GTR exist. METHODS Imaging and clinical data from 26 consecutive patients with GTR of rGBM were analyzed. The spatial pattern of recurrences was analyzed according to the RANO-HGG criteria ("response assessment in neuro-oncology criteria for high-grade gliomas"). Progression-free (PFS) and overall survival (OS) were analyzed by the Kaplan-Meier method. Furthermore, a systematic review was performed in PubMed. RESULTS All but 4 patients underwent adjuvant chemotherapy after GTR. Progression was diagnosed in 20 of 26 patients and 70% of recurrent tumors occurred adjacent to the resection cavity. The median extension beyond the edge of the resection cavity was 20 mm. Median PFS was 6 months; OS was 12.8 months. We propose a target volume containing the resection cavity and every contrast enhancing lesion as the gross tumor volume (GTV), a spherical margin of 5-10 mm to generate the clinical target volume (CTV), and a margin of 1-3 mm to generate the planning target volume (PTV). Re-irradiation of this volume is deemed to be safe and likely to prolong PFS. CONCLUSION Re-irradiation is worth considering also after GTR, as the volumes that need to be treated are limited and re-irradiation has already proven to be a safe treatment option in general. The strategy of early re-irradiation is currently being tested within the GlioCave/NOA 17/Aro 2016/03 trial.
Collapse
|
11
|
Feng E, Sui C, Wang T, Sun G. Temozolomide with or without Radiotherapy in Patients with Newly Diagnosed Glioblastoma Multiforme: A Meta-Analysis. Eur Neurol 2017; 77:201-210. [PMID: 28192785 DOI: 10.1159/000455842] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 01/04/2017] [Indexed: 01/05/2023]
Abstract
BACKGROUND/AIM The current meta-analysis evaluated the survival outcomes of newly diagnosed glioblastoma patients treated with radiotherapy (RT) alone and with RT + temozolomide (TMZ). METHODS Relevant studies were identified by an extensive literature search in Medline, Current Contents and Cochrane databases by 2 independent reviewers using the terms "glioblastoma multiforme/glioblastoma, TMZ, radiation therapy/RT and survival." RESULTS Results revealed a median survival of 13.41-19 months in the combined treatment group, as opposed to 7.7-17.1 months in the RT-alone group. Progression-free survival (PFS) was also significantly different between the 2 groups (RT + TMZ, 6.3-13 months; RT-alone, 5-7.6 months). While there was no significant difference in the 6-month survival and 6-month PFS rates between the RT + TMZ and RT groups (pooled OR 0.690; p = 0.057 and OR 0.429, p = 0.052, respectively), the 1-year survival and 1-year PFS rates showed significant difference (OR 0.469; p = 0.030 and OR 0.245, p < 0.001, respectively). CONCLUSIONS Concomitant RT + TMZ is more effective and improves the overall survival and PFS in patients with newly diagnosed glioblastoma.
Collapse
Affiliation(s)
- Enshan Feng
- Department of Neurosurgery, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | | | | | | |
Collapse
|
12
|
Mattoli MV, Massaccesi M, Castelluccia A, Scolozzi V, Mantini G, Calcagni ML. The predictive value of 18F-FDG PET-CT for assessing the clinical outcomes in locally advanced NSCLC patients after a new induction treatment: low-dose fractionated radiotherapy with concurrent chemotherapy. Radiat Oncol 2017; 12:4. [PMID: 28057034 PMCID: PMC5217210 DOI: 10.1186/s13014-016-0737-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 12/06/2016] [Indexed: 12/25/2022] Open
Abstract
Background Patients with locally advanced non-small-cell lung cancer (LA-NSCLC) have poor prognosis despite several multimodal approaches. Recently, low-dose fractionated radiotherapy concurrent to the induction chemotherapy (IC-LDRT) has been proposed to further improve the effects of chemotherapy and prognosis. Until now, the predictive value of metabolic response after IC-LDRT has not yet been investigated. Aim: to evaluate whether the early metabolic response, assessed by 18F-fluoro-deoxyglucose positron emission-computed tomography (18F-FDG PET-CT), could predict the prognosis in LA-NSCLC patients treated with a multimodal approach, including IC-LDRT. Methods Forty-four consecutive patients (35males, mean age: 66 ± 7.8 years) with stage IIIA/IIIB NSCLC were retrospectively evaluated. Forty-four patients underwent IC-LDRT (2 cycles of chemotherapy, 40 cGy twice daily), 26/44 neo-adjuvant chemo-radiotherapy (CCRT: 50.4Gy), and 20/44 surgery. 18F-FDG PET-CT was performed before (baseline), after IC-LDRT (early) and after CCRT (final), applying PET response criteria in solid tumours (PERCIST). Patients with complete/partial metabolic response were classified as responders; patients with stable/progressive disease as non-responders. Progression free survival (PFS) and overall survival (OS) were assessed using Kaplan-Meyer analysis; the relationship between clinical factors and survivals were assessed using uni-multivariate regression analysis. Results Forty-four out of 44, 42/44 and 23/42 patients underwent baseline, early and final PET-CT, respectively. SULpeak of primary tumour and lymph-node significantly (p = 0.004, p = 0.0002, respectively) decreased after IC-LDRT with a further reduction after CCRT (p = 0.0006, p = 0.02, respectively). At early PET-CT, 20/42 (47.6%) patients were classified as responders, 22/42 (52.3%) as non-responders. At final PET-CT, 19/23 patients were classified as responders (12 responders and 7 non-responders at early PET-CT), and 4/23 as non-responders (all non-responders at early PET-CT). Early responders had better PFS and OS than early non-responders (p ≤ 0.01). Early metabolic response was predictive factor for loco-regional, distant and global PFS (p = 0.02, p = 0.01, p = 0.005, respectively); surgery for loco-regional and global PFS (p = 0.03, p = 0.009, respectively). Conclusions In LA-NSCLC patients, 18F-FDG metabolic response assessed after only two cycles of IC-LDRT predicts the prognosis. The early evaluation of metabolic changes could allow to personalize therapy. This multimodality approach, including both low-dose radiotherapy that increases the effects of induction chemotherapy, and surgery that removes the disease, improved clinical outcomes. Further prospective investigation of this new induction approach is warranted.
Collapse
Affiliation(s)
- Maria Vittoria Mattoli
- Institute of Nuclear Medicine, Fondazione Policlinico Universitario Agostino Gemelli, Università Cattolica del Sacro Cuore, Largo A. Gemelli, 8, 00168, Rome, Italy
| | - Mariangela Massaccesi
- Department of Radiation Oncology, Fondazione Policlinico Universitario Agostino Gemelli, Università Cattolica del Sacro Cuore, Largo A. Gemelli, 8, 00168, Rome, Italy
| | - Alessandra Castelluccia
- Department of Radiation Oncology, Fondazione Policlinico Universitario Agostino Gemelli, Università Cattolica del Sacro Cuore, Largo A. Gemelli, 8, 00168, Rome, Italy.
| | - Valentina Scolozzi
- Institute of Nuclear Medicine, Fondazione Policlinico Universitario Agostino Gemelli, Università Cattolica del Sacro Cuore, Largo A. Gemelli, 8, 00168, Rome, Italy
| | - Giovanna Mantini
- Department of Radiation Oncology, Fondazione Policlinico Universitario Agostino Gemelli, Università Cattolica del Sacro Cuore, Largo A. Gemelli, 8, 00168, Rome, Italy
| | - Maria Lucia Calcagni
- Institute of Nuclear Medicine, Fondazione Policlinico Universitario Agostino Gemelli, Università Cattolica del Sacro Cuore, Largo A. Gemelli, 8, 00168, Rome, Italy
| |
Collapse
|
13
|
Morganti AG, Cellini F, Mignogna S, Padula GDA, Caravatta L, Deodato F, Picardi V, Macchia G, Cilla S, Buwenge M, Lullo LDI, Gambacorta MA, Balducci M, Mattiucci GC, Autorino R, Valentini V. Low-dose radiotherapy and concurrent FOLFIRI-bevacizumab: a Phase II study. Future Oncol 2016; 12:779-87. [DOI: 10.2217/fon.15.350] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Aims: Low-dose radiation therapy (LDRT) can increase biological efficacy of chemotherapy. This Phase II trial evaluates LDRT plus FOLFIRI-bevacizumab (FOLFIRI-B) in metastatic colorectal cancer. Materials & methods: Primary objective: raising the clinical complete response rate from 5 to 25%. Secondary objectives: toxicity, progression-free survival. Patients underwent 12 FOLFIRI-B cycles plus two daily LDRT fractions (20 cGy/6 h interval) on each cycle. Statistical analysis was planned on 18 patients. Results: Results on 18 patients are reported. Specifically considering irradiated sites: 15/18 patients had a partial (11/18) or complete (4/18) response. Among 11 partial responders, three became a pathological CR after surgery. Grade 3–4 toxicity was recorded in two patients (11.1%). At median follow-up of 30 months (range: 8-50), 7/18 patients progressed in irradiated sites. Conclusion: Seven out of 18 patients (38.9%) had clinical or pathological CR in lesions treated with LDRT. Further studies on this newer treatment modality seem justified.
Collapse
Affiliation(s)
- Alessio Giuseppe Morganti
- Radiation Oncology Unit, Department of Experimental, Diagnostic & Specialty Medicine – DIMES, University of Bologna, S Orsola-Malpighi Hospital, Bologna, Italy
| | - Francesco Cellini
- Radiation Oncology Department – Gemelli ART, Policlinico Universitario ‘A Gemelli’, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Samantha Mignogna
- General Oncology Unit – Fondazione di Ricerca e Cura ‘Giovanni Paolo II’, Campobasso, Italy
| | - Gilbert DA Padula
- Cancer Research Consortium of West Michigan (CRCWM), Michigan State University, MI, USA
| | - Luciana Caravatta
- Radiation Oncology Unit – Fondazione di Ricerca e Cura ‘Giovanni Paolo II’, Campobasso, Italy
| | - Francesco Deodato
- Radiation Oncology Unit – Fondazione di Ricerca e Cura ‘Giovanni Paolo II’, Campobasso, Italy
| | - Vincenzo Picardi
- Radiation Oncology Unit – Fondazione di Ricerca e Cura ‘Giovanni Paolo II’, Campobasso, Italy
| | - Gabriella Macchia
- Radiation Oncology Unit – Fondazione di Ricerca e Cura ‘Giovanni Paolo II’, Campobasso, Italy
| | - Savino Cilla
- Medical Physics Unit – Fondazione di Ricerca e Cura ‘Giovanni Paolo II’, Campobasso, Italy
| | - Milly Buwenge
- Radiation Oncology Unit – Fondazione di Ricerca e Cura ‘Giovanni Paolo II’, Campobasso, Italy
| | | | - Maria Antonietta Gambacorta
- Radiation Oncology Department – Gemelli ART, Policlinico Universitario ‘A Gemelli’, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Mario Balducci
- Radiation Oncology Department – Gemelli ART, Policlinico Universitario ‘A Gemelli’, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Gian Carlo Mattiucci
- Radiation Oncology Department – Gemelli ART, Policlinico Universitario ‘A Gemelli’, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Rosa Autorino
- Radiation Oncology Department – Gemelli ART, Policlinico Universitario ‘A Gemelli’, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Vincenzo Valentini
- Radiation Oncology Department – Gemelli ART, Policlinico Universitario ‘A Gemelli’, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
14
|
Steffens R, Semrau S, Lahmer G, Putz F, Lettmaier S, Eyüpoglu I, Buchfelder M, Fietkau R. Recurrent glioblastoma: who receives tumor specific treatment and how often? J Neurooncol 2016; 128:85-92. [PMID: 26907492 DOI: 10.1007/s11060-016-2079-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 02/15/2016] [Indexed: 02/08/2023]
Abstract
The recurrence of glioblastoma (rGBM) is inevitable and often short-term. Therefore, information on the prognosis and effectiveness of tumor-specific versus purely palliative approaches should be more in-depth than a mere list of available treatment options for patients in this situation. However, follow-up data on the course of the disease in unselected patient populations after completion of primary treatment are scarce. This single-center analysis investigated the rate and number of glioblastoma recurrences after initial radiotherapy in 189 consecutive GM patients, focusing on the incidence of early death and the frequency of tumor-specific treatment (TST) versus best-supportive care (BSC) as well as the outcomes for the different approaches. In 61 % of initial population first recurrence (rGBM) could be determined by histology or imaging. 47 % received TST. 58 % of the patients with rGBM and TST were diagnosed with a second recurrence. Up to five recurrences were treated. 35-45 % of patients died before undergoing imaging studies to confirm the next recurrence. Multivariate analysis identified male sex and KPS score as independent factors (p < 0.01) for the choice of TST over BSC. Median overall survival from the diagnosis of first recurrence was 267 days in the TST group versus 65 days in patients receiving BSC (p < 0.0001). Nearly half of all rGBM patients received second-line TST, but a remarkably high proportion died early. Gender and KPS played a role in the choice of TST over BSC for recurrence treatment.
Collapse
Affiliation(s)
- Rieke Steffens
- Department of Radiation Oncology, Friedrich Alexander University of Erlangen-Nuremberg, Universitätsstr. 27, 91054, Erlangen, Germany
| | - Sabine Semrau
- Department of Radiation Oncology, Friedrich Alexander University of Erlangen-Nuremberg, Universitätsstr. 27, 91054, Erlangen, Germany.
| | - Godehard Lahmer
- Department of Radiation Oncology, Friedrich Alexander University of Erlangen-Nuremberg, Universitätsstr. 27, 91054, Erlangen, Germany
| | - Florian Putz
- Department of Radiation Oncology, Friedrich Alexander University of Erlangen-Nuremberg, Universitätsstr. 27, 91054, Erlangen, Germany
| | - Sebastian Lettmaier
- Department of Radiation Oncology, Friedrich Alexander University of Erlangen-Nuremberg, Universitätsstr. 27, 91054, Erlangen, Germany
| | - Ilker Eyüpoglu
- Department of Neurosurgery, University Hospital Erlangen, Schwabachanlage 6, Erlangen, 91054, Germany
| | - Michael Buchfelder
- Department of Neurosurgery, University Hospital Erlangen, Schwabachanlage 6, Erlangen, 91054, Germany
| | - Rainer Fietkau
- Department of Radiation Oncology, Friedrich Alexander University of Erlangen-Nuremberg, Universitätsstr. 27, 91054, Erlangen, Germany
| |
Collapse
|
15
|
Muth C, Rubner Y, Semrau S, Rühle PF, Frey B, Strnad A, Buslei R, Fietkau R, Gaipl US. Primary glioblastoma multiforme tumors and recurrence : Comparative analysis of the danger signals HMGB1, HSP70, and calreticulin. Strahlenther Onkol 2015; 192:146-55. [PMID: 26646311 DOI: 10.1007/s00066-015-0926-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 11/11/2015] [Indexed: 11/24/2022]
Abstract
PURPOSE Glioblastoma multiforme (GBM) is the most common and aggressive brain tumor. Despite improved multimodal therapies, the tumor recurs in most cases. Diverging patient survival suggests great tumor heterogeneity and different therapy responses. Danger signals such as high-mobility group box protein 1 (HMGB1), heat shock protein 70 (HSP70), and calreticulin (CRT) are biomarker candidates, due to their association with tumor progression versus induction of antitumor immune responses. Overexpression of these danger signals has been reported for various types of tumors; however, their role in GBM is still elusive. A direct comparison of their expression in the primary tumor versus the corresponding relapse is still lacking for most tumor entities. PATIENTS AND METHODS We therefore performed an expression analysis by immunohistochemistry of the danger signals HMGB1, HSP70, and CRT in primary tumors and the corresponding relapses of 9 patients with de novo GBM. RESULTS HMGB1 was highly expressed in primary tumors with a significant reduction in the respective relapse. The extracellular HSP70 expression was significantly increased in the relapse compared to the primary tumor. CRT was generally highly expressed in the primary tumor, with a slight increase in the relapse. CONCLUSION The combination of a decreased expression of HMGB1, an increased expression of extracellular HSP70, and an increased expression of CRT in the relapse seems to be beneficial for patient survival. HMGB1, extracellular HSP70, and CRT could be taken into concerted consideration as potential biomarkers for the prognosis of patients with GBM.
Collapse
Affiliation(s)
- Carolin Muth
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Yvonne Rubner
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Sabine Semrau
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Paul-Friedrich Rühle
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Benjamin Frey
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Annedore Strnad
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Rolf Buslei
- Department of Neuropathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Rainer Fietkau
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Udo S Gaipl
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
16
|
Green AL, Ramkissoon SH, McCauley D, Jones K, Perry JA, Hsu JHR, Ramkissoon LA, Maire CL, Hubbell-Engler B, Knoff DS, Shacham S, Ligon KL, Kung AL. Preclinical antitumor efficacy of selective exportin 1 inhibitors in glioblastoma. Neuro Oncol 2015; 17:697-707. [PMID: 25366336 PMCID: PMC4482855 DOI: 10.1093/neuonc/nou303] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2014] [Accepted: 09/30/2014] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Glioblastoma (GBM) is poorly responsive to current chemotherapy. The nuclear transporter exportin 1 (XPO1, CRM1) is often highly expressed in GBM, which may portend a poor prognosis. Here, we determine the efficacy of novel selective inhibitors of nuclear export (SINE) specific to XPO1 in preclinical models of GBM. METHODS Seven patient-derived GBM lines were treated with 3 SINE compounds (KPT-251, KPT-276, and Selinexor) in neurosphere culture conditions. KPT-276 and Selinexor were also evaluated in a murine orthotopic patient-derived xenograft (PDX) model of GBM. Cell cycle effects were assayed by flow cytometry in vitro and immunohistochemistry in vivo. Apoptosis was determined by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) and caspase 3/7 activity assays. RESULTS Treatment of GBM neurosphere cultures with KPT-276, Selinexor, and KPT-251 revealed dose-responsive growth inhibition in all 7 GBM lines [range of half-maximal inhibitory concentration (IC50), 6-354 nM]. In an orthotopic PDX model, treatment with KPT-276 and Selinexor demonstrated pharmacodynamic efficacy, significantly suppressed tumor growth, and prolonged animal survival. Cellular proliferation was not altered with SINE treatment. Instead, induction of apoptosis was apparent both in vitro and in vivo with SINE treatment, without overt evidence of neurotoxicity. CONCLUSIONS SINE compounds show preclinical efficacy utilizing in vitro and in vivo models of GBM, with induction of apoptosis as the mechanism of action. Selinexor is now in early clinical trials in solid and hematological malignancies. Based on these preclinical data and excellent brain penetration, we have initiated clinical trials of Selinexor in patients with relapsed GBM.
Collapse
Affiliation(s)
- Adam L Green
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (A.L.G., J.A.P., J.H.-R.H., B.H.-E.); Division of Hematology-Oncology, Boston Children's Hospital, Boston, Massachusetts (A.L.G.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (S.H.R., L.A.R., C.L.M., D.S.K., K.L.L.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.H.R., K.L.L.); Karyopharm Therapeutics, Natick, Massachusetts (D.M., S.S.); Lurie Family Imaging Center, Dana-Farber Cancer Institute, Boston, Massachusetts (K.J.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (K.L.L.); Department of Pediatrics, Columbia University Medical Center, New York, New York (A.L.K.)
| | - Shakti H Ramkissoon
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (A.L.G., J.A.P., J.H.-R.H., B.H.-E.); Division of Hematology-Oncology, Boston Children's Hospital, Boston, Massachusetts (A.L.G.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (S.H.R., L.A.R., C.L.M., D.S.K., K.L.L.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.H.R., K.L.L.); Karyopharm Therapeutics, Natick, Massachusetts (D.M., S.S.); Lurie Family Imaging Center, Dana-Farber Cancer Institute, Boston, Massachusetts (K.J.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (K.L.L.); Department of Pediatrics, Columbia University Medical Center, New York, New York (A.L.K.)
| | - Dilara McCauley
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (A.L.G., J.A.P., J.H.-R.H., B.H.-E.); Division of Hematology-Oncology, Boston Children's Hospital, Boston, Massachusetts (A.L.G.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (S.H.R., L.A.R., C.L.M., D.S.K., K.L.L.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.H.R., K.L.L.); Karyopharm Therapeutics, Natick, Massachusetts (D.M., S.S.); Lurie Family Imaging Center, Dana-Farber Cancer Institute, Boston, Massachusetts (K.J.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (K.L.L.); Department of Pediatrics, Columbia University Medical Center, New York, New York (A.L.K.)
| | - Kristen Jones
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (A.L.G., J.A.P., J.H.-R.H., B.H.-E.); Division of Hematology-Oncology, Boston Children's Hospital, Boston, Massachusetts (A.L.G.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (S.H.R., L.A.R., C.L.M., D.S.K., K.L.L.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.H.R., K.L.L.); Karyopharm Therapeutics, Natick, Massachusetts (D.M., S.S.); Lurie Family Imaging Center, Dana-Farber Cancer Institute, Boston, Massachusetts (K.J.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (K.L.L.); Department of Pediatrics, Columbia University Medical Center, New York, New York (A.L.K.)
| | - Jennifer A Perry
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (A.L.G., J.A.P., J.H.-R.H., B.H.-E.); Division of Hematology-Oncology, Boston Children's Hospital, Boston, Massachusetts (A.L.G.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (S.H.R., L.A.R., C.L.M., D.S.K., K.L.L.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.H.R., K.L.L.); Karyopharm Therapeutics, Natick, Massachusetts (D.M., S.S.); Lurie Family Imaging Center, Dana-Farber Cancer Institute, Boston, Massachusetts (K.J.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (K.L.L.); Department of Pediatrics, Columbia University Medical Center, New York, New York (A.L.K.)
| | - Jessie Hao-Ru Hsu
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (A.L.G., J.A.P., J.H.-R.H., B.H.-E.); Division of Hematology-Oncology, Boston Children's Hospital, Boston, Massachusetts (A.L.G.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (S.H.R., L.A.R., C.L.M., D.S.K., K.L.L.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.H.R., K.L.L.); Karyopharm Therapeutics, Natick, Massachusetts (D.M., S.S.); Lurie Family Imaging Center, Dana-Farber Cancer Institute, Boston, Massachusetts (K.J.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (K.L.L.); Department of Pediatrics, Columbia University Medical Center, New York, New York (A.L.K.)
| | - Lori A Ramkissoon
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (A.L.G., J.A.P., J.H.-R.H., B.H.-E.); Division of Hematology-Oncology, Boston Children's Hospital, Boston, Massachusetts (A.L.G.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (S.H.R., L.A.R., C.L.M., D.S.K., K.L.L.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.H.R., K.L.L.); Karyopharm Therapeutics, Natick, Massachusetts (D.M., S.S.); Lurie Family Imaging Center, Dana-Farber Cancer Institute, Boston, Massachusetts (K.J.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (K.L.L.); Department of Pediatrics, Columbia University Medical Center, New York, New York (A.L.K.)
| | - Cecile L Maire
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (A.L.G., J.A.P., J.H.-R.H., B.H.-E.); Division of Hematology-Oncology, Boston Children's Hospital, Boston, Massachusetts (A.L.G.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (S.H.R., L.A.R., C.L.M., D.S.K., K.L.L.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.H.R., K.L.L.); Karyopharm Therapeutics, Natick, Massachusetts (D.M., S.S.); Lurie Family Imaging Center, Dana-Farber Cancer Institute, Boston, Massachusetts (K.J.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (K.L.L.); Department of Pediatrics, Columbia University Medical Center, New York, New York (A.L.K.)
| | - Benjamin Hubbell-Engler
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (A.L.G., J.A.P., J.H.-R.H., B.H.-E.); Division of Hematology-Oncology, Boston Children's Hospital, Boston, Massachusetts (A.L.G.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (S.H.R., L.A.R., C.L.M., D.S.K., K.L.L.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.H.R., K.L.L.); Karyopharm Therapeutics, Natick, Massachusetts (D.M., S.S.); Lurie Family Imaging Center, Dana-Farber Cancer Institute, Boston, Massachusetts (K.J.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (K.L.L.); Department of Pediatrics, Columbia University Medical Center, New York, New York (A.L.K.)
| | - David S Knoff
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (A.L.G., J.A.P., J.H.-R.H., B.H.-E.); Division of Hematology-Oncology, Boston Children's Hospital, Boston, Massachusetts (A.L.G.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (S.H.R., L.A.R., C.L.M., D.S.K., K.L.L.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.H.R., K.L.L.); Karyopharm Therapeutics, Natick, Massachusetts (D.M., S.S.); Lurie Family Imaging Center, Dana-Farber Cancer Institute, Boston, Massachusetts (K.J.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (K.L.L.); Department of Pediatrics, Columbia University Medical Center, New York, New York (A.L.K.)
| | - Sharon Shacham
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (A.L.G., J.A.P., J.H.-R.H., B.H.-E.); Division of Hematology-Oncology, Boston Children's Hospital, Boston, Massachusetts (A.L.G.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (S.H.R., L.A.R., C.L.M., D.S.K., K.L.L.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.H.R., K.L.L.); Karyopharm Therapeutics, Natick, Massachusetts (D.M., S.S.); Lurie Family Imaging Center, Dana-Farber Cancer Institute, Boston, Massachusetts (K.J.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (K.L.L.); Department of Pediatrics, Columbia University Medical Center, New York, New York (A.L.K.)
| | - Keith L Ligon
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (A.L.G., J.A.P., J.H.-R.H., B.H.-E.); Division of Hematology-Oncology, Boston Children's Hospital, Boston, Massachusetts (A.L.G.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (S.H.R., L.A.R., C.L.M., D.S.K., K.L.L.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.H.R., K.L.L.); Karyopharm Therapeutics, Natick, Massachusetts (D.M., S.S.); Lurie Family Imaging Center, Dana-Farber Cancer Institute, Boston, Massachusetts (K.J.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (K.L.L.); Department of Pediatrics, Columbia University Medical Center, New York, New York (A.L.K.)
| | - Andrew L Kung
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (A.L.G., J.A.P., J.H.-R.H., B.H.-E.); Division of Hematology-Oncology, Boston Children's Hospital, Boston, Massachusetts (A.L.G.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (S.H.R., L.A.R., C.L.M., D.S.K., K.L.L.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.H.R., K.L.L.); Karyopharm Therapeutics, Natick, Massachusetts (D.M., S.S.); Lurie Family Imaging Center, Dana-Farber Cancer Institute, Boston, Massachusetts (K.J.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (K.L.L.); Department of Pediatrics, Columbia University Medical Center, New York, New York (A.L.K.)
| |
Collapse
|