search
Back to results

A Clinical Study to Improve Brain Function and Quality of Life of Patients With Newly Diagnosed Brain Tumors (Gliomas). (ImproveCodel)

Primary Purpose

Oligodendroglioma

Status
Recruiting
Phase
Phase 3
Locations
Germany
Study Type
Interventional
Intervention
CETEG
PCV
RT
Sponsored by
University Hospital Heidelberg
About
Eligibility
Locations
Arms
Outcomes
Full info

About this trial

This is an interventional treatment trial for Oligodendroglioma focused on measuring Radiotherapy, Chemotherapy, Temozolomide

Eligibility Criteria

18 Years - undefined (Adult, Older Adult)All SexesDoes not accept healthy volunteers

Inclusion Criteria:

  • Histologically confirmed, newly diagnosed WHO grade II or III glioma.
  • Tumor carries an isocitrate dehydrogenase (IDH) mutation (determined by immunohistochemistry (IHC) and/or deoxyribonucleic acid (DNA) sequencing).
  • Tumor is co-deleted for 1p/19q (determined by copy number variations, fluorescence in situ hybridization (FISH), multiplex ligation-dependent probe amplification (MLPA) or other appropriate methods).
  • Open biopsy or resection.
  • Age ≥18 years.
  • Karnofsky Performance Index (KPI) ≥60%.
  • Life expectancy > 6 months.
  • Availability of formalin-fixed paraffin-embedded (FFPE) or fresh-frozen tissue and ethylenediamine tetraacetic acid (EDTA) blood for biomarker research.
  • Standard magnetic resonance imaging (MRI) ≤ 72 post-surgery according to the present national and international guidelines.
  • Craniotomy or intracranial biopsy site must be adequately healed.
  • ≥ 2 weeks and ≤ 3 months from surgery without any interim radio- or chemotherapy or experimental intervention.
  • Willing and able to comply with regular neurocognitive and health-related quality of life tests/questionnaires.
  • Indication for postsurgical cytostatic/-toxic therapy.
  • Written Informed consent.
  • Female patients with reproductive potential have a negative pregnancy test (serum or urine) within 6 days prior to start of therapy. Female patients are surgically sterile or agree to use adequate contraception during the period of therapy and 6 months after the end of study treatment, or women have been postmenopausal for at least 2 years.
  • Male patients are willing to use contraception.

Exclusion Criteria:

  • Participation in other ongoing interventional clinical trials.
  • Insufficient tumor material for molecular diagnostics.
  • Inability to undergo MRI.
  • Abnormal (≥ Grade 2 CTCAE v5.0) laboratory values for hematology (Hb, WBC (White Blood Cell), neutrophils, or platelets), liver (serum bilirubin, ALT (Alanine Amino Transferase), or AST (Aspartate Amino Transferase)) or renal function (serum creatinine).
  • Active tuberculosis; known HIV infection or active Hepatitis B (HBV) or Hepatitis C (HCV infection, or active infections requiring oral or intravenous antibiotics or that can cause a severe disease and pose a severe danger to lab personnel working on patients' blood or tissue (e.g. rabies).
  • Any prior anti-cancer therapy or co-administration of anti-cancer therapies other than those administered/allowed in this study. History of low-grade glioma that did not require prior treatment with chemotherapy or radiotherapy is not an exclusion criterion.
  • Immunosuppression not related to prior treatment for malignancy.
  • History of other malignancies (except for adequately treated basal or squamous cell carcinoma or carcinoma in situ) within the last 5 years unless the patient has been disease-free for 5 years.
  • Any clinically significant concomitant disease (including hereditary fructose intolerance) or condition that could interfere with, or for which the treatment might interfere with, the conduct of the study or the absorption of oral medications or that would, in the opinion of the Principal Investigator, pose an unacceptable risk to the patient in this study.
  • Any psychological, familial, sociological, or geographical condition potentially hampering compliance with the study protocol requirements and/or follow-up procedures; those conditions should be discussed with the patient before trial entry.
  • Pregnancy or breastfeeding.
  • History of hypersensitivity to the investigational medicinal product or to any drug with similar chemical structure or to any excipient present in the pharmaceutical form of the investigational medicinal product.
  • QTc (corrected QT interval) time prolongation > 500 ms.
  • Patients under restricted medication for procarbazine, lomustine, vincristine and temozolomide.
  • Liver disease characterized by:

    • ALT or AST (≥ Grade 2 CTCAE v5.0) confirmed on two consecutive measurements OR
    • Impaired excretory function (e.g., hyperbilirubinemia) or synthetic function or other conditions of decompensated liver disease such as coagulopathy, hepatic encephalopathy, hypoalbuminemia, ascites, and bleeding from esophageal varices (≥ Grade 2 CTCAE v5.0) OR
    • Acute viral or active autoimmune, alcoholic, or other types of acute hepatitis
  • Known uncorrected coagulopathy, platelet disorder, or history of non-drug induced thrombocytopenia.
  • History of autoimmune disease, including but not limited to myasthenia gravis, myositis, autoimmune hepatitis, systemic lupus erythematosus, rheumatoid arthritis, inflammatory bowel disease, vascular thrombosis associated with antiphospholipid syndrome, Wegener's granulomatosis, Sjogren's syndrome, Guillain-Barre syndrome, multiple sclerosis, vasculitis, or glomerulonephritis; autoimmune-related hypothyroidism (patients on a stable dose of thyroid replacement hormone are eligible for this study) and type I diabetes mellitus (patients on a stable dose of insulin regimen are eligible for this study).
  • Vaccination with life vaccines during treatment and 4 weeks before start of treatment.
  • Existing neuromuscular diseases, especially neural muscular atrophy with segmental demyelination (demyelinising form of Charcot-Marie-Tooth syndrome)
  • Chronic constipation and subileus
  • Combination treatment with mitomycin (risk of a pronounced bronchospasm and acute shortness of breath)
  • Hypersensitivity to dacarbazine (DTIC)

Sites / Locations

  • University Hospital Heidelberg, Department of NeurooncologyRecruiting
  • Charité, University Medicine Berlin, NeurosurgeryRecruiting
  • Knappschaftskrankenhaus Bochum GmbH, Neurology ClinicRecruiting
  • University Hospital Bonn, Neurology ClinicRecruiting
  • Chemnitz Hospital, NeurosurgeryRecruiting
  • University Hospital Cologne, NeurosurgeryRecruiting
  • University Hospital Duesseldorf, Neurooncology
  • University Hospital Frankfurt, NeurooncologyRecruiting
  • University Hospital Göttingen, NeurosurgeryRecruiting
  • University Hospital Saarland, NeurosurgeryRecruiting
  • University Hospital of Jena, NeurosurgeryRecruiting
  • University Hospital Leipzig, Radiation TherapyRecruiting
  • University Hospital Mannheim, Neurology ClinicRecruiting
  • University Clinic Muehlenkreis, MindenRecruiting
  • University Hospital rechts der Isar, Radiation OncologyRecruiting
  • University Hospital Regensburg, Neurology ClinicRecruiting
  • Helios Hospital Schwerin, NeurosurgeryRecruiting
  • University Hospital Tuebingen, NeurooncologyRecruiting
  • University Hospital Wuerzburg, NeurosurgeryRecruiting

Arms of the Study

Arm 1

Arm 2

Arm Type

Active Comparator

Experimental

Arm Label

RT PCV

CETEG

Arm Description

Radiotherapy (RT) for over approximately 5-6 weeks: at 50.4/54 Gy in 1.8 Gy fractions for grade II and at 59.4 Gy in 1.8 Gy fractions for grade III gliomas PCV cycles are 6 weeks long and given as: Day 1: Lomustine (CCNU) at 110 mg/m2 body surface orally, Days 8/29: Vincristine 1.4 mg/m2 i.v. (capped at 2 mg), Days 8-21: Procarbazine 60 mg/m2 orally (capped at 100 mg).

Six 42-day cycles of lomustine plus temozolomide according to the commonly used regimen: Day 1: Lomustine (CCNU) at 100 mg/m2 Days 2-6: Temozolomide at 100 mg/m2 (cycles 1) and in 50 mg/m2 steps to 200 mg/m2 from cycle 2 on dependent on hematological toxicity

Outcomes

Primary Outcome Measures

Qualified overall survival (qOS)
The primary efficacy endpoint is the overall survival without functional and/or cognitive and/or quality of life deterioration over a period of 90 days, coined qualified overall survival (qOS). Short-term qOS is defined as the time from randomization to a deterioration in any of the following measures: NeuroCogFX®, KPI, HRQoL, NANO scale or death.
Short-term qOS deterioration in NeuroCogFX®
A decline is a reduction in the mean percentile rank in 2 or more items (Fliessbach et al. 2010, Hoffermann et al. 2017) in two or more subset scores of established neuropsychometric test batteries determined by NeuroCogFX® (Fliessbach et al. 2010).
Short-term qOS deterioration in KPI
A decrease in the KPI from 100 or 90 to 70 or less, a decrease in KPI of at least 20 from 80 or less, or a decrease in KPS from any baseline to 50 or less. Fulfilment of one of these criteria is considered neurological deterioration unless attributable to comorbid events or changes in corticosteroid dose (van den Bent et al. 2011).
Short-term qOS deterioration in HrQoL
A worsening of at least 10 points, which is the minimal clinically relevant difference, in at least one of the five selected domains of the HrQoL (global health status (GHS), physical functioning (PF), social functioning (SF), determined in the QLQ-C30 with higher scores indicate better HRQoL; communication deficits (CD) & motor dysfunction (MD) determined by QLQ-BN20 with lower scores indicate better HRQoL) (Taphoorn et al. 2015).
Short-term qOS deterioration in NANO scale
A decline in the NANO scale defined as a ≥2 level worsening from baseline within ≥1 domain or worsening to the highest score within ≥1 domain that is felt to be related to underlying tumor progression and not attributable to a comorbid event or change in concurrent medication (Nayak et al. 2017).
Short-term qOS deterioration due to death
Death due to any cause.

Secondary Outcome Measures

Short-term qOS
Defined as qOS as described above (deterioration in NeuroCogFX®, KPI, HRQoL, NANO scale or death) but neglecting the subsequent time interval of 3 months (90 days).
Overall survival (OS)
Defined as the time from randomization until death due to any cause.
Progression-free survival (PFS)
Defined as the time from randomization to the day of first documentation of clinical or radiographic tumor progression or death of any cause.

Full Information

First Posted
March 15, 2022
Last Updated
September 22, 2022
Sponsor
University Hospital Heidelberg
Collaborators
Universitätsmedizin Mannheim, Ruhr University of Bochum
search

1. Study Identification

Unique Protocol Identification Number
NCT05331521
Brief Title
A Clinical Study to Improve Brain Function and Quality of Life of Patients With Newly Diagnosed Brain Tumors (Gliomas).
Acronym
ImproveCodel
Official Title
Improvement of Functional Outcome for Patients With Newly Diagnosed Grade II or III Glioma With Co-deletion of 1p/19q - IMPROVE CODEL: the NOA-18 Trial
Study Type
Interventional

2. Study Status

Record Verification Date
September 2022
Overall Recruitment Status
Recruiting
Study Start Date
April 7, 2021 (Actual)
Primary Completion Date
March 31, 2029 (Anticipated)
Study Completion Date
March 31, 2029 (Anticipated)

3. Sponsor/Collaborators

Responsible Party, by Official Title
Principal Investigator
Name of the Sponsor
University Hospital Heidelberg
Collaborators
Universitätsmedizin Mannheim, Ruhr University of Bochum

4. Oversight

Studies a U.S. FDA-regulated Drug Product
No
Studies a U.S. FDA-regulated Device Product
No
Data Monitoring Committee
Yes

5. Study Description

Brief Summary
Oligodendrogliomas in the novel edition of the Central Nervous System (CNS) World Health Organization (WHO) classification are now molecularly defined by isocitrate dehydrogenase (IDH)1 or IDH2 mutations and 1p/19q co-deletion. The prognosis of these molecularly defined tumors is to be determined in new series since survival data from older histology-based studies and population-based registries are confounded by the inclusion of 20-70% not molecularly matching subsets. Also, the optimal treatment is a matter of ongoing investigations. An extensive, but safe surgery is associated with improved outcome as is the addition of chemotherapy with procarbazine, CCNU (lomustine), and vincristine (PCV) to the partial brain radiotherapy (RT). However, the exact timing of postsurgical therapy especially for tumors of the WHO grade II and acknowledging some variability in grading as well as the choice of chemotherapy, temozolomide instead of PCV (CODEL: NCT00887146 randomizing CNS WHO grade 2 and 3 oligodendrogliomas to chemoradiation(CHRT)therapy with PCV or with temozolomide) or the need for primary radiotherapy RT are subjects of clinical studies (POLCA: NCT02444000 randomizing patients with newly diagnosed CNS WHO grade 3 oligodendrogliomas to standard CHRT with PCV or PCV alone). Given the young age of patients with CNS WHO grade 2 and 3 oligodendrogliomas and the relevant risk of neurocognitive, functional and quality-of-life impairment with the current aggressive standard of care treatment, chemoradiation with PCV, of the tumor located in the brain optimizing care is the major challenge. NOA-18/IMPROVE CODEL aims at improving qualified overall survival (qOS) for adult patients with CNS WHO grade 2 and 3 oligodendrogliomas by randomizing between standard chemoradiation with up to six six-weekly cycles with PCV and six six-weekly cycles with lomustine and temozolomide (CETEG), thereby delaying radiotherapy (RT) and adding the chemoradiotherapy (CHRT) concept at progression after initial radiation-free chemotherapy, allowing for an effective salvage treatment and delaying potentially deleterious side effects. QOS represents a new concept and is defined as OS without functional and/or cognitive and/or quality of life (QOL) deterioration regardless whether tumor progression or toxicity is the main cause.
Detailed Description
The primary objective of the NOA-18/IMPROVE CODEL trail is to show superiority of an initial CETEG treatment followed by partial brain radiotherapy (RT) plus PCV (RT-PCV) at progression over partial brain radiotherapy (RT) followed by procarbazine, lomustine and vincristine (PCV) chemotherapy (RT-PCV) and best investigators choice (BIC) at progression for sustained qOS. An event with respect to a sustained qOS is then defined as a functional and/or cognitive deterioration on two consecutive study visits with an interval of 3 months, tolerating a deviation of at most 1 month. Assessments are done with 3-monthly MRI, assessment of the NANO (Neurologic assessment in neuro-oncology) scale, HRQoL, and KPS (Karnofsky performance status) and annually cognitive testing. Secondary objectives are evaluation and comparison of the two groups regarding secondary endpoints (short-term qOS, PFS, OS, complete and partial response rate). The trial is planned to be conducted at 21 NOA (Neurooncology Working Party of the German Cancer Society) study sites in Germany.

6. Conditions and Keywords

Primary Disease or Condition Being Studied in the Trial, or the Focus of the Study
Oligodendroglioma
Keywords
Radiotherapy, Chemotherapy, Temozolomide

7. Study Design

Primary Purpose
Treatment
Study Phase
Phase 3
Interventional Study Model
Parallel Assignment
Masking
None (Open Label)
Allocation
Randomized
Enrollment
406 (Anticipated)

8. Arms, Groups, and Interventions

Arm Title
RT PCV
Arm Type
Active Comparator
Arm Description
Radiotherapy (RT) for over approximately 5-6 weeks: at 50.4/54 Gy in 1.8 Gy fractions for grade II and at 59.4 Gy in 1.8 Gy fractions for grade III gliomas PCV cycles are 6 weeks long and given as: Day 1: Lomustine (CCNU) at 110 mg/m2 body surface orally, Days 8/29: Vincristine 1.4 mg/m2 i.v. (capped at 2 mg), Days 8-21: Procarbazine 60 mg/m2 orally (capped at 100 mg).
Arm Title
CETEG
Arm Type
Experimental
Arm Description
Six 42-day cycles of lomustine plus temozolomide according to the commonly used regimen: Day 1: Lomustine (CCNU) at 100 mg/m2 Days 2-6: Temozolomide at 100 mg/m2 (cycles 1) and in 50 mg/m2 steps to 200 mg/m2 from cycle 2 on dependent on hematological toxicity
Intervention Type
Drug
Intervention Name(s)
CETEG
Other Intervention Name(s)
Lomustine (CCNU) and Temozolomide
Intervention Description
At progression, patients without prior radiotherapy will undergo radiotherapy and PCV as an adjunct chemotherapy if bone marrow reserve allows in the experimental arm.
Intervention Type
Drug
Intervention Name(s)
PCV
Other Intervention Name(s)
Procarbazine, Lomustine and Vincristine
Intervention Description
In the comparator arm there is the option for second radiotherapy or even reuse of the full radiochemotherapy regimen as it had been given at diagnosis (BIC).
Intervention Type
Radiation
Intervention Name(s)
RT
Other Intervention Name(s)
Radiotherapy
Intervention Description
Radiotherapy at 50.4/54 Gy in 1.8 Gy fractions for grade II and 59.4 Gy in 1.8 Gy fractions for grade III gliomas
Primary Outcome Measure Information:
Title
Qualified overall survival (qOS)
Description
The primary efficacy endpoint is the overall survival without functional and/or cognitive and/or quality of life deterioration over a period of 90 days, coined qualified overall survival (qOS). Short-term qOS is defined as the time from randomization to a deterioration in any of the following measures: NeuroCogFX®, KPI, HRQoL, NANO scale or death.
Time Frame
From 3 months after start of treatment, every 3 months until maximum of 10 years or date of death from any cause, whichever came first.
Title
Short-term qOS deterioration in NeuroCogFX®
Description
A decline is a reduction in the mean percentile rank in 2 or more items (Fliessbach et al. 2010, Hoffermann et al. 2017) in two or more subset scores of established neuropsychometric test batteries determined by NeuroCogFX® (Fliessbach et al. 2010).
Time Frame
Every 12 months, after a decline within 1 week and after 90 days
Title
Short-term qOS deterioration in KPI
Description
A decrease in the KPI from 100 or 90 to 70 or less, a decrease in KPI of at least 20 from 80 or less, or a decrease in KPS from any baseline to 50 or less. Fulfilment of one of these criteria is considered neurological deterioration unless attributable to comorbid events or changes in corticosteroid dose (van den Bent et al. 2011).
Time Frame
From 3 months after start of treatment, every 3 months until maximum of 10 years or date of death from any cause, whichever came first.
Title
Short-term qOS deterioration in HrQoL
Description
A worsening of at least 10 points, which is the minimal clinically relevant difference, in at least one of the five selected domains of the HrQoL (global health status (GHS), physical functioning (PF), social functioning (SF), determined in the QLQ-C30 with higher scores indicate better HRQoL; communication deficits (CD) & motor dysfunction (MD) determined by QLQ-BN20 with lower scores indicate better HRQoL) (Taphoorn et al. 2015).
Time Frame
From 3 months after start of treatment, every 3 months until maximum of 10 years or date of death from any cause, whichever came first.
Title
Short-term qOS deterioration in NANO scale
Description
A decline in the NANO scale defined as a ≥2 level worsening from baseline within ≥1 domain or worsening to the highest score within ≥1 domain that is felt to be related to underlying tumor progression and not attributable to a comorbid event or change in concurrent medication (Nayak et al. 2017).
Time Frame
From 3 months after start of treatment, every 3 months until maximum of 10 years or date of death from any cause, whichever came first.
Title
Short-term qOS deterioration due to death
Description
Death due to any cause.
Time Frame
From start of randomization until death from any cause
Secondary Outcome Measure Information:
Title
Short-term qOS
Description
Defined as qOS as described above (deterioration in NeuroCogFX®, KPI, HRQoL, NANO scale or death) but neglecting the subsequent time interval of 3 months (90 days).
Time Frame
From 3 months after start of treatment until maximum of 10 years or date of death from any cause, whichever came first.
Title
Overall survival (OS)
Description
Defined as the time from randomization until death due to any cause.
Time Frame
From start of randomization until death from any cause
Title
Progression-free survival (PFS)
Description
Defined as the time from randomization to the day of first documentation of clinical or radiographic tumor progression or death of any cause.
Time Frame
From randomization until the day of first documentation of clinical or radiographic tumor progression or death of any cause, whichever occurs first

10. Eligibility

Sex
All
Minimum Age & Unit of Time
18 Years
Accepts Healthy Volunteers
No
Eligibility Criteria
Inclusion Criteria: Histologically confirmed, newly diagnosed WHO grade II or III glioma. Tumor carries an isocitrate dehydrogenase (IDH) mutation (determined by immunohistochemistry (IHC) and/or deoxyribonucleic acid (DNA) sequencing). Tumor is co-deleted for 1p/19q (determined by copy number variations, fluorescence in situ hybridization (FISH), multiplex ligation-dependent probe amplification (MLPA) or other appropriate methods). Open biopsy or resection. Age ≥18 years. Karnofsky Performance Index (KPI) ≥60%. Life expectancy > 6 months. Availability of formalin-fixed paraffin-embedded (FFPE) or fresh-frozen tissue and ethylenediamine tetraacetic acid (EDTA) blood for biomarker research. Standard magnetic resonance imaging (MRI) ≤ 72 post-surgery according to the present national and international guidelines. Craniotomy or intracranial biopsy site must be adequately healed. ≥ 2 weeks and ≤ 3 months from surgery without any interim radio- or chemotherapy or experimental intervention. Willing and able to comply with regular neurocognitive and health-related quality of life tests/questionnaires. Indication for postsurgical cytostatic/-toxic therapy. Written Informed consent. Female patients with reproductive potential have a negative pregnancy test (serum or urine) within 6 days prior to start of therapy. Female patients are surgically sterile or agree to use adequate contraception during the period of therapy and 6 months after the end of study treatment, or women have been postmenopausal for at least 2 years. Male patients are willing to use contraception. Exclusion Criteria: Participation in other ongoing interventional clinical trials. Insufficient tumor material for molecular diagnostics. Inability to undergo MRI. Abnormal (≥ Grade 2 CTCAE v5.0) laboratory values for hematology (Hb, WBC (White Blood Cell), neutrophils, or platelets), liver (serum bilirubin, ALT (Alanine Amino Transferase), or AST (Aspartate Amino Transferase)) or renal function (serum creatinine). Active tuberculosis; known HIV infection or active Hepatitis B (HBV) or Hepatitis C (HCV infection, or active infections requiring oral or intravenous antibiotics or that can cause a severe disease and pose a severe danger to lab personnel working on patients' blood or tissue (e.g. rabies). Any prior anti-cancer therapy or co-administration of anti-cancer therapies other than those administered/allowed in this study. History of low-grade glioma that did not require prior treatment with chemotherapy or radiotherapy is not an exclusion criterion. Immunosuppression not related to prior treatment for malignancy. History of other malignancies (except for adequately treated basal or squamous cell carcinoma or carcinoma in situ) within the last 5 years unless the patient has been disease-free for 5 years. Any clinically significant concomitant disease (including hereditary fructose intolerance) or condition that could interfere with, or for which the treatment might interfere with, the conduct of the study or the absorption of oral medications or that would, in the opinion of the Principal Investigator, pose an unacceptable risk to the patient in this study. Any psychological, familial, sociological, or geographical condition potentially hampering compliance with the study protocol requirements and/or follow-up procedures; those conditions should be discussed with the patient before trial entry. Pregnancy or breastfeeding. History of hypersensitivity to the investigational medicinal product or to any drug with similar chemical structure or to any excipient present in the pharmaceutical form of the investigational medicinal product. QTc (corrected QT interval) time prolongation > 500 ms. Patients under restricted medication for procarbazine, lomustine, vincristine and temozolomide. Liver disease characterized by: ALT or AST (≥ Grade 2 CTCAE v5.0) confirmed on two consecutive measurements OR Impaired excretory function (e.g., hyperbilirubinemia) or synthetic function or other conditions of decompensated liver disease such as coagulopathy, hepatic encephalopathy, hypoalbuminemia, ascites, and bleeding from esophageal varices (≥ Grade 2 CTCAE v5.0) OR Acute viral or active autoimmune, alcoholic, or other types of acute hepatitis Known uncorrected coagulopathy, platelet disorder, or history of non-drug induced thrombocytopenia. History of autoimmune disease, including but not limited to myasthenia gravis, myositis, autoimmune hepatitis, systemic lupus erythematosus, rheumatoid arthritis, inflammatory bowel disease, vascular thrombosis associated with antiphospholipid syndrome, Wegener's granulomatosis, Sjogren's syndrome, Guillain-Barre syndrome, multiple sclerosis, vasculitis, or glomerulonephritis; autoimmune-related hypothyroidism (patients on a stable dose of thyroid replacement hormone are eligible for this study) and type I diabetes mellitus (patients on a stable dose of insulin regimen are eligible for this study). Vaccination with life vaccines during treatment and 4 weeks before start of treatment. Existing neuromuscular diseases, especially neural muscular atrophy with segmental demyelination (demyelinising form of Charcot-Marie-Tooth syndrome) Chronic constipation and subileus Combination treatment with mitomycin (risk of a pronounced bronchospasm and acute shortness of breath) Hypersensitivity to dacarbazine (DTIC)
Central Contact Person:
First Name & Middle Initial & Last Name or Official Title & Degree
Wolfgang Wick, Prof. Dr.
Phone
+49 6221 56
Ext
7075
Email
wolfgang.wick@med.uni-heidelberg.de
First Name & Middle Initial & Last Name or Official Title & Degree
Antje Wick, PD Dr.
Phone
+49 6221 56
Ext
7075
Email
antje.wick@med.uni-heidelberg.de
Overall Study Officials:
First Name & Middle Initial & Last Name & Degree
Wolfgang Wick, Prof. Dr.
Organizational Affiliation
University Hospital Heidelberg
Official's Role
Principal Investigator
Facility Information:
Facility Name
University Hospital Heidelberg, Department of Neurooncology
City
Heidelberg
State/Province
Baden-Württemberg
ZIP/Postal Code
69120
Country
Germany
Individual Site Status
Recruiting
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Wolfgang Wick, Prof. Dr.
Facility Name
Charité, University Medicine Berlin, Neurosurgery
City
Berlin
ZIP/Postal Code
10117
Country
Germany
Individual Site Status
Recruiting
Facility Name
Knappschaftskrankenhaus Bochum GmbH, Neurology Clinic
City
Bochum
ZIP/Postal Code
44892
Country
Germany
Individual Site Status
Recruiting
Facility Name
University Hospital Bonn, Neurology Clinic
City
Bonn
ZIP/Postal Code
53127
Country
Germany
Individual Site Status
Recruiting
Facility Name
Chemnitz Hospital, Neurosurgery
City
Chemnitz
ZIP/Postal Code
09116
Country
Germany
Individual Site Status
Recruiting
Facility Name
University Hospital Cologne, Neurosurgery
City
Cologne
ZIP/Postal Code
50937
Country
Germany
Individual Site Status
Recruiting
Facility Name
University Hospital Duesseldorf, Neurooncology
City
Duesseldorf
ZIP/Postal Code
40225
Country
Germany
Individual Site Status
Not yet recruiting
Facility Name
University Hospital Frankfurt, Neurooncology
City
Frankfurt
ZIP/Postal Code
60528
Country
Germany
Individual Site Status
Recruiting
Facility Name
University Hospital Göttingen, Neurosurgery
City
Göttingen
ZIP/Postal Code
37075
Country
Germany
Individual Site Status
Recruiting
Facility Name
University Hospital Saarland, Neurosurgery
City
Homburg
ZIP/Postal Code
66421
Country
Germany
Individual Site Status
Recruiting
Facility Name
University Hospital of Jena, Neurosurgery
City
Jena
ZIP/Postal Code
07747
Country
Germany
Individual Site Status
Recruiting
Facility Name
University Hospital Leipzig, Radiation Therapy
City
Leipzig
ZIP/Postal Code
04103
Country
Germany
Individual Site Status
Recruiting
Facility Name
University Hospital Mannheim, Neurology Clinic
City
Mannheim
ZIP/Postal Code
68167
Country
Germany
Individual Site Status
Recruiting
Facility Name
University Clinic Muehlenkreis, Minden
City
Minden
ZIP/Postal Code
32429
Country
Germany
Individual Site Status
Recruiting
Facility Name
University Hospital rechts der Isar, Radiation Oncology
City
Munich
ZIP/Postal Code
81675
Country
Germany
Individual Site Status
Recruiting
Facility Name
University Hospital Regensburg, Neurology Clinic
City
Regensburg
ZIP/Postal Code
93053
Country
Germany
Individual Site Status
Recruiting
Facility Name
Helios Hospital Schwerin, Neurosurgery
City
Schwerin
ZIP/Postal Code
19049
Country
Germany
Individual Site Status
Recruiting
Facility Name
University Hospital Tuebingen, Neurooncology
City
Tuebingen
ZIP/Postal Code
72076
Country
Germany
Individual Site Status
Recruiting
Facility Name
University Hospital Wuerzburg, Neurosurgery
City
Würzburg
ZIP/Postal Code
97080
Country
Germany
Individual Site Status
Recruiting

12. IPD Sharing Statement

Plan to Share IPD
No
Citations:
PubMed Identifier
34185076
Citation
Louis DN, Perry A, Wesseling P, Brat DJ, Cree IA, Figarella-Branger D, Hawkins C, Ng HK, Pfister SM, Reifenberger G, Soffietti R, von Deimling A, Ellison DW. The 2021 WHO Classification of Tumors of the Central Nervous System: a summary. Neuro Oncol. 2021 Aug 2;23(8):1231-1251. doi: 10.1093/neuonc/noab106.
Results Reference
background
PubMed Identifier
33293629
Citation
Weller M, van den Bent M, Preusser M, Le Rhun E, Tonn JC, Minniti G, Bendszus M, Balana C, Chinot O, Dirven L, French P, Hegi ME, Jakola AS, Platten M, Roth P, Ruda R, Short S, Smits M, Taphoorn MJB, von Deimling A, Westphal M, Soffietti R, Reifenberger G, Wick W. EANO guidelines on the diagnosis and treatment of diffuse gliomas of adulthood. Nat Rev Clin Oncol. 2021 Mar;18(3):170-186. doi: 10.1038/s41571-020-00447-z. Epub 2020 Dec 8. Erratum In: Nat Rev Clin Oncol. 2022 May;19(5):357-358.
Results Reference
background
PubMed Identifier
20449630
Citation
Fliessbach K, Rogowski S, Hoppe C, Sabel M, Goeppert M, Helmstaedter C, Calabrese P, Schackert G, Tonn JC, Simon M, Schlegel U. Computer-based assessment of cognitive functions in brain tumor patients. J Neurooncol. 2010 Dec;100(3):427-37. doi: 10.1007/s11060-010-0194-9. Epub 2010 May 7.
Results Reference
background
PubMed Identifier
27836394
Citation
Hoffermann M, Bruckmann L, Mahdy Ali K, Zaar K, Avian A, von Campe G. Pre- and postoperative neurocognitive deficits in brain tumor patients assessed by a computer based screening test. J Clin Neurosci. 2017 Feb;36:31-36. doi: 10.1016/j.jocn.2016.10.030. Epub 2016 Nov 9.
Results Reference
background
PubMed Identifier
26014298
Citation
Taphoorn MJ, Henriksson R, Bottomley A, Cloughesy T, Wick W, Mason WP, Saran F, Nishikawa R, Hilton M, Theodore-Oklota C, Ravelo A, Chinot OL. Health-Related Quality of Life in a Randomized Phase III Study of Bevacizumab, Temozolomide, and Radiotherapy in Newly Diagnosed Glioblastoma. J Clin Oncol. 2015 Jul 1;33(19):2166-75. doi: 10.1200/JCO.2014.60.3217. Epub 2015 May 26.
Results Reference
background
PubMed Identifier
28453751
Citation
Nayak L, DeAngelis LM, Brandes AA, Peereboom DM, Galanis E, Lin NU, Soffietti R, Macdonald DR, Chamberlain M, Perry J, Jaeckle K, Mehta M, Stupp R, Muzikansky A, Pentsova E, Cloughesy T, Iwamoto FM, Tonn JC, Vogelbaum MA, Wen PY, van den Bent MJ, Reardon DA. The Neurologic Assessment in Neuro-Oncology (NANO) scale: a tool to assess neurologic function for integration into the Response Assessment in Neuro-Oncology (RANO) criteria. Neuro Oncol. 2017 May 1;19(5):625-635. doi: 10.1093/neuonc/nox029.
Results Reference
background
PubMed Identifier
20231676
Citation
Wen PY, Macdonald DR, Reardon DA, Cloughesy TF, Sorensen AG, Galanis E, Degroot J, Wick W, Gilbert MR, Lassman AB, Tsien C, Mikkelsen T, Wong ET, Chamberlain MC, Stupp R, Lamborn KR, Vogelbaum MA, van den Bent MJ, Chang SM. Updated response assessment criteria for high-grade gliomas: response assessment in neuro-oncology working group. J Clin Oncol. 2010 Apr 10;28(11):1963-72. doi: 10.1200/JCO.2009.26.3541. Epub 2010 Mar 15.
Results Reference
background
PubMed Identifier
27370396
Citation
Wick W, Roth P, Hartmann C, Hau P, Nakamura M, Stockhammer F, Sabel MC, Wick A, Koeppen S, Ketter R, Vajkoczy P, Eyupoglu I, Kalff R, Pietsch T, Happold C, Galldiks N, Schmidt-Graf F, Bamberg M, Reifenberger G, Platten M, von Deimling A, Meisner C, Wiestler B, Weller M; Neurooncology Working Group (NOA) of the German Cancer Society. Long-term analysis of the NOA-04 randomized phase III trial of sequential radiochemotherapy of anaplastic glioma with PCV or temozolomide. Neuro Oncol. 2016 Nov;18(11):1529-1537. doi: 10.1093/neuonc/now133. Epub 2016 Jul 1. Erratum In: Neuro Oncol. 2016 Nov;18(11):e1.
Results Reference
background
PubMed Identifier
35692047
Citation
Wick A, Sander A, Koch M, Bendszus M, Combs S, Haut T, Dormann A, Walter S, Pertz M, Merkle-Lock J, Selkrig N, Limprecht R, Baumann L, Kieser M, Sahm F, Schlegel U, Winkler F, Platten M, Wick W, Kessler T. Improvement of functional outcome for patients with newly diagnosed grade 2 or 3 gliomas with co-deletion of 1p/19q - IMPROVE CODEL: the NOA-18 trial. BMC Cancer. 2022 Jun 13;22(1):645. doi: 10.1186/s12885-022-09720-z.
Results Reference
derived

Learn more about this trial

A Clinical Study to Improve Brain Function and Quality of Life of Patients With Newly Diagnosed Brain Tumors (Gliomas).

We'll reach out to this number within 24 hrs