ArticlePDF Available

Phase 2 clinical trial of VAL-083 as first-line treatment in newly-diagnosed MGMT-unmethylated glioblastoma multiforme (GBM): Halfway report

Authors:

Abstract and Figures

Background and Aim: Approximately 60% of glioblastoma multiforme (GBM) patients possess an unmethylated O-6-methylguanine-DNA methyltransferase (MGMT) gene, which confers a limited response to standard-of-care treatment with temozolomide (TMZ), resulting in a lower survival. Dianhydrogalactitol (VAL-083) is a novel bi-functional DNA-targeting agent that induces interstrand cross-links at N7-guanine, leading to DNA double-strand breaks and ultimately cell death. VAL-083 circumvents MGMT-mediated repair of the O6 guanine alkylator TMZ. A Phase 2 study has been initiated for VAL-083 in newly diagnosed MGMT unmethylated GBM. Subjects and Methods: The study has two parts: part 1 is a dose–escalation and induction format to enroll up to ten patients in which they received VAL-083 at 20, 30, or 40 mg/m2 per day for 3 days every 21 days concurrently with standard radiation treatment and VAL-083 for up to eight additional cycles. Part 2 comprises an expansion phase to enroll up to twenty additional patients. This study was performed with approval by the Institutional Review Board of Sun Yat-sen University Cancer Center (B2016-058-01) on January 13, 2017, and registered with the ClinicalTrials.gov (NCT03050736) on February 13, 2017. Results: After completion of dose escalation, VAL-083, 30 mg/m2 per day, in combination with radiation therapy, was generally safe and well tolerated. At the cutoff date, 23 patients had been enrolled, 14 of whom had been treated in the expansion phase. Consistent with prior studies, myelosuppression was the most common adverse event. Pharmacokinetic assessment indicated that the levels of VAL-083 were as high in the cerebrospinal fluid as in plasma, 2 h postinfusion. Of the 22 patients who had reached their four precycle magnetic resonance imaging assessments, 12 were assessed with disease progression, with a median progression-free survival of 9.9 (95% confidence interval 7.3–12.0) months for all the patients studied. Conclusion: These preliminary data support VAL-083 as a potentially valuable treatment option for newly diagnosed GBM.
Content may be subject to copyright.
© 2020 Glioma | Published by Wolters Kluwer - Medknow 167
Original Article
Phase 2 clinical trial of VAL-083 as first-line treatment in
newly-diagnosed MGMT-unmethylated glioblastoma multiforme
(GBM): Halfway report
Chengcheng Guo1,*, Qunying Yang1,*, Jiawei Li1, Shaoxiong Wu2, Meiling Deng2, Xiaojing Du2, Ke Sai1, Xiaobing Jiang1, Zhenghe Chen1, Ji Zhang1, Fuhua Lin1,
Jian Wang1, Yinsheng Chen1, Chao Ke1, Xiangheng Zhang1, Xue Ju1, Yonggao Mou1, Jeffrey Bacha3,4, Anne Steino3,4, Sarath Kanekal3,4, Claire Kwan3,4,
Gregory Johnson3,4, Richard Schwartz3,4, John Langlands3,4, Dennis Brown3,4, Zhong-ping Chen1
1Department of Neurosurgery/Neuro‑Oncology, Sun Yat‑sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of
Oncology in South China, Guangzhou, Guangdong Province, China.
2Department of Radiation Oncology, Sun Yat‑sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in
South China, Guangzhou, Guangdong Province, China.
3Delmar Pharmaceuticals, Inc., Vancouver, Canada.
4Delmar Pharmaceuticals, Inc., Menlo Park, CA, USA.
*Contributed Equally.
Background and Aim: Approximately 60% of glioblastoma multiforme (GBM) patients possess an unmethylated O-6-methylguanine-DNA
methyltransferase (MGMT) gene, which confers a limited response to standard-of-care treatment with temozolomide (TMZ), resulting in a
lower survival. Dianhydrogalactitol (VAL-083) is a novel bi-functional DNA-targeting agent that induces interstrand cross-links at N7-guanine,
leading to DNA double-strand breaks and ultimately cell death. VAL-083 circumvents MGMT-mediated repair of the O6 guanine alkylator
TMZ. A Phase 2 study has been initiated for VAL-083 in newly diagnosed MGMT unmethylated GBM. Subjects and Methods: The study
has two parts: part 1 is a dose–escalation and induction format to enroll up to ten patients in which they received VAL-083 at 20, 30, or
40 mg/m2 per day for 3 days every 21 days concurrently with standard radiation treatment and VAL-083 for up to eight additional cycles.
Part 2 comprises an expansion phase to enroll up to twenty additional patients. This study was performed with approval by the Institutional
Review Board of Sun Yat-sen University Cancer Center (B2016-058-01) on January 13, 2017, and registered with the ClinicalTrials.
gov (NCT03050736) on February 13, 2017. Results: After completion of dose escalation, VAL-083, 30 mg/m2 per day, in combination with
radiation therapy, was generally safe and well tolerated. At the cutoff date, 23 patients had been enrolled, 14 of whom had been treated in the
expansion phase. Consistent with prior studies, myelosuppression was the most common adverse event. Pharmacokinetic assessment indicated
that the levels of VAL-083 were as high in the cerebrospinal uid as in plasma, 2 h postinfusion. Of the 22 patients who had reached their
four precycle magnetic resonance imaging assessments, 12 were assessed with disease progression, with a median progression-free survival
of 9.9 (95% condence interval 7.3–12.0) months for all the patients studied. Conclusion: These preliminary data support VAL-083 as a
potentially valuable treatment option for newly diagnosed GBM.
Keywords: Adjuvant, chemotherapy, dianhydrogalactitol,
glioblastoma multiforme, O-6-methylguanine-DNA
methyltransferase, phase 2, Stupp regimen, temozolomide
Access this article online
Quick Response Code:
Website:
www.jglioma.com
DOI:
10.4103/glioma.glioma_25_19
Address for correspondence: Prof. Zhong‑ping Chen,
Department of Neurosurgery/Neuro‑Oncology, Sun Yat‑sen University
Cancer Center, Collaborative Innovation Center for Cancer Medicine,
State Key Laboratory of Oncology in South China, Guangzhou 510060,
Guangdong Province, China.
E‑mail: chenzhp@sysucc.org.cn;
Dr. Dennis Brown,
DelMar Pharmaceuticals, Inc., Vancouver, Canada.
Email: dbrown@delmarpharma.com
This is an open access journal, and arcles are distributed under the terms of the Creave
Commons Aribuon‑NonCommercial‑ShareAlike 4.0 License, which allows others to
remix, tweak, and build upon the work non‑commercially, as long as appropriate credit
is given and the new creaons are licensed under the idencal terms.
For reprints contact: reprints@medknow.com
How to cite this article: Guo C, Yang Q, Li J, Wu S, Deng M, Du X, et al.
Phase 2 clinical trial of VAL-083 as rst-line treatment in newly-diagnosed
MGMT-unmethylated glioblastoma multiforme (GBM): Halfway report.
Glioma 2019;2:167-73.
Abstract
Received: 09-December-2019 Revised: 23-December-2019
Accepted: 27-December-2019 Published: 23-January-2020
[Downloaded free from http://www.jglioma.com on Friday, January 24, 2020, IP: 10.232.74.22]
Guo, et al.: VAL‑083 for the newly diagnosed GBM with MGMT unmethylation
Glioma ¦ Volume 2 ¦ Issue 4 ¦ October‑December 2019
168
intRoduCtion
Glioblastoma multiforme (GBM) is the most common malignant
and aggressive brain cancer with an incidence of 2–3/100,000
persons/year. Patients with GBM have a poor prognosis and
a 5-year overall survival (OS) rate of 9.8%. Tumor resection
represents the rst line in glioblastoma treatment, and is the rst
step in the management of patients.[1-3] Current standard (Stupp
regimen) was established by the EORTC-NCIC trial, including
the usage of temozolomide (TMZ) with concurrent radiation
followed by the six cycles of adjuvant TMZ chemotherapy after
the maximal surgical tumor resection.[4] The Stupp regimen
signicantly improves the survival of newly diagnosed GBM
patients, with a median OS of 14.6 months when TMZ is
combined with radiation compared to those receiving radiation
alone (12.1 months), and the 2-year survival was 26.5%
versus 10.4%. Furthermore, patients with an unmethylated
promoter for the gene encoding O-6-methylguanine-DNA
methyltransferase (MGMT) had more aggressive prognosis
and resistance to TMZ, with a median progression-free
survival (PFS) of 10.3 months in patients with GBM with
methylated  MGMT promoter versus 5.3 months in patients
with unmethylated MGMT promoter.[5] Overall, 68.9% of the
patients with GBM with methylated MGMT promoter were
progression free at 6 months versus 40.0% of patients with
GBM with an unmethylated MGMT promoter. The median
OS was also longer in patients with GBM with methylated
MGMT promoter than those with unmethylated MGMT
promoter; 21.7 months versus 12.7 months, respectively.
This difference in outcome for GBM patients with methylated
versus unmethylated MGMT promoter has been conrmed
more recently in a study that demonstrated a median PFS in
patients with GBM of 6.9 months for those with unmethylated
MGMT promoter versus 11.6 months for those with methylated
MGMT.[5,6] In Stupp study,[5] the median OS was also longer
in patients with GBM with methylated MGMT promoter than
those with unmethylated MGMT promoter, 20.9 months versus
16.0 months, respectively. Given the aggressive biological
nature of GBM, particularly in patients with unmethylated
MGMT promoter, there is an urgent need to develop effective
new therapies.
Dianhydrogalactitol (VAL-083) is a “first-in-class”
bi-functional DNA-targeting agent that introduces interstrand
DNA cross-links at the N7-position of guanine leading
to DNA double-strand breaks and cancer cell death. This
involves S-phase-dependent DNA double-strand breaks and
homologous recombination DNA repair.[7,8] VAL-083 has
demonstrated clinical activity against a range of cancers,
including GBM and ovarian cancer in clinical trials sponsored
by the U.S. National Cancer Institute.[9-12] The antitumor
activity of VAL-083 has been shown to be unaffected by the
common mechanisms of chemoresistance, including MGMT,
in cancer cell models and animal studies.[13,14]
As a result, this clinical trial in newly diagnosed GBM patients
with an unmethylated promoter for the gene encoding MGMT
was designed to determine if rst-line treatment with VAL-083
in combination with radiotherapy can provide improvement
in efcacy over the historical standard-of-care TMZ plus
radiotherapy.
subjeCts and methods
Study design
This was a single-arm, open-label study to determine the safety
and the maximal tolerated dose of VAL-083 in combination
with a standard-of-care radiation regimen when used to treat
newly diagnosed GBM in patients with unmethylated promoter
of the MGMT gene. Pharmacokinetic properties of VAL-083
in this population and tumor responses to treatment were also
evaluated. This study was performed after approval by the
Institutional Review Board of Sun Yat-sen University Cancer
Center (B2016-058-01) on January 13, 2017. All patients had
the ability to sign and provide written informed consent prior
to any study-related procedure. This study was registered with
the ClinicalTrials.gov (NCT03050736) on February 13, 2017.
The study was conducted in two parts:
Part 1: Dose confirmation comprised a series of three
cohorts to conrm the recommended dose of VAL-083 in
combination with a standard-of-care radiation regimen.
Standard radiotherapy was dened as conformal, fractionated
focal radiation at a dose of 2 Gy/fraction given once daily
5 days/week (Monday through Friday) over a period of
6 weeks (the induction period), for a total dose of 60 Gy.
VAL-083 (DelMar Pharmaceuticals, Inc., Menlo Park, CA,
USA) was administered concurrently with radiation therapy,
intravenously daily for 3 days, commencing on the 1st day
of radiotherapy (study days 1–3), with a repeated 3-day
cycle (cycle #2) administered on study days 22–24 of radiation
therapy. If radiation therapy was delayed, cycle #2 of VAL-083
was also delayed so that it was given concurrently with
radiation therapy. VAL-083 was administered such that the
intravenous VAL-083 infusion was completed approximately
60 min prior to the radiation therapy.
After completion of the 6-week radiotherapy regimen, on
approximately study day 43 or thereafter, the patients commenced
adjuvant maintenance therapy with VAL-083 alone, administered
intravenously daily for 3 days every 21 days (cycles 3–10), at the
same assigned dose, for up to eight maintenance cycles, such that
the patients could receive a maximum of 10 cycles of VAL-083
during induction as adjuvant therapy.
Dose escalation of VAL-083 proceeded in three sequential
cohorts, consisting patients receiving 20, 30, and
40 mg/m2 per day for 3 days every 21 days. Dose escalation was
followed in accordance with the scheme outlined in Table 1.
All patients who had received at least one dose of VAL-083
were evaluable for safety and determination of the maximal
tolerated dose.
For Part 2 (expansion stage), the same dosing and radiation
schedule as in Part 1 was utilized. The dose of VAL-083 studied
[Downloaded free from http://www.jglioma.com on Friday, January 24, 2020, IP: 10.232.74.22]
Guo, et al.: VAL‑083 for the newly diagnosed GBM with MGMT unmethylation
Glioma ¦ Volume 2 ¦ Issue 4 ¦ October‑December 2019 169
in Part 2 was determined from Part 1 (30 mg/m2 per day for
3 days every 21 days).
Per protocol, patients in either part of the study continued to
receive VAL-083 at the assigned dosage and were followed
for the safety and indications of activity, as long as the patient
continued to demonstrate response or stable disease and
tolerated therapy, for a maximum of eight cycles. Patients
responding to VAL-083 therapy at the end of the prescribed
treatment period were permitted to continuously receive
VAL-083.
In both parts of the study, baseline cranial magnetic resonance
imaging (MRI) was obtained within 3 weeks prior to
commencing radiation therapy, 3 weeks after completing
radiation therapy (prior to VAL-083 cycle 4), and then every
3 months, while the patient remained in the study, or more
frequently if clinically indicated. The same method was utilized
for all response assessments.
Patients
Patients, 18–70 years of age, with documented unmethylated
MGMT gene promoter status and histologically proven
newly diagnosed supratentorial GBM were enrolled in
our study. In addition, patients who had received no prior
chemotherapy, radiation therapy, or immunotherapy for their
brain tumor; those had an interval of ≥2 weeks but ≤7 weeks
after surgery or biopsy before the rst administration of study
treatment; those who recovered from the effects of surgery,
postoperative infection, and other complications before study
registration and stable or decreasing dose of steroids >5 days
prior to randomization; those who fell under World Health
Organization Karnofsky performance status[15] ≥70%, expected
survival ≥2 months, and normal hematologic, renal, and hepatic
functions (absolute neutrophil count ≥1500/mm3; platelet
count ≥100,000/mm3; hemoglobin ≥10 g/dL; liver function
values <2.5 times the upper limit of normal for the laboratory;
bilirubin <2 times the upper limit of normal for the laboratory;
and serum creatinine ≤1.5 times the upper limit of normal or
creatinine clearance >50 mL/min [measured or calculated by
the Cockcroft–Gault formula])[16] at screening were included
in the study. Contraindications to participation included active
or uncontrolled infection, other coexistent malignant disease,
and pregnancy or lactation.
Patients were excluded from trial participation if they had prior
chemotherapy and radiation; had used Gliadel® wafer during
the operation; had a history of active peptic ulcer within the last
6 months before enrollment; were pregnant or breastfeeding;
had uncontrolled hypertension; had inability to undergo MRI
evaluation; those on current alcohol dependence or drug abuse;
had known hypersensitivity to study treatment; those who were
unable or unwilling to fulll the study requirements; or those
unwilling to provide informed consent.
All patients provided written informed consent prior to
participating in the current study. All patients were conrmed
to be MGMT unmethylated prior to enrollment.
MGMT assay
MGMT methylation-specic polymerase chain reaction (MSP)
was used to conrm the methylation status of the MGMT
promoter region. Genomic DNA was extracted from
parafn-embedded specimens according to the manufacturer’s
instructions (TianGen DNA Mini Kit, Beijing, China).
The methylation status was determined by performing the
bisulte modication, which converts unmethylated but not
methylated cytosines to uracils. The genomic DNA (50 ng)
was amplied using primers designed to detect the methylated
or unmethylated sequences by MSP. MSP was performed
according to the manufacturer’s protocol (EZ DNA
Methylation Gold kit, Zymo Research, Irvine, CA, USA).
The primers were specific for either the methylated or
modied unmethylated DNA, as previously described.[17]
DNA obtained from normal peripheral blood lymphocytes
served as the negative control, and enzymatically methylated
DNA from peripheral blood lymphocytes was used as the
positive control. Ten microliters of each 50-μL MSP product
was directly loaded onto nondenaturing 6% polyacrylamide
gels, stained with ethidium bromide, and examined under
ultraviolet illumination.
Safety and dose-limiting toxicity assessment
All adverse events were graded utilizing the National Cancer
Institute Common Toxicity Criteria version 4.0.[18] Patients in
the Part 1 stage were evaluable for dose-limiting toxicity (DLT)
if they completed cycle one or experienced a DLT during the
rst two cycles of treatment with VAL-083 during the period
of radiation therapy. The following treatment-related adverse
events were considered a DLT during cycles 1 and 2: any
Grade 4 thrombocytopenia, or Grade 3 thrombocytopenia
with hemorrhage; absolute neutrophil count nadir <500/μL
or platelet count <50,000/μL, lasting for >5 days; absolute
neutrophil count <500/μL with fever (febrile neutropenia);
treatment delays of >3 weeks for hematologic toxicity; any
Table 1: Study cohort’s dose-escalation scheme
If dose-limiting
toxicity observed in
Action
Cohort 1 (20 mg/m2
per day) (n=1)
0 of 1 patient Proceed with enrollment in the next cohort
1 of 1 patient Enroll ve additional patients
1 of 6 patients Proceed with enrollment in the next cohort
≥2 of 6 patients Do not dose escalate
Cohort 2 (30 mg/m2
per day) (n=3)
0 of 3 patients Proceed with enrollment in the next cohort
1 of 3 patients Enroll three additional patients
1 of 6 patients Proceed with enrollment in the next cohort
≥2 of 6 patients Do not dose escalate
Cohort 3 (40 mg/m2
per day) (n=6)
≤1 of 6 patients Proceed with enrollment in Part 2
≥2 of 6 patients Consider lower dose for Part 2
[Downloaded free from http://www.jglioma.com on Friday, January 24, 2020, IP: 10.232.74.22]
Guo, et al.: VAL‑083 for the newly diagnosed GBM with MGMT unmethylation
Glioma ¦ Volume 2 ¦ Issue 4 ¦ October‑December 2019
170
Grade 3 or 4 nonhematologic toxicity due to treatment with
the exception of alopecia, nausea, and vomiting; Grade 3 or
4 nausea or vomiting while receiving an optimal antiemetic
regimen for prophylaxis and management; and treatment
delays of >3 weeks for toxicity.
Clinical and radiographic assessment
At baseline, all patients received a physical examination and
clinical assessment, and medical history was obtained. These
assessments involved vital signs, performance status, and
routine laboratory studies including hematology, coagulation,
kidney function and liver function, urinalysis, pregnancy test,
chest X-ray, electrocardiogram, and whole-brain MRI. Vital
signs and laboratory assessments were conducted prior to each
treatment cycle. Whole-brain MRI was repeated prior to cycle
4 and every 3 months thereafter. Tumors were evaluated by the
investigator using the Response Assessment in NeuroOncology
criteria.[19] Response was measured by a reduction in tumor size.
Pharmacokinetics analysis
The objectives were to determine the pharmacokinetic prole
and dose–exposure relationship of VAL-083 for injection on
day 1, when given as an infusion over 60 min. Cerebrospinal
uid (CSF) analysis was undertaken to obtain a single-point
concentration of VAL-083 in CSF. On cycle 1, day 1, at each
dose level, blood was collected predose; 15 ± 5, 30 ± 5,
60 ± 10, 120 ± 10, 240 ± 15, and 360 ± 15 min after the end
of intravenous injection of study drug; and immediately prior
to cycle 1, day 2 dosing. For CSF, cycle 1, day 3 blood for
VAL-083 plasma concentration was collected 15 ± 5 min
after the end of intravenous injection of study drug. Plasma
pharmacokinetic parameters were determined for cycle
1 day 1 at each dose level via noncompartmental analysis
using  WinNonlin version 2.0 (Pharsight Co., Certara, NJ,
USA) or newer version. Predose plasma concentration (Ctrough)
determined directly from the concentration–time profile,
day 1; maximum observed concentration (Cmax) on day
1; time of observed Cmax (Tmax) on day 1; area under the
concentration–time curve from predose (time 0) to the time of
the last quantiable concentration (area under the curve [AUC]
AUClast) on day 1 calculated using the linear-log trapezoidal
rule; area under the concentration–time curve extrapolated to
innity calculated using the linear-log trapezoidal rule (AUCinf)
on day 1; total oral body clearance at steady state (CL/F)
calculated as dose/AUC day 1; mean residence time calculated
(AUMC/AUC) where AUMC is area under the moment curve;
Vz: the apparent volume of distribution during the terminal
phase; Lambda z (λz): the terminal elimination rate constant
determined by selection of at least three decreasing data points
on the terminal phase of the concentration–time curve on day
1; and terminal elimination half-life (T1/2) on day 1 determined
from 0.693/λz.
Statistical analysis
MedCalc Ver 9.3.1 (MedCalc Inc., Mariakerke, Belgium) was
used in all statistical analyses. The primary safety objective of
the study was to determine the maximal tolerated dose and to
describe the safety and tolerability of VAL-083 in combination
with a standard-of-care radiation regimen.
The efficacy objective in both parts of the study was to
determine the activity of VAL-083 in newly diagnosed
unmethylated MGMT GBM patients. Outcome assessment
will be performed based on tumor response to treatment,
PFS, PFS at 6 months, and overall survival (OS), compared
to historical results in the target population. The median PFS
and OS were estimated by the Kaplan–Meier analysis. For
disease response, the frequency of best response (complete
response, partial response, and stable disease) of the treatment
was determined. Pharmacokinetic parameters were computed
by noncompartmental analysis.
Results
Patient demographics
Demographics for all patients enrolled in the study are described
in Table 2. The median age was 53.5 (range: 21.1–65.0) years;
8/23 (35%) were male; the mean body surface area was
1.69 ± 0.17 m2; and the median Karnofsky performance status
was 90 (range 70–100).
As of November 2, 2019, a total of 23 patients had been treated
in the study. Dose escalation cohorts evaluating doses of 20,
30, and 40 mg/m2 per day on days 1, 2, and 3 of a 21-day
cycle were completed. As myelosuppression was observed
at the dose of 40 mg/m2 per day, the dose of VAL-083 was
reduced to 30 mg/m2 daily on days 1, 2, and 3 every 21 days,
administered concurrently with radiation therapy. This dose
was selected for the Part 2 of the study. To date, 14 patients
have been treated in the expansion phase with a starting dose
of 30 mg/m2 per day.
Safety
Myelosuppression (decreased platelet, neutrophil, lymphocyte,
and white cell counts) was conrmed to be the most common
adverse event. Additional adverse events included anemia,
decreased red cell count and increased liver function enzymes,
and fatigue. Hematological adverse events generally resolved
spontaneously; serious adverse events possibly related to
VAL-083 were reported in 4/23 (17%) of patients. Three
DLTs were in patients who completed the rst two cycles of
treatment [Table 3].
Pharmacokinetics
Pharmacokinetic proles were determined on day 1 of cycle
1 for each patient. Maximum concentration and AUC were
broadly linear with respect to dose; the terminal elimination
half-life was 0.8 h. Data obtained to date indicate that overall
the concentration of VAL-083 was as high in CSF as in plasma
at 2 h postinfusion [Table 4].
Efficacy
The best response was determined by the investigator for
patients who had completed their rst planned assessment
prior to cycle 4. At November 2, 2019, 19 patients received
[Downloaded free from http://www.jglioma.com on Friday, January 24, 2020, IP: 10.232.74.22]
Guo, et al.: VAL‑083 for the newly diagnosed GBM with MGMT unmethylation
Glioma ¦ Volume 2 ¦ Issue 4 ¦ October‑December 2019 171
at least one assessment prior to initiating cycle 4 and beyond.
There were 9/19 (43%) patients assessed as complete response,
8/19 (48%) assessed as stable disease, and 2/19 (10%) assessed
as progressive disease. There were two patients who have
not yet reached prior-to-cycle 4 assessment and two patients
discontinued or died before the rst planned assessment time
point (prior to cycle 4).
As of the cutoff date of November 2, 2019, for all patients
including completed and active treatment patients, the median
number of cycles of VAL-083 received was 8, and nine patients
received >10 cycles.
For the 22 patients who had completed at least their rst
assessment at the cutoff date, 12 were assessed with disease
progression. The median PFS for patients who had shown
disease progression by MRI assessment is summarized in
Table 5.
disCussion
Glioblastoma is intrinsically inltrative and destructive in the
brain with poor prognosis, with a median OS of 14.6 months
with radiation and TMZ chemotherapy combined.[14]
Standard therapy for patients with malignant gliomas has
traditionally involved maximal surgical resection/debulking
of the primary tumor (if feasible), followed by radiation
therapy with concurrent and the adjuvant TMZ of 5 days for
28 days.[10] However, radical resection surgery is virtually
impossible, due to the inltrative nature of the tumor and
critical nature of the surrounding central nervous system
tissue.
Treatment of GBM has limited new promising approaches
over the Stupp regimen, especially in patients with an
unmethylated promoter for the gene encoding MGMT.
The MGMT gene resides on chromosome 10q26 and is
responsible for this DNA repair protein, which removes
alkyl groups from the O6 position of guanine. The prognostic
value of MGMT promoter methylation status has been
determined in previous clinical trials.[5,6,20] The 2-year OS
in MGMT-unmethylated patients was 22.7% compared to
46% in MGMT-methylated patients.[5] Attempts to improve
the efcacy have used dose–dense usage of TMZ, trying
to modulate the MGMT expression. However, the median
OS (14.6 months for the Stupp regimen vs. 13.3 months for
the dose–dense regimen, P = 0.44) and the PFS (5.1 months
for the Stupp regimen vs. 6.0 months for the dose–dense
regimen, P = 0.15) did not provide any overall improvement
in survival outcomes.[21]
Responses to the antivascular endothelial growth factor
antibody, bevacizumab, have led to signicant improvements
in PFS. However, bevacizumab treatment in patients with
recurrent glioblastoma does not confer a survival advantage
despite prolonged PFS.[21,22] Furthermore, bevacizumab has
been reported to induce a more invasive tumor phenotype,[23]
and a meta-analysis of ve recurrent GBM trials concluded
that outcome following bevacizumab failure is poor.[24] The
median OS in bevacizumab-failed GBM has been reported to
be approximately 2–5 months.[25]
Table 2: Study patient demographics
All patients (n=23) 20 mg/m2 per day (n=1) 30 mg/m2 per day (n=19) 40 mg/m2 per day (n=3)
Age (years)
Mean±SD 48.9±12.5 33.9 50.3±13.2 46.0±4.4
Median (minimum–maximum) 53.5 (21.1–65.0) 33.9 54.7 (21.1–65.0) 44.1 (42.9–51.1)
Sex, n (%)
Male 8 (35) 1 (100) 12 (63) 2 (67)
Female 15 (65) 0 7 (37) 1 (33)
Body surface area (m2)
Mean±SD 1.69±0.17 1.64 1.67±0.17 1.70±0.27
Median (minimum–maximum) 1.67 (1.45–2.01) 1.64 1.68 (1.45–2.01) 1.67 (1.45–1.99)
Height (cm)
Mean±SD 166.5±10.1 162.0 166.4±10.3 168.7±12.5
Median (minimum–maximum) 165.0 (150.0–183.0) 162.0 168.0 (150.0–183.0) 163.0 (160–183.0)
Weight (kg)
Mean±SD 62.8±10.4 60 63.1±10.2 61.8±15.8
Median (minimum–maximum) 62.0 (46.0–80.0) 60 62.0 (49.5–80.0) 62.0 (46.0–77.5)
Karnofsky Performance Status 90 (55.25–72.25) 90 90 80
Median (minimum–maximum)
VAL-083 was administered intravenously on days 1, 2, and 3 every 21 days. SD: Standard deviation
Table 3: Frequency of dose-limiting toxicities in patients
receiving VAL-083
VAL-083 dose
(mg/m2 per day)
Number of patients
completed 42-day
treatment (2 cycles)
Number of patients
with dose-limiting
toxicities [n (%)]
20 1 0
30 18 2 (11)
40 3 1 (33)
VAL-083 was administered intravenously on days 1, 2, and 3 every 21 days
[Downloaded free from http://www.jglioma.com on Friday, January 24, 2020, IP: 10.232.74.22]
Guo, et al.: VAL‑083 for the newly diagnosed GBM with MGMT unmethylation
Glioma ¦ Volume 2 ¦ Issue 4 ¦ October‑December 2019
172
A Phase II study of bevacizumab and erlotinib, and epidermal
growth factor receptor, a tyrosine kinase inhibitor, based on
the Stupp regimen, showed that the sequential treatment did
not increase the OS for the unmethylated GBM patients with
13.2 months of OS and 9.2 months of PFS.[26] Neither the “CORE”
trial including Cilengitide[27] nor the “Glarius” trial[28] including
the Stupp regimen sequential with bevacizumab and irinotecan
showed any benecial result for unmethylated GBM patients.
VAL-083 is water soluble and is administered by intravenous
infusion. Studies in rodents have demonstrated that VAL-083
crosses the blood–brain barrier, where it accumulates in the
brain tumor tissue preferentially.[29] In the current study,
VAL-083 has been measured in the CSF of patients 2 h after
the end of infusion, and levels have been found to be at least
as high as those in plasma at the same time point.
The main adverse event in patients receiving VAL-083 has
been myelosuppression. The maximum dose evaluated in
this study was 40 mg/m2 per day and was associated with
myelosuppression and lower overall tolerability. As a result,
30 mg/m2 per day for 3 days in a 3-week cycle was used in
the dose–escalation part of this study.
In the ongoing evaluation of the study data, VAL-083
has shown favorable efficacy in this patient population
with respect to median PFS. During our mid-point data
review (November 2, 2019), the overall median PFS for
VAL-083 was 9.9 (95% condence interval [CI]: 7.3–12.0)
months with 12/22 (54%) patients progressed and for those
receiving the intended treatment dose of 30 mg/m2 was
10.4 (95% CI: 6.0–12.0) months, with 9/18 (50%) patients
progressed. In the Stupp regimen, the median PFS was
5.3–6.9 months in unmethylated MGMT GBM patients.[5,6]
These median PFS times are longer than the PFS previously
reported and are close to the PFS for the methylated MGMT
TMZ patients (10.3–11.6 months).[5,6]
While we look forward to the completion of enrollment of
patients in this study, these preliminary results support our
optimism that VAL-083 can provide a valuable option than
currently available treatments for patients with unmethylated
MGMT glioblastoma.
ConClusion
VAL-083 at 30 mg/m2 per day for 3 days every 21 days in
combination with radiation therapy is generally safe and well
tolerated, and multiple treatment cycles in the adjuvant setting
have been achieved. Adverse events have been shown to be
consistent with those of prior studies. Levels of VAL-083
measured in the CSF at 2 h postinfusion were as high as those
measured in plasma, demonstrating signicant penetration to
the brain. VAL-083 at 30 mg/m2 per day in combination with
radiotherapy has demonstrated benet with respect to disease
progression over standard-of-care TMZ in the same setting.
These preliminary data support the premise that VAL-083 has the
potential to provide a valuable treatment option for such patients.
Financial support and sponsorship
Nil.
Institutional review board statement
This study was approved by the Institutional Review Board
of Sun Yat-sen University Cancer Center (B2016-058-01) on
January 13, 2017, China, and registered with the ClinicalTrials.
gov (NCT03050736) on February 13, 2017.
Declaration of participant consent
The authors certify that they have obtained the appropriate
participant consent form. In the forms, the participants
have given their consent for the participants’ images and
other clinical information to be reported in the journal. The
participants understood that their names and initials would
not be published and due efforts would be made to conceal
their identity.
Conflicts of interest
Jeffrey Bacha was not afliated with DelMar Pharmaceuticals
Ltd. when submitting this article for publication.
Table 4: Concentration of VAL-083 in plasma and cerebral spinal fluid
VAL-083 dose
(mg/m2 per day)
nVAL-083 concentration (ng/mL) Ratio at 2 h
cerebrospinal
fluid/plasma
Plasma maximum
concentration
Plasma at 2 h
postdose
Cerebrospinal fluid
at 2 h postdose
20 1 481.0 110.0 154 1.40
30 6 574.9±261.5 97.2±20.9 123.0±27.8 1.19±0.37
40 3 898.7±69.6 169.7±41.9 189.7±69.9 1.13±0.41
VAL-083 administered intravenously via a 60-min infusion on days 1, 2, and 3 every 21 days
Table 5: Frequency of disease progression and median progression-free survival in patients receiving VAL-083
All patients (n=22) 20 mg/m2 per day (n=1) 30 mg/m2 per day (n=18) 40 mg/m2 per day (n=3)
Number of patients progressed, n (%) 12 (55) 1 (100) 9 (50) 2 (67)
Median PFS (months)a (95% CI) 9.9 (7.3–12.0) 3.0 10.4 (6.0–12.0) 9.9 (9.3–9.9)
VAL-083 was administered intravenously on days 1, 2, and 3 every 21 days. Median PFS determined by Kaplan-Meier analysis with patients censored at
the last known date alive. Median PFS determined at cutoff date of November 2, 2019. CI: Condence interval, PFS: Progression-free survival
[Downloaded free from http://www.jglioma.com on Friday, January 24, 2020, IP: 10.232.74.22]
Guo, et al.: VAL‑083 for the newly diagnosed GBM with MGMT unmethylation
Glioma ¦ Volume 2 ¦ Issue 4 ¦ October‑December 2019 173
RefeRenCes
1. National Comprehensive Cancer Network. NCCN Clinical Practice
Guidelines in Oncology: Central Nervous System Cancers. Available
from: http://www.nccn.org/professionals/physician_gls/pdf/cns.
pdf. [Last accessed on 2019 Nov 02].
2. Stupp R, Tonn JC, Brada M, Pentheroudakis G; ESMO Guidelines
Working Group. High-grade malignant glioma: ESMO Clinical
Practice Guidelines for diagnosis, trea tment and follow-up. Ann
Oncol 2010;21 Suppl 5:v190-3.
3. Tonn JC, Thon N, Schnell O, Kreth FW. Personalized surgical therapy.
Ann Oncol 2012;23 Suppl 10:x28-32.
4. Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ,
et al. Radiotherapy plus concomitant and adjuvant temozolomide for
glioblastoma. N Engl J Med 2005;352:987-96.
5. Hegi ME, Diserens AC, Gorlia T, Hamou MF, de Tribolet N, Weller M,
et al. MGMT gene silencing and benet from temozolomide in
glioblastoma. N Engl J Med 2005;352:997-1003.
6. Tanguturi SK, Trippa L, Ramkissoon SH, Pelton K, Knoff D, Sandak D,
et al. Leveraging molecular datasets for biomarker-based clinical trial
design in glioblastoma. Neuro Oncol 2017;19:908-17.
7. Zhai B, Steino A, Bacha J, Brown D, Daugaard M. Molecular
mechanisms of dianhydrogalactitol (VAL-083) in cancer treatment.
Cancer Res 2016;76:2985.
8. Zhai B, Steinø A, Bacha J, Brown D, Daugaard M. Dianhydrogalactitol
induces replication-dependent DNA damage in tumor cells preferentially
resolved by homologous recombination. Cell Death Dis 2018;9:1016.
9. Eagan RT, Ames MM, Powis G, Kovach JS. Clinical and pharmacologic
evaluation of split-dose intermittent therapy with dianhydrogalactitol.
Cancer Treat Rep 1982;66:283-7.
10. Eagan RT, Dinapoli RP, Hermann RC Jr, Groover RV, Layton DD Jr,
Scott M. Combination carmustine (BCNU) and dianhydrogalactitol in
the treatment of primary brain tumors recurring after irradiation. Cancer
Treat Rep 1982;66:1647-9.
11. Institóris E, Szikla K, Otvös L, Gál F. Absence of cross-resistance
between two alkylating agents: BCNU vs. bifunctional galactitol.
Cancer Chemother Pharmacol 1989;24:311-3.
12. Schabel FM Jr., Trader MW, Laster WR Jr, Wheeler GP, Witt MH.
Patterns of resistance and therapeutic synergism among alkylating
agents. Antibiot Chemother (1971) 1978;23:200-15.
13. Fouse SD, Steino A, Butowski N, Bacha JA, Kanekal S, Dos Santos N,
et al. Dianhydrogalactitol inhibits the growth of glioma stem and
non-stem cultures, including temozolomide-resistant cell lines, in vitro
and in vivo. Neuro Oncol 2015;16:v83.
14. Hu K, Fotovati A, Chen J, Triscott J, Bacha J, Brown D, et al. Abstract
811: VAL083, a novel N7 alkylating agent, surpasses temozolomide
activity and inhibits cancer stem cells providing a new potential
treatment option for glioblastoma multiforme. Cancer Res 2012.
15. Schag CC, Heinrich RL, Ganz PA. Karnofsky performance
status revisited: Reliability, validity, and guidelines. J Clin Oncol
1984;2:187-93.
16. Cockcroft DW, Gault MH. Prediction of creatinine clearance from
serum creatinine. Nephron 1976;16:31-41.
17. Qiu ZK, Shen D, Chen YS, Yang QY, Guo CC, Feng BH, et al.
Enhanced MGMT expression contributes to temozolomide resistance
in glioma stem-like cells. Chin J Cancer 2014;33:115-22.
18. US Department of Health and Human Services. National Cancer
Institute. Common Terminology Criteria for Adverse Events (CTCAE)
Version 4.0. National Institutes of Health/National Cancer Institute.
2009.
19. Wen PY, Cloughesy TF, Ellingson BM, Reardon DA, Fine HA,
Abrey L, et al. Report of the Jumpstarting Brain Tumor Drug
Development Coalition and FDA clinical trials neuroimaging
endpoint workshop (January 30, 2014, Bethesda MD). Neuro Oncol
2014;16 Suppl 7:vii36-47.
20. Li D, Chen Y, Guo C, Zhang X, Sai K, Ke C, et al. Real-world
management and survival outcomes of patients with newly diagnosed
gliomas from a single institution in China: A retrospective cohort study.
Glioma 2019;2:96-104.
21. Gilbert MR, Wang M, Aldape KD, Stupp R, Hegi ME, Jaeckle KA,
et al. Dose-dense temozolomide for newly diagnosed glioblastoma:
A randomized phase III clinical trial. J Clin Oncol 2013;31:4085-91.
22. Gilbert MR, Dignam JJ, Armstrong TS, Wefel JS, Blumenthal DT,
Vogelbaum MA, et al. A randomized trial of bevacizumab for newly
diagnosed glioblastoma. N Engl J Med 2014;370:699-708.
23. de Groot JF, Mandel JJ. Update on anti-angiogenic treatment for
malignant gliomas. Curr Oncol Rep 2014;16:380.
24. Reardon DA, Desjardins A, Peters K, Gururangan S, Sampson J,
Rich JN, et al. Phase II study of metronomic chemotherapy with
bevacizumab for recurrent glioblastoma after progression on
bevacizumab therapy. J Neurooncol 2011;103:371-9.
25. Iwamoto FM, Abrey LE, Beal K, Gutin PH, Rosenblum MK, Reuter VE,
et al. Patterns of relapse and prognosis after bevacizumab failure in
recurrent glioblastoma. Neurology 2009;73:1200-6.
26. Raizer JJ, Giglio P, Hu J, Groves M, Merrell R, Conrad C, et al. A phase
II study of bevacizumab and erlotinib after radiation and temozolomide
in MGMT unmethylated GBM patients. J Neurooncol 2016;126:185-92.
27. Nabors LB, Fink KL, Mikkelsen T, Grujicic D, Tarnawski R, Nam DH,
et al. Two cilengitide regimens in combination with standard treatment
for patients with newly diagnosed glioblastoma and unmethylated
MGMT gene promoter: Results of the open-label, controlled,
randomized phase II CORE study. Neuro Oncol 2015;17:708-17.
28. Schäfer N, Proescholdt M, Steinbach JP, Weyerbrock A, Hau P,
Grauer O, et al. Quality of life in the GLARIUS trial randomizing
bevacizumab/irinotecan versus temozolomide in newly diagnosed,
MGMT-nonmethylated glioblastoma. Neuro Oncol 2018;20:975-85.
29. Levin VA, Freeman-Dove MA, Maroten CE.
Dianhydrogalactitol (NSC-132313): Pharmacokinetics in normal and
tumor-bearing rat brain and antitumor activity against three intracerebral
rodent tumors. J Natl Cancer Inst 1976;56:535-9.
[Downloaded free from http://www.jglioma.com on Friday, January 24, 2020, IP: 10.232.74.22]
... Val-083 has a small therapeutic window, displaying toxicity at relatively low doses when administered in vivo (14,15). However, a recent halfway report of the phase II clinical trial NCT03050736 has shown that after dose escalation, Val-083 in combination with radiotherapy was generally safe and well tolerated (16). ...
... It was recently granted orphan drug designation by the U.S. FDA for the treatment of glioma, medullo-blastoma and ovarian cancers, as well as a fast-track status for the treatment of recurrent GBM. Recent data on newly diagnosed GBM showed Val-083 safety in combination with radiotherapy (16). Moreover, it has been recently shown to be more effective than TMZ at similar doses in patient-derived organoids, and consistent with our data, the effect was independent on their MGMT promoter methylation status (40). ...
Article
Glioblastoma (GBM) is the most frequent and aggressive primary tumor type in the central nervous system in adults. Resistance to chemotherapy remains one of the major obstacles in GBM treatment. Identifying and overcoming the mechanisms of therapy resistance is instrumental to develop novel therapeutic approaches for GBM patients. To determine the major drivers of temozolomide (TMZ) sensitivity, we performed shRNA screenings in GBM lines with different MGMT status. We then evaluated dianhydrogalactitol (Val-083), a small alkylating molecule that induces interstrand DNA crosslinking, as a potential treatment to bypass TMZ-resistance mechanisms. We found that loss of Mismatch Repair (MMR) components and MGMT expression are mutually exclusive mechanisms driving TMZ resistance in vitro. Treatment of established GBM cells and tumorsphere lines with Val-083 induces DNA damage and cell cycle arrest in G2/M phase, independently of MGMT or MMR status, thus circumventing conventional resistance mechanisms to TMZ. Combination of TMZ and Val-083 shows a synergic cytotoxic effect in tumor cells in vitro, ex vivo and in vivo. We propose this combinatorial treatment as a potential approach for GBM patients.
... MGMT's activity, which includes the induction of DNA methylation, impedes the replication of tumour cells, undermining the efficacy of TMZ. Additionally, the limited capacity of TMZ to cross the BBB and its associated toxicity at therapeutic doses underscore the need for alternative therapeutic approaches [9][10][11][12][13]. The ongoing challenges in GBM treatment, such as limited BBB penetration, resistance mechanisms, dosage constraints, and toxicity, necessitate the exploration of novel therapeutic strategies. ...
Article
Full-text available
Background/Objectives: Glioblastoma multiforme (GBM), an aggressive and deadly brain tumour, presents significant challenges in achieving effective treatment due to its resistance to current therapies and poor prognosis. This study aimed to synthesise and evaluate 23 novel analogues of 3,4-dihydroquinolin-2(1H)-one, designed to enhance druggability and solubility, and to investigate their potential as VEGFR2 inhibitors for GBM treatment. Methods: The synthesised compounds were analysed using in silico methods, including molecular docking and dynamics studies, to assess their interactions with key residues within the VEGFR2 binding pocket. In vitro evaluations were performed on U87-MG and U138-MG GBM cell lines using MTT assays to determine the IC50 values of the compounds. Results: Among the tested compounds, 4u (IC50 = 7.96 μM), 4t (IC50 = 10.48 μM), 4m (IC50 = 4.20 μM), and 4q (IC50 = 8.00 μM) demonstrated significant antiproliferative effects against both the U87-MG and U138-MG cell lines. These compounds exhibited markedly higher efficacy compared to temozolomide (TMZ), which showed IC50 values of 92.90 μM and 93.09 μM for U87-MG and U138-MG, respectively. Molecular docking and dynamics studies confirmed strong interactions between the compounds and VEGFR2 kinase, supporting their substantial anti-cancer activity. Conclusions: This study highlights the promising potential of 3,4-dihydroquinolin-2(1H)-one analogues, particularly 4m, 4q, 4t, and 4u, as VEGFR2-targeting therapeutic agents for GBM treatment. Further detailed research is warranted to validate and expand upon these findings.
... Dianhydrogalactitol (VAL-083) is another kind of DNA-alkylating agent independent of MGMT repair and temozolomide pathway. In the halfway report of this phase II clinical trial targeting umMGMT nGBM, 10 patients remained progression free, while another 12 patients progressed with PFS of 9.9 months [47]. It turns out that VAL-083 may be promising for umMGMT patients in substitute of temozolomide. ...
Article
Full-text available
Glioblastoma (GBM) is a highly malignant brain tumor with a dismal prognosis. Standard therapy for GBM comprises surgical resection, followed by radiotherapy plus concomitant and adjuvant temozolomide (TMZ) therapy. The methylation status of the O6-methylguanine DNA methyltransferase (MGMT) promoter is one of the most essential predictive biomarkers for patients with GBM treated with TMZ. Patients with an unmethylated MGMT promoter (umMGMT), who comprise 60% of patients with GBM, present an even worse prognosis because of TMZ resistance. Radiotherapy with various fractionation, chemotherapy compensating for TMZ, targeted therapy against diverse oncogenic pathways, immunotherapy of vaccine or immune checkpoint inhibitor, and tumor treating fields have been studied in umMGMT GBM patients. However, most efforts have yielded negative results or merely minimal improvements. Therefore, effective patient subgroup selection concerning precision medicine has become the focus. By assigning different treatments to the corresponding patient subgroups, a better curative effect and subsequently prolonged survival can be achieved. In this review, we re-evaluate the value of standard TMZ therapy and summarize the new clinical strategies and attempts to treat patients with umMGMT, which yielded positive and negative results, to provide alternative treatment options and discuss future directions of umMGMT GBM treatment.
... VAL-083 demonstrates cytotoxicity in glioma cell lines and xenograft models and remained active in glioma cell lines with high MGMT expression [85]. Interim results from a phase II trial in newly diagnosed MGMT unmethylated glioblastoma (NCT03050736) and phase I/II study in recurrent glioblastoma (NCT01478178) show a favorable safety profile [86,87]. A phase II study of VAL-083 is currently underway with MGMT unmethylated glioblastoma in the adjuvant or recurrent setting (NCT02717962) [88]. ...
Article
Full-text available
Purpose of Review This review discusses current and investigative strategies for targeting DNA repair in the management of glioma. Recent Findings Recent strategies in glioma treatment rely on the production of overwhelming DNA damage and inhibition of repair mechanisms, resulting in lethal cytotoxicity. Many strategies are effective in preclinical glioma models while clinical feasibility remains under investigation. The presence of glioma biomarkers, including IDH mutation and/or MGMT promoter methylation, may confer particular susceptibility to DNA damage and inhibition of repair. These biomarkers have been adopted as eligibility criteria in the design of multiple ongoing clinical trials. Summary Targeting DNA repair mechanisms with novel agents or therapeutic combinations is a promising approach to the treatment of glioma. Further investigations are underway to optimize this approach in the clinical setting.
Article
Importance: High-grade gliomas (HGGs) constitute the most common and aggressive primary brain tumor, with 5-year survival rates of 30.9% for grade 3 gliomas and 6.6% for grade 4 gliomas. The add-on efficacy of interferon alfa is unclear for the treatment of HGG. Objectives: To compare the therapeutic efficacy and toxic effects of the combination of temozolomide and interferon alfa and temozolomide alone in patients with newly diagnosed HGG. Design, setting, and participants: This multicenter, randomized, phase 3 clinical trial enrolled 199 patients with newly diagnosed HGG from May 1, 2012, to March 30, 2016, at 15 Chinese medical centers. Follow-up was completed July 31, 2021, and data were analyzed from September 13 to November 24, 2021. Eligible patients were aged 18 to 75 years with newly diagnosed and histologically confirmed HGG and had received no prior chemotherapy, radiotherapy, or immunotherapy for their HGG. Interventions: All patients received standard radiotherapy concurrent with temozolomide. After a 4-week break, patients in the temozolomide with interferon alfa group received standard temozolomide combined with interferon alfa every 28 days. Patients in the temozolomide group received standard temozolomide. Main outcomes and measures: The primary end point was 2-year overall survival (OS). Secondary end points were 2-year progression-free survival (PFS) and treatment tolerability. Results: A total of 199 patients with HGG were enrolled, with a median follow-up time of 66.0 (95% CI, 59.1-72.9) months. Seventy-nine patients (39.7%) were women and 120 (60.3%) were men, with ages ranging from 18 to 75 years and a median age of 46.9 (95% CI, 45.3-48.7) years. The median OS of patients in the temozolomide plus interferon alfa group (26.7 [95% CI, 21.6-31.7] months) was significantly longer than that in the standard group (18.8 [95% CI, 16.9-20.7] months; hazard ratio [HR], 0.64 [95% CI, 0.47-0.88]; P = .005). Temozolomide plus interferon alfa also significantly improved median OS in patients with O6-methylguanine-DNA methyltransferase (MGMT) unmethylation (24.7 [95% CI, 20.5-28.8] months) compared with temozolomide (17.4 [95% CI, 14.1-20.7] months; HR, 0.57 [95% CI, 0.37-0.87]; P = .008). Seizure and influenzalike symptoms were more common in the temozolomide plus interferon alfa group, with 2 of 100 (2.0%) and 5 of 100 (5.0%) patients with grades 1 and 2 toxic effects, respectively (P = .02). Finally, results suggested that methylation level at the IFNAR1/2 promoter was a marker of sensitivity to temozolomide plus interferon alfa. Conclusions and relevance: Compared with the standard regimen, temozolomide plus interferon alfa treatment could prolong the survival time of patients with HGG, especially the MGMT promoter unmethylation variant, and the toxic effects remained tolerable. Trial registration: ClinicalTrials.gov Identifier: NCT01765088.
Article
Glioma is often referred to as one of the most dreadful central nervous system (CNS)-specific tumors with rapidly-proliferating cancerous glial cells, accounting for nearly half of the brain tumors at an annual incidence rate of 30-80 per a million population. Although glioma treatment remains a significant challenge for researchers and clinicians, the rapid development of nanomedicine provides tremendous opportunities for long-term glioma therapy. However, several obstacles impede the development of novel therapeutics, such as the very tight blood-brain barrier (BBB), undesirable hypoxia, and complex tumor microenvironment (TME). Several efforts have been dedicated to exploring various nanoformulations for improving BBB permeation and precise tumor ablation to address these challenges. Initially, this article briefly introduces glioma classification and various pathogenic factors. Further, currently available therapeutic approaches are illustrated in detail, including traditional chemotherapy, radiotherapy, and surgical practices. Then, different innovative treatment strategies, such as tumor-treating fields, gene therapy, immunotherapy, and phototherapy, are emphasized. In conclusion, we summarize the article with interesting perspectives, providing suggestions for future glioma diagnosis and therapy improvement.
Chapter
This chapter provides a timely overview of new drugs under development for glioblastoma therapy. First, we describe the challenges to drug development that are specific to glioblastoma and inherent issues that explain the high failure rate of late phase clinical trials against these tumors. We identify measures that have the potential to overcome current challenges especially if applied early during drug development, preclinical testing, and clinical translation. We focus on agents that target tumor cell-autonomous mechanisms in primary glioblastoma that represents 90%–95% of glioblastoma diagnoses. Druggable targets in glioblastoma cells are described in a topology-based fashion including agents and strategies directed at (i) DNA, DNA repair, and DNA integrity including novel approaches to DNA alkylation, PARP, DNA protein kinase, and telomerase inhibition; (ii) epigenetic determinants of chromatin structure and gene transcription, including histone deacetylase and methylase inhibitors; (iii) modulators of cell cycle and apoptosis, including CDK4/6 inhibitors, drugs that interfere with the mitotic spindle, MDM2, Wee-1 and protein phosphatase 2A inhibitors and caspase activators; (iv) protein homeostasis, including proteasome and neddylation inhibitors; and (v) upstream cellular signaling pathways, including EGFR, PI3K/AKT/mTOR and MEK inhibitors. We also briefly discuss drug delivery and synthetic lethality approaches that may open new avenues for otherwise potentially effective drugs.
Article
Full-text available
Background and Aim: Guidelines recommend adjuvant treatment for patients with high-grade gliomas and low-grade gliomas with high risk of progression. In clinical practice, however, treatments may not conform to these suggested guidelines. In this study, we reviewed the treatments and outcomes in patients with gliomas at Sun Yat-Sen University Cancer Center (SYSUCC), China. Materials and Methods: Medical records and radiologic images of 1215 glioma patients who underwent surgery at the center from 2000 to 2017 were retrospectively reviewed, and their clinicopathological characteristics, treatment method, and overall survival (OS) were analyzed. The study was approved by the Ethics Committee of SYSUCC on February 20, 2019 (approval No. GZR2019-219). Results: A total of 1001 patients diagnosed with glioma (initially World Health Organization 2007 criteria, then 2016 criteria) were enrolled, including 90 patients with Grade I, 307 Grade II, 239 Grade III, and 365 Grade IV gliomas. A total of 331 of 604 patients with high-grade glioma (54.8%) and 159 of 397 with low-grade glioma (40.1%) received postsurgical radiotherapy, and 285 patients with high-grade tumors (47.1%) and 80 with low-grade tumors (20.2%) received adjuvant chemotherapy. The median OS was 17.5 months for Grade IV and 43.1 months for Grade III gliomas. The median OS of patients with low-grade glioma was not reached. The 5-year survival rates of patients with Grades I, II, III, and IV gliomas were 94.7%, 73.7%, 45.7%, and 18.6%, respectively. Multivariate analysis identified onset age, preoperative seizure, tumor location, pathological subtype, resection extent, and postsurgical treatment as independent predictors of OS in patients with high-grade gliomas. Patients with high-grade glioma who received postsurgical treatment had better survival than those without adjuvant therapy (Grade III: 52.6 vs. 20.3 months, P = 0.012; Grade IV: 22.6 vs. 12.1 months, P < 0.001). Among patients with diffuse low-grade gliomas, age, performance status, preoperative seizure, Ki-67 index, tumor subtype, and resection extent were associated with clinical outcomes. Conclusion: Glioma patients are not always treated according to guidelines. Although standard care may lead to favorable prognoses, individualized treatments may be more acceptable and result in better outcomes and should thus be considered in routine clinical practice.
Article
Full-text available
1,2:5,6-Dianhydrogalactitol (DAG) is a bifunctional DNA-targeting agent causing N7-guanine alkylation and inter-strand DNA crosslinks currently in clinical trial for treatment of glioblastoma. While preclinical studies and clinical trials have demonstrated antitumor activity of DAG in a variety of malignancies, understanding the molecular mechanisms underlying DAG-induced cytotoxicity is essential for proper clinical qualification. Using non-small cell lung cancer (NSCLC) as a model system, we show that DAG-induced cytotoxicity materializes when cells enter S phase with unrepaired N7-guanine DNA crosslinks. In S phase, DAG-mediated DNA crosslink lesions translated into replication-dependent DNA double-strand breaks (DSBs) that subsequently triggered irreversible cell cycle arrest and loss of viability. DAG-treated NSCLC cells attempt to repair the DSBs by homologous recombination (HR) and inhibition of the HR repair pathway sensitized NSCLC cells to DAG-induced DNA damage. Accordingly, our work describes a molecular mechanism behind N7-guanine crosslink-induced cytotoxicity in cancer cells and provides a rationale for using DAG analogs to treat HR-deficient tumors.
Article
Full-text available
A formula has been developed to predict creatinine clearance (Ccr) from serum creatinine (Scr) in adult males: (see article)(15% less in females). Derivation included the relationship found between age and 24-hour creatinine excretion/kg in 249 patients aged 18-92. Values for Ccr were predicted by this formula and four other methods and the results compared with the means of two 24-hour Ccr's measured in 236 patients. The above formula gave a correlation coefficient between predicted and mean measured Ccr's of 0.83; on average, the difference predicted and mean measured values was no greater than that between paired clearances. Factors for age and body weight must be included for reasonable prediction.
Article
Full-text available
Survival for glioblastoma (GBM) patients with an unmethyated MGMT promoter in their tumor is generally worse than methylated MGMT tumors, as temozolomide (TMZ) response is limited. How to better treat patients with unmethylated MGMT is unknown. We performed a trial combining erlotinib and bevacizumab in unmethylated GBM patients after completion of radiation (RT) and TMZ. GBM patients with an unmethylated MGMT promoter were trial eligible. Patient received standard RT (60 Gy) and TMZ (75 mg/m(2) × 6 weeks) after surgical resection of their tumor. After completion of RT they started erlotinib 150 mg daily and bevacizumab 10 mg/kg every 2 weeks until progression. Imaging evaluations occurred every 8 weeks. The primary endpoint was overall survival. Of the 48 unmethylated patients enrolled, 46 were evaluable (29 men and 17 women); median age was 55.5 years (29-75) and median KPS was 90 (70-100). All patients completed RT with TMZ. The median number of cycles (1 cycle was 4 weeks) was 8 (2-47). Forty-one patients either progressed or died with a median progression free survival of 9.2 months. At a follow up of 33 months the median overall survival was 13.2 months. There were no unexpected toxicities and most observed toxicities were categorized as CTC grade 1 or 2. The combination of erlotinib and bevacizumab is tolerable but did not meet our primary endpoint of increasing survival. Importantly, more trials are needed to find better therapies for GBM patients with an unmethylated MGMT promoter.
Article
Full-text available
Background: Survival outcomes for patients with glioblastoma remain poor, particularly for patients with unmethylated O(6)-methylguanine-DNA methyltransferase (MGMT) gene promoter. This phase II, randomized, open-label, multicenter trial investigated the efficacy and safety of 2 dose regimens of the selective integrin inhibitor cilengitide combined with standard chemoradiotherapy in patients with newly diagnosed glioblastoma and an unmethylated MGMT promoter. Methods: Overall, 265 patients were randomized (1:1:1) to standard cilengitide (2000 mg 2×/wk; n = 88), intensive cilengitide (2000 mg 5×/wk during wk 1-6, thereafter 2×/wk; n = 88), or a control arm (chemoradiotherapy alone; n = 89). Cilengitide was administered intravenously in combination with daily temozolomide (TMZ) and concomitant radiotherapy (RT; wk 1-6), followed by TMZ maintenance therapy (TMZ/RT→TMZ). The primary endpoint was overall survival; secondary endpoints included progression-free survival, pharmacokinetics, and safety and tolerability. Results: Median overall survival was 16.3 months in the standard cilengitide arm (hazard ratio [HR], 0.686; 95% CI: 0.484, 0.972; P = .032) and 14.5 months in the intensive cilengitide arm (HR, 0.858; 95% CI: 0.612, 1.204; P = .3771) versus 13.4 months in the control arm. Median progression-free survival assessed per independent review committee was 5.6 months (HR, 0.822; 95% CI: 0.595, 1.134) and 5.9 months (HR, 0.794; 95% CI: 0.575, 1.096) in the standard and intensive cilengitide arms, respectively, versus 4.1 months in the control arm. Cilengitide was well tolerated. Conclusions: Standard and intensive cilengitide dose regimens were well tolerated in combination with TMZ/RT→TMZ. Inconsistent overall survival and progression-free survival outcomes and a limited sample size did not allow firm conclusions regarding clinical efficacy in this exploratory phase II study.
Article
Background: The GLARIUS trial which investigated the efficacy of bevacizumab (BEV)/irinotecan (IRI) as compared to standard temozolomide (TMZ) in the first-line therapy of MGMT-nonmethylated glioblastoma showed that progression-free survival was significantly prolonged by BEV/IRI while overall survival was similar in both arms. The present report focusses on quality of life (QoL) and Karnofsky performance score (KPS) during the whole course of the disease. Patients and methods: Patients (n=170) received standard radiotherapy and were randomized (2:1) for BEV/IRI or standard TMZ. At least every three months KPS was determined and QoL was measured using the EORTC-QLQ C30 and BN20 questionnaires. A generalized estimating equation model (GEE) evaluated differences in the course of QoL and KPS over time. Also, the time to first deterioration and the time to postprogression deterioration was analyzed separately. Results: In all dimensions of QoL and KPS, GEE analyses and time to first deterioration analyses did not detect significant differences between the treatment arms. At progression, 82% of patients receiving second-line therapy in the standard arm received BEV second-line therapy. For the dimensions motor dysfunction and headaches, time to postprogression deterioration was prolonged in the standard arm receiving crossover second-line BEV in the vast majority of patients at the time of evaluation. Conclusions: GLARIUS did not find indications for a BEV-induced detrimental effect on QoL in first-line therapy of MGMT-nonmethylated GBM patients. Moreover, GLARIUS provided some indirect corroborative data supporting the notion that BEV may have beneficial effects upon QoL in relapsed GBM.
Article
Background.: Biomarkers can improve clinical trial efficiency, but designing and interpreting biomarker-driven trials require knowledge of relationships among biomarkers, clinical covariates, and endpoints. We investigated these relationships across genomic subgroups of glioblastoma (GBM) within our institution (DF/BWCC), validated results in The Cancer Genome Atlas (TCGA), and demonstrated potential impacts on clinical trial design and interpretation. Methods.: We identified genotyped patients at DF/BWCC, and clinical associations across 4 common GBM genomic biomarker groups were compared along with overall survival (OS), progression-free survival (PFS), and survival post-progression (SPP). Significant associations were validated in TCGA. Biomarker-based clinical trials were simulated using various assumptions. Results.: Epidermal growth factor receptor (EGFR)(+) and p53(-) subgroups were more likely isocitrate dehydrogenase (IDH) wild-type. Phosphatidylinositol-3 kinase (PI3K)(+) patients were older, and patients with O6-DNA methylguanine-methyltransferase (MGMT)-promoter methylation were more often female. OS, PFS, and SPP were all longer for IDH mutant and MGMT methylated patients, but there was no independent prognostic value for other genomic subgroups. PI3K(+) patients had shorter PFS among IDH wild-type tumors, however, and no DF/BWCC long-term survivors were either EGFR(+) (0% vs 7%, P = .014) or p53(-) (0% vs 10%, P = .005). The degree of biomarker overlap impacted the efficiency of Bayesian-adaptive clinical trials, while PFS and OS distribution variation had less impact. Biomarker frequency was proportionally associated with sample size in all designs. Conclusions.: We identified several associations between GBM genomic subgroups and clinical or molecular prognostic covariates and validated known prognostic factors in all survival periods. These results are important for biomarker-based trial design and interpretation of biomarker-only and nonrandomized trials.
Article
Dianhydrogalactitol (VAL-083) is a unique bi-functional alkylating agent causing methylation of N7-guanine and inter-strand DNA crosslinks. VAL-083 is a small water-soluble molecule that readily crosses the blood-brain-barrier. In China, VAL-083 is approved as a chemotherapeutic drug for the treatment of chronic myelogenous leukemia (CML) and lung cancer. In the United States, VAL-083 has been evaluated in more than 40 National Cancer Institute (NCI)-sponsored phase I and phase II clinical trials. Preclinical studies and clinical trial data suggested antineoplastic effects of VAL-083 in a variety of malignancies, including lung cancer, brain tumors, leukemia, cervical cancer, and ovarian cancer. Here we report new insight into VAL-083 mechanism of action by showing that VAL-083 leads to irreversible cell cycle arrest and cell death caused by replication-dependent DNA damage. In lung (H2122, H1792, H23 and A549) and prostate (PC3 and LNCaP) cancer cell lines, VAL-083 treatment caused irreversible cell cycle arrest in late S and G2 phase as measured by propidium iodide (PI) and immunofluorescent (IF) staining in synchronized cultures. Importantly, VAL-083 was cytotoxic to all cell lines tested (IC50 range 3.06 - 25.7 μM). Western blot and IF analyses of DNA repair markers were employed to investigate the DNA damage response induced by VAL-083 in cancer cells. VAL-083 treatment led to phosphorylation of the proximal DNA double-strand break (DSB) sensor Ataxia Telangiectasia Mutated kinase (ATM), the single-strand DNA-binding Replication Protein A (RPA32), and the histone variant H2A.X (γH2A.X). Importantly, the DNA damage was specific to cells in S phase indicating that VAL-083-induced DNA cross-links translates into more severe DNA lesions during replication. Furthermore, S/G2 phase cell cycle arrest and increased phosphorylation of γH2A.X in cancer cells persisted after pulse-treatment with VAL-083, indicating irreversible DNA lesions. Taken together, VAL-083 displayed broad anti-neoplastic activity in lung and prostate cancer cells through the induction of replication-dependent DNA damage. Elucidation of the molecular mechanisms underlying VAL-083 cytotoxicity in cancer cells will offer help in identifying and predicting efficacy of combination treatments. Citation Format: Beibei Zhai, Anne Steino, Jeffrey Bacha, Dennis Brown, Mads Daugaard. Molecular mechanisms of dianhydrogalactitol (VAL-083) in cancer treatment. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 2985.
Article
The standard of care for glioblastoma multiforme (GBM) patients is surgical resection followed by temozolomide (TMZ) and irradiation (XRT). However, TMZ-resistance has emerged as a significant unmet medical need, as DNA repair enzyme 0-6-methylguanine DNA methyltransferase (MGMT) removes the methyl-group adducts caused by TMZ. Dianhydrogalactitol (VAL-083) is a structurally unique alkylating agent causing DNA crosslinks at N7 position of guanine. Because VAL-083's N7 adducts are not subject to MGMT mediated repair, it may be an effective chemotherapeutic in the treatment of TMZ-resistant GBM. VAL-083 crosses the blood brain barrier and accumulates in brain tumor tissue. We have recently shown that TMZ activity is similar in cancer stem cells (CSC) and their paired non-CSC from primary GBM tissues, and that the activity is MGMT-dependent. We thus sought to investigate how our CSC and non-CSC panel would respond to VAL-083 alone or in combination with XRT. We further investigated the activity of VAL-083 in in vivo models of drug-resistant GBM in comparison to TMZ. Rag2 mice bearing intracranial human GBM xenograft tumors of either MGMT-positive and TMZ-resistant origin (BT74), or MGMT-negative and TMZ-sensitive origin (U251) were treated. VAL-083 was given i.p. 3 times/week x 3 weeks, and the efficacy of VAL-083 in controlling tumor growth compared to TMZ (30 mg/kg). Disease progression was evaluated by overall survival, clinical observations and body weight measurements. Our in vitro results show that VAL-083 is a potent inhibitor of all tested primary GBM cultures, irrespective of MGMT status. VAL-083 causes cell cycle arrest and loss of cell viability in TMZ-resistant cells, and at lower concentrations than TMZ in TMZ-sensitive cells. Furthermore, VAL-083 is not affected by cell culture condition (Stem vs. Non-Stem). Low dose VAL-083 combined with XRT exhibited an additive effect in all cultures tested, suggesting that VAL-083 might act as a radiosensitizer. In the in vivo U251 model, the median survival time for mice treated with 4 mg/kg VAL-083 was significantly increased to 72 days compared to 48 days for controls (p<0.0001). Median survival time for 3 mg/kg VAL-083 was 54 days. Body weight loss was observed in mice treated with 5 mg/kg and treatment was stopped after 4 doses after which the animals recovered and their median survival was 57 days. Animals treated with TMZ were terminated at day 102 at the end of the study. BT74: study is ongoing, data will be presented at the meeting.In conclusion, VAL-083 is highly efficacious against both stem and non-stem GBM cell cultures in vitro, the activity is independent of MGMT and VAL-083 appears to act as a radiosensitizer in GBM. In vivo xenograft GBM models further validate the benefits of VAL-083 in the treatment of GBM and support ongoing clinical research with VAL-083, which is currently in a clinical trial for GBM patients with recurrent disease. Citation Format: Shaun D. Fouse, Anne Steino, Nicholas Butowski, Jeffrey A. Bacha, Sarath Kanekal, Nancy Dos Santos, Dennis M. Brown, Joseph F. Costello. Dianhydrogalactitol inhibits the growth of glioma stem and non-stem cultures, including temozolomide-resistant cell lines, in vitro and in vivo. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 2562. doi:10.1158/1538-7445.AM2015-2562
Article
Glioblastoma (GBM) remains one of the most difficult tumors to treat in part because many new agents fail to cross the blood brain barrier (BBB) and secondly due to intrinsic drug resistance. Temozolomide (TMZ) is a front-line therapy for the treatment of GBM, however, it is often ineffective due to drug inactivation by O6-methylguanine-DNA methyltransferase (MGMT). Cancer stem cells (CSC) are a subpopulation of the tumor that resist therapy and give rise to relapse. Here we described VAL083 a novel alkylating agent that creates N7 methylation on DNA, which was initially intriguing because it crosses the BBB. We addressed how it compared to TMZ, whether it could be used to overcome MGMT-driven drug resistance and if it has activity against CSCs. Addressing these questions provides further preclinical support for VAL083, which is currently undergoing human clinical trials in the USA against refractory GBM. VAL083 inhibited U251 and SF188 cell growth in monolayer and as neurospheres better then TMZ and caused apoptosis after 72 hrs. In a 10-day colony formation assay, VAL083 (5uM) suppressed SF188 growth by ∼95%. T98G cells are classically TMZ resistant and express MGMT yet VAL083 inhibited their growth in monolayer after 72 hrs in a dose-dependent manner (IC50<5 uM). VAL083 also inhibited the growth of CSCs by 100% in neurosphere growth assays. In summary, VAL083 has better in vitro efficacy than TMZ against brain tumor cells, can overcome resistance associated with MGMT, and targets brain tumor CSCs demonstrating that it has the potential to surpass the standard-of-care. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 811. doi:1538-7445.AM2012-811