This study has been transitioned to CTIS with ID 2024-510691-20-00 check the CTIS register for the current data. The primary objective of Arm 1 is to evaluate the overall response rate (ORR) as determined by an independent radiology review committee…
ID
Source
Brief title
Condition
- Nervous system neoplasms malignant and unspecified NEC
Synonym
Research involving
Sponsors and support
Intervention
Outcome measures
Primary outcome
ORR, defined as the proportion of patients with best overall confirmed response
of complete response (CR) or partial response (PR) as determined by the
Response Assessment in Neuro-Oncology (RANO) criteria
Secondary outcome
Type, frequency, and severity of AEs and laboratory abnormalities
Pharmacokinetic profile of DAY101 (e.g., area under the concentration-time
curve [AUC], Cmin, etc.)
Change from baseline QT interval corrected for HR by Fridericia*s formula
(ΔQTcF)
Change from baseline PR interval (ΔPR)
Change from baseline QRS interval (*QRS)
Change from baseline heart rate (*HR)
ECG waveform morphology
Measured by the proportion of patients with best overall confirmed response of
CR or PR by RANO criteria
Measured by the proportion of patients with best overall confirmed response of
CR or PR by RAPNO-LGG criteria
Measured by the time following initiation of DAY101 to progression or death in
patients treated with DAY101
Measured by the length of response in patients with best overall confirmed
response of CR or PR by RANO criteria
Measured by the time to first response following initiation of DAY101 in
patients with best overall confirmed response of CR or PR by RANO criteria
Measured on the proportion of patients with best overall response of CR, PR, or
SD lasting 12 months or more following initiation of DAY101
Measured by Teller Acuity Cards®II
Molecular analysis of cells obtained from archival tissue
Background summary
Relapsed or progressive pLGG is a serious, life-altering, and potentially fatal
disease with significant disease- and treatment-associated morbidity. There is
currently no standard of care for patients who progress after initial therapy
and no FDA-approved therapies. As noted in Section 1.1, a significant body of
data clearly shows that a majority of patients with pLGG also have an
alteration in BRAF that drives tumor growth: either a KIAA1549:BRAF wild-type
fusion, or a BRAF V600 mutation. Type I BRAF inhibitors are mechanistically
only able to inhibit the BRAF V600 mutations. Using a Type I RAF inhibitor in a
patient with a BRAF fusion has the potential to cause paradoxical activation of
the RAS/RAF/MAPK pathway and tumor growth. Because the majority of BRAF
alterations in patients with pLGG are BRAF wild-type fusions, use of Type I RAF
inhibitors is not indicated. Currently approved MEK inhibitors can block MAPK
signaling resulting from either BRAF V600 mutations or the KIAA1549:BRAF
wild-type fusion, but do not directly inhibit BRAF. In addition, MEK inhibitors
are associated with significant cardiac, dermatologic, and ophthalmologic
toxicities. The Type II BRAF inhibitor DAY101 has been shown preclinically to
be able to inhibit both BRAF V600 and BRAF fusion alterations, and has been
shown to have significantly greater CNS penetration than any of the available
Type I RAF inhibitors or MEK inhibitors. To date, DAY101 has been administered
to 9 pediatric patients with pLGG in the ongoing Phase 1 study PNOC014. Eight
of the 9 patients harbored a RAF fusion (7 harbored the classic KIAA1549:BRAF
fusion and 1 harbored a SRGAP3-RAF1 gene fusion). Five of 8 (63%) patients with
a RAF fusion have achieved a CR or PR with a median time-to-response of 10.5
weeks. DAY101 has been well-tolerated thus far in pediatric patients, with no
dose-limiting toxicities (DLTs) having been observed, despite dosing at or
above the BSA-adjusted adult MTD.
The purpose of this study is to evaluate the efficacy and safety of DAY101 as
monotherapy in pediatric patients with a recurrent or progressive LGG harboring
a known activating BRAF alteration. The primary endpoint is ORR as defined by
the RANO criteria, determined by an IRC.
Study objective
This study has been transitioned to CTIS with ID 2024-510691-20-00 check the CTIS register for the current data.
The primary objective of Arm 1 is to evaluate the overall response rate (ORR)
as determined by an independent radiology review committee (IRC) and measured
by the proportion of patients with best overall confirmed response of complete
response (CR) or partial response (PR) by Response Assessment in Neuro-Oncology
(RANO) criteria following treatment with DAY101 in pediatric patients aged 6
months to 25 years, inclusive with a relapsed or progressive low-grade glioma
harboring a known activating v-raf murine sarcoma viral oncogene homolog B
(BRAF) alteration.
ARM 2 (LOW-GRADE GLIOMA EXTENSION) OBJECTIVES
Primary Objective:
The primary objective of Arm 2 is to assess the safety and tolerability of
DAY101 in pediatric patients aged 6 months to 25 years, inclusive, with a
relapsed or progressive low-grade glioma harboring a known or expected to be
activating RAF alteration.
ARM 3 (ADVANCED SOLID TUMOR) OBJECTIVES
Primary Objective:
The primary objective of Arm 3 is to evaluate the preliminary efficacy of
DAY101 as measured by the ORR as determined by an IRC following treatment with
DAY101 in pediatric patients aged 6 months to 25 years, inclusive, with a
relapsed or progressive advanced solid tumor harboring a known or expected to
be activating RAF fusion.
Study design
STUDY DESIGN:
This is a Phase 2, multicenter, multi-arm, open-label study evaluating DAY101
in pediatric patients with
low-grade gliomas and advanced solid tumors. The study will consist of the
following treatment arms:
• Arm 1 (Low-Grade Glioma): Patients aged 6 months to 25 years, inclusive, with
recurrent or
progressive low-grade glioma harboring a known activating BRAF alteration,
including BRAF V600 mutations and KIAA1549:BRAF fusions. Patients with BRAF
alterations will be identified through molecular assays as routinely performed
at Clinical Laboratory Improvement Amendments (CLIA) of 1988 or other similarly
certified laboratories.
• Arm 2 (Low-Grade Glioma Extension): Patients aged 6 months to 25 years,
inclusive, with
recurrent or progressive low-grade glioma harboring a known or expected to be
activating RAF alteration (e.g., BRAF or CRAF/RAF1 fusion or BRAF V600
mutations). Patients with RAF alterations will be identified through molecular
assays as routinely performed at CLIA-certified or other similarly certified
laboratories. Opening of Arm 2 to enrollment will be based on the
recommendation of the Data Safety Monitoring Board (DSMB) as described in the
protocol.
• Arm 3 (Advanced Solid Tumor): Patients aged 6 months to 25 years, inclusive,
with advanced
solid tumors harboring a known or expected to be activating RAF fusion (e.g.,
BRAF or CRAF/RAF1 fusion). Patients with RAF fusions will be identified through
molecular assays as routinely performed at CLIA-certified or other similarly
certified laboratories.
For all arms, treatment cycles will repeat every 28 days in the absence of
disease progression or unacceptable toxicity. Patients will continue on DAY101
until radiographic evidence of disease progression as determined by treating
investigator, unacceptable toxicity, decision to enter a *drug holiday* period,
patient withdrawal of consent, or death.
Patients will be treated with DAY101 for a planned period of 26 cycles
(approximately 24 months), after which they may continue on DAY101 or, at any
point, opt to enter a *drug holiday* discontinuation period. Patients will
continue to undergo routine periodic radiographic evaluations per
protocol-defined timelines during the discontinuation period and may be
re-treated with DAY101 if there is radiographic evidence of disease progression
after drug discontinuation.
Patients who have radiographic evidence of disease progression may be allowed
to continue DAY101 if, in the opinion of the investigator, and approved by the
Sponsor, the patient is deriving clinical benefit from continuing study
treatment. Patients being treated beyond progression with DAY101 will be
reconsented prior to continuation of therapy. Disease assessments for patients
being treated beyond progression should continue as per regular schedule.
The study will consist of a screening period, a treatment period, an end of
treatment (EOT) visit, a safety follow-up visit, and long-term follow-up
assessments. Ongoing safety, disease stability/progression, survival, and
subsequent anticancer therapies will be assessed in the long-term follow-up
period.
DAY101 will be administered at the recommended Phase 2 dose (RP2D) of 420 mg/m2
(not to exceed
600 mg), orally (PO) once weekly (QW) (Days 1, 8, 15, and 22 of a 28-day
cycle). Body surface area (BSA) will be determined by the Mösteller Formula
[*(height × weight)/3600)]. DAY101 is administered as an oral tablet or
reconstituted liquid suspension formulation.
Intervention
DAY101 for oral dosing is provided as an immediate-release tablet in one
strength, 100 mg. The 100-mg tablets are red to yellowish red oval tablets and
are labeled DAY101.
Study burden and risks
Blood draws.
2 year study duration with around 30 visits.
Study procedures.
Keeping of Dosing Diaries.
Risks of the procedures.
Side effects of the medication.
Sierra Point Parkway Suite 501 2000
Brisbane CA 94005
US
Sierra Point Parkway Suite 501 2000
Brisbane CA 94005
US
Listed location countries
Age
Inclusion criteria
1. Patients must be age 6 months to 25 years, inclusive, with:
a) Arm 1 (Low-Grade Glioma): A relapsed or progressive low-grade glioma with a
documented known activating BRAF alteration, as identified through molecular
assays as routinely performed at CLIA or other similarly certified laboratories
b) Arm 2 (Low-Grade Glioma Extension): A relapsed or progressive low-grade
glioma with a documented known or expected to be activating BRAF mutation or
RAF fusion, as identified through molecular assays as routinely performed at
CLIA-certified or other similarly certified laboratories
c) Arm 3 (Advanced Solid Tumor): Locally advanced or metastatic solid tumor with
a documented known or expected to be activating RAF fusion, as identified
through molecular assays as routinely performed at CLIA-certified or other
similarly certified laboratories, that has relapsed or progressed or was
nonresponsive to available therapies and for which no standard or available
systemic curative therapy exists
2. Patients must have histopathologic verification of malignancy at either
original diagnosis
or relapse.
3. Patients must have received at least one line of prior systemic therapy and
have
documented evidence of radiographic progression.
4. Patients must have evaluable and/or measurable disease (imaging must be
performed
within 28 days of the initiation of treatment) as specified below:
a) Arm 1 (Low-Grade Glioma): Must have at least one measurable lesion as defined
by RANO criteria (T1-weighted lesion that can be reproducibly measured in at
least 2 dimensions of at least 10 mm, visible on 2 or more axial slices that
are preferably, at most, 5 mm apart with 0-mm skip)
b) Arm 2 (Low-Grade Glioma Extension): Must have evaluable (either
unidimensionally measurable lesions, masses with margins not clearly defined,
or lesions with maximal perpendicular diameters less than 10 mm) and/or
measurable disease as defined by RANO criteria
c) Arm 3 (Advanced Solid Tumor): Must have at least one measurable lesion as
defined by RECIST v1.1 (>= 10 mm by CT/MRI scan [slice thickness <= 5 mm],
>= 20 mm by chest X-ray, or >= 10 mm with calipers by clinical exam, or
pathologic lymph nodes with a short axis of >= 15 mm by CT scan/MRI)
5. Radiation therapy to the measurable lesion(s) must be completed at least six
months prior
to administration of DAY101. Patients who have documented radiographic
progression less than six months from radiotherapy in one or more measurable
lesions are eligible.
6. Patients must have Karnofsky (those 16 years and older) or Lansky (those
younger than
16 years) performance score of at least 50. Patients who are unable to walk
because of paralysis, but who are able to sit in a wheelchair, will be
considered ambulatory for the purpose of assessing the performance score.
7. Patients must have fully recovered from the acute toxic effects of all prior
anticancer
chemotherapy and have undergone the following washout periods, as applicable:
a) Myelosuppressive chemotherapy: At least 21 days must have elapsed after the
last
dose of myelosuppressive chemotherapy (42 days if prior nitrosourea).
b) Radiation therapy (XRT): At least 14 days must have elapsed after the last
dose
fraction of XRT.
c) Stem cell transplant or adoptive cell therapy: At least 100 days must have
elapsed
after cell infusion.
d) Investigational agent or any other anticancer therapy not defined above: At
least
four weeks prior to planned start of DAY101, or five half-lives, whichever is
shorter.
8. Chronic toxicities from prior anticancer therapy must be stable and at Common
Terminology Criteria for Adverse Events (CTCAE) version 5.0 Grade <= 2.
a) Ongoing retinopathy must be <= Grade 1.
9. Patients must have fully recovered from any prior surgery.
10. An archival tumor tissue sample must be available. If an archival tumor
tissue sample is
not available, a fresh biopsy should be performed at baseline. For patients
enrolling to Arm 2 (Low-Grade Glioma Extension) that do not have archival tumor
tissue, enrollment may be considered on a case-by-case basis following a
discussion between the investigator and the Day One Medical Monitor.
11. Patients must have adequate hematologic function, as defined by the
following:
a) Absolute neutrophil count >= 1000/mm3
b) Platelet count >= 75.0 ** 109/L (transfusions allowed per institutional
guidelines;
last transfusion > 2 weeks prior to C1D1)
c) Hemoglobin >= 10.0 g/dL (transfusions allowed per institutional guidelines;
last
transfusion > 4 weeks prior to C1D1)
d) Hematopoietic growth factors: At least 14 days after the last dose of a
long-acting
growth factor (e.g., Neulasta®) or seven days for short-acting growth factor
12. Patients must have adequate hepatic and renal function, defined by the
following:
a) Total bilirubin <= 1.5 ** upper limit of normal (ULN) for age (patients with
documented Gilbert*s Disease may be enrolled with Sponsor approval and total
bilirubin <= 2.0 ** ULN)
b) Serum glutamic-pyruvic transaminase (SGPT)/alanine aminotransferase (ALT)
<= 2.5 ** ULN
c) Serum glutamic-oxaloacetic transaminase (SGOT)/aspartate transaminase (AST)
<= 2.5 ** ULN
d) Serum creatinine within normal limits, or estimated glomerular filtration
rate
>= 60 mL/min/1.73 m2 based on local institutional practice for determination
13. Thyroid function tests must be consistent with stable thyroid function.
Patients on a stable
dose of thyroid replacement therapy for a minimum of three weeks before
starting DAY101 are eligible.
14. Patients must have left ventricular ejection fraction (LVEF) of >= 50% as
measured by
echocardiogram (ECHO) or multiple-gated acquisition (MUGA) scan, or fractional
shortening (FS) >= 25% (Tissot et al., 2018) as measured by ECHO, within 28 days
before the first dose of DAY101. If normal practice at the institution is to
provide the LVEF result as a range of values, then the upper value of the range
will be used to determine the result.
15. Patients receiving steroids for tumor-associated symptoms must be on a
stable dose (e.g.,
no initial/loading dose, no increase or decrease) for 14 days prior to C1D1.
16. Patients must be able to comply with treatment, laboratory monitoring, and
required
clinic visits for the duration of study participation.
17. Male and female patients with reproductive potential must be willing to use
a highly
effective birth control method for the duration of treatment and for 180 days
following the last dose of study drug. Highly effective birth control methods
are described in Appendix K.
18. Patients must have ability to swallow tablets or liquid or be willing to
comply with
administration of a nasal or gastric tube for gastric access.
19. Parent/guardian of child or adolescent patient must have the ability to
understand, agree
to, and sign the study ICF and applicable pediatric assent form before
initiation of any protocol-related procedures; patient must have the ability to
give assent, as applicable, at the time of parental/guardian consent.
Exclusion criteria
Patient*s tumor has an additional previously known or expected to be activating
molecular alteration(s) (e.g., histone mutation, IDH1/2 mutations, FGFR
mutations or fusions, MYBL alterations, NF-1 somatic or germline mutations).
2. Patient has symptoms of clinical progression without radiographically
recurrent or
radiographically progressive disease.
Patient has known or suspected diagnosis of neurofibromatosis type 1 (NF-1)
via genetic
testing or current diagnostic criteria.
4. Patient has history of any major disease, other than the primary malignancy
under study,
that might interfere with safe protocol participation.
5. Patient has a history or current evidence of central serous retinopathy
(CSR), retinal vein
occlusion (RVO), or ophthalmopathy present at baseline that would be considered
a risk factor for CSR or RVO. Ophthalmological findings secondary to
long-standing optic pathway glioma (such as visual loss, optic nerve pallor, or
strabismus) will NOT be considered significant abnormalities for the purposes
of this study.
6. Patient has major surgery within 14 days (two weeks) prior to C1D1 (does not
include
central venous access, cyst fenestration or cyst drainage, or
ventriculoperitoneal shunt placement or revision).
7. Patient has clinically significant active cardiovascular disease, or history
of myocardial
infarction, or deep vein thrombosis/pulmonary embolism within six months prior
to C1D1, ongoing cardiomyopathy, or current prolonged QT interval corrected for
heart rate by Fridericia*s formula (QTcF) interval > 470 milliseconds based on
triplicate electrocardiogram (ECG) average.
8. Patient is currently enrolled in any other investigational treatment study.
Participation in
a concurrent observational or bio-sampling study is allowed.
9. Patient has active systemic bacterial, viral, or fungal infection.
10. Patient has nausea and vomiting >= National Cancer Institute (NCI) Common
Terminology Criteria for Adverse Events (CTCAE) v5.0 Grade 2, malabsorption
requiring supplementation, or significant bowel or stomach resection that would
preclude adequate absorption of DAY101.
11. Patient is neurologically unstable despite adequate treatment (e.g.,
uncontrolled seizures). 12. Patient is currently being treated with a strong
CYP2C8 inhibitor or inducer other than
those allowed per Section 5.3.2. Medications that are substrates of CYP2C8 are
allowed
but should be used with caution.
13. Patient is pregnant or lactating.
14. Patient has a history of any drug reaction with eosinophilia and systemic
symptoms
(DRESS) syndrome or Stevens Johnsons syndrome (SJS), or hypersensitivity to the
investigational medicinal product or to any drug with similar chemical
structure or to any other excipient present in the pharmaceutical form of the
investigational medicinal product.
15. There are other unspecified reasons that, in the opinion of the
investigator, make the
patient unsuitable for enrollment.
Design
Recruitment
Medical products/devices used
Followed up by the following (possibly more current) registration
No registrations found.
Other (possibly less up-to-date) registrations in this register
No registrations found.
In other registers
Register | ID |
---|---|
EU-CTR | CTIS2024-510691-20-00 |
EudraCT | EUCTR2020-003657-30-NL |
ClinicalTrials.gov | NCT04775485 |
CCMO | NL77024.041.21 |