This study has been transitioned to CTIS with ID 2024-513759-34-00 check the CTIS register for the current data. Primary ObjectivesPhase 2The primary objectives of Phase 2 are:• To evaluate short- and long-term safety and tolerability of SNDX 5613…
ID
Source
Brief title
Condition
- Leukaemias
Synonym
Research involving
Sponsors and support
Intervention
Outcome measures
Primary outcome
Phase 2
Primary endpoints in Phase 2 are:
• CR+CRh rate.
• Frequency, duration, and severity of TEAEs, TRAEs, and SAEs.
• Incidence and shifts of clinically significant clinical laboratory
abnormalities.
• Change from baseline in other observations related to safety, including ECGs,
vital signs, ophthalmologic examination findings, and performance status.
Secondary outcome
Phase 2
Secondary endpoints in Phase 2 are:
• • Transfusion independence, defined as any transfusion-free period lasting
for at least 56 consecutive days, during which the patient is either on
SNDX-5613 therapy or after cessation of SNDX-5613 therapy but prior to the
start of new therapy.
• CRc rate (ie, CR+CRh+CRi+CRp).
• ORR (CRc+MLFS+PR).
• TTR.
• DOR.
• EFS.
• OS.
• PK parameters: Cmax, Tmax, AUC0-t, AUC0-24, CL/F, Vz/F, and t1/2.
Exploratory Endpoints
Phase 2
The exploratory endpoint in Phase 2 is:
• Response relationship with correlative biomarkers in blood and bone marrow.
• PK parameters: Cmax, Tmax, AUC0-t, AUC0-24, CL/F, Vz/F, t1/2.
Background summary
Acute leukemias generally result from acquired mutations in hematopoietic
progenitor cells. Chromosomal abnormalities are often discrete mutational
features in leukemia. Many of these chromosomal abnormalities are due to
specific rearrangements that lead to the formation of fusion genes, which
become drivers for tumorigenesis and tumor development (Wang 2017). A specific
example involves the KMT2A gene (Wang 2017) . Rearrangements at the KMT2A locus
(11q23) can lead to the formation of oncogenic gene fusions that characterize
KMT2Ar acute leukemias.
The KMT2A protein is a key regulator of development and is the mammalian
homologue of Drosophila trithorax. It is an important epigenetic regulator of
HOX gene expression (Del Rizzo 2011). Rearrangements at the KMT2A locus create
chimeric proteins that fuse the N-terminus of KMT2A to variable C-terminal
domains derived from different rearrangement partners. Currently, more than 90
different fusion partners are known (Meyer 2018). Expression of these fusions
enables an aberrant transcription program characterized by overexpression of
HOX and other developmental genes. This transcription program suppresses
differentiation and enhances proliferation, leading to the KMT2Ar acute
leukemias.
Rearrangements involving the KMT2A locus (11q23) are routinely diagnosed using
fluorescence in situ hybridization (FISH). Depending on the progenitor cell of
origin, KMT2Ar can phenotypically appear as acute lymphoblastic leukemia (ALL),
AML, or mixed phenotype acute leukemia (MPAL). These rearrangements are rare
and KMT2Ar has a combined annual incidence of ~4000 cases per year in the
United States (US), Europe and Japan. Approximately 10% of all leukemias harbor
KMT2A rearrangements (Winters 2017).
The relapse risk for KMT2Ar patients is high after conventional chemotherapy
and stem cell transplantation, with an overall 5-year survival rate of only
approximately 35% (Muntean 2012). No therapies are currently available that
specifically target KMT2Ar leukemia. SNDX 5613 may provide a novel, targeted
treatment for KMT2Ar acute leukemias.
Study objective
This study has been transitioned to CTIS with ID 2024-513759-34-00 check the CTIS register for the current data.
Primary Objectives
Phase 2
The primary objectives of Phase 2 are:
• To evaluate short- and long-term safety and tolerability of SNDX 5613.
• To assess the CR+CRh rate.
Secondary Objectives
Phase 2
Secondary objectives of Phase 2 are:
• • To assess postbaseline transfusion independence.
• To assess the CRc rate (CR+CRh+CRi+CRp).
• To assess the overall response rate (ORR) (CRc+MLFS+partial remission [PR]).
• To assess the time to response (TTR), duration of response (DOR), and event
free survival (EFS).
• To assess overall survival (OS).
• To characterize PK parameters of SNDX 5613 and relevant metabolites.
Exploratory Objectives
Phase 2
The exploratory objective of Phase 2 is:
• To evaluate SNDX 5613 pharmacodynamic, safety, and efficacy relationship with
correlative biomarkers, which may include immunophenotyping of circulating PBMC
and/or bone marrow, gene expression, mutational analysis, and MRD.
Study design
Study SNDX 5613-0700 is a Phase 1/2, open-label, dose-escalation, and expansion
study of SNDX 5613. Patients aged >=6 months (or >=12 years in Germany only), and
weighing >=35 kg if intended to receive SNDX-5613 in combination with
cobicistat,,with R/R acute leukemia are planned to be enrolled. This Phase 1/2
study was opened in September 2019 and at the time of this amendment, Phase 1
Arms A F are closed to enrollment. The Phase 2 portion initiated with the
identification of a recommended Phase 2 dose (RP2D) for patients receiving a
strong cytochrome P450 (CYP)3A4 inhibitor azole. At the time of this amendment,
Phase 2 Cohorts 2A and 2B are closed to enrollment. Phase 2 Cohort 2C is
currently enrolling.
The RBA assessment has also been completed, and the PK between the capsule and
tablet formulations are comparable with no change in the safety profile
observed in patients receiving tablet formulation, indicating that the
formulation switch should not result in a clinically meaningful difference. All
US patients who are able to swallow tablets have been switched to the tablet
formulation. In addition, the low fat meal food effect evaluation has been
completed; the effect of a low fat meal on PK was minimal. A description of the
Phase 1 Arms A-F elements of the protocol remains in place for reference.
Phase 1
SNDX 5613 will be investigated on an every-12-hour (q12h) schedule and a
3-times-a-day (TID) schedule; alternative dose schedules may be explored as
guided by emerging data. SNDX 5613 will be administered orally (PO) in 28-day
cycles, with the first study drug dose administered on Cycle 1, Day 1 (C1D1).
Reasons for discontinuation of Study Intervention are listed in Section 7.1.1.
Upon enrollment, patients will be assigned to one of 6 dose-escalation arms as
described below:
• Arm A: Patients must not be receiving any strong CYP3A4 inhibitor/inducers or
fluconazole. Patients who were receiving a strong CYP3A4 inhibitor/inducer or
fluconazole must have discontinued the medication at least 7 days prior to
enrollment. Patients will receive SNDX-5613 q12h.
• Arm B: Patients must be receiving above (strong CYP3A4 inhibitors) for
antifungal prophylaxis for at least 7 days prior to enrollment and while on
SNDX-5613 treatment. Patients must not be receiving any other strong CYP3A4
inhibitors/inducers.
• Arm C: Patients must receive a daily dose of cobicistat from C1D2 on.
Patients must not be receiving any other strong or moderate CYP3A4
inhibitors/inducers. Patients who were receiving a moderate/strong CYP3A4
inhibitor/inducer must have discontinued the medication at least 7 days prior
to enrollment. Patients will receive SNDX-5613 q12h.
• Arm D: Patients must be receiving fluconazole (moderate CYP3A4 inhibitor) for
antifungal prophylaxis for at least 7 days prior to enrollment and while on
SNDX-5613 treatment. Patients must not be receiving any other strong or
moderate CYP3A4 inhibitors/inducers. Patients will receive SNDX-5613 q12h.
• Arm E: Patients must not be receiving any weak, moderate, or strong CYP3A4
inhibitors/inducers. Patients who were receiving a CYP3A4 inhibitor/inducer
must have discontinued the medication at least 7 days prior to enrollment.
Patients will receive SNDX-5613 TID.
• Arm F: Patients must be receiving isavuconazole (moderate CYP3A4 inhibitor)
for antifungal prophylaxis for at least 7 days prior to enrollment and while on
SNDX-5613 treatment. Patients must not be receiving any other weak, moderate,
or strong CYP3A4 inhibitors/inducers. Patients will receive SNDX-5613 q12h.
Exclusion criteria addressing the use of moderate and strong CYP3A4 inhibitors
in each arm can be found in Section 5.2 Exclusion Criterion 15.
Throughout the study, the Safety Review Committee (SRC) will monitor safety and
efficacy parameters as specified in the SRC Charter.
An Independent Data Monitoring Committee (IDMC) has been established for the
phase 2 study and specific guidelines on the operation and purpose of the IDMC
is documented in a Charter.
Intervention
SNDX 5613 capsules (25 and 113 mg free base equivalents), SNDX-5613 tablets
(25, 110, and 160 mg), or oral solution will be taken orally or administered
through NG/G- or duodenal tube q12h or TID at the designated dose according to
patients* cohort assignments (further information is available in the Pharmacy
Manual). For patients receiving SNDX-5613 in a q12h regimen, study drug is to
be administered on an empty stomach, at least 2 hours after a meal and 1 hour
before the next meal.
In Phase 2, all patients will receive SNDX-5613 at the RP2D, as determined
based on data from Phase 1.
Patients will receive SNDX 5613 PO either q12h or TID in 28-day cycles, with
the first study drug dose administered on C1D1. More dosing schedules may be
explored based on emerging data. Reasons for discontinuation of Study
Intervention are listed in Section 7.1.1.
Study burden and risks
Overall Phase I Safety Summary:
• Grade 3 TRAE observed: anemia, diarrhea, generalized weakness, hypokalemia,
increased fatigue, platelet count decreased, QTc prolongation
• Related SAE observed: QTc prolongation, differentiation syndrome
Conclusion:
• The intermediate dose levels of 276 mg in Arm A and 163 mg in Arm B are both
safe and tolerable.
• The currently available safety data suggests a clinical QTc signal (which was
predicted based on in vitro and toxicology studies). Otherwise SNDX-5613 has
been well tolerated.
35 Gatehouse Drive Building D, Floor 3
Waltham MA 02451
US
35 Gatehouse Drive Building D, Floor 3
Waltham MA 02451
US
Listed location countries
Age
Inclusion criteria
Diagnosis
1. Patients in phase 1 Arm A and Arm B must have active acute leukemia
harboring KMT2A rearrangement or NPM1 mutation (bone marrow blasts >=5% or
reappearance of blasts in peripheral blood) as defined by the National
Comprehensive Cancer Network (NCCN) guidelines in the NCCN Clinical Practice
Guidelines in Oncology (NCCN Guidelines®) for Acute Lymphoblastic Leukemia
(Version 1.2020) and Acute Myeloid Leukemia (Version 3.2020) (National
Comprehensive Cancer Network 2020; National Comprehensive Cancer Network 2020).
Patients in phase 1 Arm C, Arm D, Arm E and Arm F must meet one of the
following 2 criteria:
• active acute leukemia (bone marrow blasts >=5% or reappearance of blasts in
peripheral blood) as defined by the National Comprehensive Cancer Network
(NCCN) in the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®)
for Acute Lymphoblastic Leukemia (Version 1.2020) and Acute Myeloid Leukemia
(Version 3.2020) (National Comprehensive Cancer Network 2020; National
Comprehensive Cancer Network 2020).
• acute leukemia harboring an KMT2A rearrangement, NUP98 rearrangement, or NPM1
mutation that have detectable disease in the bone marrow not meeting criterion
for active leukemia as described above.
2. Phase 1: see more details in the protocol.
• Note that phase 1 is not applicable for the Netherlands.
3. 3. Phase 2: Documented R/R active acute leukemia (bone marrow blasts >=5% or
reappearance of blasts in peripheral blood) as defined by the NCCN Guidelines®
for Acute Lymphoblastic Leukemia (Version 1.2020) and Acute Myeloid Leukemia
(Version 3.2020) (National Comprehensive Cancer Network 2020; National
Comprehensive Cancer Network 2020).
Cohort 2A: Documented R/R ALL/MPAL with a KMT2A rearrangement.
Cohort 2B: Documented R/R AML with a KMT2A rearrangement.
Cohort 2C: Documented R/R AML with NPM1m.
Mutational status is to be reviewed locally to determine patient eligibility in
Phase 2 and confirmed centrally. Central confirmation of KMT2Ar status will be
obtained by fluorescence in situ hybridization (11q23 MLL-Break Apart FISH
testing Flagship Biosciences, Morrisville, NC). NPM1 mutational status
confirmation will be obtained by PCR based amplification and sequencing (Focus
Myeloid panel, Flagship Biosciences, Morrisville, NC). All assays will be
conducted in a CLIA certified laboratory. Patients whose mutational status
cannot be confirmed centrally, or whose final pathology or flow reports do not
confirm the presence of >=5% bone marrow blasts, will be replaced to ensure a
sufficient number of patients for the primary efficacy analyses.
Disease Status
4. Recurrent or refractory AML/ALL or MPAL, as defined by standardized criteria
(for example, European LeukemiaNet criteria [Döhner 2017]; International
Working Group criteria [Cheson 2003]) after standard of care therapy, including
but not limited to one or two cycles of intensive chemotherapy, or venetoclax
combinations.. Patients with persistent leukemia after initial therapy or with
recurrence of leukemia at any time after achieving a response during or after
the course of treatment (including allogeneic [HSCT] are eligible. Refractory
or relapsed leukemia is defined by presence of >=5% blasts in the bone marrow
and/or persistence or reappearance of peripheral blasts. Patients who have <5%
blasts in the bone marrow at baseline may be replaced to ensure a sufficient
number of patients for the efficacy analyses.
In addition, all patients must have:
• White blood cell (WBC) count below 25,000/µL at time of enrollment. Patients
may receive cytoreduction prior to enrollment per Inclusion Criterion 11 and 14.
Age/Weight
5. Male or female patient aged >=6 months. Patients intend to receive SNDX-5613
in combination with cobicistat must weigh >=35 kg.
Performance Level
6. Eastern Cooperative Oncology Group (ECOG) performance status score 0-2 (if
aged >=18 years); Karnofsky Performance Scale of >=50 (if aged >=16 years and <18
years); Lansky Performance Score of >=50 (if aged >=12 years and <16 years).
Prior Therapy:
7. Any prior treatment-related toxicities resolved to <=Grade 1 prior to
enrollment, with the exception of <=Grade 2 neuropathy or alopecia.
8. Radiation Therapy: At least 60 days from prior total body irradiation (TBI),
craniospinal radiation and/or >=50% radiation of the pelvis, or at least 14 days
from local palliative radiation therapy (small port).
9. Stem Cell Infusion: At least 60 days must have elapsed from HSCT and at
least 4 weeks must have elapsed from donor lymphocyte infusion (DLI).
10. Immunotherapy: At least 42 days since prior immunotherapy, including tumor
vaccines, and at least 21 days since receipt of chimeric antigen receptor
therapy or other modified T or NK cell therapy.
11. Antileukemia Therapy: At least 14 days, or 5 half-lives, whichever is
shorter, since the completion of antileukemic therapy (for example, but not
limited to, small molecule or cytotoxic/myelosuppressive therapy), with the
following exceptions:
• Hydroxyurea for cytoreduction can be initiated and continued concomitantly
with SNDX-5613.
• Phase 1 patients may receive intrathecal chemotherapy at the time of
diagnostic lumbar puncture at least 24 hours prior to the start of SNDX-5613
and may continue prophylactic intrathecal chemotherapy beginning in Cycle 2, at
the treating physician*s discretion.
• Phase 2 and Phase 1 backfill patients may continue to receive prophylactic
intrathecal chemotherapy at any time at the treating physician*s discretion.
12. Hematopoietic Growth Factors: At least 7 days since the completion of
therapy with short-acting hematopoietic growth factors and 14 days with
long-acting growth factors.
13. Biologics (eg, monoclonal antibody therapy): At least 90 days or 5
half-lives, whichever is shorter, since the completion of therapy with an
antineoplastic biologic agent.
14. Steroids: At least 7 days since systemic glucocorticoid therapy, unless
receiving physiologic dosing (equivalent to <=10 mg prednisone daily for
patients >=18 years or <=10 mg/m2/day for patients <18 years) or cytoreductive
therapy.
Adequate Organ Function Requirements within 10 Days of Treatment Initiation
15. Estimated glomerular filtration (GFR) based on local institutional practice
for age-appropriate determination (eg, Schwartz formula for pediatric patients
or Cockcroft Gault formula for adults): GFR >=60 mL/min/1.73m2.
16. Adequate liver function defined as:
• Total bilirubin <1.5 × the upper limit of normal (ULN) for age or normal
conjugated bilirubin.
• Alanine aminotransferase (ALT) and aspartate aminotransferase (AST) <3 × ULN
(unless attributed to leukemic involvement).
Reference tables for normal liver function criteria by age and sex are provided
in Section 10.9.
Other Adequate Organ Function Requirements.
17. Adequate cardiac function defined as ejection fraction (EF) of >50% by
echocardiogram or multigated acquisition (MUGA) scan.
Contraception
18. If a female of childbearing potential, willing to use a highly effective
method of contraception or double barrier method from the time of enrollment
through 120 days following the last study drug dose. (See Section 10.4 for
details regarding contraception.)
19. If male who can father a child, agrees to use barrier contraception from
the time of enrollment through 120 days following the last study drug dose. See
Section 10.4 for details regarding contraception.)
Exclusion criteria
Diagnosis
1. Diagnosis of active acute promyelocytic leukemia.
2. Isolated extramedullary relapse (Phase 2 only).
3. Active CNS disease (cytologic, such as any blasts on cytospin, or
radiographic). Patients who have cleared CNS disease by at least one negative
tap prior to dosing may be enrolled, and prophylactic intrathecal chemotherapy
may be continued while on trial.
The following patients are required to have a lumbar puncture or Ommaya
reservoir tap during the screening period:
• Signs and symptoms of CNS disease
•
• AML with monocytic phenotype.
• WBC >=50,000 /µL at presentation.
• History of CNS or any extramedullary disease.
• ALL or MPAL.
Infection
4. Detectable human immunodeficiency virus (HIV) viral load within the previous
6 months. Patients with a known history of HIV 1/2 antibodies must have viral
load testing prior to study enrollment.
5. Hepatitis B (defined as hepatitis B virus [HBV] surface antigen positive and
HBV core antibody positive or positive HBV deoxyribonucleic acid [DNA],
6. Hepatitis C (defined as positive hepatitis C [HCV] antibody with reflex to
positive HCV ribonucleic acid [RNA]).
Pregnancy and Breast-Feeding
7. Pregnant or nursing women. Negative serum pregnancy tests are required
during Screening and a negative serum or urine pregnancy test is required
within 72 hours prior to receiving the first study drug administration, in
females of childbearing potential. If the urine test is positive or cannot be
confirmed as negative, a serum pregnancy test will be required.
Concurrent Conditions
8. Cardiac Disease:
• Any of the following within the 6 months prior to study entry: myocardial
infarction, uncontrolled/unstable angina, congestive heart failure (New York
Heart Association Classification Class >=II), life-threatening, uncontrolled
arrhythmia, cerebrovascular accident, or transient ischemic attack.
• QTc using Fridericia*s correction (QTcF) >450 msec (Section 8.2.7).
9. Gastrointestinal Disease:
• Any gastrointestinal issue of the upper GI tract likely to affect oral drug
absorption or ingestion (gastric bypass, gastroparesis, etc).
• Cirrhosis with a Child-Pugh score of B or C.
10. Graft-Versus-Host Disease (GVHD): Signs or symptoms of acute or chronic
GVHD >Grade 0 within 4 weeks of enrollment. All transplant patients must have
been off all systemic immunosuppressive therapy and calcineurin inhibitors for
at least 4 weeks prior to enrollment. Patients may be on physiological doses of
steroids.
11. Concurrent malignancy in the previous 2 years with the exception of basal
cell carcinoma of the skin, squamous cell carcinoma of the skin, or carcinoma
in situ (eg, breast carcinoma, cervical cancer in situ, melanoma in situ)
treated with potentially curative therapy, or concurrent low-grade lymphoma,
that is asymptomatic and lacks bulky disease and shows no evidence of
progression, and for which the patient is not receiving any systemic therapy or
radiation.
12. Concurrent malignancy must be in complete remission (CR) or no evidence of
disease (NED) during this timeframe.
13. History of or any concurrent condition, therapy, laboratory abnormality, or
allergy to excipients (see formulation details in Investigator Brochure) that
in the Investigator*s opinion might confound the results of the study,
interfere with the patient*s participation for the full duration of the study,
or is not in the best interest of the patient to participate.
Concomitant Medications and Interventions
14. Any commercially available or investigational antileukemic therapy other
than SNDX-5613, with the following exceptions:
• Short-term administration of corticosteroid and/or hydroxyurea for
cytoreduction.
• In the Phase 2 portion and Phase 1 backfill, intrathecal chemotherapy for CNS
prophylaxis is permitted, at the treating physician*s discretion. In the Phase
1 DLT evaluable cohort, CNS prophylaxis may continue starting in C2D1.
15. In Phase 1 some Exclusions apply. see more details in the protocol.
• Note that phase I is not applicable for the Netherlands.
In Phase 2: Patients who will not receive SNDX 5613 with co administration of a
strong CYP3A4 inhibitor must discontinue all strong CYP3A4 inhibitors at least
7 days prior to first dose of SNDX 5613. They will receive the SNDX 5613 RP2D
for patients who are not on a strong CYP3A4 inhibitor, and they may continue to
receive moderate or weak CYP3A4 inhibitors, including fluconazole and
isavuconazole.
Patients who will receive SNDX 5613 with co administration of a strong CYP3A4
inhibitor azole antifungal (itraconazole, ketoconazole, posaconazole, or
voriconazole) must have started the azole antifungal treatment at least 24
hours prior to enrollment. They will receive the SNDX 5613 RP2D for patients
who are on a strong CYP3A4 inhibitor.
Note that the RP2D for SNDX 5613 for patients on a strong CYP3A4 inhibitor
versus patients who are not on a strong CYP3A4 inhibitor is outlined in Table
9. Refer to Appendix 10.6 for examples of strong CYP3A4 inhibitors/inducers.
16. Participation in Phase 1 and Phase 2: Patients requiring the concurrent use
of medications known or suspected to prolong the QT/QTc interval, with the
exception of drugs with low risk of QT/QTc prolongation that are used as
standard supportive therapies (eg, diphenhydramine, famotidine, ondansetron,
Bactrim) and the azoles permitted whichthe relevant arms of Phase 1 and in
phase 2. Please see Appendix 10.7 for examples of medications that may be
appropriate substitutes for such medications.
17. Receipt of an investigational agent within 30 days of starting SNDX-5613
unless Inclusion Criteria 8, 9, 10, and 13 apply, then the longer period must
be applied. Patients may continue with noninterventional follow-up from
previous clinical studies.
18. Patients who have had prior exposure to a menin inhibitor.
19. Any concurrent systemic treatment to prevent GVHD.
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 | CTIS2024-513759-34-00 |
EudraCT | EUCTR2020-004104-34-NL |
CCMO | NL77798.041.21 |