This study has been transitioned to CTIS with ID 2024-515744-23-00 check the CTIS register for the current data. The primary objectives of the study are to:Determine the OBD(s) and RP2D's) of BMF-219 monotherapy administered daily based on…
ID
Source
Brief title
Condition
- Leukaemias
Synonym
Research involving
Sponsors and support
Intervention
Outcome measures
Primary outcome
Determine the OBD and RP2D of BMF-219 monotherapy (all cohorts).
Note that the OBD and RP2D may differ between Arm A and Arm B in Cohort 1.
-The OBD/RP2D will be determined based on evaluation of all available
pharmacokinetics (PK)/pharmacodynamics (PD), target engagement, safety, and
tolerability data.
- Escalation to the maximum tolerated dose (MTD) will not be performed if the
OBD/RP2D can be identified at a lower level. Specifically, the maximum dose to
be administered will not exceed more than one dose level above the OBD. Should
dose limiting toxicity (DLT) be encountered at a dose level below the OBD/RP2D,
the MTD will be defined as the highest dose that is not expected to cause DLT
in more than 20% of subjects.
Secondary outcome
Evaluate the safety as expressed by treatment-emergent adverse events (TEAEs)
and serious adverse events (SAEs).
- Adverse Events (AEs) and SAEs will be graded according to the NCI -CTCAE v5.0.
Determine maximum observed plasma concentration (Cmax), time to reach maximum
observed concentration (Tmax), and area under plasma-concentration time curve
from time 0 to time of last quantifiable concentration (AUC0-last) of BMF-219.
-Cmax, Tmax, and AUC0-last of BMF-219.
Evaluate the efficacy of BMF-219 as measured by complete response rate (CRR)
(Cohort 1) or objective response rate (ORR) (Cohorts 2, 3, and 4) per
Investigator assessment as per corresponding response criteria
• Complete response rate (CRR) (all cohorts)
• Complete response rate composite (CRRc) (Cohorts 1 and 4)
• ORR (all cohorts)
Assess additional evidence of antitumor activity per Investigator assessment as
per corresponding response criteria.
• Duration of complete response (DOCR) (all cohorts)
• Duration of complete response composite (DOCRc) (Cohorts 1 and 4)
• Duration of response (DOR) (all cohorts)
• Disease control rate (DCR) (Cohorts 2, 3, and 4)
• Duration of disease control (DDC) (Cohorts 2, 3, and 4)
• Time to progression (TTP) (Cohorts 2, 3, and 4)
• Relapse-free survival (RFS)/progression-free survival (PFS) (all cohorts)
• Time to Response (all cohorts)
• Time to Complete Response (TTCR) (all cohorts)
• Overall survival (OS) (all cohorts)
Background summary
Biomea Fusion is developing BMF-219, an orally bioavailable, covalent, small
molecule, menin inhibitor for the treatment of acute leukemia and other
menin-dependent malignancies. BMF-219 disrupts a key oncogenic interaction with
MLL1-fusion proteins resulting from gene rearrangements in the MLL1 gene locus,
and with MLL1-wt proteins in the presence of NPM1 and other mutations,
respectively, as illustrated in Figure 2 below. Disruption of this interaction
with small-molecule inhibitors or interference of menin protein expression via
genetic intervention effectively suppresses uncontrolled cancer cell growth and
induces both apoptotic death and cell differentiation as evidenced by induction
of differentiation markers in treated leukemia cells (Krivtsov et al, 2019;
Klossowski et al, 2020; Dzama et al, 2020).
Nonclinical studies of BMF-219 performed by Biomea Fusion have confirmed the
critical nature of menin function in DLBCL and MM. Specifically, single-agent
BMF-219 treatment also dramatically reduced tumor growth ex vivo in two primary
DLBCL specimens, one cMYC-amplified (from a subject who initially responded,
then progressed on R-EPOCH) and the other a triple-hit tumor (from a subject
who initially responded, then progressed on R-CHOP). Of note, BMF-219 was as
effective in inhibiting the growth of these primary DLBCL specimens ex vivo as
in its inhibition of MLL1 fusion-positive and NPM1-mutant primary AML specimens
ex vivo.
BMF-219 was shown in parallel studies employing the MLL1 fusion-containing
acute leukemia cell line MOLM13 to extinguish both cMYC and BCL2 transcripts to
barely detectable levels at sub-micromolar concentrations. Consistent with
these findings, cMYC target genes were substantively downregulated in AML PDX
mouse models treated with BMF-219. Elucidation of the mechanism(s) by which
menin inhibition modulates cMYC and BCL2 transcription in malignancies with
varied genetic backgrounds is the focus on ongoing nonclinical studies at
Biomea Fusion.
In contrast to the robust anti-DLBCL activity observed with BMF-219, reversible
menin inhibitors showed minimal-to-modest efficacy in the same in vitro and ex
vivo models.
Study objective
This study has been transitioned to CTIS with ID 2024-515744-23-00 check the CTIS register for the current data.
The primary objectives of the study are to:
Determine the OBD(s) and RP2D's) of BMF-219 monotherapy administered daily
based on evaluation of all available PK/ PD, target engagement, safety, and
tolerability data. Note that the OBD and RP2D may differ between arms and/or
cohorts.
The secondary objectives of the study are to:
1. Evaluate the safety as expressed by treatment-emergent adverse events
(TEAEs) and serious adverse events (SAEs). [Time Frame: During treatment and up
to approximately 28 days after treatment discontinuation, or until immediately
before the initiation of another anticancer therapy, whichever occurs first.]
(all cohorts)
2. Determine Cmax, Tmax and AUC0-last. [Time Frame: Blood samples for
determination of BMF-219 concentration will be collected during Cycle 1 and
Cycle 2] (all cohorts)
3. Evaluate the efficacy of BMF-219 as measured by CRR (Cohort 1) or ORR
(Cohorts 2, 3, and 4) per Investigator assessment based on:
* Cohort 1: per modified Cheson (2003) criteria in AML or the NCCN Clinical
Practice Guidelines, ALL (Version 1.2022) (Appendix 14.11)
* Cohort 2: Revised criteria for response assessment of lymphoma (Cheson, 2014)
* Cohort 3: International Myeloma Working Group (IMWG) response criteria
(Kumar, 2016)
* Cohort 4: iwCLL guidelines (Hallek, 2018)
4. Assess additional evidence of antitumor activity as measured by the
following based on the applicable guidelines above:
* Cohort 1: CRR, complete response rate composite (CRRc), DOCR, DOCRc, DOR,
time to relapse, RFS, time to response, TTCR, and OS
* Cohort 2: CRR, DOCR, DOR, DCR, DDC, TTP, PFS, time to response, TTCR, and OS
* Cohort 3: CRR, DOCR, DOR, DCR, DDC, TTP, PFS, time to response, TTCR, and OS
* Cohort 4: CRR, CRRc, DOCR, DOCRc, DOR, DCR, DDC, TTP, PFS, time to response,
TTCR, and OS
The following exploratory objectives are also to be examined:
1. Characterize the PD effects of BMF-219 in subjects with AL (Cohort 1), DLBCL
(Cohort 2), MM (Cohort 3), and CLL/SLL (Cohort 4) by the assessment of:
* Changes in the patterns of gene expression in BMF-219-treated tumor cells
2. Evaluate both gene mutation status and global gene expression profiles in
BMF-219-treated leukemia cells, including subclonal population analysis to
explore predictors of anti-leukemia activity and/or resistance (Cohort 1)
3. Evaluate DLBCL histology, DLBCL subtype (germinal center B-cell [GCB] or
non-GCB), *double hit* (DH-DLBCL) and *triple hit,* (TH-DLBCL) as well as
*double expressor* (DEDLBCL) and *triple expressor* (TH-DLBCL) status in all
subjects to explore predictors of anti-tumor activity and resistance (Cohort 2)
4. Correlational studies to evaluate anti-myeloma response as related to:
* Cytogenetic and fluorescence in situ hybridization (FISH) prognostic markers
including p53 abnormalities and chromosomal aberrations (e.g., del 17p,
t(4;14), t(14;16), del 13) and other MM cytogenetic classifications
* Gene expression and plasma protein levels
* Time since initial diagnosis of active myeloma
* Lytic lesions as measured by imaging (Cohort 3)
5. Assess minimal residual disease (MRD) status in R/R AL subjects who achieve
CR or Cri (Cohort 1), and R/R MM subjects who achieve CR, and stringent
complete response (sCR) (Cohort 3), and in R/R CLL/SLL subjects who achieve
CR/Cri or PR (Cohort 4)
6. Food-effect studies in subjects with DLBCL (Cohort 2). MM (Cohort 3) and
CLL/SLL (Cohort 4) enrolled at Dose Levels 2, 3 and 4 during dose escalation
and in the expansion cohorts
7. Assess the effects of BMF-219 on glycemic control for subjects with diabetes
by measuring blood glucose, change in hemoglobin A1c (HbA1c) levels and blood
C-peptide levels (all cohorts)
Study design
This Phase 1, FIH, multi-site, open-label, dose-escalation and expansion study
of BMF-219, acovalent, small molecule, menin inhibitor, will determine the
safety and tolerability, PK/PD, and clinical activity of escalating doses of
BMF-219 administered orally daily in 28-day cycles. using
an ATD in adult subjects (>=18 years of age) with R/R acute leukemia including
ALL, AML, and AMPL (Cohort 1), R/R DLBCL (Cohort 2), R/R MM (Cohort 3), and R/R
CLL/SLL (Cohort 4). The dose escalation will follow a modified Fibonacci
sequence. The study schema is shown in
Figure 3 of the study protocol.
• In Cohort 1, the 3+3 dose escalation portion will enroll at least 2/3 of
subjects who are menin inhibitor naïve with confirmed MLL1r (KMT2A) genetic
alteration and no more than 1/3 of subjects who are menin inhibitor experienced
with confirmed MLL1r (KMT2A) genetic alterations (Subcohorts are described in
Table 9 of the study protocol). The dose expansion portion of Cohort 1 will
enroll subjects who are menin-inhibitor naïve grouped based on their mutational
status and subjects who are menin-inhibitor experienced agnostic of mutational
status. One or more dose levels/dosages (QD or BD) may be explored in a
parallel or staggered fashion and one or more dose levels administered under
different fed conditions may also be explored. Cohort 1 will selectively enroll
subjects who are not (Arm A) or are (Arm B) receiving drugs that are moderate
or strong inhibitors of CYP3A4 activity (see Section 8.12.3 of the study
protocol).
• In Cohorts 2, 3, and 4, the dose escalation and dose expansion portions will
enroll subjects agnostic of genetic mutational status. A food effect substudy
will be conducted in Cohorts 2, 3, and 4. Cohorts 2, 3, and 4 will selectively
enroll subjects who are not receiving drugs that are moderate or strong
inhibitors of CYP3A4 activity (see Section 8.12.3 of the study protocol).
Within each cohort, the dose-escalation portion will identify the OBD(s) and/or
RP2D(s) of BMF-219. Thereafter, in the expansion cohort portion for each of the
4 indications, subjects will be enrolled and treated at their respective OBD or
RP2D to further evaluate the safety, tolerability, and clinical activity of
BMF-219.
Intervention
For acute leukemia (Cohort 1), the starting dose of BMF-219 to be administered
to the first cohort of subjects in the dose escalation portion is 325 mg QD
(Arm A: Subjects who are not receiving a moderate or strong CYP3A4 inhibitor)
or 75 mg QD (Arm B: Subjects who are receiving a moderate or strong CYP3A4
inhibitor) administered orally.
For DLBCL (Cohort 2), MM (Cohort 3) and CLL/SLL (Cohort 4) the starting dose of
BMF-219 will be 325 mg QD. These cohorts will undergo independent single-arm
dose escalations. All subjects participating in Arm A (Cohorts 1, 2, 3 or 4)
will use the same escalation scheme, to identify the OBD/RP2D of BMF-219 for
each indication, and they must not be receiving drugs that inhibit CYP3A4
activity.
Study burden and risks
The burden and risk consist mainly of extra time spent compared to standard
treatment and side effects, and the risks of medical evaluation, including
biopsy and MRI/CT scans.
Middlefield Road 900
Redwood City CA 94063
US
Middlefield Road 900
Redwood City CA 94063
US
Listed location countries
Age
Inclusion criteria
1. Read, understood, and provided written informed consent and, if applicable,
Health Insurance Portability and Accountability Act (HIPAA) authorization by
subject or legal guardian after the nature of the study has been fully
explained and must be willing to comply with all study requirements and
procedures including DLBCL tumor biopsies (Cohort 2), serial bone marrow and
peripheral blood sampling.
2. Males and females of age: >= 18 years
3. All subjects must have histologically or pathologically confirmed diagnosis
of their malignancy and/or measurable R/R disease, as follows:
a. Cohort 1 only: Refractory or relapsed acute leukemia defined as > 5% blasts
in the bone marrow or reappearance of blasts in the peripheral blood (as
defined by the NCCN in the NCCN Clinical Practice Guidelines in Oncology [NCCN
Guidelines®] for Acute Lymphoblastic Leukemia [Version 2.2021] and Acute
Myeloid Leukemia [Version 3.2021] ) Specific mutational statuses may be
required for allocation to a specific subcohort.
b. Cohort 2 only: Previously treated, pathologically confirmed de novo DLBCL,
or DLBCL transformed from previously indolent lymphoma (e.g., follicular
lymphoma) with documented clinical or radiological evidence of progressive or
persistent disease. At study entry, subjects must have measurable disease as
per the revised criteria for response assessment of lymphoma (Cheson, 2014).
c. Cohort 3 only: Measurable MM based on at least one (1) of the following:
i. Serum M-protein >= 0.5 g/dL by serum protein electrophoresis (SPEP) (for an
IgA-based myeloma, preferably by a quantitative serum IgA level)
ii. Urinary M-protein excretion >= 200 mg/24 hours
iii. Free light chain MM: Serum free light chain (sFLC) >= 10 mg/dL (100 mg/L),
provided serum FLC ratio is abnormal
iv. Of note, subjects without measurable disease in serum or urine, but with
plasmacytoma(s) >= 2.0 cm are eligible
d. Cohort 4 only: Previously treated CLL/SLL with active disease meeting any of
the following
conditions per the iwCLL 2018 criteria
i. Evidence of progressive marrow failure as manifested by the development of,
or worsening of, anemia and/or thrombocytopenia
ii. Massive (i.e., >= 6 cm below the left costal margin) or progressive or
symptomatic splenomegaly
iii. Massive nodes (i.e., >= 10 cm in longest diameter) or progressive or
symptomatic lymphadenopathy
iv. Progressive lymphocytosis with an increase of >= 50% over a 2-month period,
or lymphocyte doubling time (LDT) < 6 months. LDT can be obtained by linear
regression extrapolation of absolute lymphocyte counts obtained at intervals of
2 weeks over an observation period of 2 to 3 months; subjects with initial
blood lymphocyte counts < 30 × 109/L may require a longer observation period to
determine the LDT. Factors contributing to lymphocytosis other than CLL/SLL
(e.g., infections, steroid administration) should be excluded
v. Autoimmune complications including anemia or thrombocytopenia poorly
responsive to corticosteroids
vi. Symptomatic or functional extranodal involvement (e.g., skin, kidney, lung,
spine)
vii. Disease-related symptoms as defined by any of the following:
a. Unintentional weight loss >= 10% within the previous 6 months
b. Significant fatigue (i.e., ECOG PS 2 or worse; cannot work or unable to
perform usual activities)
c. Fevers >= 100.5°F or 38.0°C for 2 or more weeks without evidence of
infection
d. Night sweats for >= 1 month without evidence of infection
4. Subjects must be refractory or must have progressed on, or following
discontinuation of the most recent anti-cancer therapy, with the following
considerations:
a. Cohort 1 only: Have failed or are ineligible for any approved standard of
care therapies, including HSCT
b. Cohort 2 only: Must have received at least 2 previous systemic regimens for
the treatment of their de novo or transformed DLBCL (i.e., transformed from a
previously diagnosed indolent lymphoma [e.g., follicular lymphoma]) including:
i. at least 1 course of anthracycline-based chemotherapy (unless absolutely
contraindicated due to cardiac dysfunction, in which case other active agents
such as etoposide, bendamustine, or gemcitabine must have been given), and
ii. at least 1 course of anti-CD20 immunotherapy (e.g., rituximab), unless
contraindicated due to severe toxicity
Note: Subjects who were considered ineligible for standard multi-agent
immunochemotherapy must have received at least 2 prior treatment regimens
including at least 1 course of anti-CD20 antibodies and must have been approved
by the Medical Monitor. Prior stem cell transplantation is allowed; induction,
consolidation, stem cell collection, preparative regimen, and transplantation ±
maintenance are considered a single line of therapy. CAR-T therapy is allowed,
and it is considered a prior line of therapy. Subjects with either persistent
or progressive disease after discontinuing the most recent line of therapy may
be eligible for participation.
c. Cohort 3 only: Must have received at least 3 anti-MM regimens including
proteasome inhibitor (e.g., bortezomib or carfilzomib) and immunomodulatory
drug (IMiD) (e.g., lenalidomide or pomalidomide) therapy. Note:
Relapsed-and-refractory MM is defined as relapse of disease in subjects who
must have achieved minimal response (MR) or better, which either becomes
non-responsive while on salvage therapy, or progresses within 60 days of last
therapy.
d. Cohort 4 only: Must have received at least 2 prior systemic treatment
regimens.
5. ECOG PC of 0-2 and an estimated expected life expectancy of > 3 months in
the opinion of the Investigator.
6. Adequate liver function: serum bilirubin <= 1.5x upper limit of normal (ULN)
except for Gilbert*s syndrome or non-hepatic origin such as hemolysis (who must
have a total bilirubin < 3x ULN); aspartate aminotransferase (AST) and alanine
aminotransferase (ALT) <= 2x ULN (those subjects with known liver involvement of
their disease and ALT and AST < 5x ULN may be enrolled, subject to Medical
Monitor approval).
7. Adequate renal function: estimated creatinine clearance (eCrCl) >= 60 mL/min
(Cohort 1) or eCrCl >= 30 mL/min (Cohorts 2, 3, and 4) using the Cockcroft-Gault
equation
8. Subjects in Cohorts 2 ,3 and 4 must have the following hematologic
parameters independent of transfusion and/or blood product support at least 5
days prior to laboratory testing:
a. Absolute neutrophil count (ANC) >= 500 /mm3
b. Platelet count >= 50,000 /mm3 (Cohorts 2 and 3) / >= 30,000 /mm3 (Cohort 4)
(see Section 7.1).
c. Hemoglobin >= 8.0 g/dL.
Note: subjects who have cytopenias due to significant bone marrow infiltration
do not have to meet hematologic eligibility criteria. (Significant bone marrow
infiltration is defined as > 50% disease involvement.)
9. Both men and women of childbearing potential or their partners must use
adequate birth control measures during the course of the trial and for at least
90 days after discontinuing study treatment. Subjects and/or partners who are
surgically sterile or postmenopausal are exempt from this requirement.
• Females are to be not pregnant, non-lactating, and can be postmenopausal
(defined as amenorrheic for at least 1 year while not taking oral
contraceptives [OCPs] without an alternative cause). Females of childbearing
potential must have a negative pregnancy test at screening and be willing to
have additional pregnancy tests during the study and must agree to use adequate
contraception during the study and for approximately 90 days following the last
administration of investigational product to avoid pregnancy. Adequate
contraception is defined as oral, intravaginal, transdermal, implantable or
injectable contraceptives, intrauterine devices, surgical sterilization
(achieved through hysterectomy, oophorectomy, or bilateral salpingectomy or
tubal ligation in addition to/or a combination of an intrauterine
hormone-releasing system (IUS) and spermicid
Exclusion criteria
1. Certain disease subtypes or occurrences, as follows:
a. Cohort 1: APL, CML in blast crisis, iEMR.
b. Cohort 2: PMBCL, DLBCL transformed from diseases other than indolent NHL,
Burkitt Lymphoma
c. Cohort 3: Active plasma cell leukemia, myeloma with amyloidosis, systemic
light chain amyloidosis
d. Cohort 4: Known or suspected history of Richter*s transformation
2. WBC count > 50,000/ µL (uncontrollable with cytoreductive therapy) (Cohort 1
only).
3. Known central nervous involvement, as follows:
a. Cohort 1: Clinically active CNS leukemia. Previously controlled CNS leukemia
is acceptable, however
b. Cohort 2: Active CNS lymphoma or meningeal involvement
c. Cohort 3: Active CNS MM
d. Cohort 4: Active CNS leukemia
4. Prior menin inhibitor therapy (exept for subjects in Cohort 1).
5. Known positive test for human immunodeficiency virus, hepatitis C, or
hepatitis B surface antigen. Of note: HBV core Ab positive but HBV DNA negative
subjects with no prior history of reactivation with prior CD20 monoclonal
antibody exposure and prophylaxis would be allowed with reinstitution of
appropriate prophylaxis; HCV Ab positive after treatment with anti-hepatitis C
medications and viral load negative for at least 6 months would be eligible. If
the subject is known to be cytomegalovirus (CMV) IgG or CMV IgM positive, the
subject must be evaluated for the presence of CMV DNA by PCR. Subjects who are
known to be CMV IgG or CMV IgM positive but who are CMV DNA negative by PCR are
eligible. Antiviral prophylaxis should be considered per institutional protocol.
6. Subjects with a pre-existing disorder predisposing them to a serious or
life-threatening infection (e.g., cystic fibrosis, congenital or acquired
immunodeficiency, bleeding disorder, or cytopenias not related to acute
leukemia, DLBCL, MM, or CLL/SLL).
7. An active uncontrolled acute or chronic systemic fungal, bacterial, or viral
infection.
8. Significant cardiovascular disease including unstable angina pectoris,
uncontrolled hypertension or arrhythmia, history of cerebrovascular accident
including transient ischemic attack within 6 months prior to the first dose of
the study treatment, congestive heart failure (New York Heart Association
[NYHA] Class III or IV) related to primary cardiac disease, ischemic or severe
valvular heart disease, or a myocardial infarction within 6 months prior to the
first dose of study treatment. Additional cardiovascular exclusions include any
evidence of pericardial effusion or LVEF < 45% assessed by echocardiogram
(ECHO), multi-gated acquisition (MUGA), or local standard.
9. Mean QTcF or QTcB of > 470 millisecond (ms) on triplicate ECGs performed
within 5 minutes of each other.
10. Major surgery within 4 weeks prior to the first dose of study treatment.
Surgery requiring local/epidural anesthesia (excluding biopsies) must be
completed at least 72 hours before study drug administration and the subject
should be recovered.
11. Unable to swallow tablets or have gastrointestinal disease or dysfunction
that may interfere with oral absorption of study treatment, such as:
a. Chronic diarrhea or ingestion (e.g., short-gut syndrome, gastroparesis,
etc.).
b. Cirrhosis with a Child-Pugh score of B or C.
c. Post gastrectomy
12. GVHD: Signs or symptoms of acute GVHD of any severity or chronic GVHD other
than disease limited to skin that is adequately controlled with topical
steroids alone within 3 weeks of enrollment. All transplant subjects must have
been off all systemic immunosuppressive therapy and calcineurin inhibitors for
at least 3 weeks prior to enrollment. The use of topical steroids for cutaneous
GVHD is allowed and stable systemic steroid doses less than or equal to 20 mg
of prednisone or equivalent daily is permitted with Medical Monitor approval.
13. Concurrent malignancy in the previous 2 years with the exception of
adequately treated non-melanomatous skin cancer, or carcinoma in situ (e.g.,
breast carcinoma, cervical cancer in situ, melanoma in situ) treated with
potentially curative therapy; superficial bladder cancer not treated with
intravesical chemotherapy or BCG within 6 months, localized prostate cancer and
PSA <1.0 mg/dL on 2 consecutive measurements at least 3 months apart with the
most recent one being within 4 weeks of study entry. Concurrent malignancy must
be in complete response or no evidence of disease during this timeframe.
14. Any underlying medical condition (including uncontrolled diabetes and
muscular glycogenosis) that, in the Investigator*s opinion, will make the
administration of study treatment hazardous or obscure the interpretation of
toxicity determination or AEs.
15. Women who are pregnant or lactating. All female subjects with reproductive
potential must have a negative pregnancy test prior to starting study treatment.
16. Known recent (within the past year) or ongoing alcohol or drug abuse.
17. Live, attenuated vaccine within 4 weeks before the first dose of study
treatment.
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 | CTIS2024-515744-23-00 |
EudraCT | EUCTR2022-002798-27-NL |
ClinicalTrials.gov | NCT05153330 |
CCMO | NL82560.091.22 |