This study has been transitioned to CTIS with ID 2023-508369-34-00 check the CTIS register for the current data. The purpose of this first-in-human (FIH) study is to characterize the safety, tolerability, pharmacokinetics (PK), pharmacodynamics (PD…
ID
Source
Brief title
Condition
- Mesotheliomas
Synonym
Research involving
Sponsors and support
Intervention
Outcome measures
Primary outcome
Safety: Incidence and severity of Adverse Events (AEs) and Serious Adverse
Events (SAEs), including clinically significant changes in laboratory
parameters, vital signs, and ECGs. Incidence and nature of dose limiting
toxicities (DLTs) during the first cycle of dosing
Tolerability: Dose interruptions, reductions and dose intensity
See also section 2 of the protocol.
Secondary outcome
Overall Response Rate (ORR), disease control rate (DCR), progression-free
survival (PFS), duration of response (DOR) as per RECIST v1.1*
Plasma concentration vs. time profiles and derived PK parameters for IAG933
such as Cmax, Tmax, Cmin, AUC, T1/2, Racc
ORR, DCR, PFS, DOR as per RECIST v1.1*
OS
Background summary
The Hippo pathway is an evolutionarily conserved signaling pathway that plays
essential roles in embryonic development, control of organ size, epithelial
homeostasis, tissue regeneration, and wound healing. Numerous studies have
identified roles for the Hippo pathway in cancer cell migration, invasion, and
metastasis.
The core components of the Hippo pathway include neurofibromin 2 (NF2),
mammalian sterile 20-like kinases 1 and 2 (MST1/2), large tumor suppressor
kinase 1/2 (LATS1/2), two human paralogs, Yes-associated protein (YAP) and
transcriptional coactivator with PDZ-binding motif (TAZ, also known as WW
Domain Containing Transcription Regulator 1), and transcriptional enhancer
associate domain (TEAD) family members (Pan 2010). The kinase cascade of the
Hippo pathway regulates the activity of the transcriptional coactivators YAP
and TAZ, which interact with TEADs to induce target gene transcription
(Moroishi et al 2015).
When the Hippo pathway is *on*, MST1/2 phosphorylate and activate LATS1/2,
which in turn phosphorylate YAP/TAZ on multiple serine residues, resulting in
the binding of 14-3-3, cytoplasmic retention and sequestration, which is then
followed by ubiquitination and degradation. When the Hippo pathway is *off*,
dephosphorylated YAP/TAZ translocate into the nucleus, bind TEAD, and drive
transcription of target genes that are critical for cell growth, proliferation,
and survival (Pan 2010).
Hyperactivation of YAP and/or TAZ (and subsequent hyperactivity of the
YAP/TAZ-TEAD transcriptional complex) is commonly seen in a number of human
cancers (Anon 2017, Muramatsu et al 2011, Eun et al 2017) and high expression
and nuclear localization of YAP/TAZ has been associated with poor prognosis and
resistance to treatment (Jia 2003, Zanconato et al 2016). High nuclear
expression of YAP/TAZ has been found in a variety of cancers including
high-grade breast carcinomas and triple-negative breast cancer (Cordenonsi et
al 2011, Diaz-Martin et al 2015), as well as in head and neck cancers where it
has correlated with tumor recurrence, resistance to radio- and immunotherapy
and poor outcomes (Lee et al 2015).
In esophageal cancers, YAP amplification (Anon 2017), and high nuclear
localization is a predictor of poor prognosis and resistance to therapy
(Muramatsu et al 2011), while YAP-regulated gene expression has been associated
with poor prognosis in colorectal cancer (Muramatsu et al 2011, Eun et al
2017). TAZ amplifications have been found in lung squamous cell carcinoma (Jia
2003, Sun et al 2019, Dey et al 2020) and in ovarian cancers (Lamar 2012) In
addition, YAP/TAZ may also be implicated in the development and progression of
metastases. Studies indicated that ectopic expression of YAP has potent
pro-metastatic activity, particularly nuclear-localized mutants of YAP (Lamar
2012). This activity relies on TEAD binding, suggesting that YAP/TEAD
inhibition may offer therapeutic potential in aggressive cancers (Dey et al
2020).
See also section 1 of the protocol.
Study objective
This study has been transitioned to CTIS with ID 2023-508369-34-00 check the CTIS register for the current data.
The purpose of this first-in-human (FIH) study is to characterize the safety,
tolerability, pharmacokinetics (PK), pharmacodynamics (PD) and preliminary
anti-tumor activity of IAG933 in adult patients with advanced mesothelioma or
other solid tumors harboring certain molecular alterations in the Hippo
pathway. The dose escalation part will include patients with advanced
mesothelioma, or solid tumors bearing NF2/LATS1/LATS2 truncating mutations
(LATS1/LATS2 mutations will only be included in the dose escalation part) or
functional YAP/TAZ fusions. The main purpose of dose escalation is to determine
the maximum tolerated dose(s) (MTDs) and/or recommended dose(s) (RDs) and
dosing schedule(s) of single agent IAG933. The RD(s) will be further explored
in select indications in the expansion part of the study. The purpose of dose
expansion is to assess preliminary anti-tumor activity and further explore
safety and tolerability of IAG933 at the RD(s). In addition, the study will
assess PD changes induced by IAG933 and characterize the PK/PD relationship.
Finally, a food effect evaluation of IAG933 may be performed in an exploratory
food effect cohort to investigate the effect of food on IAG933 exposure.
See also section 1 of protocol.
Study design
This study is a FIH, open-label, phase I, multi-center study of IAG933 as a
single agent, consisting of a dose escalation part followed by a dose expansion
part. Dose escalation will be conducted in adult patients with advanced
mesothelioma or other solid tumors with specified dysregulations in the Hippo
pathway. Upon determination of the MTDs and/or RDs for expansion, the study
will continue with an expansion part in defined patient populations.
Initiation of dosing between the first patients (up to the first 3) in a cohort
at a daily dose higher than any daily dose previously tested and shown to be
safe will be staggered by at least 48 hours.
The study treatment will be administered until the patient experiences
unacceptable toxicity, progressive disease, and/or has treatment discontinued
at the discretion of the Investigator or the patient, or due to withdrawal of
consent.
An exploratory food effect cohort(s) may be included to assess IAG933 PK
properties, safety and tolerability in patients under fed conditions.
See also section 3 of the protocol
Intervention
For this study, *investigational drug* and *study treatment* refer to IAG933.
The investigational drugs used in this study are listed in Table 6-1 in the
protocol. Additional drug strengths may become available and be used in this
study.
See also section 6 of the protocol.
Study burden and risks
Risks and sideeffects assocated with the treatment (IAG933) and the tests
necessary to track the patients suchs as blooddraws, imaging and tumor
biopsies.
The burden of participtating consists of the following:
For dosing 3don/4doff:
Cycle 1: 1 visit of 3 days and 5 visits of 1 day
Cycle 2: 4 visits
Cycle 3-6: 2 visits
From cycle 7: 1 visit
For continous dosing:
Cycle 1: 1 visits of 3 days, 4 visits of 1 day
Cycle 2: 4 visits
Cycle 3-6: 2 visits
From cycle 7: 1 visit
Most visits will be 2-4 hours long, PK days (day 1 and day 15 of cycle 1) will
be at minimum 12 hours long. The start of the study in both schedules has tests
on day 1, 2 and 3. Patients can go home at the end of day 1 and 2, or can use
alternative accomodations near the hospital if they do not need to be observed
by research staff due to adverse events.
During the visits, the following tests might be performed, the frequency and
number of tests will depend on the type of visit and the schedule for dosing:
physical exams, blooddraws, urine collection, Holter research, ECGs,
echocardiogram, imaging, pregnancy tests (for women who can get pregnant),
tumor and skin biopsies.
Haaksbergweg 16
Amsterdam 1101 BX
NL
Haaksbergweg 16
Amsterdam 1101 BX
NL
Listed location countries
Age
Inclusion criteria
1.Signed informed consent must be obtained prior to participation in the study.
2. Male or female patients must be >= 18 years of age
3. (dose escalation) Patients with histologically or cytologically confirmed
diagnosis of advanced (unresectable or metastatic) mesothelioma or other solid
tumors. Patients with solid tumors other than mesothelioma must have local
available data for loss-of-function NF2/LATS1/LATS2 genetic alterations
(truncating mutation or gene deletion; LATS1/LATS2 mutations will only be
included in the dose escalation part), or functional YAP/TAZ fusions (see
Appendix 4 for requirements for molecular alterations ). Patients with
malignant EHE can be enrolled with only histological confirmation of the
disease. Patients must have failed available standard therapies, be intolerant
of or ineligible for standard therapy, or for whom no standard therapy exists.
4. Dose expansion part: the following patients will be enrolled into 3
different treatment groups:
Group 1: Advanced (unresectable or metastatic) MPM patients who have failed
available standard therapies for advanced/metastatic disease, be intolerant or
ineligible to receive such therapy, or for whom no standard therapy exists.
Group 2: Advanced (unresectable or metastatic) solid tumor patients with
available local data for NF2 truncating mutation or deletions (refer to
Appendix 4 for more details). Patient must have failed available standard
therapies, be intolerant or ineligible to receive such therapy, or for whom no
standard therapy exists.
Group 3: Advanced (unresectable or metastatic) solid tumor patients with
available local data for functional YAP/TAZ fusions (refer to Appendix 4 for
more details). EHE patients can be included with only histological confirmation
of the disease. Patient must have failed available standard therapies, be
intolerant or ineligible to receive such therapy, or for whom no standard
therapy exists.
5. Presence of at least one measurable lesion according to mRECIST v1.1 (for
mesothelioma patients, refer to Appendix 2), RECIST v1.1 (for patients with
other solid tumors, refer to Appendix 1), or RANO (for patients with primary
brain tumors, refer to Appendix 3).
6. Patient must have a site of disease amenable to biopsy and be a candidate
for tumor biopsy according to the treating institution*s guidelines. Patient
must be willing to undergo a new tumor biopsy at screening/baseline, and again
during therapy on this study. Archival tissue obtained within 3 months and
after last systemic treatment may be used at screening. Exceptions may be
considered after documented discussion with Novartis.
7. Eastern Cooperative Oncology Group (ECOG) performance status of 0 or 1.
Exclusion criteria
1. Treatment with any of the following anti-cancer therapies prior to the first
dose of study treatment within the stated timeframes:
a. <= 4 weeks for thoracic radiotherapy to lung fields or limited field
radiation for palliation within <= 2 weeks prior to the first dose of study
treatment. An exception to this exists for patients who have received
palliative radiotherapy to bone, who must have recovered from
radiotherapy-related toxicities but for whom a 2-week washout period is not
required.
b. <= 4 weeks or <= 5 half-lives (whichever is shorter) for chemotherapy or
biological therapy (including monoclonal antibodies) or continuous or
intermittent small molecule therapeutics or any other investigational agent.
c. <= 6 weeks for cytotoxic agents with risk of major delayed toxicities, such
as nitrosoureas and mitomycin C.
d. <= 4 weeks for immuno-oncologic therapy, such as CTLA4, PD-1, or PD-L1
antagonists
e. Prior treatment with TEAD inhibitor at any time
2. For mesothelioma patients: use of non-invasive antineoplastic therapy (e.g.,
tumor treating fields, brand name Optune LuaTM) within 2 weeks of the tumor
assessment at screening.
3. Malignant disease, other than that being treated in this study. Exceptions
to this exclusion include the following: malignancies that were treated
curatively and have not recurred within 2 years prior to study entry;
completely resected basal cell and squamous cell skin cancers; any malignancy
considered to be indolent and that has never required therapy; and completely
resected carcinoma in situ of any type.
4. Presence of symptomatic CNS metastases, or CNS tumors or metastases that
require local CNS-directed therapy (such as radiotherapy within 3 months of
tumor assessment at screening or surgery), or increasing doses of
corticosteroids 2 weeks prior to study entry.
Patients with treated symptomatic brain tumors should be neurologically stable
(for 4 weeks post-treatment and prior to study entry) and at a dose of <= 10 mg
per day prednisone or equivalent for at least 2 weeks before administration of
any study treatment
5. Patients who have undergone major surgery <= 4 weeks prior to first dose of
study treatment
6. History of allogeneic bone marrow or solid organ transplant.
7. Insufficient renal function at Screening:
a. Serum creatinine > 1.5 x ULN
b. Estimated glomerular filtration rate (eGFR) < 50 mL/min/1.73m2 (calculated
using the Cockcroft-Gault formula, or the CKD-EPI Creatinine-Cystatin C formula
as listed in Appendix 7).
c. Urine protein-creatinine ratio > 0.5 g/g (56.5 mg/mmol)
8. Clinically significant cardiac disease or risk factors at screening,
including any of the following:
a. Clinically significant and/or uncontrolled heart disease, including coronary
artery disease, uncontrolled hypertension, clinically significant arrhythmia,
and congestive heart failure (NYHA grade >= 2).
b. Acute myocardial infarction or unstable angina pectoris within 6 months
prior to study entry.
c. Left ventricular ejection fraction (LVEF) < 50% as determined by
Cardiovascular magnetic resonance imaging (cardiac magnetic resonance imaging
(MRI)) or trans-thoracic echocardiography (TTE).
d. Resting QTcF >=450 msec (male) or >=460 msec (female) at screening, or QTc not
assessable
e. Resting heart rate (physical exam or 12 lead ECG) < 50 bpm
f. PR interval >200ms, Mobitz type II second degree AV block, high-grade AV
block or third degree (complete) AV block
g. Risk factors for Torsades de Pointes (TdP), including uncorrected
hypokalemia or hypomagnesemia, history of cardiac failure, or history of
clinically significant/symptomatic bradycardia, or any of the following:
i. History of familial long QT syndrome, known family history of TdP or family
history of idiopathic sudden death
ii. Concomitant QT prolonging medication(s) with a known risk of QT
prolongation that cannot be discontinued or replaced by safe alternative
medications at least 7 days prior to the start of study treatment and for the
duration of the study (see Appendix 5)
9. Insufficient bone marrow function at screening:
a. Absolute Neutrophil Count (ANC) < 1.5 x 109/L
b. Hemoglobin < 9.0 g/dL without transfusion support within 7 days prior to
start of study treatment
c. Platelet count < 75 x 109/L without transfusion support within 7 days prior
to start of study treatment
10. Insufficient hepatic function at screening:
a. Serum total bilirubin > 1.5 x upper limit of normal (ULN). An exception is
for patients with Gilbert*s syndrome, who are excluded if total bilirubin > 3.0
x ULN and direct bilirubin >1.5 x ULN.
b. Aspartate aminotransferase (AST) > 3 x ULN or > 5 x ULN if liver metastases
are present.
c. Alanine aminotransferase (ALT) > 3 x ULN or > 5 x ULN if liver metastases
are present.
11. Patients who have the following laboratory values outside of the laboratory
normal limits (treatment may be given during screening to correct values):
a. Potassium
b. Magnesium
c. Total calcium (corrected for low serum albumin)
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 | CTIS2023-508369-34-00 |
EudraCT | EUCTR2021-000383-30-NL |
ClinicalTrials.gov | NCT04857372 |
CCMO | NL79021.078.22 |