This study has been transitioned to CTIS with ID 2024-513627-16-01 check the CTIS register for the current data. Dose escalationPrimary• To determine the RP2D of single-agent petosemtamab in mCRC patients who have progressed on chemotherapy, with or…
ID
Source
Brief title
Condition
- Miscellaneous and site unspecified neoplasms malignant and unspecified
Synonym
Research involving
Sponsors and support
Intervention
Outcome measures
Primary outcome
Dose escalation:
• To determine the preliminary RP2D of single-agent petosemtamab in mCRC
patients who have progressed on chemotherapy, with or without
an anti-vascular endothelial growth factor (VEGF) therapy, and with an
anti-EGFR therapy (if RASwt)
Dose expansion (single agent- non-randomized expansion cohorts):
• To determine the overall response rate (ORR) per RECIST 1.1 (per
investigator review)
Dose expansion (single agent, randomized expansion in HNSCC 2/3L):
• To descriptively characterize all relevant clinical safety and efficacy data
within the study
• To characterize the exposure-safety relationship of petosemtamab administered
at 1100 mg and 1500 mg Q2W in terms of TEAEs
Dose expansion (combination):
• To determine the overall response rate (ORR) per RECIST 1.1 (per
investigator review)
• To characterize safety and tolerability for combinations.
Dose expansion (combination, 2L mCRC cohort)
• To characterize safety and tolerability for petosemtamab in
combination with FOLFIRI/FOLFOX
Secondary outcome
Dose escalation:
safety and tolerability
PK
immunogenicity
biomarkers in tumor samples relevant to EGFR and LGR5 and early tumor response
profile
preliminary antitumor activity
Dose exp. (single agent/combination):
antitumor activity in terms of PFS, DOR RECIST 1.1 (per investigator)
and in exp. cohorts with antitumor activity observed (per central review)
OS
safety/tolerability of single-agent peto. and confirm the RP2D
PK of peto. (single agent) and of peto. in combo with pembro.FOLFOX/FOLFIRI
immunogenicity of peto.
biomarkers relevant to EGFR and LGR5 and tumor response of peto.
Dose exp. (single agent, randomized exp. in HNSCC 2/3L):
exposure-efficacy relationship of peto. at 1100 mg and 1500 mg Q2W:
sum of lesions and in terms of Grade 3-4 TEAEs, IRRs and non-IRR TEAEs
other safety
PK
immunogenicity
antitumor activity: ORR, DOR, PFS RECIST 1.1 (per
investigator/centralreview)
biomarkers in samples relevant to EGFR and LGR5 as well as the early
tumor response profile
Background summary
Colorectal cancer (CRC) is the third most-diagnosed cancer in Europe and the
United States, and 30% of patients with CRC present with metastatic disease
(Schmoll et al, 2012; Torre et al, 2015). Epidermal growth factor receptor
(EGFR) is frequently mutated or overexpressed in CRC and many other cancers
including the lung, colon, head and neck, and pancreas among others. Antibodies
targeting the extracellular domain of EGFR and small molecule inhibitors
targeting the intracellular kinase domain of EGFR are used extensively in the
standard of care management of these cancers.
Leucine-rich repeat-containing G-protein coupled receptor 5 (LGR5) is a
G-protein-coupled, seven-transmembrane-domain receptor for R-spondins which are
potent WNT signal enhancers and stem cell growth factors (de Lau et al, 2011).
It is present in intestinal stem cells found in the base of intestinal crypts,
and other normal differentiated cell types including brain, endometrium,
muscle, ovary and pancreas. LGR5 is highly regulated and maintains a small
number of native stem cells in their undifferentiated state in the base of the
colonic crypt. It is also expressed in primary and metastatic colon cancer in
animal models (de Sousa e Melo et al, 2017; Shimokawa et al, 2017) and
LGR5-positive cells have been shown to play a role of in establishing and
maintaining liver metastasis. Cancer stem cells (CSCs) of various origins
including colorectal cancer (CRC) stem cells are LGR5-positive. WNT has also
been shown to be an oncogenic driver in many cancers, and is the most commonly
altered pathway in CRC. Dysregulation of the WNT/β-catenin axis results in
uncontrolled proliferation and dissemination of CSCs that form the primary
tumor and ultimately metastatic lesions. LGR5 signal enhancement is proposed to
be important in stem cell renewal, maintaining cells in the stem cell program,
and the upregulation of LGR5 expression observed in adenocarcinomas (Martin et
al, 2017) is consistent with the link between WNT signal strength, stem cell
signature, and colon CSC behavior.
Rationale for developing MCLA-158 in EGFR-driven tumors
MCLA-158 is a common light chain human full-length IgG1 bispecific antibody
with enhanced ADCC activity, targeting the EGFR and the LGR5, two receptors
which are critical for the growth and survival of CSCs. The convergence of the
WNT and EGFR signaling pathways provides both oncogenic and mitogenic drivers
in CRC stem cells. CRC stem cells play a central role not only in tumor
initiation and growth, but also in tumor relapse and resistance to anticancer
therapies.
Via its bispecific mode of action, it is hypothesized that MCLA-158 induces
antitumor activity by simultaneously binding and blocking oncogenic drivers and
proliferative signaling in CRC stem cells. MCLA-158*s enhanced
antibody-dependent cell-mediated cytotoxicity (ADCC) properties are expected to
augment these antitumor effects via the recruitment of immune effector cells
that directly kill tumors. MCLA-158 has demonstrated strong inhibitory growth
activity in patient-derived tumor organoids and was significantly more active
in organoids derived from CRC tumors than from normal tissue. Importantly
normal tissue organoids are dependent on R-spondin for growth but the activity
of MCLA-158 in normal organoids was no different to that of the EGFR-targeting
antibody cetuximab. The bispecific format of MCLA-158 confers greater potency
than combining each targeting arm against LGR5 and EGFR as separate monoclonal
antibodies. In vivo experiments with patient-derived and organoid-derived
xenografts have demonstrated activity in RAS wildtype (RASwt) and RAS mutant
(RASmut) CRC models. Beyond CRC, activity has been observed in patient-derived
xenograft (PDX) models that co-express elevated LGR5 and EGFR mRNA levels
according to RNA sequencing data. MCLA-158 demonstrated significant antitumor
activity in the majority of PDX models of head and neck squamous cancer,
gastric adenocarcinoma, and esophageal squamous cell carcinoma, in both the
KRASwt and KRASmut settings. While MCLA-158 inhibits EGFR signaling, the LGR5
arm acts as a simple docker of the antibody targeting it to CRC cells, and does
not appear to block R-spondin binding to LGR5 or modulate the WNT signaling
pathway, consistent with its lack of activity in normal tissue organoids.
Planned early MCLA-158 development
The clinical strategy for the development of MCLA-158 involves a phase 1/2
study with an initial dose-finding part for single-agent MCLA-158 in mCRC
patients progressing on oxaliplatin- and irinotecan-based chemotherapies ±
antiangiogenic therapy to determine the recommended phase 2 dose (RP2D). In an
expansion part, activity, safety, and tolerability of MCLA-158 at the
single-agent R2PD will be evaluated in cohorts of selected solid tumor
indications with dependency on EGFR signaling. Eligible indications may include
locally advanced unresectable or metastatic head and neck squamous cell
carcinoma (HNSCC), anogenital squamous cell carcinoma (SCC), non-small cell
lung cancer (NSCLC; SCC and non-SCC), gastric/gastroesophageal junction
adenocarcinoma with EGFR amplification or high EGFR expression, endometrial
adenocarcinoma, pancreatic adenocarcinoma, and CRC KRASwt.
Study objective
This study has been transitioned to CTIS with ID 2024-513627-16-01 check the CTIS register for the current data.
Dose escalation
Primary
• To determine the RP2D of single-agent petosemtamab in mCRC patients who have
progressed on chemotherapy, with or without an anti-VEGF therapy, and with an
anti-EGFR therapy (if RASwt)
Secondary
• To characterize the safety and tolerability of petosemtamab
• To evaluate preliminary antitumor activity
• To characterize the PK of petosemtamab
• To characterize the immunogenicity of petosemtamab
• To correlate biomarkers in tumor samples relevant to EGFR and LGR5 as well as
the early tumor response profile of petosemtamab
Exploratory
• To explore additional biomarkers in tumor and blood samples (circulating free
deoxyribonucleic acid [DNA], cancer cells and proteins) predictive of response
or resistance to therapy.
• To evaluate the mutational status and other genetic alterations or gene
expression status of a panel of genes on (archival) tumor biopsies as potential
selection biomarkers for patient stratification.
• To evaluate potential pharmacodynamics (PD) biomarkers in response to therapy
• To evaluate further biomarkers to investigate the drug (ie,
mode-of-action-related effect and/or safety) and/or the pathomechanism of the
disease
Dose Expansion Single agent
Primary
• To determine the ORR per RECIST 1.1 (per investigator review)
Secondary
• To evaluate antitumor activity in terms of PFS and DOR per RECIST 1.1 (per
investigator review)
• To evaluate antitumor activity in terms of ORR and DOR per RECIST 1.1 in
expansion cohorts with preliminary antitumor activity observed (per central
review)
• To evaluate OS
• To characterize safety and tolerability of single-agent petosemtamab and
confirm the RP2D
• To characterize the PK of petosemtamab
• To characterize the immunogenicity of petosemtamab
• To correlate biomarkers in tumor samples relevant to EGFR and LGR5 as well as
the early tumor response profile of petosemtamab
Exploratory
• To explore additional biomarkers in tumor and blood samples (circulating free
nucleic acids, cancer cells, and proteins) predictive of response or resistance
to therapy
• To evaluate the mutational status and other genetic alterations or gene
expression status of a panel of genes on (archival) tumor biopsies as potential
selection biomarkers for patient stratification
• To evaluate potential PD biomarkers in response to therapy
• To evaluate further biomarkers to investigate the drug (ie,
mode-of-action-related effect and/or safety) and/or the pathomechanism of the
disease
Dose Expansion Combination
Primary
• To determine the ORR per RECIST 1.1 (per investigator review)
• To characterize safety and tolerability for combinations
Secondary
• To evaluate antitumor activity in PFS and DOR per RECIST 1.1 (per
investigator review)
• To evaluate antitumor activity in terms of ORR and DOR per RECIST 1.1 (per
central review)
• To evaluate OS
• To characterize safety and tolerability of petosemtamab in combination with
pembrolizumab as well as to further evaluate RP2D
• To characterize the PK of petosemtamab in combination with pembrolizumab
• To characterize the immunogenicity of petosemtamab
• To correlate biomarkers in tumor samples relevant to EGFR and LGR5 as well as
the early tumor response profile of petosemtamab
Exploratory
• To explore additional biomarkers in tumor and blood samples (circulating free
nucleic acids, cancer cells, and proteins) predictive of response or resistance
to therapy
• To evaluate the mutational status and other genetic alterations or gene
expression status of a panel of genes on (archival) tumor biopsies as potential
selection biomarkers for patient stratification
• To evaluate potential PD biomarkers in response to therapy
• To evaluate further biomarkers to investigate the drug (ie,
mode-of-action-related effect and/or safety) and/or the pathomechanism of the
disease
Dose expansion (combination, 2L mCRC cohort)
• To characterize safety and tolerability for petosemtamab in
combination with FOLFIRI/FOLFOX
Study design
This is a FIH phase 1/2 open-label multicenter study combining an initial dose
escalation part with a dose expansion part (in both single-agent and
combination cohorts). The initial dose escalation part was completed, and the
preliminary RP2D was established at 1500 mg Q2W (see Section 3.1.1).
At the time of implementation of this amendment, patients who are already on
treatment or in follow up will continue to follow assessments in protocol
amendment v 6.0 (dated 04 MAY 2023), while new patients treated under this
amendment will be governed by this protocol amendment.
Single-agent or combination cohorts of selected solid tumor indications for
which there is evidence of EGFR dependency and potential sensitivity to EGFR
inhibition will be evaluated in a dose expansion part of the study. The
eligible solid tumor indications are specified in the petosemtamab development
section above.
Tumor samples will be analyzed in central laboratories (unless otherwise
stated), using a Sponsor-approved test in accordance with local clinical
testing regulations, for potential biomarkers retrospectively and by
immunohistochemistry (IHC) if available at the investigational site. Patients
must sign a screening informed consent form (ICF) before the sample is
submitted for analysis
When the ICF is signed, a screening period of 28 days will start (see Section
3.3). All screening procedures should take place within 28 days prior to the
patient's first dose, including the baseline tumor biopsy (see Section 12.2.1).
Petosemtamab will be administered IV as a flat dose over an infusion period of
2 to approximately 6 h, Q2W, with 4-week cycles (28 days). As of 22 January
2021, 1500 mg Q2W is the primary dose level under investigation in the dose
expansion cohorts with demonstration of clinical activity.
Approximately 567 patients will be assigned to 1 of 7 possible dose expansion
treatment arms: 4 arms using petosemtamab as a single agent and 3 arms using
petosemtamab in combination with other agents as shown in the above diagram and
explained further in Section 3.1.1.
Safety, PK, immunogenicity, and antitumor activity of single-agent petosemtamab
will be characterized in all cohort patients, and retrospective biomarker
analyses including EGFR and LGR5 status will be performed.
A Safety Monitoring Committee (SMC) composed of the principal investigators,
the Sponsor*s medical expert(s), and safety and PK representatives, will review
data according to the SMC charter at designated timepoints throughout the study
and advise on DLTs, addition of extra patients at a given dose, and dose
escalation or de-escalation. The SMC or Sponsor may propose opening dose
expansion cohorts and advise on safety decisions, including the modification of
dosing increments, de-escalations, and dosing frequency (see Sections 3.1.6 and
9.14).
The Sponsor will monitor safety on an ongoing basis, and instruct sites on
measures for monitoring IRRs.
Dose escalation safety evaluation
The planned dose escalation part of the study has been completed. Allometric
scaling of a nonclinical PK model was used to predict petosemtamab exposure in
humans. The petosemtamab starting dose was 5 mg (flat dose) IV, Q2W, with
4-week cycles. Up to 11 dose levels were investigated: 5, 20, 50, 90, 150, 225,
335, 500, 750, 1100 and 1500 mg (flat dose), Q2W. The administered dose, dose
increments, and frequency of dosing for each patient and each cohort was
subject to change based on patient safety, PK and PD data, and upon
recommendation of the SMC.
An accelerated Simon design was implemented to evaluate safety during dose
escalation.
To be evaluable for a DLT, patients had to receive the intended petosemtamab
dose for the first 28-day cycle, or experience a DLT. Patients not evaluable
for DLT were replaced (see Section 3.1.4.1).
Dose expansion and combination safety evaluation
For the dose expansion (both single-agent and combination) cohorts, safety
evaluation will follow the guidelines
Intervention
INVESTIGATIONAL THERAPY AND REGIMEN MCLA-158: IV infusion every 2 weeks, with a
starting dose of 5 mg (flat dose). The following dose levels were planned: 5,
20, 50, 90, 150, 225, 335, 500, 750, 1100 and 1500 mg (flat dose). Dose
escalation was halted once the RP2D to investigate further in the expansion
part was selected. The administered dose, dose increments, and frequency of
dosing for each patient and each cohort is subject to change based on patient
safety, PK and PD data, and upon recommendation of the SMC. As of 22 January
2021, 1500 mg Q2W is the dose level under investigation in the expansion part
with demonstration of clinical activity. Infusions must be administered over a
minimum of 4 hours during Cycle 1. Details of MCLA-158 administration over the
minimum 4 hours are provided in separate Study Specific Dosing Instructions.
Subsequent infusions after Cycle 1 can be reduced to 2 hours at the
investigator*s discretion and in the absence of IRRs. A mandatory premedication
regimen (dexamethasone, antihistamines, paracetamol) is administered for all
infusions in Cycle 1, initiated within 24 hours prior to each infusion with
optional premedication with dexamethasone for subsequent infusions in the
absence of severe IRRs. A cycle is considered 4 weeks. For each patient, a
6-hour observation period will be implemented following infusion start for the
initial MCLA-158 infusion, a 4-hour period for the second infusion, and a
minimum of 2 hours for all subsequent administrations, corresponding to at
least the duration of the infusion. Treatment adaptation • MCLA-158 infusion
will be interrupted immediately in the event of an infusion-related reaction
(IRR) and symptomatic treatment will be administered. For mild to moderate
events (grade 1-2) after marked clinical improvement, the infusion can be
resumed at a 50% infusion rate. For grade 3 events, it is up to the
investigator*s judgment whether to resume the infusion at the 50% rate or to
stop MCLA-158. For grade 4 events, MCLA-158 must be stopped definitively. •
Dose reductions, interruptions and symptomatic treatment will be implemented in
the event of dermatologic toxicities, diarrhea/vomiting, hypomagnesemia,
thrombocytopenia, and neutropenia (see protocol section 5.3.3). • MCLA-158
administration can be delayed to manage AEs for a maximum of 2 infusions (i.e.,
up to 6 weeks between two consecutive infusions). Treatment duration Study
treatment will be administered until confirmed progressive disease (as per
RECIST 1.1), unacceptable toxicity, withdrawal of consent, patient
non-compliance, investigator decision (e.g., clinical deterioration), or
MCLA-158 interruption >6 consecutive weeks. Patients will be followed up for
safety for at least 30 days following the last MCLA-158 infusion and until
recovery or stabilization of all related toxicities, and for disease
progression and survival status for 12 months. In protocol version 5 a
combination cohort with Pembrolizumab and MCLA-158 is added. Patients with
HNSCC in first lmine will be treated in this cohort.
Study burden and risks
· Risk of infusion-related reactions (IRRs) and/or hypersensitivity
Antibodies like petosemtamab can cause a reaction when given by infusion. These
are called IRRs. IRRs can include hypotension (low blood pressure), shortness
of breath, skin redness, nausea, headache, fever, chills, tremor, excessive
sweating, itching, tachycardia, and/or vomiting. These reactions might be mild,
moderate, or serious. Out of the 79 patients treated, 72% experienced an IRR.
Most of these IRRs were mild to moderate with 6 patients discontinuing
treatment due to their IRR. IRRs typically occur during or after the first
infusion of the therapeutic antibodies, and may improve or stop altogether on
later infusions.
· Skin side effects
Study drugs like petosemtamab, which target an area on cells called the EGFR
receptor, can cause skin related side effects. Out of the 79 patients treated,
35% experienced rash, 25% dermatitis acneiform, 19% redness, 14% itching, 10%
dry skin and skin fissures, 8% bumps around your hair follicles (which may
become sores), 6% skin toxicity and 5% excessive sweating. Most of these events
were mild to moderate with 1 out of 79 patients experiencing severe dermatitis
acneiform, bumps around hair follicles, skin toxicity and excessive sweating.
· Gastrointestinal side effects
Study drugs like petosemtamab, which target an area on cells called the EGFR
receptor, can also cause side effects affecting the gastrointestinal system.
Out of the 79 patients treated 18% experienced nausea, 9% diarrhea and
inflammation of the mucous membranes of the mouth and 5% vomiting. All these
events were mild to moderate. 1 out of 79 patients experienced severe abdominal
discomfort.
· Other side effects
Out of the 79 patients treated, 17% experienced a decrease in blood magnesium
levels, 8% skin infections around the nail, 6% decrease in white blood cells,
weakness and fatigue. 5% had patches of red bumps with pus and 5% experienced a
syncope.
Other potential risks seen in drugs that target the EGFR receptor are shown
below:
· Other EGFR-related risks
Other potential risks seen include conjunctivitis (*pink eye*), swelling of the
eyelids, increased tear production (*watery eyes*), decrease of potassium
levels in the blood, and respiratory symptoms such as scarring in the lungs or
other related lung conditions. To date, some of these risks have been observed,
and assessed as related, in patients treated at 1500mg Q2W. Events included dry
eye (3 patients) and lacrimation increased, eye pruritus, scleral hyperemia,
vision blurred and keratopathy in 1 patient each.
· LGR5-related risks
As described previously, petosemtamab also attaches to an area on cells called
the LGR5 receptor, which is part of a system called the WNT pathway. There is
not enough information available about side effects caused by antibodies
targeting the LGR5 in humans so the likelihood of side effects is difficult to
predict. As LGR5 is present on both tumoral and normal intestinal stem cells,
as well as other cells that may be critical for normal tissue function,
targeting LGR5 cells as a therapeutic anticancer strategy may also impact
normal tissue functioning. Concerns of effects on intestinal stem cells
(Gastrointestinal toxicity), bone turnover (risk of bone fracture) and
hematopoiesis (myelosuppression) will be taken into consideration in clinical
studies with petosemtamab.
Pregnancy and Reproductive Risks
The potential risks of MCLA-158 on human reproductive function and on the human
fetus have not yet been studied. Women must not therefore become pregnant or
breast-feed while participating in the study. As a result, if you (or your
sexual partner) are of child-bearing potential, you must use an effective
method of contraception throughout the clinical study and for 6 months after
the last dose of MCLA-158. Effective methods of contraception include: true
abstinence, or a sole partner who is vasectomized, or a combination of two of
the following: intrauterine device/system, a condom with spermicidal
foam/gel/film/cream/suppository, and an occlusive cap (diaphragm or
cervical/vault caps) with spermicidal foam, gel, film, cream or suppository.
The use of hormonal methods of contraception are not considered effective
enough for this clinical study.
If you do become pregnant during the clinical study, you must inform your
investigating doctor and you will no longer be able to take part in this
clinical study. If you or your partner becomes pregnant during the trial or
within six (6) months of stopping treatment, your trial team will discuss all
the options available to you. The outcome and progress of any pregnancy will be
followed by your trial team, and you will be asked questions about the
pregnancy and baby, if appropriate.
As MCLA-158 is a new drug being developed there may be other side effects that
are not yet known.
5. Other side effects associated with medical procedures used during the
clinical study
Blood draws, where samples of your blood are taken to run laboratory tests, are
standard practice in your illness and are not specific to this clinical study.
However, there may be more blood draws than usual which may make you more
susceptible to side effects (pain, bruising, inflammation and swelling of the
vein, bleeding or even an infection at the puncture site).
CT scans, magnetic resonance imaging (MRI) and bone scans are normally carried
out to diagnose and assess your disease and those done in this clinical study
are not expected to be different. However, there may be more of these
procedures than you are used to.
A CT scan is a series of x-rays put together by a computer. CT scans will
expose you to small amounts of radiation. Although repeated radiation may
damage body tissues and slightly increase chances of having cancer, the risk
from the imaging being done for this clinical study is not considered to be
significant.
Magnetic resonance imaging (MRI) is a technique that uses a magnetic field and
radio waves to create detailed images of the organs and tissues within your
body.
CT scans and MRI scans involve dyes (called *contrast medium*) being injected
into one of your veins to help the organs in your body and your cancer to show
up on the scans. There is a risk that you may have an allergic reaction to the
dye. This reaction may be mild (such as a skin rash or hives) or severe (such
as breathing difficulties and shock). In rare instances, severe or fatal
allergic reactions have been reported (0.001-0.0002% of cases).
Both CT and MRI scans mean lying still in a confined space for a period of
time, so you should consider this, if you suffer from claustrophobia.
A bone scan is a nuclear medicine test which helps find cancer that has started
in or has spread to the bones. This means that the procedure uses a very small
amount of a radioactive substance, called a tracer. The tracer is injected into
a vein. The tracers in the radioactive dye used in a bone scan produce very
little radiation exposure. Even the risk of having an allergic reaction to the
tracers is low. Bone scans will only be performed if your cancer has spread to
your bones.
A multigated acquisition (MUGA) scan is a nuclear medicine imaging test that
checks how well the heart is pumping during rest or exercise. A MUGA scan uses
a radioactive material (radiopharmaceutical) that targets the heart, along with
a gamma camera and a computer, to create images of the blood flowing through
the heart.
A MUGA scan is also called nuclear ventriculography, radionuclide angiography
or cardiac blood pool scan.
The dose of x-rays or radioactive materials used in nuclear medicine imaging
can be different for every test. The dose depends on the type of procedure and
body part being examined. In general, the dose of radioactive material given is
small and you are exposed to low levels of radiation during the test. The
benefit
Uppsalalaan (Floors 3rd and 4th) 17
Utrecht 3584 CT
NL
Uppsalalaan (Floors 3rd and 4th) 17
Utrecht 3584 CT
NL
Listed location countries
Age
Inclusion criteria
1. Signed ICF before initiation of any study procedures
2. Age >=18 years at signing of ICF
3. Histologically or cytologically confirmed solid tumors with evidence of
metastatic or locally advanced disease not amenable to standard therapy with
curative intent:
• Expansion cohorts: patients with locally advanced unresectable or metastatic
disease for the following indications:
SINGLE AGENT
o 2L/3L HNSCC PATIENTS: patients who have progressed on or after, or are
intolerant to, anti-PD-(L)1 and platinum therapy as monotherapy or in
combination with other agents and no previous exposure to EGFR inhibitors.
Patients treated with platinum-containing therapy only in the adjuvant setting,
or in the context of multimodal therapy for locally advanced disease, should
have disease progression within 6 months of the last dose of platinum
containing therapy. Patients should not have received more than 2 prior lines
of treatment in recurrent or metastatic disease.
* Human papillomavirus (HPV) status determined by p16 IHC or molecular HPV test
for all oropharyngeal tumors should be reported when available.
* The eligible HNSCC primary tumor locations are oropharynx, oral cavity,
hypopharynx, and larynx.
o Cancers of the anogenital tract with squamous cell histology (ie, cervical,
vaginal, vulvar, penile, anal)
o Skin SCC
o NSCLC non-SCC and NSCLC SCC
o GEA with histologically confirmed EGFR amplification (fluorescence in situ
hybridization [FISH] score EGFR/ chromosome 7 (CEP7) ratio >=2.0, or tumor NGS
EGFR copy >=8, or ctDNA >=4, or EGFR IHC H-score >=200)
o PA
o mCRC in 3L+: Patients should be free of mutations in RAS family genes (i.e.
KRAS, NRAS, Harvey Rat Sarcoma virus [HRAS], or RAF family genes (i.e. BRAF,
A-Rapidly Accelerated Fibrosarcoma [ARAF}, Rapidly Accelerated Fibrosarcoma-1
[RAF1]), determined by central ctDNA NGS prescreening]). Note: If the patient
was treated with an EGFR inhibitor in 1L or 2L, then the patient should have
shown CR/PR. In addition, the patient at study entry should have at least 6
months of interval since the last administration of EGFR inhibitor.
Other indications may be considered, such as malignant salivary gland tumors.
• Note 1: Patients with NSCLC must receive all recommended standard therapies
driven by the histological subtype and tumor molecular profile.
• Note 2: Patients with other indications must have been previously treated
with 1 or 2 lines of the standard approved therapy (when applicable) in the
locally advanced/unresectable or metastatic setting.
COMBINATION
o 1L HNSCC: patients eligible to receive pembrolizumab as 1L monotherapy with
tumors expressing PD-L1, CPS >=1, as determined by an FDA-approved test in the
US, or by an approved equivalent test in other countries; patients should not
have previous systemic therapy administered in the recurrent or metastatic
setting, although previous systemic therapy as part of multimodal treatment for
locally advance disease is allowed if ended >=6 months prior to signing the ICF.
The eligible HNSCC primary tumor locations are oropharynx, oral cavity,
hypopharynx, and larynx. Previous treatments with anti-PD-(L)1 or anti-EGFR
therapies are not allowed.
o 2L mCRC: Patients should have been previously diagnosed with histologically
or cytologically confirmed unresectable or metastatic adenocarcinoma of the
colon or rectum. Patients must be RAS/RAF WT as determined using NGS on tumor
tissue (primary or metastatic) or other appropriate tumor tissue based assay,
to be confirmed by the sponsor. Patients must be naive to prior anti-EGFR
therapy. Radiographically confirmed disease progression must have occurred
during or within 6 months of prior 1L chemotherapy.
o Cohort to be treated with petosemtamab and FOLFIRI: patients should have had
only 1 prior chemotherapy regimen for the metastatic setting, consisting of 1L
fluoropyrimidine-oxaliplatin-based chemotherapy ± bevacizumab. Note:
FOLFOX-based adjuvant treatment would be considered front-line if PD occurred
within 6 months of completion of adjuvant therapy.
o Cohort to be treated with petosemtamab and FOLFOX: patients should have had
only 1 prior chemotherapy regimen for the metastatic setting, consisting of 1L
fluoropyrimidine-irinotecan-based chemotherapy ± bevacizumab.
4. A baseline new tumor sample (formalin-fixed paraffin-embedded [FFPE] core
needle biopsy) from a metastatic or primary site. If the patient has an
available tumor sample as an FFPE block with sufficient material (at least 20
slides with >20% tumor content) and has not received further anticancer
treatment since sample collection, then a new tumor biopsy at baseline is not
necessary. Archival FFPE slides are not acceptable. Archival tumor material
(ie, FFPE block) from before the last line of treatment received is only
acceptable if the patient has not been treated with anti-human epidermal growth
factor receptor-2 (HER-2) or anti EGFR (3L+ mCRC only) therapies, but should be
confirmed by the Sponsor.
5. Amenable for biopsy (if safe/feasible)
6. Measurable disease as defined by RECIST v1.1 by radiologic methods
7. ECOG PS of 0 or 1
8. Life expectancy >=12 weeks, as per investigator
9. Left ventricular ejection fraction (LVEF) >=50% by echocardiogram (ECHO) or
multigated acquisition scan (MUGA)
10. Adequate organ function:
• ANC >=1.5 X 109/L
• Hemoglobin >=9 g/dL
• Platelets >=100 x 109/L
• Serum magnesium, sodium, corrected total calcium, phosphate, and potassium
within normal ranges (or corrected with supplements or appropriate treatment).
• Alanine aminotransferase (ALT), aspartate aminotransferase (AST) <=2.5 x upper
limit of normal (ULN) and total bilirubin <=1.5 x ULN (unless due to known
Gilbert*s syndrome who are excluded if total bilirubin >3.0 x ULN or direct
bilirubin >1.5 x ULN); in cases of liver involvement, ALT/AST <=5 x ULN and
total bilirubin <=1.5 x ULN will be allowed, unless due to known Gilbert*s
syndrome when total bilirubin <=3.0 x ULN or direct bilirubin <=1.5 x ULN will be
allowed.
• Serum creatinine <=1.5 x ULN or creatinine clearance (CrCl) >=60 mL/min
calculated according to the Cockroft and Gault formula or Modification of Diet
in Renal Disease (MDRD) formula for patients aged >65 years (Appendix 2,
Section 20.2)
• Serum albumin >=3 g/dL
• International normalized ratio (INR) or prothrombin time (PT) <=1.5 x ULN
unless patient is receiving anticoagulant therapy and in therapeutic range of
intended used anticoagulant
• Activated partial thromboplastin time (APTT) or partial thromboplastin time
(PTT) <=1.5 x ULN unless patient is receiving anticoagulant therapy and is in
therapeutic range of intended used anticoagulant
11. Willing to undergo testing for human immunodeficiency virus (HIV) if not
tested within the past 6 months. Known HIV-positive patients are eligible
provided the cluster of differentiation 4 (CD4+) PROPERTY OF MERUS N.V.
CONFIDENTIAL Page 94 of 166
count is >=300/µL, viral load is undetectable, and the patient is currently
receiving highly active antiretroviral therapy (HAART).
Exclusion criteria
1. Central nervous system metastases that are untreated or symptomatic, or
require radiation, surgery, or continued steroid therapy to control symptoms
within 14 days of study entry
2. Known leptomeningeal involvement
3. Participation in another clinical trial or treatment with any
investigational drug within 4 weeks prior to study entry
4. Any systemic anticancer therapy within 4 weeks or 5 half-lives, whichever is
shorter, of the first dose of study treatment. For cytotoxic agents that have
major delayed toxicity (eg, mitomycin C, nitrosoureas), or anticancer
immunotherapies, a washout period of 6 weeks is required.
5. Requirement for immunosuppressive medication (eg, methotrexate,
cyclophosphamide)
6. Major surgery or radiotherapy within 3 weeks of the first dose of study
treatment. Patients who received prior radiotherapy to >=25% of bone marrow are
not eligible, irrespective of when it was received.
7. Persistent Grade >1 clinically significant toxicities related to prior
antineoplastic therapies (except for alopecia); stable sensory neuropathy Grade
<=2 National Cancer Institute-Common Terminology Criteria for Adverse Events
(NCI-CTCAE) v4.03 is allowed.
8. History of hypersensitivity reaction to any of the excipients of
petosemtamab, human proteins, or any non-IMP treatment required for this study
9. Uncontrolled hypertension (systolic BP >150 mmHg and/or diastolic BP >100
mmHg) with appropriate treatment; unstable angina; history of congestive heart
failure of Class II-IV New York Heart Association (NYHA) criteria, or serious
cardiac arrhythmia requiring treatment (except atrial fibrillation, paroxysmal
supraventricular tachycardia); or history of myocardial infarction within 6
months of study entry
10. History of prior malignancies with the exception of excised cervical
intraepithelial neoplasia or nonmelanoma skin cancer, or curatively treated
cancer deemed at low risk for recurrence with no evidence of disease for >=3
years.
11. Current dyspnea at rest of any origin, or other diseases requiring
continuous oxygen therapy, including patients with a history of ILD (eg,
pneumonitis or pulmonary fibrosis), or evidence of ILD on baseline chest
computerized tomography (CT) scan
12. Current serious illness or medical conditions including, but not limited
to, uncontrolled active infection, clinically significant pulmonary, metabolic,
or psychiatric disorders
13. Patients with known infectious diseases:
• Active hepatitis B infection (hepatitis B surface antigen [HbsAg] positive)
without receiving antiviral treatment. Note:
o Patients who are HbsAg positive must receive antiviral treatment with
lamivudine, tenofovir, entecavir, or other antiviral agents, starting at least
>=7 days before the initiation of study treatment.
o Patients with antecedents of hepatitis B (eg, anti-hepatitis B core
(anti-HBc) positive, HbsAg, and hepatitis B virus [HBV]-DNA negative) are
eligible.
• Positive test for hepatitis C virus (HCV) RNA. Note: Patients in whom HCV
infection resolved spontaneously (ie, positive HCV antibodies without
detectable HCV RNA), or who achieved a sustained response after antiviral
treatment and show absence of detectable HCV RNA >=6 months (with the use of
interferon [IFN]-free regimens) or >=12 months (with the use of IFN-based
regimens) after cessation of antiviral treatment, are eligible.
14. Pregnant or breastfeeding patients; patients of childbearing potential must
use highly effective contraception methods prior to study entry, for the
duration of study participation, and for 6 months after the last dose of
petosemtamab.
15. 1L HNSCC combination cohort: has a diagnosis of immunodeficiency, or is
receiving systemic steroid therapy or any form of immunosuppressive therapy
within 7 days prior to the first dose. Corticosteroids used as premedication
for allergic reactions or IRRs specified in the protocol are allowed.
16. 1L HNSCC combination cohort: active autoimmune disease that has required
systemic immune suppressive treatment in the past 2 years; replacement therapy
(eg, thyroxine, insulin, or physiologic corticosteroid replacement therapy for
adrenal or pituitary insufficiency, etc.) is not considered immune suppressive
treatment.
17. 1L HNSCC combination cohort: has had an allogeneic tissue/solid organ
transplant
18. 1L HNSCC combination or HNSCC single-agent cohort: patients may not have a
primary tumor site of nasopharynx (any histology)
19. Patients previously treated with anti-EGFR inhibitors are not eligible for
this study (except in 3L+ mCRC, see Inclusion Criterion [IC] 3).
20. mCRC cohorts: mCRC with a RAS/RAF mutation identified by local ctDNA or
tumor test at screening, or identified as such in disease history, are not
eligible for this study.
21. 2L mCRC cohorts: Patients with an active inflammatory bowel disease, or
other bowel disease causing chronic diarrhea (defined as NCI-CTCAE Grade >=2),
are not eligible for this study.
22. 2L mCRC cohorts: Patients with peripheral sensory neuropathy with
functional impairment (defined as NCI-CTCAE Grade >=3) are not eligible for this
study.
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 | CTIS2024-513627-16-01 |
EudraCT | EUCTR2017-004745-24-NL |
ClinicalTrials.gov | NCT03526835 |
CCMO | NL81045.041.22 |