The primary objectives of this trial are:- To determine the safety and tolerability of increasing doses of PRECIOUS-01 after intravenous (i.v.) administration in subjects with solid tumors;- To assess the effect of increasing doses of PRECIOUS-01 on…
ID
Source
Brief title
Condition
- Miscellaneous and site unspecified neoplasms malignant and unspecified
Synonym
Research involving
Sponsors and support
Intervention
Outcome measures
Primary outcome
The primary endpoints of the trial are:
- Safety profiles: incidence of treatment-emerging Adverse Events (AEs) and
Serious Adverse Events (SAE), and laboratory abnormalities graded according to
the National Cancer Institute (NCI) Common Terminology Criteria for Adverse
Events (CTCAE) v5.0 reporting severity and relatedness (see Appendix I);
occurrence of DLTs; incidence of treatment discontinuations and treatment
modifications due to AEs and laboratory abnormalities; changes in vital signs,
electrocardiogram (ECG) and Eastern Cooperative Oncology Group performance
status (ECOG PS); deaths;
- The immune modulating effect directly in the tumor is planned to be assessed
at baseline and after Cycle 3 specifically to analyze the immunological
composition in tumor biopsies using an established immunohistochemistry (IHC)
assay for the detection of CD3, CD8, FoxP3, CD45RO and CD20 positive cells;
these results will support the decision on the RP2D as part of the planned
trial.
Secondary outcome
The secondary endpoints of the trial are:
- The RP2D, which will be evaluated using all data on safety and the immune
modulating effect per dose level tested (if MTD is reached at one of the tested
dose levels, then this dose will be used)
- Evaluation of the immune modulating activity at each dose level (focused on
the presence of NY-ESO-1-specific T cells, iNKT cell activation, DC activation,
cytokine responses, and anti-NY-ESO-1-specific antibody responses) using
peripheral blood.
Background summary
In the rapidly evolving treatment landscape of advanced solid tumors,
immunotherapeutic approaches have revolutionized cancer care for patients.
Despite these advances, there is an undiminished need for the development of
novel immunotherapeutic treatment modalities that can orchestrate an effective
anti-tumor immune response in a highly targeted way, while circumventing
immunosuppressive characteristics of the tumor microenvironment.
In this trial, we investigate a new class of immunomodulatory nanomedicines:
PLGA-nanoparticles loaded with tumor antigen NY- ESO-1 peptides in combination
with the invariant Natural Killer T (iNKT) cell activator IMM60.
PLGA-nanoparticles
Biodegradable PLGA-nanoparticles function as a delivery platform for
immunomodulators and tumor antigens to induce a specific anti-tumor immune
response. PLGA has minimal (systemic) toxicity and is used in various
drug-carrying platforms as encapsulating agent. Uptake of the nanoparticle by
Dendritic Cells (DC) and subsequent prolonged presentation of PLGA-
encapsulated peptides on Major Histocompatibility Complex (MHC) class I and II
molecules are able to generate functional and tumor-antigen-specific CD8+ and
cluster of differentiation (CD)4+ T cell responses in preclinical studies.
NY-ESO-1
NY-ESO-1 is a cancer-testis antigen expressed during embryogenesis and in the
testis, an immune privileged site. Furthermore, NY-ESO-1 expression is observed
in several advanced cancers: lung (2-32%), melanoma (40%), bladder (32-35%),
prostate (38%), ovarian (30%), esophageal (24-33%), and gastric cancers
(8-12%). Clinical trials have shown the safety and tolerability of Good
Manufacturing Practices (GMP)-grade NY-ESO-1 peptides in patients with cancer.
By studying published NY-ESO-1 epitopes and their respective Human Leukocyte
Antigen (HLA) alleles using a cancer antigenic peptide database, we defined two
long peptides of 27 amino acids and one short peptide of 9 amino acids for
nanoparticle encapsulation (85-111, 117-143, and 157- 165). In combination, all
three peptides are able to cover more than 80% of the European population for
both class I and class II HLA alleles. Administration of NY-ESO-1-specific
peptides leads to antigen-specific T-cell responses against NY-ESO-1-positive
tumors. The peptides planned to be investigated in the current trial were
selected based on their previously published immunogenicity (i.e., the capacity
to induce T cell responses).
iNKT cell activator ThrCer6
iNKT cells recognize glycolipid molecules presented with CD1d molecules on DCs
via their invariant T cell receptor (TCR). DCs are considered the most potent
Antigen-Presenting Cells (APCs) of the immune system and induce
antigen-specific cytotoxic (CD8+) T cell responses to tumor antigens. Upon
CD1d-dependent interaction with DCs, iNKT cells will be stimulated to secrete
pro-inflammatory cytokines (e.g., interferon [IFN]-* and interleukin [IL]-12).
Moreover, CD40-CD40 ligand (CD40L) mediated interaction between DCs and iNKT
cells during glycolipid presentation leads to DC maturation demonstrated by the
upregulation of co-stimulatory molecules.
αGalCer consists of a galactose connected via an α-linkage to a ceramide-group.
Once presented with CD1d on antigen presenting cells, it functions as a potent
activator of iNKT cells. ThrCer6 is an alpha-Galactosylceramide (αGalCer)-
derived iNKT cell activator that possesses a superior iNKT cell activation
profile compared to αGalCer in vitro as well as in vivo. Unlike αGalCer, it
does not subject to glycosidase-mediated degradation. Induction of
antigen-specific CD8+ T cell responses at lower concentrations of ThrCer6 than
αGalCer were observed in mice due to a prolonged bioavailability. ThrCer6
maintained the capacity to induce DC maturation in vivo to a similar extent as
αGalCer. Furthermore, iNKT cell-mediated anti-tumor effects include IFN-*-
dependent Natural Killer (NK) cell responses, maturation of DCs, expansion of
antigen-specific CD8+ T cell responses, inhibition of metastatic behavior and
improved survival in cancer mouse models.
Co-encapsulation in nanoparticles: iNKT cell activator and tumor antigen
Preclinical studies in murine cancer models showed αGalCer/CD1d-dependent
superiority of nanoparticles that co-encapsulated αGalCer+Ovalbumin (OVA) in
terms of CD8+-specific T cell induction compared with nanoparticles with OVA
combined with either nanoparticles with αGalCer or soluble αGalCer. These
findings appear independent of CD4+ T cell help. Co-encapsulation of a potent
iNKT cell activator such as αGalCer or ThrCer6, with a tumor antigen, such as
NY-ESO-1, can result in detectable cytotoxic T lymphocyte (CTL) responses
against the respective antigen. With this in mind, in vitro experiments with
peripheral blood mononuclear cells were performed to first assess feasibility
of co-encapsulation. These experiments demonstrated the feasibility of
encapsulating three NY-ESO-1 peptides (the same peptides to be used in this
trial) together with IMM60 in the same particle. Peptides were processed and
presented by multiple HLA types. In mice expressing the human HLA-A2 molecule,
activity of nanoparticle-encapsulated IMM60 was evident by DC maturation and
IFN-γ secretion. Also, CD4+ and CD8+ T cell responses against NY-ESO-1 peptides
were observed with the same nanoparticles co-encapsulating NY-ESO-1 peptides.
Study objective
The primary objectives of this trial are:
- To determine the safety and tolerability of increasing doses of PRECIOUS-01
after intravenous (i.v.) administration in subjects with solid tumors;
- To assess the effect of increasing doses of PRECIOUS-01 on the composition
and spatial heterogeneity of immune cells directly in the tumor
The secondary objectives of this trial are:
- To determine the Recommended Phase 2 Dose (RP2D) of PRECIOUS-01 for a
subsequent Phase II trial. - To assess immunological responses during and after
therapy with PRECIOUS-01 in blood.
The exploratory objective of this trial is:
- To evaluate the clinical outcome of PRECIOUS-01 using Response Evaluation
Criteria in Solid Tumors (RECIST) v1.1.
Further exploratory objectives will be determined based on results of ongoing
preclinical studies.
Study design
This is an open-label, first-in-human Phase I dose escalation trial to
investigate the safety and tolerability of increasing doses of PRECIOUS-01
administered i.v. in subjects with solid tumors.
Eligible subjects will receive three i.v. infusions of PRECIOUS-01 at a
3-weekly interval in three dose-finding cohorts (low: 0.4 mg/kg, intermediate:
0.8 mg/kg, and high: 1.6 mg/kg fixed doses). Subjects will be monitored for
safety and the occurrence of Dose-Limiting Toxicities (DLTs). It is planned to
follow a 3+3 design for the dose escalation steps. Three subjects will be
enrolled sequentially per cohort. If the maximum tolerated dose (MTD) is not
reached in the planned dose escalation cohorts, the RP2D will be based on the
observed safety and immune-modulatory activity as pharmacodynamic parameter
supporting the RP2D. The sample size is based on the determination of the
MTD/RP2D. In order to collect sufficient information regarding changes in
immune-related parameters as readout for pharmacodynamics of the particles
(e.g., NY-ESO-1-specific CD8+ T cells, iNKT cell activation, and NY-ESO-1
antibody responses in serum) it is planned to extend the two highest dosing
cohorts to a total of six subjects.
Based on accumulating data during the trial and no observed toxicity in cohort
2 after three subjects for DLT observation, no further subjects may be enrolled
at that dose level. Instead, cohort 3 would start. If no toxicity is observed
in the six planned subjects in cohort 3 and based on the recommendation of the
Dose Steering Board (DSB), three additional subjects may be enrolled at the
high dose level for a total of nine subjects. If a toxicity is observed in the
first six subjects of cohort 3, three subjects will be enrolled in cohort 2
(for a total of six subjects at that dose level). The total number of subjects
will not change.
The starting dose is 10% of the No Observed Adverse Effect Level (NOAEL) in the
toxicology study. Doses will be escalated after a review of the safety data
from the previous cohort by a DSB upon completion of the DLT observation
period. Pre-defined DLT criteria will be used.
Intervention
PRECIOUS-01 will be administered i.v. three times at 3-weekly intervals (Day 1,
Day 22 and Day 43).
Doses are specified as follows: low (0.4 mg/kg PRECIOUS-01), intermediate (0.8
mg/kg PRECIOUS-01) and high (1.6 mg/kg PRECIOUS-01). PRECIOUS-01 will be
administered as fixed doses (based on a 70-kg average body weight).
Study burden and risks
Risks
PRECIOUS-01 has not yet been given to patients, therefore side effects to
PRECIOUS-01 are unknown. The particle has a low potential risk of inducing
allergic reactions. Possible AEs associated with an allergic reaction can
include fever, chills, headache, weakness, nausea, vomiting, diarrhea, low
blood pressure, respiratory symptoms, and rashes. Additionally, since the mode
of action of PRECIOUS-01 includes potential stimulation of the immune response,
there is a likelihood of immune-related AEs. Generally, such AEs have been
observed after specific blocking of inhibitory receptors expressed on T cells
(e.g. Programmed Cell Death Protein 1 [PD-1]) rather than after induction of
tumor-specific immune responses. If PRECIOUS-01 elicits a particularly strong
T-cell immune response in a few subjects predisposed to develop autoimmunity,
immune-related AEs may nonetheless be observed.
Toxicology findings recorded in preclinical studies include hepatocellular
necrosis, vascular/perivascular mononuclear cell infiltration and thrombosis,
all partially recovered. The hepatocellular necrosis correlated with an
increase in alanine aminotransferase (ALT) and aspartate aminotransferase (AST)
levels. To mitigate the thromboembolic risks observed in preclinical studies
with PRECIOUS-01, subjects who have active thromboembolic events that require
anti-coagulation were excluded from the trial. Additionally, all subjects will
be closely monitored for these associated laboratory parameters on Days 1, 7,
and 14 of Cycle 1 and on Days 1 and 7 during Cycle 2 and 3. If needed, the
frequency of monitoring for these changes will be appropriately adapted during
the trail per subject.
The toxicology results also suggest an increase in IFN-γ and TNF-α production,
which can potentially be associated with fever, chills, dizziness and bone
marrow toxicity. The levels of IFN-γ and TNF-α will be carefully monitored as
detailed in the Schedule of Assessments.
Administration of PRECIOUS-01 was associated with lower white blood cell and
platelet count, increased red blood cell mass, reticulocytes and circulating
enzyme levels. These were fully recoverable.
Additional to potential risks related to administration of PRECIOUS-01, there
are also procedural risks related to blood sampling, other possible
subcutaneous injections and diagnostic procedures. Blood sampling and
subcutaneous injections may cause pain, bleeding, bruising, and/or infections
at the site of cannula insertion. Rare complications due to blood sampling are
syncope, thrombophlebitis, as well as accidental punctures of an artery or
nerve.
Benefits
This is the first clinical trial with PRECIOUS-01. The primary aim of this
trial is to obtain safety, and tolerability data when PRECIOUS-01 is
administered to subjects with NY-ESO-1-positive solid tumors. In addition to
the minimal risk using PRECIOUS- 01 (please see IB), preclinical experience
with the compound indicates a potential benefit that might be expected in terms
of an immune response against the tumor that might lead to a beneficial
clinical outcome.
The safety and efficacy data, together with the immunological data obtained
from this trial, will help establish the treatment regimen and a Recommended
Phase II Dose (RP2D) that can form the basis for a recommended dose suitable
for subsequent clinical trials.
Benefit/risk ratio
The anticipated risks, based on the non-clinical experience with PRECIOUS-01,
are expected to be manageable. The importance of the objective of this trial is
considered to outweigh the risks and burdens to the subjects. Measures are
implemented to minimize burdens and risks for subjects. Subjects will be
monitored closely for the occurrence of any significant clinical events and
treatment will only continue if it is considered safe and appropriate to do so.
The benefit/risk assessment is favorable and justifies the planned trial.
Geert Grooteplein 26
Nijmegen 6525GA
NL
Geert Grooteplein 26
Nijmegen 6525GA
NL
Listed location countries
Age
Inclusion criteria
1. Age >=18 years at time of signing informed consent.
2. Performance status (ECOG <= 1) (Appendix II).
3. Estimated life expectancy of at least 6 months.
4. Histologically or cytologically confirmed advanced and /or metastatic solid
tumor with progressive disease at baseline, for whom no standard treatment is
available. Suitable solid tumor indications include non-small cell lung cancer
(NSCLC), melanoma, epithelial ovarian cancer, bladder cancer, breast cancer,
and synovial sarcoma, adenoid cystic carcinoma, cervical cancer, endometrial
cancer, lung cancer, pancreatic cancer, prostate cancer, myxoid and round cell
liposarcoma, neuroblastoma, vulvar cancer, esophageal cancer, hepatocellular
cancer, and head and neck cancer.
5. Subject with evaluable disease per RECIST v1.1.
6. Adequate hematologic, renal and liver function as defined by laboratory
values performed within 14 days of start of treatment:
a. Hemoglobin (Hb) >= 6 mmol/L;
b. Absolute Lymphocyte Count (ALC) > 0.8 x 109/L;
c. Absolute Neutrophil Count (ANC) >= 1.5 × 109/L;
d. Platelet count > 100 x 109/L;
e. Serum creatinine <= 1.5 x ULN or calculated creatinine clearance >= 60 mL/min
(as determined by MDRD [Modification of Diet in Renal Disease]) for patients
with serum creatinine levels > 1.5 x ULN;
f. Serum bilirubin < 25 µmol/L;
g. Aspartate Aminotransferase (AST), and Alanine Aminotransferase (ALT) <= ULN
unless related to liver metastasis (in which case levels should be < 3 x ULN).
h. Alkaline Phosphatase (ALP) <= ULN unless related to liver or bone marrow
metastases (in which case levels should be <= 3 x ULN).
7. Previous therapy-derived toxicities should be resolved to Grade < 2
according to CTCAE v5.0 (Appendix I), with exceptions for alopecia.
8. All subjects of childbearing potential (defined as < 2 years after last
menstruation or not surgically sterile) must have a negative highly sensitive
pregnancy test at screening (urine/serum) and agree to use a highly effective
method for contraception according to the EU Clinical Trial Facilitation Group
guidance from time of signing the informed consent form (ICF) until at least
120 days after the last administration of PRECIOUS-01. The partners of subjects
with childbearing potential must also apply contraceptive methods, and are
recommended not to donate sperm.
9. Before registration, ability of subject to give written informed consent
according to International Council for Harmonisation (ICH) Good Clinical
Practice (GCP), and national rules/local regulations.
10. Expected adequacy of follow-up.
Exclusion criteria
1. Second malignancy in the previous 2 years, with the exception of adequately
treated in situ carcinoma of the cervix uteri and basal or squamous cell
carcinoma of the skin,
2. Clinical suspicion or radiological evidence of active brain metastases.
Patients with brain metastases that have been treated previously and are proven
stable (computed tomography [CT] or magnetic resonance imaging [MRI] < 30 days)
and without steroids for > 3 months are allowed.
3. Subjects with thromboembolic events within the past year.
4. Subjects suffering from melanoma, non-Hodgkin lymphoma, or renal cell
carcinoma who have a serum Lactic Acid Dehydrogenase (LDH) > ULN.
5. Subjects on any other anticancer therapy (cytotoxic, biologic or
investigational agents), unless at least 4 weeks (or 5 half-lives, whichever is
shorter, 6 weeks for mitomycin-C or nitrosoureas), have elapsed since the last
dose before the first administration of PRECIOUS-01. At least 4 weeks should
have elapsed since receiving palliative radiotherapy. Chronic treatment with
non-investigational gonadotropin-releasing hormone analogs or other hormonal or
supportive care is permitted.
6. Subjects with major surgery within 4 weeks before initiating treatment or
with minor surgical procedure within 7 days before initiating treatment (except
for port-a-cath or central line i.v. placement, or biopsy), or anticipation of
the need for major surgery during the course of the trial treatment.
7. Concomitant use of oral or i.v. immunosuppressive drugs. Inhaled, topical or
intranasal steroids and adrenal replacement steroids < 10 mg/day (prednisone
equivalent) are permitted in the absence of auto-immune disease.
8. Uncontrolled infectious disease, i.e., negative testing for human
immunodeficiency virus (HIV), Hepatitis B Virus (HBV), Hepatitis C Virus (HCV)
and syphilis (Treponema Pallidum Hemagglutination Assay [TPHA]).
9. (Systemic) autoimmune disease such as, but not limited to, inflammatory
bowel disease, multiple sclerosis and lupus. Subjects with type 1 diabetes
mellitus, hypothyroidism after autoimmune thyroiditis and skin disorders
(eczema and psoriasis) are not excluded.
10. History of clinically significant cardiovascular disease (<= 6 months prior
to Day 1 on trial) such as stroke, Transient Ischemic Attack (TIA), unstable
angina, New York Heart Association (NYHA) Grade II or greater congestive heart
failure, myocardial infarction, uncontrolled hypertension, cardiac arrhythmia
requiring medication, relevant pathological ECG findings or uncontrolled
hypertension (systolic > 150 mm Hg and/or diastolic > 100 mm Hg).
11. Serious (bleeding and clotting) condition(s) that may interfere with safe
administration of PRECIOUS-01.
12. Abnormal or clinically significant coagulation parameters at the discretion
of the Clinical Investigator, i.e.:
a. Prothrombin Time - International Normalized Ratio (PT-INR)
b. Activated Partial Thromboplastin Time (APTT)
c. Subjects being treated with anticoagulants are excluded if the coagulation
parameters are outside the therapeutic intervals as described in the Summary of
Product Characteristics (SmPC) for the administered treatment.
13. Evidence of any other conditions (such as psychological/familial
sociological/geographical issues, psychiatric illness, infectious diseases,
physical examination or laboratory findings) that may interfere with the
planned treatment, affect subject compliance or place the subject at high risk
from treatment-related complications. These conditions must be discussed with
the subject before registration in the trial.
14. History of severe allergic episodes and/or Quincke*s edema.
15. Prior allogeneic tissue/solid organ transplant, stem cell or bone marrow
transplant.
16. Known hypersensitivity to any component of PRECIOUS-01.
17. Pregnant or lactating women. A highly sensitive pregnancy test
(urine/serum) must be performed within 7 days prior to start of trial treatment
for confirmation in case of childbearing potential.
Design
Recruitment
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 |
---|---|
EudraCT | EUCTR2017-002567-41-NL |
CCMO | NL72876.091.20 |