This study has been transitioned to CTIS with ID 2023-509908-15-00 check the CTIS register for the current data. All primary and secondary objectives will compare zanubrutinib (also known as BGB-3111) plus rituximab followed by zanubrutinib…
ID
Source
Brief title
Condition
- Lymphomas non-Hodgkin's B-cell
Synonym
Research involving
Sponsors and support
Intervention
Outcome measures
Primary outcome
The primary efficacy analysis of PFS will be conducted as assessed by
independent central review, per the 2014 Lugano Classification for NHL.
Progression-free survival will be compared between the 2 arms using a log-rank
test stratified by MIPI score (low vs. intermediate or high), age (>= 70 years
vs. < 70 years), and geographic region (North America/Europe and Asia-Pacific
region.
The primary hypothesis testing for PFS by independent central review is for
non-inferiority. The non inferiority of zanubrutinib plus rituximab (R)
followed by zanubrutinib monotherapy to bendamustine plus rituximab (BR)
followed by observation will be tested for the Intent-to-Treat analysis set
under the prespecified margin of 1.17 (hazard ratio of zanubrutinib + R to
BR). The primary objective of the study is met if the non inferiority is
demonstrated. The null and alternative hypotheses for testing PFS non
inferiority of Arm A to Arm B are as follows:
H0N1: Hazard ratio (HR) (Arm A/Arm B) >= 1.17
HaN1: HR (Arm A/Arm B) < 1.17
The HR and its 2-sided 95% confidence interval will be estimated from a
stratified Cox regression model. The p-value to test H0N1 will be based on Wald
test for the treatment effect from the Cox regression. The distribution of
PFS, including median PFS and PFS rate at selected timepoints such as 12 and 24
months, will be estimated using the Kaplan-Meier method for each arm.
There will be 2 interim analyses of PFS determined by independent central
review, and performed when approximately 157 and 210 events (60% and 80% of the
target number of events at final analysis) from the Arms A and B are observed.
It is estimated that it will take approximately 46 and 61 months from the study
start to observe 157 and 210 events, respectively, from 500 patients. The
final analysis of PFS will take place after 262 events are observed, which is
estimated to be approximately 81 months from study start.
If non-inferiority is demonstrated at either an interim or final analysis,
superiority of zanubrutinib + R followed by zanubrutinib monotherapy to BR
followed by observation will be tested next. The monitoring boundary for the
superiority test is based on the O*Brien-Fleming type alpha spending function.
The monitoring boundary for the non-inferiority test at the interim analyses is
simply the monitoring boundary for the superiority test as described by the
hazard ratio. These boundaries are depicted in Table 7 and Table 8. The
p-value will be used for the primary inference.
Justification of Non-Inferiority Margin
The non-inferiority margin was conservatively derived using the 95%-95% fixed
margin method based on the Food and Drug Administration (FDA) Guidance for
Industry: Non-Inferiority Clinical Trials to Establish Effectiveness (FDA,
2016). The PFS HR of bendamustine plus rituximab compared to rituximab plus
cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP) in STIL
frontline study (Rummel et al, 2013) in the MCL subgroup was 0.49 (95% CI
0.28-0.79). The PFS HR of bendamustine plus R-CHOP/rituximab plus
cyclophosphamide, vincristine, and prednisone (R-CVP) in BRIGHT study (Flinn et
al, 2014) in the MCL subgroup was 0.4 (95% CI 0.21-0.75). Both studies are
well controlled with disease status measured by radiology assessment and
conducted in first-line patients, same as this study. The MCL subgroups were
stratified for randomization in both studies with sample size of 94 (for BRIGHT
study) and 74 (for STIL study). The PFS status in MCL subgroups were followed
enough to estimate the median PFS for both arms with enough precision. The
efficacy of bendamustine plus rituximab (M1) in PFS hazard ratio scale was
estimated as 0.676 from the results of these 2 studies by a fixed effects
meta-analysis. Requiring 60% of M1 to be retained in zanubrutinib plus
rituximab, a non-inferiority margin of 1.17 is generated. The margin is also
clinically relevant.
Secondary outcome
Secondary efficacy endpoints will be analyzed and compared between the 2
treatment groups if the non inferiority test of the primary endpoint is
significant either at the interim or final analyses. PFS, ORR, DOR, rate of CR
or complete metabolic response, and time to response based on the assessment by
independent central review as well as investigator assessment will be analyzed.
• PFS by investigator assessment will be calculated based on
investigator-assessed tumor responses. PFS by investigator assessment will be
analyzed using the same analysis methods as the primary endpoint of PFS by
independent central review.
• ORR will be estimated as the crude proportion of patients in each treatment
group who achieve partial response (PR) or higher. Associated 95%
Clopper-Pearson confidence intervals will be calculated by treatment group.
The odds ratio (and 95% confidence intervals), which will be provided as a
measure of the relative treatment effect, will be estimated using the
stratified Cochran-Mantel-Haenszel method.
• Duration of response: The distribution of DOR, including median and other
quartiles, will be estimated using the Kaplan-Meier method for each treatment
group. Hypothesis testing comparing DOR between the 2 treatment groups will
not be performed.
• Overall survival: OS between the treatment groups will be compared using the
same methods employed for the PFS comparison.
• Rate of complete response or complete metabolic response will be analyzed
using the same methods employed for ORR analysis.
• Time to response will be summarized for each treatment group using sample
statistics, such as sample mean, median, and standard deviation.
• Patient-reported outcomes: The EORTC QLQ-C30 and EQ-5D-5L questionnaires will
be utilized. The scores and their changes from baseline will be summarized and
compared between the 2 treatment groups.
Background summary
Mantle cell lymphoma (MCL) is diagnosed in 0.51-0.55/100,000 persons per year
in the United States of America, making up 6% of all non-Hodgkin lymphoma
(NHL), with a similar incidence rate worldwide (Smedby and Hjalgrim, 2011; Wang
et al, 2015). MCL is a cancer of older persons with a median age at diagnosis
of approximately 70 years (Sharman et al, 2017, Dreyling et al, 2017) and a
male preponderance of roughly 2.5:1 (Barista et al, 2001; Smedby and Hjalgrim,
2011). Patients usually present with advanced disease; 70% are diagnosed in
stage IV. Most commonly, there is generalized lymphadenopathy and organomegaly.
Approximately 10-20% of patients present with bone marrow involvement, and B
symptoms (fever, night sweats, and weight loss) are present in approximately
40% of patients. Extranodal involvement of the gastrointestinal tract,
particularly the colon, is common, but central nervous system involvement is
uncommon (Skarbnik and Goy, 2015).The diagnosis is based on
pathological assessment with typical immunophenotype of CD5+, CD20+, CD43+,
CD23-/+,cyclin D1+, and CD10-/+ (NCCN Guideline, 2018).
In addition to the classic MCL immunophenotype of CD20+, the pathognomonic
feature of MCL is overexpression of cyclin D1, which is a consequence of
juxtaposition of the proto-oncogene CCND1 on chromosome 11q13 to the
immunoglobulin (Ig) heavy chain gene at chromosome 14q32. Cyclin D1 can be
confirmed by immunohistochemistry in almost 100% of specimens. The molecular
consequence of cyclin D1 overexpression is deregulation of cell cycle control
by overcoming the suppressor effect of the retinoblastoma 1 (RB1) and the cell
cycle inhibitor p27. Frequent companion anomalies include secondary chromosomal
and molecular alterations targeting proteins that regulate the cell cycle and
senescence (B lymphoma Mo-MLV insertion region 1 homolog [BMI1], inhibitor of
kinase 4a [INK4a], alternate reading frame [ARF], cyclin D-dependent kinase
[CDK]4, and RB1), and interfere with the cellular response to deoxyribonucleic
acid (DNA) damage (ataxia telangiectasia mutated [ATM]), checkpoint kinase 2
[CHK2] and p53) (Rickert, 2013). Concomitantly, β2 microglobulin and lactate
dehydrogenase (LDH) are increased in 56% and 45% of patients, respectively;
these markers should be monitored for disease activity and are also prognostic.
The Mantle Cell Lymphoma International Prognostic Index (MIPI) is composed of
the parameters of the Eastern Cooperative Oncology Group (ECOG) performance
status, age, leukocyte count, and LDH, and classifies patients into low,
intermediate, and high-risk groups, with corresponding median overall survival
(OS) time of 72 months, 51 months, and 29 months, respectively (Vose, 2013).
The histological pattern of MCL is also prognostic, with complete response (CR)
rates of cyclophosphamide, doxorubicin, vincristine, and prednisone (CHOP) for
mantle-zone/nodular/diffuse patterns of 73%, 25%, and 19%, respectively, and 3
year survival rates of 100%, 50%, and 55%, respectively (Barista, 2001).
Another prognostic factor is the expression of the proliferation-associated
antigen Ki-67; 60% of patients have over 10% of their tumors staining positive
for Ki-67, with highly proliferative cases showing a much poorer outcome than
tumors with low proliferation.
Management of MCL has been with chemo-immunotherapeutic combinations, most
commonly rituximab-CHOP (R-CHOP) or bendamustine plus rituximab (BR)
combinations that result in high response rates; however, patients invariably
relapse (Rummel et al, 2013; Kluin-Nelemans et al, 2012). Younger, fit
patients have the option of high intensity chemotherapy, such as
hyper-fractionated cyclophosphamide, vincristine, doxorubicin, dexamethasone
(hyper- CVAD), or induction chemotherapy followed by stem cell transplantation,
with many patients achieving prolonged remissions (Dreyling et al, 2005;
Romaguera et al, 2005). Among the available therapies, the BR combination has
demonstrated better tolerability with lower rates of hematological toxicities,
infections, peripheral neuropathy, alopecia, and stomatitis when compared with
R-CHOP. Furthermore, the BR combination also showed better efficacy in that
median progression-free survival of BR is significantly longer than R-CHOP
(69.5 months versus 31.2 months; p < 0.0001) in treatment-naive MCL patients
(Rummel et al, 2013; Flinn et al, 2014). Combination therapy with bendamustine
and rituximab has become one of the most used first-line regimens for MCL,
given the recent evidence showing longer progression-free survival with this
combination than with R-CHOP (35.4 versus. 22.1 months); and a better safety
profile (Martin et al, 2021). However, elderly patients and those with
comorbidities that preclude treatment with standard chemoimmunotherapy regimens
and stem cell transplantation have few options, and treatment is largely
palliative. No standard approach to treatment has been defined for these
patients (Dreyling et al, 2017).
Additionally, there is as yet no conclusive evidence that rituximab maintenance
therapy following a combination of rituximab and bendamustine treatment
provides a benefit in MCL. In an ongoing study of 120 patients with previously
untreated stage II (with bulky disease > 7 cm), III or IV MCL treated with up
to 6 cycles of rituximab and bendamustine followed by 1:1 randomization to
either rituximab maintenance (375 mg/m2 every 2 months for a total of 2 years)
or observation only arms, after a median time of observation of 4.5 years, no
significant difference in progression-free survival (PFS) could be observed
between the 2 arms (p=0.130, 47 events, hazard ratio [HR] 0.64, 95% CI:
0.73-1.14). The median for rituximab maintenance was not yet reached, whereas
for the observation arm the median was 54.7 months (95% CI: 40.1. - not yet
reached). There was no difference in OS between arms (p = 0.271, 27 events, HR
1.53, 95% CI 0.73 - 3.32) with a median of 69.6 months for rituximab
maintenance versus a median not yet reached in the observation arm (Rummel et
al, 2016). National Comprehensive Cancer Network (NCCN) guidelines currently
recommend against rituximab maintenance following bendamustine-based therapy
for mantle cell lymphoma (NCCN v5.2018).
Study objective
This study has been transitioned to CTIS with ID 2023-509908-15-00 check the CTIS register for the current data.
All primary and secondary objectives will compare zanubrutinib (also known as
BGB-3111) plus rituximab followed by zanubrutinib monotherapy versus
bendamustine plus rituximab followed by observation only.
Primary:
• To compare efficacy, as measured by progression-free survival (PFS)
determined by independent central review
Secondary:
• To evaluate efficacy, as measured by the following:
- Progression-free survival determined by investigator assessment
- Overall response rate (ORR), as determined by independent central review and
by investigator assessment
- Duration of response (DOR), as determined by independent central review and
by investigator assessment
- Overall survival (OS)
- Rate of complete response (CR) or complete metabolic response determined by
independent central review and by investigator assessment
- Time to response, as determined by independent central review and by
investigator assessment
- Patient reported outcomes
- To evaluate safety and tolerability
Exploratory:
- To evaluate the pharmacokinetics (PK) of zanubrutinib (zanubrutinib plus
rituximab arm only) when co administered with rituximab
- To evaluate the correlation of certain pathologic features (eg, Ki-67) and
molecular characteristics (including but not limited to mutations in genes and
pathways of the cell cycle and senescence, DNA damage response, and cell
survival) with rate of CR or complete metabolic response, ORR, DOR, and PFS
- To evaluate the mechanisms of resistance to zanubrutinib
Study design
This is an international, multicenter, Phase 3, open-label, randomized,
active-controlled study of zanubrutinib plus rituximab followed by zanubrutinib
monotherapy versus bendamustine plus rituximab followed by observation in
approximately 500 patients with previously untreated mantle cell lymphoma (MCL)
who are ineligible for stem cell transplantation due to age or comorbidities.
The primary efficacy endpoint is PFS as determined by independent central
review. Disease response will be assessed per the Lugano Classification for
Non-Hodgkin Lymphoma (NHL) (Cheson et al, 2014), hereafter referred to as
Lugano Classification for NHL.
Central randomization (1:1) will be used to assign patients to one of the
following study drug treatments:
• Arm A: zanubrutinib plus rituximab for 6 cycles, followed by zanubrutinib
monotherapy
• Arm B: bendamustine plus rituximab for 6 cycles, followed by observation
Randomization will be stratified by MCL International Prognostic Index (MIPI)
score (low vs. intermediate or high), age (>= 70 years vs. < 70 years), and
geographic region (North America/Europe and Asia-Pacific region).
Study treatment must commence within 5*days after randomization. Each cycle of
zanubrutinib and rituximab or bendamustine and rituximab consists of 28 days.
Study drug treatments will be administered as follows, depending on treatment
assignment:
In Arm A, zanubrutinib will be administered as two 80-mg capsules orally twice
a day (160 mg twice a day) with or without food. Rituximab will be
administered intravenously at a dose of 375 mg/m2 on Day 1 of Cycles 1 to 6
only. Zanubrutinib will be administered prior to the start of rituximab
infusion. For patients considered by the investigator to be at high risk for
infusion reaction, rituximab may be administered by split dosing over more than
one day for the first and/or subsequent cycles as per institutional guidelines.
(NOTE: Rituximab split dosing is not available in Japan based on local
labeling). Following completion of Cycle 6, patients in Arm A will continue on
zanubrutinib monotherapy until disease progression, withdrawal of consent,
death, lost to follow-up, or end of study, whichever occurs first. Following
disease progression, subjects will be followed further for survival and
subsequent MCL therapies.
In Arm B, bendamustine will be administered intravenously at a dose of 90
mg/m2/day on Days 1 and 2 of Cycles 1 to 6, and rituximab will be administered
intravenously at a dose of 375 mg/m2 on Day 1 of Cycles 1 to 6. Following
completion of Cycle 6, patients in Arm B will receive no further treatment but
will continue to be observed (including assessments for safety and efficacy)
until disease progression, withdrawal of consent, death, lost to follow up, or
end of study, whichever occurs first. Following disease progression, subjects
will be followed further for survival and subsequent MCL therapies.
Intervention
• Arm A: zanubrutinib plus rituximab for 6 cycles, followed by zanubrutinib
monotherapy
• Arm B: bendamustine plus rituximab for 6 cycles, followed by observation
Rituximab is given by intravenous (IV) infusion
Study burden and risks
Zanubrutinib is an experimental agent and therefore it is not known if
zanubrutinib will have any direct benefit to the patient. Taking part in this
medical scientific research may or may not improve the patient's health. Even
if there is no direct benefit others may benefit from what is learned from this
study.
Disadvantages of participation in the study may be:
- possible side effects/complications of the intervention;
- possible adverse effects/discomforts of the measurements in the study.
Participation in the study also means:
- additional time;
- an additional or an extended hospitalization;
- additional testing;
- that the patient has appointments to attend;
Gateway Drive, 3rd Floor 1840
San Mateo CA 94404
US
Gateway Drive, 3rd Floor 1840
San Mateo CA 94404
US
Listed location countries
Age
Inclusion criteria
- >=*70 years of age at the time of informed consent, OR
>= 60 and < 70 years of age with comorbidities precluding autologous stem cell
transplantation
- Histologically confirmed diagnosis of MCL based on the World Health
Organization 2016 classification
- No prior systemic treatments for MCL
- Presence of measurable disease
- Availability of archival tissue confirming diagnosis of MCL, or willing to
undergo fresh tumor biopsy
- ECOG performance status of 0, 1, or 2
- Life expectancy of >=*3 months
- Adequate organ function (refer to Protocol Synopsis for more details)
- Female patients of childbearing potential must practice highly effective
methods of contraception
- Male patients are eligible if abstinent, vasectomized or if they agree to the
use of barrier contraception in combination with other methods
- Ability to provide written informed consent and ability to understand and
comply with the requirements of the study
- For all patients irrespective of their age, Creatinine clearance of >= 30
mL/min
Exclusion criteria
- Known central nervous system involvement by lymphoma
- Prior hematopoietic stem cell transplantation
- Prior exposure to a BTK inhibitor, rituximab, or bendamustine
- Patients for whom the goal of therapy is tumor debulking prior to stem cell
transplant
- Prior malignancy within the past 3 years, except for curatively treated basal
or squamous cell skin cancer, superficial bladder cancer, carcinoma in situ of
the cervix or breast, or localized Gleason score 6 prostate cancer
- Clinically significant cardiovascular disease (for more details refer to the
Protocol)
- History of severe bleeding disorder such as hemophilia A, hemophilia B, von
Willebrand disease, or history of spontaneous bleeding requiring blood
transfusion or other medical intervention
- History of stroke or intracranial hemorrhage within 6 months before first
dose of study drug
- Unable to swallow capsules or disease significantly affecting
gastrointestinal function
- Active fungal, bacterial and/or viral infection requiring systemic therapy
- Underlying medical conditions that, in the investigator*s opinion, will
render the administration of study drug hazardous or obscure the interpretation
of safety or efficacy results
- Known infection with human immunodeficiency virus (HIV), or serologic status
reflecting active hepatitis B or C infection (Refer to the Protocol for more
details)
- Major surgery within 4 weeks of the first dose of study drug
- Pregnant or lactating women
- Vaccination with a live vaccine within 35 days prior to the first dose of
study drug
- Ongoing alcohol or drug addiction
- Hypersensitivity to zanubrutinib, bendamustine, or rituximab or any of the
other ingredients of the study drugs
- Requires ongoing treatment with a strong CYP3A inhibitor or inducer
- Concurrent participation in another therapeutic clinical trial.
- Patients enrolled in Germany only, who are severe immunocompromised
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 | CTIS2023-509908-15-00 |
EudraCT | EUCTR2019-000413-36-NL |
ClinicalTrials.gov | NCT04002297 |
CCMO | NL75419.078.20 |