Skip to main content
Erschienen in: Advances in Therapy 1/2022

Open Access 25.11.2021 | Original Research

Population Pharmacokinetics and Exposure–Response Analyses for Venetoclax in Combination with R-CHOP in Relapsed/Refractory and Previously Untreated Patients with Diffuse Large B Cell Lymphoma

verfasst von: Divya Samineni, Weize Huang, Leonid Gibiansky, Hao Ding, Rong Zhang, Chunze Li, Arijit Sinha, Richa Rajwanshi, Kathryn Humphrey, Alexandra Bazeos, Ahmed Hamed Salem, Dale Miles

Erschienen in: Advances in Therapy | Ausgabe 1/2022

Abstract

Introduction

Outcomes remain poor in patients with diffuse large B cell lymphoma (DLBCL) who overexpress BCL-2 protein. We present population pharmacokinetics (PopPK) and exposure–response (ER) analyses for venetoclax (a selective BCL-2 inhibitor) administered with rituximab-cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP) in patients with relapsed/refractory (R/R) and previously untreated (1L) non-Hodgkin lymphoma (NHL) from the phase 1b/2 CAVALLI study, to confirm dose selection for future studies.

Methods

Analyses included 216 patients with R/R or 1L NHL treated for eight 21-day cycles with 400–800 mg venetoclax (cycle 1: days 4–10; cycles 2–8: days 1–10) in combination with R for eight cycles and CHOP for 6–8 cycles. A legacy PopPK model for venetoclax was used to describe the observed data and provide post hoc PK parameters. Venetoclax steady-state exposure (AUCss) was used to predict clinical efficacy, safety, or tolerability. To isolate the effect of venetoclax, ER analyses referenced data from the R-CHOP arm of a historical control study, GOYA, in 1L DLBCL.

Results

There was no significant association between venetoclax AUCss and progression-free survival or complete response either for all-comers or the BCL-2-immunohistochemistry-positive subpopulation. No statistically significant trends were observed with venetoclax AUCss and the key grade ≥ 3 adverse events and serious adverse events. Similar dose intensities were observed for venetoclax and R-CHOP components across venetoclax exposures, suggesting venetoclax did not impact delivery of the R-CHOP backbone.

Conclusions

The PopPK and ER analyses, in addition to the positive benefit–risk observed in the clinical data, support the selection of 800 mg venetoclax given with R-CHOP for future studies in BCL-2-immunohistochemistry-positive patients with 1L DLBCL.

Trial Registration

ClinicalTrials.gov Identifier NCT02055820.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1007/​s12325-021-01919-z.
Key Summary Points
The pharmacokinetics (PK) of venetoclax in the CAVALLI study were adequately described using a previously developed legacy population pharmacokinetics (PopPK) model, with study-specific (CAVALLI) effects on apparent clearance (CL/F) and apparent central volume of distribution (V2/F) added to the PopPK model to account for the possible differences between the population of CAVALLI study patients relative to the population of patients from the prior analysis.
Encouraging outcomes for efficacy were achieved without compromising delivery of rituximab-cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP) therapy; increased venetoclax exposures did not translate into improved efficacy for investigator-assessed progression free survival (INV-PFS) and positron emission tomography-computed tomography assessed complete response (PET-CR) rates in all-comers or BCL-2-IHC-positive subpopulations.
Venetoclax plus R-CHOP demonstrated increased but manageable toxicity versus R-CHOP alone, with adverse events expected on the basis of the mechanism of action of venetoclax and consistent with the toxicity profile of R-CHOP. Data did not suggest that higher doses of venetoclax would cause additional toxicity compared with lower doses of venetoclax within the CAVALLI study.

Introduction

The B cell lymphoma 2 protein (BCL-2) family proteins comprise the sentinel network that regulates the mitochondrial or intrinsic apoptotic response [1]. A recognized hallmark of cancer development, overexpression of anti-apoptotic BCL-2 family proteins mediates resistance to chemotherapy in experimental models of lymphoma, and is associated with poorer prognosis in patients with diffuse large B cell lymphoma (DLBCL) treated with cyclophosphamide, doxorubicin, vincristine, and prednisone (CHOP) [4]. Venetoclax, a selective inhibitor of BCL-2 [5], is approved globally for monotherapy or combination therapy in patients with chronic lymphocytic leukemia (CLL) or small lymphocytic lymphoma (SLL), as well as treatment-naïve unfit patients with acute myeloid leukemia [6, 7]. Venetoclax has also demonstrated significant clinical activity across a broad range of non-Hodgkin lymphoma (NHL) subtypes [8].
CAVALLI is a phase 1b/2 multicenter, open-label study investigating the safety and efficacy of venetoclax plus an anti-CD20 antibody (rituximab [R] or obinutuzumab [GA101; G]) in combination with standard CHOP chemotherapy in patients with NHL. The phase 1b portion of the study included two parallel treatment arms, with planned venetoclax doses ranging from 200 to 800 mg orally at two different dosing schedules: once daily (21 days per cycle) or a noncontinuous dosing schedule of 10 doses in a 21-day cycle (10/21-day dosing), plus standard cycles of R-CHOP or G-CHOP in relapsed/refractory (R/R) or previously untreated patients with NHL [9]. A greater than anticipated incidence of hematologic toxicity was evidenced by dose-limiting toxicities (DLTs) at the initial starting dose of 200 mg administered daily in a 21-day cycle for eight cycles. The phase 1b part of CAVALLI established the recommended phase 2 dose of venetoclax as 800 mg in a 10/21-day dosing schedule, i.e., days 4–10 of cycle 1 and days 1–10 of cycles 2–8, in combination with R for eight cycles and CHOP [9] for 6–8 cycles in patients with first-line (1L) DLBCL. The incidence of hematologic toxicity observed in patients following this schedule was lower than in patients following the once-daily dosing schedule; as such, the noncontinuous dosing schedule was preferred, given that it allowed administration of higher doses of venetoclax. The G-CHOP arm was not expanded to phase 2 in the light of results from the phase 3 GOYA study in 1418 patients with previously untreated DLBCL, which showed no improvement in progression-free survival (PFS) with G-CHOP over R-CHOP in 1L therapy [9, 10].
After 30.8 months’ median follow-up, venetoclax plus R-CHOP continued to be associated with improved investigator-assessed (INV)-PFS in the all-comer 1L DLBCL population (adjusted Cox regression hazard ratio of 0.61; 95% confidence interval [CI] of 0.43–0.87), compared with matched controls from the GOYA study. Promising efficacy (PFS) was evident in the poor prognostic BCL-2-immunohistochemistry (IHC)-positive subpopulation (adjusted Cox proportional hazards [CPH] ratio of 0.55; 95% CI of 0.34–0.89 in CAVALLI versus the R-CHOP arm of GOYA) [11].
As a complementary report to the initial efficacy, safety, and biomarker analyses of CAVALLI phase 2 [11], the present study investigated population pharmacokinetics (PopPK) and exposure–response (ER) characteristics of venetoclax from CAVALLI to confirm the dose selection of venetoclax in combination with R-CHOP chemoimmunotherapy for future studies.

Methods

Compliance with Ethics Guidelines

All procedures involving human participants in the CAVALLI and GOYA studies were in accordance with the ethical standards of the institutional review board and with the 1964 Helsinki declaration and its later amendments. The CAVALLI study was approved by the institutional review boards/ethics committees at all participating institutions including Comite de Protection des Personnes, WIRB, and CEIC Hospital Universitario (Table S1 in the supplementary material). This article does not contain any studies with animals performed by any of the authors. Informed consent was obtained from all individual participants included in the study. This article does not contain any identifying information about participants and therefore consent was not sought from participants for publication.

Studies

Fifty-six patients with R/R or 1L NHL (arm A with R-CHOP, N = 24; arm B with G-CHOP, N = 32) were enrolled in the phase 1b portion and 211 patients with 1L DLBCL were enrolled in the phase 2 portion of the CAVALLI study, previously described by Zelenetz et al. [9]. The data cutoff date for the PopPK and ER analyses in this manuscript for CAVALLI was April 12, 2019.
To isolate the venetoclax effect in the CAVALLI study, the R-CHOP arm of the phase 3 GOYA trial (patients with International Prognostic Index [IPI] 2–5) was used as a historical control for comparative analyses of efficacy, safety, and tolerability. In GOYA, 1418 patients with 1L DLBCL were randomized to receive eight 21-day cycles of G or R plus 6–8 cycles of standard CHOP [10]. Overall, patient characteristics are similar for CAVALLI and GOYA studies. For the efficacy analyses, double-robust methods were applied to the adjustment of the baseline prognostic covariates in the CAVALLI and GOYA studies [12]. The data cutoff date for GOYA was April 29, 2016.

Analytical Methods

Plasma concentrations of venetoclax were determined using validated liquid chromatography methods with tandem mass spectrometry. The lower limit of quantification (LLOQ) for venetoclax was 2.08 ng/mL or 2.18 ng/mL [13]. BCL-2 protein expression was assessed by IHC (BCL-2 cutoff, 50% medium/high expression) and BCL-2 translocations by fluorescence in situ hybridization (FISH) [12]. Intensive PK sampling was performed for venetoclax on cycle 1 day 4 and cycle 2 day 1 at pre- and up to 8 h post-dose and at pre-dose on cycle 1 day 8 and cycle 2 day 10.

PopPK Modeling and Evaluation

A previously developed PopPK legacy model for venetoclax in 505 subjects with R/R CLL/SLL, R/R NHL, and healthy volunteers [13] enrolled in eight phase 1/2 studies was used. Parameter values were fixed at prior estimates to describe the observed PK and provide post hoc predicted PK parameters for venetoclax in the CAVALLI study. The appropriateness of the legacy model for predicting exposures in CAVALLI was evaluated using diagnostic plots, visual predictive checks (VPCs), and normalized prediction distribution errors plots [1416].

Determination of Venetoclax Exposure

Individual venetoclax values for area under the curve (AUC) at steady state (AUCss nominal) were estimated using empirical Bayesian post hoc estimates of the relevant PK parameters, the protocol-assigned dose, and covariate values on day 1 as follows:
$$ {\text{AUC}}_{{{\text{ss}}\,{\text{nominal}}}} = \, D_{{{\text{nom}}}} \times {\text{F1}}/\left( {{\text{CL}}/{\text{F}}} \right), $$
(1)
where Dnom is the nominal dose (protocol assigned dose), CL/F is apparent clearance, and F1 is relative bioavailability. Given the dependence of bioavailability on dose that was defined in the legacy PopPK model, Dnom was used to compute the F1 parameter for the exposure measure. Values of covariates at baseline were used to compute CL/F. Patients without evaluable PK data and those not included in the PopPK analysis were excluded.

ER Analyses

To isolate the venetoclax effect, the ER analyses referenced data from patients in the R-CHOP arm of the GOYA trial [10]. Three types of ER analyses were conducted using CAVALLI study data: (1) exposure–efficacy and (2) exposure–dose intensity analyses (using phase 2 patients [1L DLBCL, n = 199] receiving 800 mg venetoclax on the 10/21-day dosing regimen), and (3) exposure–safety analysis (using phase 1b/2 patients receiving 400, 600, or 800 mg venetoclax on the 10/21-day dosing regimen). Further, venetoclax AUCss nominal (Eq. 1) was used as the exposure metric for predicting clinical response.
Exposure–efficacy relationships were conducted using (1) Kaplan–Meier (KM) analysis stratified by venetoclax exposure AUCss nominal, (2) CPH modeling, and (3) logistic regression analysis. Logistic regression and CPH models were used to assess the impact of exposure on the key efficacy endpoints of positron emission tomography-computed tomography-assessed complete response (PET-CR) and investigator-assessed PFS (INV-PFS), respectively, in both all-comers and BCL-2-positive patients. PET-CR was assessed at the end of treatment (EOT) visit, 6–8 weeks from day 1 of the last cycle received. If a patient has not experienced clinical response or death at the data cutoff date, PFS was censored at the day of the last tumor assessment.
Relationships between venetoclax exposure and PFS were first characterized using base Cox models that characterize the marginal effect of venetoclax exposure on PFS without considering covariates. A significance level of α = 0.05 was used to evaluate the exposure coefficient. The hazard function in the CPH model was expressed as:
$$\lambda \left(t\right)={\lambda }_{0}\left(t\right) \mathrm{exp} \left({\beta }^{\tau }\chi \right),$$
(2)
where λ0(t) is the baseline hazard function and \(\chi \) is a vector of predictor variables (continuous exposure [AUCss nominal] included in the base model). The parameter vector β is estimated by maximum partial-likelihood.
Logistic regression models were implemented to assess correlations between venetoclax exposure (AUCss nominal); the probability of PET-CR and treatment-emergent safety endpoints, i.e., grade ≥ 3 adverse events (AEs) like neutropenia, thrombocytopenia, infections, and febrile neutropenia; and serious AEs (SAEs), in the intent-to-treat population. These analyses include the treatment-emergent grade ≥ 3 AEs reported through 30 days after the last dose of venetoclax or CHOP, or 90 days after the last dose of R, whichever is later; SAEs reported until the cutoff date of July 13, 2018 were included. A significance level of α = 0.05 was used to evaluate the exposure coefficient. A covariate analysis was also carried out, in which a forward addition (with α = 0.01) stepwise procedure was implemented.
Covariates tested in the ER analyses for efficacy and safety included demographics (sex, age, body mass index [BMI]), and baseline disease characteristics: Eastern Cooperative Oncology Group performance status (ECOG PS; 0–1 vs 2 +), IPI (high [4, 5] vs non-high [2, 3]), bulky disease (> 7.5 mm: yes vs no), stage (IV vs I–III), lactate dehydrogenase (low/normal vs high), and cell-of-origin (COO), based on likely associations with the clinical efficacy and/or safety endpoints. Missing continuous covariates were imputed by study using the median value of the covariate, and the missing categorical covariates by the most frequent population value. There were no covariates with missing data exceeding 15% of study data. The covariate analysis was conducted using the forward addition procedure. A significance level of α = 0.01 (objective function change of 6.63 points for one parameter) was used. Significance levels of covariate effects (associated p values) were presented.
Relative dose intensity (RDI) for the venetoclax or R-CHOP components was calculated from the first day of venetoclax or R-CHOP treatment until the date of the planned EOT as follows:
$$\frac{(\mathrm{Total\; actual\; dose \;to \;actual \;EOT})/(\mathrm{No}.\mathrm{ of \;actual \;days \;to \;actual \;EOT}) }{(\mathrm{Total \;planned \;dose \;to \;planned \;EOT})/(\mathrm{Total \;days \;to \;planned \;EOT})}.$$
Thus, dose holds and reductions with penalty for early treatment discontinuations due to AEs were accounted for by imputing the actual doses as zero until EOT. The effect of venetoclax exposure (AUCss nominal) on dose intensity of individual R-CHOP and venetoclax components was also evaluated.

Software

PopPK analysis was carried out using nonlinear mixed effects modeling with NONMEM software, Version 7.3.0 (ICON Development Solutions) [17]. The first-order conditional estimation method with interaction (FOCEI) was used for all NONMEM model runs. All ER analyses were performed using R software, version 3.3.3, for Windows (R project, http://​www.​r-project.​org/​). The function glm() with logit link was used for the logistic regression analysis, and coxph() of the survival package was used for CPH modeling.

Results

The baseline demographics and covariates are shown in Table 1.
Table 1
Continuous and categorical covariates at baseline
Continuous covariates
Measure
Result
Patients
Number
232
Age, years
Mean (SD)
61.2 (12.6)
Median (range)
64 (18–85)
Body weight, kg
Mean (SD)
77.8 (16.8)
Median (range)
77.0 (46.0–182)
Categorical covariates
Level
Number (%)
Sex
Male
129 (55.6)
Female
103 (44.4)
Study population
DLBCL
218 (94.0)
FL
10 (4.3)
Transformed lymphoma
2 (0.9)
Other
2 (0.9)
Race
White
165 (71.1)
Asian
5 (2.2)
Black/African American
4 (1.7)
Native Hawaiian or Pacific Islander
3 (1.3)
Unknown
55 (23.7)
Dose
200 mg
7 (3.0)
400 mg
3 (1.3)
600 mg
8 (3.4)
800 mg
214 (92.2)
ECOG PS
0
103 (44.4)
1
94 (40.5)
2
35 (15.1)
IPI
0–1 (Low)
26 (11.2)
2 (Low intermediate)
86 (37.1)
3 (High intermediate)
69 (29.7)
4–5 (High)
51 (22.0)
Stage
I
5 (2.2)
II
30 (12.9)
III
44 (19.0)
IV
153 (65.9)
COO
ABC
51 (27.1)
GCB
113 (60.1)
Unclassified
24 (12.8)
BCL-2 IHC
Positive
120 (60.3)
Negative
79 (39.7)
MYC IHC
Positive
146 (72.6)
Negative
55 (27.4)
DP
DP
91 (45.7)
Non-DP
108 (54.3)
DH
DH
7 (4.5)
Non-DH
150 (95.5)
C3AHIB
Not administered
5 (2.2)
Weak but no moderate or strong
152 (66.7)
Moderate but no strong
65 (28.5)
Strong
6 (2.6)
OATP3HIB
Not administered
225 (98.7)
Administered
3 (1.3)
Region
North America
80 (34.5)
Western Europe
100 (43.1)
Eastern Europe
37 (15.9)
Other
15 (6.5)
Missing continuous covariates were imputed by the median value of the covariate. Missing categorical covariates were imputed by the most frequent value in the population. There were no covariates with missing data fraction exceeding 15% of study data
BCL-2 B cell lymphoma 2 protein, C3AHIB CYP3A inhibitor flag, COO cell-of-origin, DH doubt hit, DLBCL diffuse large B cell lymphoma, DP double positive, ECOG PS Eastern Cooperative Oncology Group performance status, FL follicular lymphoma, IHC immunohistochemistry, OATP3HIB OATP1B3 inhibitor flag, SD standard deviation

PopPK Analysis

Among the 232 patients who received venetoclax in combination with R-CHOP in CAVALLI, 223 (97.8%) had at least one evaluable PK sample, i.e., had at least one quantifiable PK sample, and were included in the analysis (1150 samples in total); all the 32 patients enrolled on the venetoclax + G-CHOP arm in phase 1b had at least one evaluable PK sample (179 samples in total). Exclusion of PK samples obtained more than 10 days after the last dose, those with concentrations below the LLOQ, and/or those with missing time left 1100 quantifiable samples from 260 patients for use in the PopPK analysis. This resulted in a proportion of missing samples of 19%.
The legacy model was applied to the CAVALLI study data. All model parameters were fixed, including the inter-individual and residual variability. The basic diagnostic plots are shown in Fig. 1. The legacy model generally fitted the new data from the CAVALLI study. However, there were some biases (described in the supplementary material); therefore, the base model was improved by adding a multiplicative “study effect” covariate to each of CL/F and apparent central volume of distribution (V2/F) to account for possible differences in PK between CAVALLI and the historical studies used to generate the legacy model.
The study effect covariate was shown to be significant on both parameters. The effect on CL/F was estimated at 1.63 (95% confidence interval [CI] 1.49–1.76) and that on V2/F at 2.33 (95% CI 2.17–2.54). Another model using a similar study effect on bioavailability (instead of separate effects on CL/F and V2/F) was not as good. The parameter estimates and the variability for venetoclax PK are shown in Table 2.
Table 2
Parameter estimates for the final current model
Parameter
Final model
Legacy model estimate
RSE (%)
Variability
Shrinkage
CL/F (L/day)
447
3.51
θCL/F, strong CYP3A inhibitor
0.18
6.03
θCL/F, moderate CYP3A inhibitor
0.84
4.24
θCL/F,OATP1B3 inhibitor
0.85
2.54
θCL/F, GO27878
1.63
4.25
V2/F (L)
118
13.7
θV2/F, CLL/SLL/NHL
1.71
12.5
θV2/F,SEX=female
0.68
6.68
θV2/F,GO27878
2.33
4.67
Q/F (L/day)
97.2
5.54
V3/F (L)
119
4.16
ka(1/day)
3.72
4.09
θF1,FOOD=fasting
0.34
0.97
θF1,FOOD=moderate fat
1.31
8.24
θF1,FOOD=high fat
1.43
1.3
θF1,FOOD=fed
1.23
4.29
F1,DOSE nonlinearity (400 mg as reference)
− 0.18
2.38
ω2CL/F
0.15
9.35
CV = 39.1%
19.0%
ω2V2/F
0.21
6.34
CV = 45.3%
27.7%
ω2F1
0.10
13.37
CV = 31.2%
28.3%
σ2prop
0.22
1.67
σ2add
3.07 × 10–7
39.7
t1/2 (day)
1.05
95% CI 95% confidence interval, C3AHIB CYP3A inhibitor indicator variable, CL clearance, CL/F apparent clearance, CV coefficient of variation, F1 relative bioavailability, ka first-order absorption rate constant, OATP3HIB OATP1B3 indicator variable, Q/F apparent inter-compartmental clearance, RSE residual standard error, V2/F apparent central volume of distribution, V3/F apparent peripheral volume of distribution, ω2 inter-individual variances, σ2 residual variances
In the final model evaluation, goodness-of-fit of the data was confirmed (Fig. 2; described further in the supplementary material, Figs. S1 and S2), with no apparent model deficiencies.

Exposure–Efficacy

The 800 mg venetoclax dose was evaluated for impact on efficacy in the phase 2 part of CAVALLI. To support the previously published clinical findings [11], exposure–efficacy analysis was conducted in both BCL-2 positive (N = 101) and all-comer (regardless of biomarker status, N = 199) patients with 1L DLBCL enrolled in CAVALLI phase 2, who received venetoclax at 800 mg (10-day dosing) plus R-CHOP. KM plots stratified by quartiles of exposure indicated no apparent exposure–INV-PFS relationships (Fig. 3a, b). Furthermore, CPH models using exposure (AUCss nominal) as a continuous variable were tested for both the BCL-2-IHC-positive subpopulation and all-comers population; the exposure parameters of the base CPH model were not a significant predictor of INV-PFS (P > 0.05). Moreover, none of the tested covariates showed significant impact on INV-PFS (Table 3).
Table 3
Base Cox proportional hazard models for investigator-assessed PFS in all-comers and BCL-2-positive population
Model
Parameter
β
SE
RSE (%)
p value
All-comers
 Base PFS-AUCss nominal model
AUCss nominal
− 0.00181 (μg × day/mL)
0.37
20,359.24
0.79
BCL-2-positive population
 Base PFS-AUCss nominal model
AUCss nominal
− 0.2108 (μg × day/mL)
0.46
21,761
0.65
AUCss venetoclax individual predicted steady-state area under the curve following the cohort-assigned dose, BCL-2 B cell lymphoma 2 protein, HR hazard ratio computed as exp(β), HR95CI 95% confidence intervals on hazard ratio, PFS progression-free survival, RSE relative standard error of β estimate (%), SE standard error of β estimate
Similar efficacy (PET-CR rate assessed at EOT) was reported for the all-comers population (69% CAVALLI vs 63% GOYA) and the BCL-2-positive subpopulation (BCL-2 by IHC: 65% CAVALLI vs 60% GOYA). The exposure–efficacy analyses using logistic regression were conducted in both BCL-2-IHC-positive patients (BCL-2 IHC: n = 101 and BCL-2 FISH: n = 40) and the all-comers (N = 199) population. No clear association was observed between venetoclax AUCss nominal and PET-CR rates in both the all-comers and BCL-2-IHC-positive subpopulations. Although this analysis did not show a clear correlation between venetoclax AUCss nominal and PET-CR rate, a wide CI was observed around the model prediction in the BCL-2-IHC-positive subpopulations (Fig. 4a–c). Given that all patients were treated at the 800 mg venetoclax dose and also the wide CI, it was not possible to confidently predict efficacy at other doses. None of the tested covariates significantly affected CR rates.

Exposure–Safety

Exposure–safety analyses were conducted across all the 10-day venetoclax dosing regimens for the R-CHOP combination in both phase 1b and phase 2 portions of the CAVALLI study, and included both patients with 1L and R/R NHL (N = 3 at 400 mg, N = 8 at 600 mg, N = 205 at 800 mg), using logistic regression analyses. Grade 3–4 AEs occurred in 86.1% of patients in the phase 2 portion of CAVALLI (179/208) versus 66.1% in GOYA (373/564), mainly driven by a higher rate of cytopenia, febrile neutropenia, and infections. No statistically significant exposure–safety trends were associated with clinical manifestations of toxicity (i.e., grade ≥ 3 febrile neutropenia, grade ≥ 3 infections, and/or SAEs), or grade ≥ 3 neutropenia or grade ≥ 3 thrombocytopenia (Fig. 5).
Of the tested covariates, high (4, 5) IPI scores were significantly correlated with increased incidence of grade ≥ 3 febrile neutropenia (p < 0.001); increased age was significantly associated with increased incidence of grade ≥ 3 infections (p < 0.01) and grade ≥ 3 thrombocytopenia (p < 0.0001); and ECOG PS scores ≥ 2 were significantly correlated with increased incidence of grade ≥ 3 thrombocytopenia (p < 0.0001; Table 4). Despite the significant relationships observed for the patient-level covariates, no substantial impact on the exposure–safety relationship for venetoclax was evident upon including these covariates into the final logistic regression models. The observed AEs when venetoclax was given in combination with R-CHOP were manageable and consistent with the known safety profile and mechanism of action of venetoclax.
Table 4
Logistic regression analysis for adverse events
Parameter
Estimate
SE
RSE (%)
95% CI
p value
Grade ≥ 3 neutropenia
 Intercept
 − 0.20
0.44
223.9
 − 1.07; 0.67
0.66
 Slope of AUCss nominal (μg × day/mL)
1.76
0.76
42.9
0.28; 3.25*
0.325
Grade ≥ 3 infection
 Intercept
 − 3.65
1.06
29.07
 − 5.73; − 1.57
0.00058
 Slope of AUCss nominal (μg × day/mL)
 − 0.20
0.72
353.88
 − 1.62; 1.21*
0.874
 Age
0.04
0.02
36.89
0.01; 0.07
0.0067
Grade ≥ 3 febrile neutropenia
 Intercept
 − 0.81
0.42
51.53
 − 1.63; 0.01
0.052
 Slope of AUCss nominal (μg × day/mL)
 − 0.46
0.66
144.05
 − 1.75; 0.84*
0.0608
 IPI (high) [4, 5]a
1.20
0.33
27.36
0.55; 1.84
0.00026
Grade ≥ 3 thrombocytopenia
 Intercept
 − 8.70
1.59
18.33
 − 11.82; − 5.57
0.000000049
 Slope of AUCss nominal (μg × day/mL)
0.59
0.75
127.03
 − 0.88; 2.05*
0.521
 Age
0.11
0.02
20.66
0.06; 0.15
0.0000013
 ECOG (2+)b
1.65
0.45
27.62
0.75; 2.54
0.00029
Serious adverse events
 Intercept
 − 3.07
0.87
28.36
 − 4.77; − 1.36
0.00042
 Slope of AUCss nominal (μg × day/mL)
 − 0.51
0.63
123.64
 − 1.75; 0.73*
0.628
 Age
0.05
0.01
25.93
0.02; 0.07
0.00011
95% CI 95% confidence interval on the parameter estimate, AUCss venetoclax individual predicted area under the curve at steady state based on the cohort-assigned dose, RSE relative standard error of the parameter estimate (%), SE standard error
*95% CI of the exposure parameter spans zero, indicating statistical nonsignificance
aIPI (high) [4, 5] represents patients with IPI scores of 4 and 5 and in the covariate analysis were compared with those with the IPI scores of 0, 1, and 2
bECOG (2+) represents patients with ECOG PS score ≥ 2 and in the covariate analysis were compared with those with ECOG PS score < 2
*95% CI of the exposure parameter spans zero, indicating statistical nonsignficance

Exposure–Dose Intensity

The 800 mg 10-day dose regimen of venetoclax was evaluated for potential impact on the R-CHOP components for the population of patients with 1L DLBCL. The percentage of patients with greater than 90% RDI for the R-CHOP components in the CAVALLI study was generally similar to the R-CHOP arm in the GOYA study across exposure cohorts [9, 10]. Importantly, the exposure–dose intensity analyses did not show any relationship between venetoclax AUCss nominal and RDIs for the individual components of venetoclax and R-CHOP in the CAVALLI study (Fig. 6).

Discussion

In the CAVALLI study, evidence of the positive benefit–risk profile for venetoclax administered at a dose of 800 mg in a 10/21-day cycle was demonstrated, particularly for BCL-2-IHC-positive patients with 1L DLBCL, when compared with historical data from the R-CHOP arm of the GOYA study [10].
The PK of venetoclax in CAVALLI were adequately described using a previously developed legacy PopPK model. However, upon evaluation of the legacy model (when applied to the CAVALLI data) using diagnostic plots, lower exposures for the patients enrolled in CAVALLI were noticeable compared with the patients with R/R NHL previously evaluated in the legacy model at similar dose levels. It is known that increased fat intake increases venetoclax exposure [18] and therefore it could be speculated that the lower exposures in the CAVALLI study may be attributed to the lower fat content in the meal-type, i.e., 23% and 3.5% of fat calories in the meal as recommended in the CAVALLI protocol, compared with the 30% of fat calories following the completion of the standard low-fat breakfast for the patients with R/R NHL included in the legacy PopPK model. Moreover, we were not able to consider these differences in fat content of 23% and 3.5% in the PopPK model as both meals were classified as low-fat meal type in the CAVALLI study. Subsequently, a study (CAVALLI) effect of 1.63 on CL/F and 2.33 on V2/F (values higher than those reported for the historical studies) was added in the updated PopPK model to account for the possible differences between the population of CAVALLI study patients relative to the population of patients from the prior analysis.
Furthermore, in contrast to the legacy model, the present PopPK model did not include the co-administration of R as a covariate on CL/F of venetoclax. In the legacy model, concomitant R administration was estimated to increase the venetoclax CL/F by 1.21-fold based on a small sample size of 50 subjects receiving venetoclax + R [13]. However, subsequent analysis in a much larger patient population (the phase 3 MURANO study, where 181 patients with R/R CLL were randomized to receive venetoclax plus R) showed a minimal 7% increase in the CL/F for venetoclax when co-administered with R, supporting removal of the R/G covariate from the model [19]. The legacy model and phase 3 MURANO study suggest that neither R, which leads to a rapid depletion of CD20+ B cells, nor indication type (whether CLL, SLL, or NHL; all of which are characterized by varying levels of circulating B cell lymphocytes) has any apparent impact on venetoclax PK [13, 19]. Taken together, this indicates minimal impact of B cell subtypes on venetoclax PK (although this was not specifically tested as a covariate in the PopPK model).
Previous studies have evaluated the venetoclax ER relationships in subjects with CLL, NHL, or multiple myeloma [2024]. ER analyses were performed to confirm the dose selection of venetoclax in combination with R-CHOP chemotherapy for future studies. PopPK-predicted AUCss nominal was used as an exposure metric for predicting clinical response, i.e., efficacy, safety, or tolerability of venetoclax. We used AUCss nominal as the exposure metric for the ER analysis rather than average plasma concentration to the time of the event (Caverage) to avoid bias due to a correlation between lower exposures from dose reductions and response, both of which are more likely the longer a patient is on study. Moreover, AUCss nominal isolated the impact of assigned target venetoclax dose and associated steady-state exposure on safety and efficacy, and was not subject to confounding by complex interactions between time and treatment-related or disease-related changes to venetoclax or R-CHOP dosing. To isolate the treatment effect of venetoclax, the ER analyses referenced clinical data from the R-CHOP arm of GOYA.
Promising efficacy for PFS and improved efficacy for PET-CR rate assessed at EOT in CAVALLI compared with matched GOYA controls were consistent with the venetoclax mechanism of action [12]. However, the graphical and CPH analysis of the patients treated at the 800 mg dose in the phase 2 portion of the CAVALLI study showed no statistically significant relationship between venetoclax exposure and PFS in both all-comers or BCL-2-IHC-positive subpopulations, which suggests no evidence of additional efficacy with increasing venetoclax exposure in this trial. This is further confirmed by the logistic regression analyses using PET-CR rates, where increased venetoclax exposure did not translate into improved efficacy in both the all-comers or BCL-2-IHC-positive subpopulations at the 800 mg (10/21-day cycle) dose.
Although no statistically significant exposure-PFS or exposure-PET-CR trends were observed in these analyses, the results are not considered sufficiently strong enough to confidently predict efficacy at alternative venetoclax doses, as the majority of patients in the exposure-efficacy analyses were studied at a single venetoclax dose level of 800 mg and a wide CI was observed around exposure-efficacy predictions for the BCL-2-IHC-positive subpopulations.
Furthermore, compared with CLL, where the labeled venetoclax dose is 400 mg, a venetoclax dose resulting in a higher steady-state exposure in NHL may be required because of a potential decreased sensitivity to venetoclax of lymphocytes located in the lymph node compared with the blood as reflected in the lower half maximum effective concentration (EC50) values of venetoclax for circulating lymphocyte counts (0.00863 μg/mL) compared with tumor size (0.146 μg/mL) [21]. This increased sensitivity of circulating CLL cells than CLL cells within a tumor mass may be attributed to the reduced blood flow and subsequent delivery of systemic venetoclax within a tumor, and due to survival signals cells receive when in direct contact with other cells within the tumor microenvironment. It is therefore not surprising that DLBCL sensitivity is lower compared with CLL, as the bulk of the disease is non-circulating (in lymph nodes and other tissues in the body).
As long as delivery of the potentially curative standard of care R-CHOP therapy is not compromised, the addition of venetoclax is considered to pose a low risk for negatively impacting outcome in patients with 1L DLBCL. In the current study, patients with higher venetoclax exposures showed similar dose intensities for the individual components of venetoclax and R-CHOP combination therapy compared with patients with lower venetoclax exposures, suggesting that patients with higher initial venetoclax exposure did not have higher probability to maintain or reduce their venetoclax or R-CHOP backbone dose. This finding supports the proposal that addition of venetoclax to R-CHOP did not compromise delivery of R-CHOP therapy.
Logistic regression analysis of exposure-safety data from the patients in the CAVALLI study showed no statistically significant associations between venetoclax exposure and the probability of clinical manifestations of toxicity (i.e., grade ≥ 3 febrile neutropenia, grade ≥ 3 infections, and/or SAEs), or grade ≥ 3 neutropenia or grade ≥ 3 thrombocytopenia. The prognostic impact of the tested covariates has been previously reported. [2528]
Although a higher incidence of AEs was generally seen for patients with 1L DLBCL in the CAVALLI study compared with the GOYA historical control (R-CHOP), the reported AEs were manageable and predictable on the basis of the mechanism of action of venetoclax, or consistent with the known safety profile of R-CHOP [29, 30]. The interpretation of these events could be confounded by the additional mandatory mid-cycle laboratory tests and clinical evaluations required for the CAVALLI study but not required in the GOYA trial. Taken together, there was limited evidence to suggest that administering 800 mg of venetoclax would cause additional toxicity compared with the lower venetoclax doses administered to patients in the CAVALLI study; however, it is possible that higher doses could lead to increased safety events.

Conclusions

The early promising efficacy data from the CAVALLI study of venetoclax treatment in combination with R-CHOP, together with the acceptable early safety and tolerability in patients treated with this combination, support the selection of venetoclax at 800 mg in cycle 1 on days 4–10 and cycles 2–8 on days 1–10 in combination with R-CHOP for 21-day cycles in the BCL-2-IHC-positive 1L DLBCL population in future studies.

Acknowledgements

Venetoclax is being developed in collaboration between Genentech, Inc. and AbbVie Inc. The authors thank the patients who participated in the CAVALLI and GOYA studies and their families, as well as the study co-investigators, research nurses, and coordinators at each of the clinical sites. The authors also thank Sandhya Girish for her input into data analyses and manuscript development.

Funding

Genentech, Inc. and AbbVie Inc. provided financial support for the CAVALLI study and participated in the design, study conduct, analysis and interpretation of data. The GOYA study was supported by research funding from F. Hoffmann-La Roche Ltd, with scientific support from the Fondazione Italiana Linfomi. The analyses presented here were supported by Genentech, Inc. The journal’s Rapid Service and Open Access fees were funded by F. Hoffmann-La Roche Ltd.

Medical Writing and Editorial Assistance

Medical writing and editorial support were provided by Anshin Biosolutions and funded by Genentech, Inc. Under the direction of the authors, further editorial assistance was provided by Rachel Dobb PhD of Ashfield MedComms, an Ashfield Health company, and funded by F. Hoffmann-La Roche Ltd.

Authorship

All named authors meet the International Committee of Medical Journal Editors (ICMJE) criteria for authorship for this article, take responsibility for the integrity of the work as a whole, and have given their approval for this version to be published.

Author Contributions

All persons who meet authorship criteria are listed as authors, and all authors certify that they have participated sufficiently in the work to take public responsibility for the content, including participation in the concept, design, analysis, writing, or revision of the manuscript.

Disclosures

Divya Samineni, Weize Huang, Hao Ding, Rong Zhang, Chunze Li, Sandhya Girish, and Dale Miles are employees of Genentech, Inc., and may hold stock or stock options with the Roche Group. Richa Rajwanshi was previously an employee of Genentech Inc and may have held stock or stock options with the Roche Group; new affiliation is Amgen, Thousand Oaks, CA, USA. Kathryn Humphrey and Alexandra Bazeos are employees of Roche Products Ltd and may hold stock or stock options with the Roche Group. Arijit Sinha was previously an employee of Roche Products Ltd and may have held stock or stock options with the Roche group; new affiliation is AstraZeneca, Melbourn Science Park, Melbourn, Herts, UK. Ahmed Hamed Salem is an employee of AbbVie Inc. and may hold stock or stock options with AbbVie. Leonid Gibiansky is an employee of QuantPharm LLC.

Compliance with Ethics Guidelines

All procedures performed in studies involving human participants were in accordance with the ethical standards of the institutional review board and with the 1964 Helsinki declaration and its later amendments. This article does not contain any studies with animals performed by any of the authors.
Informed consent was obtained from all individual participants included in the study.
This article does not contain any identifying information about participants and therefore consent was not sought from participants for publication.

Data Availability

Qualified researchers may request access to individual patient level data through the clinical study data request platform (https://​vivli.​org/​). Further details on Roche’s criteria for eligible studies are available here (https://​vivli.​org/​members/​ourmembers/​). For further details on Roche's Global Policy on the Sharing of Clinical Information and how to request access to related clinical study documents, visit https://​www.​roche.​com/​research_​and_​development/​who_​we_​are_​how_​we_​work/​clinical_​trials/​our_​commitment_​to_​data_​sharing.​htm.
Open AccessThis article is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License, which permits any non-commercial use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by-nc/​4.​0/​.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

e.Med Allgemeinmedizin

Kombi-Abonnement

Mit e.Med Allgemeinmedizin erhalten Sie Zugang zu allen CME-Fortbildungen und Premium-Inhalten der allgemeinmedizinischen Zeitschriften, inklusive einer gedruckten Allgemeinmedizin-Zeitschrift Ihrer Wahl.

Anhänge

Supplementary Information

Below is the link to the electronic supplementary material.
Literatur
11.
Zurück zum Zitat Franck Morschhauser F, Feugier P, Flinn IW, et al. A phase 2 study of venetoclax plus R-CHOP as first-line treatment for patients with diffuse large B-cell lymphoma. Blood. 2021;137(5):600–9.CrossRef Franck Morschhauser F, Feugier P, Flinn IW, et al. A phase 2 study of venetoclax plus R-CHOP as first-line treatment for patients with diffuse large B-cell lymphoma. Blood. 2021;137(5):600–9.CrossRef
12.
Zurück zum Zitat Zelenetz AD, Feugier P, Flinn, I et al. Improved outcomes with venetoclax plus rituximab, cyclophosphamide, doxorubicin, vincristine and prednisolone (R-CHOP) in previously untreated patients with BCL-2-positive diffuse large B-cell lymphoma (DLBCL): updated desults from the phase II CAVALLI Study. (Abstract). 61st ASH Annual meeting; December 7−10. Orlando, Florida. 2019 Zelenetz AD, Feugier P, Flinn, I et al. Improved outcomes with venetoclax plus rituximab, cyclophosphamide, doxorubicin, vincristine and prednisolone (R-CHOP) in previously untreated patients with BCL-2-positive diffuse large B-cell lymphoma (DLBCL): updated desults from the phase II CAVALLI Study. (Abstract). 61st ASH Annual meeting; December 7−10. Orlando, Florida. 2019
16.
Zurück zum Zitat Yano Y, Beal SL, Sheiner LB. Evaluating pharmacokinetic/pharmacodynamic models using the posterior predictive check. J Pharmacokinet Pharmacodyn. 2001;28(2):171–92.CrossRef Yano Y, Beal SL, Sheiner LB. Evaluating pharmacokinetic/pharmacodynamic models using the posterior predictive check. J Pharmacokinet Pharmacodyn. 2001;28(2):171–92.CrossRef
17.
Zurück zum Zitat Beal S, Sheiner LB, Boeckmann A, Bauer RJ. NONMEM User’s Guides (1989–2011). Ellicott City, MD, USA: Icon Development Solutions; 2011 Beal S, Sheiner LB, Boeckmann A, Bauer RJ. NONMEM User’s Guides (1989–2011). Ellicott City, MD, USA: Icon Development Solutions; 2011
22.
Zurück zum Zitat Freise KJ, Jones AK, Menon RM, et al. Relationship between venetoclax exposure, rituximab coadministration, and progression-free survival in patients with relapsed or refractory chronic lymphocytic leukemia: demonstration of synergy. Hematol Oncol. 2017;35(4):679–84. https://doi.org/10.1002/hon.2373.CrossRefPubMed Freise KJ, Jones AK, Menon RM, et al. Relationship between venetoclax exposure, rituximab coadministration, and progression-free survival in patients with relapsed or refractory chronic lymphocytic leukemia: demonstration of synergy. Hematol Oncol. 2017;35(4):679–84. https://​doi.​org/​10.​1002/​hon.​2373.CrossRefPubMed
23.
Zurück zum Zitat Freise KJ, Jones AK, Verdugo ME, Menon RM, Maciag PC, Salem AH. Moving beyond maximum tolerated dose for targeted oncology drugs: use of clinical utility index to optimize venetoclax dosage in multiple myeloma patients. Clin Pharmacol Ther. 2017;102(6):970–6. https://doi.org/10.1002/cpt.712.CrossRefPubMed Freise KJ, Jones AK, Verdugo ME, Menon RM, Maciag PC, Salem AH. Moving beyond maximum tolerated dose for targeted oncology drugs: use of clinical utility index to optimize venetoclax dosage in multiple myeloma patients. Clin Pharmacol Ther. 2017;102(6):970–6. https://​doi.​org/​10.​1002/​cpt.​712.CrossRefPubMed
Metadaten
Titel
Population Pharmacokinetics and Exposure–Response Analyses for Venetoclax in Combination with R-CHOP in Relapsed/Refractory and Previously Untreated Patients with Diffuse Large B Cell Lymphoma
verfasst von
Divya Samineni
Weize Huang
Leonid Gibiansky
Hao Ding
Rong Zhang
Chunze Li
Arijit Sinha
Richa Rajwanshi
Kathryn Humphrey
Alexandra Bazeos
Ahmed Hamed Salem
Dale Miles
Publikationsdatum
25.11.2021
Verlag
Springer Healthcare
Erschienen in
Advances in Therapy / Ausgabe 1/2022
Print ISSN: 0741-238X
Elektronische ISSN: 1865-8652
DOI
https://doi.org/10.1007/s12325-021-01919-z

Weitere Artikel der Ausgabe 1/2022

Advances in Therapy 1/2022 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Battle of Experts: Sport vs. Spritze bei Adipositas und Typ-2-Diabetes

11.05.2024 DDG-Jahrestagung 2024 Kongressbericht

Im Battle of Experts traten zwei Experten auf dem Diabeteskongress gegeneinander an: Die eine vertrat die Auffassung „Sport statt Spritze“ bei Adipositas und Typ-2-Diabetes, der andere forderte „Spritze statt Sport!“ Am Ende waren sie sich aber einig: Die Kombination aus beidem erzielt die besten Ergebnisse.

Vorsicht, erhöhte Blutungsgefahr nach PCI!

10.05.2024 Koronare Herzerkrankung Nachrichten

Nach PCI besteht ein erhöhtes Blutungsrisiko, wenn die Behandelten eine verminderte linksventrikuläre Ejektionsfraktion aufweisen. Das Risiko ist umso höher, je stärker die Pumpfunktion eingeschränkt ist.

Triglyzeridsenker schützt nicht nur Hochrisikopatienten

10.05.2024 Hypercholesterinämie Nachrichten

Patienten mit Arteriosklerose-bedingten kardiovaskulären Erkrankungen, die trotz Statineinnahme zu hohe Triglyzeridspiegel haben, profitieren von einer Behandlung mit Icosapent-Ethyl, und zwar unabhängig vom individuellen Risikoprofil.

Gibt es eine Wende bei den bioresorbierbaren Gefäßstützen?

In den USA ist erstmals eine bioresorbierbare Gefäßstütze – auch Scaffold genannt – zur Rekanalisation infrapoplitealer Arterien bei schwerer PAVK zugelassen worden. Das markiert einen Wendepunkt in der Geschichte dieser speziellen Gefäßstützen.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.