throbber
Leukemia (2020) 34:2903–2913
`https://doi.org/10.1038/s41375-020-0996-5
`
`ARTICLE
`
`Acute myeloid leukemia
`Safety and efficacy of BAY1436032 in IDH1-mutant AML:
`phase I study results
`
`4 ● Courtney DiNardo 5 ●
`1 ● Neil Palmisiano 2 ● Ioannis Mantzaris3 ● Alice Mims
`Michael Heuser
`Lewis R. Silverman6 ● Eunice S. Wang 7 ● Walter Fiedler8 ● Claudia Baldus9 ● Sebastian Schwind 10 ●
`Timothy Pardee 11 ● Alexander E. Perl12 ● Charles Cai13 ● Stefan Kaulfuss14 ● Eleni Lagkadinou14 ●
`Christine Rentzsch14 ● Markus Wagner14 ● Gary Wilkinson14 ● Bingyan Wu13 ● Michael Jeffers13 ● Isabelle Genvresse14 ●
`Alwin Krämer15
`
`Received: 23 June 2020 / Revised: 15 July 2020 / Accepted: 21 July 2020 / Published online: 30 July 2020
`© The Author(s) 2020. This article is published with open access
`
`Abstract
`The mutant IDH1 (mIDH1) inhibitor BAY1436032 demonstrated robust activity in preclinical AML models, supporting
`clinical evaluation. In the current dose-escalation study, BAY1436032 was orally administered to 27 mIDH1 AML subjects
`across 4 doses ranging from 300 to 1500 mg twice-daily. BAY1436032 exhibited a relatively short half-life and apparent
`non-linear pharmacokinetics after continuous dosing. Most subjects experienced only partial target inhibition as indicated by
`plasma R-2HG levels. BAY1436032 was safe and a maximum tolerated dose was not identified. The median treatment
`duration for all subjects was 3.0 months (0.49–8.5). The overall response rate was 15% (4/27; 1 CRp, 1 PR, 2 MLFS), with
`responding subjects experiencing a median treatment duration of 6.0 months (3.9–8.5) and robust R-2HG decreases. Thirty
`percent (8/27) achieved SD, with a median treatment duration of 5.5 months (3.1–7.0). Degree of R-2HG inhibition and
`clinical benefit did not correlate with dose. Although BAY1436032 was safe and modestly effective as monotherapy, the low
`overall response rate and incomplete target inhibition achieved at even the highest dose tested do not support further clinical
`development of this investigational agent in AML.
`
`1234567890();,:
`1234567890();,:
`
`Supplementary information The online version of this article (https://
`doi.org/10.1038/s41375-020-0996-5) contains supplementary
`material, which is available to authorized users.
`
`* Michael Heuser
`heuser.michael@mh-hannover.de
`
`1 Department of Hematology, Hemostasis, Oncology, and Stem Cell
`Transplantation, Hannover Medical School, Hannover, Germany
`2 Department of Medical Oncology, Sidney Kimmel Cancer Center,
`Thomas Jefferson University, Philadelphia, PA, USA
`3 Department of Medical Oncology, Montefiore Medical Center,
`Albert Einstein College of Medicine, Bronx, NY, USA
`4 Division of Hematology, Department of Medicine, The Ohio State
`University Comprehensive Cancer Center, Columbus, OH, USA
`5 Department of Leukemia, The University of Texas M.D. Anderson
`Cancer Center, Houston, TX, USA
`
`6
`
`Tisch Cancer Institute, Division of Hematology/Oncology,
`Icahn School of Medicine at Mount Sinai, New York, NY, USA
`7 Department of Medicine, Roswell Park Comprehensive Cancer
`Center, Buffalo, NY, USA
`8 Department of Hematology and Oncology, University Hospital
`Hamburg-Eppendorf, Hamburg, Germany
`
`9 Division of Hematology and Oncology, Charité University
`Hospital Berlin, Berlin, Germany
`10 Division of Hematology and Oncology, University Hospital
`Leipzig, Leipzig, Germany
`11 Section on Hematology and Oncology, Comprehensive Cancer
`Center of Wake Forest Baptist Health, Winston-Salem, NC, USA
`12 Division of Hematology-Oncology, Perelman School of Medicine,
`Abramson Cancer Center of the University of Pennsylvania,
`Philadelphia, PA, USA
`13 Pharmaceuticals Division, Bayer HealthCare Pharmaceuticals,
`Inc., Whippany, NJ, USA
`14 Pharmaceuticals Division, Bayer AG, Berlin, Germany
`15 Clinical Cooperation Unit Molecular Hematology/Oncology,
`German Cancer Research Center (DKFZ) and Department of
`Internal Medicine V, University of Heidelberg,
`Heidelberg, Germany
`
`Rigel Exhibit 1041
`Page 1 of 11
`
`

`

`2904
`
`Introduction
`
`Somatic hotspot mutations in isocitrate dehydrogenase 1
`(IDH1) have been identified in a variety of cancers, with a
`frequency of ~7% in acute myeloid leukemia (AML) [1–6].
`Tumor-associated IDH1 mutations (mIDH1) change the
`conserved arginine at codon 132 in the enzymatic active site
`to a variety of alternative amino acids (R132X), and in
`doing so confer a neomorphic activity to this enzyme.
`Whereas wild-type IDH1 (wtIDH1) catalyzes the conver-
`sion of isocitrate to α-ketoglutarate (α-KG), mIDH1 con-
`verts α-KG to R-2-hydroxyglutarate (R-2HG). Subjects
`with mIDH1 AML show elevated R-2HG levels, which
`inhibits α-KG-dependent enzymes, thereby leading to epi-
`genetic alterations and ultimately impaired hematopoietic
`differentiation [7–14].
`BAY1436032 is an oral small-molecule inhibitor of
`mIDH1 that is active in preclinical models of mIDH1 cancer
`[15–17]. Most mIDH1 inhibitors, including BAY1436032,
`reportedly interact with an allosteric site on the mutant
`enzyme, although an inhibitor which interacts directly with
`the active site was recently described [18]. Preclinical
`focusing on mIDH1 AML found that
`experiments
`BAY1436032 inhibits R-2HG production and colony
`growth in vitro, while promoting leukemic blast clearance,
`myeloid differentiation, and survival in animal models [16].
`Supported by these encouraging preclinical findings,
`BAY1436032 was evaluated in a phase I clinical study in
`subjects with mIDH1 AML (NCT03127735), the results of
`which are presented herein. Objectives of the study include
`determination of the maximum tolerated dose (MTD) and
`the recommended phase II dose (RP2D), and evaluation of
`the safety, tolerability, pharmacokinetics (PK), pharmaco-
`dynamics and clinical activity of BAY1436032.
`
`Materials and methods
`
`Study design
`
`This study was an open-label, nonrandomized, multicenter
`phase I trial. Subjects were screened at 13 hospital sites in 2
`countries (USA and Germany). The study was to consist of
`dose-escalation followed by dose-expansion. The MTD
`identified in dose-escalation was to be used in the dose-
`expansion. If MTD was not reached in dose-escalation, a
`dose for the expansion would be selected based on available
`PK,
`pharmacodynamic,
`safety,
`and
`efficacy
`data.
`BAY1436032 was administered twice-daily (BID) in con-
`tinuous 28-day cycles. In dose-escalation, up to 9 evaluable
`subjects could be enrolled per cohort, with a minimum of
`3 dose-limiting toxicity (DLT)-free evaluable subjects
`required prior
`to escalating to the next highest dose.
`
`M. Heuser et al.
`
`Bayesian dose-DLT modeling was performed to help guide
`dosing decisions [19]. MTD was defined as the highest dose
`of BAY1436032 that could be given such that ≤25% of
`subjects were predicted to experience a DLT.
`DLTs were differentially defined for nonhematopoietic
`versus hematopoietic toxicities. Nonhematopoietic toxi-
`cities of ≥grade 3 occurring during the first cycle of treat-
`ment were to be considered DLTs with the following
`exceptions: (1) Alopecia and nausea controlled by medical
`management; (2) Tumor lysis syndrome if successfully
`managed clinically and resolved within 7 days without any
`end-organ damage; (3) Differentiation syndrome (DS) if
`successfully managed clinically and resolved within 7 days
`without any end-organ damage; (4) Asymptomatic ≥grade 3
`electrolyte abnormalities not considered clinically sig-
`nificant by the investigator. Missing >20% of doses of study
`drug due to any drug-related toxicity, or delay in the start of
`cycle 2 by more than 14 days due to any drug-related
`toxicity, were also considered DLTs. For certain toxicities
`such as laboratory assessments without a clear clinical
`correlate, a discussion between the investigator and the
`sponsor determined whether the adverse event (AE) should
`be assessed as a DLT.
`thrombocytopenia of
`toxicities,
`For hematological
`≥grade 3 with clinically significant bleeding, or grade 4
`neutropenia persisting 42 days after the start of treatment in
`the absence of active AML, were considered DLTs.
`DLTs identified during the first cycle of treatment were
`used to guide dose-escalation decisions and to determine the
`MTD, and if safety issues appeared in subsequent cycles
`they were also to be considered. Hydroxyurea was per-
`mitted during the first cycle if white blood cell (WBC)
`20 × 109/L and was
`exceeded
`also
`permitted
`for
`treatment of DS.
`The study protocol was approved by the institutional
`review board of participating institutions and complied with
`the Declaration of Helsinki, current Good Clinical Practice
`guidelines, and local laws and regulations. Written informed
`consent was provided by all participants prior
`to the
`initiation of any study-specific procedure. Data were entered
`into clinical research forms by the study investigators and
`their staff. The study was sponsored by Bayer AG.
`
`Subjects
`
`Male and female subjects of ≥18 years of age with an
`Eastern Cooperative Oncology Group (ECOG) performance
`status of ≤2 and advanced AML were eligible. Information
`regarding ELN 2010 risk classification was collected during
`screening [20]. Subjects were required to harbor a missense
`mutation in IDH1-R132X based on local testing reported by
`study investigators, with sponsor review of test results prior
`to enrollment. IDH1 mutational status from a bone marrow
`
`Rigel Exhibit 1041
`Page 2 of 11
`
`

`

`Safety and efficacy of BAY1436032 in IDH1-mutant AML: phase I study results
`
`2905
`
`sample collected during screening was retrospectively
`evaluated at Foundation Medicine. Subjects were to be
`relapsed or refractory to at least 1 previous line of therapy,
`or intolerable to or unable to receive established therapies,
`and could have received any number and type of prior
`therapies prior
`to enrollment, except
`those targeting
`mIDH1. A cohort sample size of 3 to 9 DLT-evaluable
`subjects in dose-escalation was chosen based on experience
`and simulation results from adaptive Bayesian dose-DLT
`model. This number of subjects is anticipated to provide
`sufficient safety information to help guide dose escalation
`decisions in a reasonable time frame without exposing an
`excess number of subjects to potentially toxic or inactive
`doses of study drug. Dose-expansion was not conducted in
`this study.
`
`Study assessments
`
`The primary objectives of the study were to determine the
`safety,
`tolerability, MTD,
`and/or RP2D dose
`of
`BAY1436032 administered in a twice-daily dosing sche-
`dule in subjects with mIDH1 advanced AML. Secondary
`objectives were to evaluate PK and to assess pharmacody-
`namic effects and evidence of clinical efficacy.
`
`Safety
`
`Safety and tolerability were evaluated by analysis of
`adverse events, physical examinations, vital signs, ECOG
`performance status, and various laboratory assessments.
`For safety monitoring, subjects were scheduled for clinic
`visits every week for the initial three cycles of treatment,
`after which time visits could be reduced to every-other-
`week with investigator and sponsor agreement. Cardiac
`function was assessed with triplicate 12-lead electro-
`cardiograms (ECG) at screening, C1-D1, C1-D2, C1-
`D15, D1 of every subsequent cycle, and at treatment end.
`Severity of adverse events and toxicities were graded by
`investigators according to the National Cancer Institute
`Common Terminology Criteria for Adverse Events (NCI-
`CTCAE) version 4.03. AEs are presented by the Medical
`Dictionary for Regulatory Activities (MedDRA) v21.1.
`
`Efficacy
`
`Disease assessments and response evaluations from bone
`marrow aspirate or biopsy were scheduled at screening, C2-
`D1, C3-D1, D1 of every second cycle thereafter, and at
`treatment end (if not done on D1 of the last cycle). Per-
`ipheral blood was analyzed at each of these time points and
`at additional
`times between bone marrow assessments.
`Clinical efficacy was assessed by investigators using the
`modified 2003 International Working Group response
`
`criteria for AML [21] with some changes based on 2017
`European Leukemia Net recommendations [22]. Response
`categories included: complete remission (CR), morphologic
`CR with incomplete hematological recovery (CRh), mor-
`phologic CR with incomplete platelet recovery (CRp),
`morphologic leukemia-free state (MLFS), partial remission
`(PR), stable disease (SD), and progressive disease (PD).
`Overall response rate included CR, CRh, CRp, MLFS, and
`PR. Following study completion,
`investigators provided
`survival information.
`
`Pharmacokinetics
`
`Plasma samples were collected at various times on C1-D1,
`C1-D2, C1-D8, and C1-D15 and stored frozen for PK
`assessments. Collection times on these days are indicated on
`Supplementary Fig. S1 (C1-D8 PK results are not shown).
`The evening dose of BAY1436032 was withheld on C1-D1
`to facilitate assessment of the 24-h single-dose time point on
`C1-D2. Quantitative analysis of BAY1436032 (free acid) in
`plasma was performed as described in Supplementary
`Methods.
`
`Pharmacodynamics
`
`For quantification of R-2HG, plasma samples were col-
`lected at
`the following time points and stored frozen:
`screening, C1-D1 (pre-dose and post-dose), C1-D8 (pre-
`dose), C1-D15 (pre-dose and post-dose), C1-D22 (pre-
`dose), pre-dose on D1 and D15 of each cycle thereafter, and
`at
`treatment end. Plasma R-2HG concentrations were
`measured by Eurofins as described in Supplementary
`Methods.
`
`Retrospective mutational analysis
`
`Mutational analysis was performed on bone marrow aspi-
`rates or biopsies collected during screening and on a subset
`of samples collected during BAY1436032 treatment. Test-
`ing was performed by Foundation Medicine using the
`FoundationOne Heme panel which detects alterations in
`>400 tumor-associated genes via next-generation sequen-
`cing. Information provided by Foundation Medicine inclu-
`ded the allelic frequency and the likely pathogenic nature of
`each alteration identified.
`
`Results
`
`Subjects
`
`Thirty-three mIDH1 AML subjects signed informed con-
`sent, 27 received BAY1436032 treatment and 6 failed
`
`Rigel Exhibit 1041
`Page 3 of 11
`
`

`

`2906
`
`M. Heuser et al.
`
`Fig. 1 Subject disposition. The chart shows an overview of subjects who signed consent to participate in the study, their allocation into dose-
`escalation treatment cohorts, and reasons for their discontinuation from the study.
`
`screening for various reasons (e.g., presence of an uncon-
`trolled infection). The first subject started treatment on June
`28, 2017 and the last study visit was on December 5, 2018.
`All subjects were treated within dose-escalation, and a
`planned dose-expansion was not pursued. BAY1436032
`tablets were orally administered BID, with the evening dose
`withheld on the first day of treatment to facilitate PK ana-
`lysis. Administration was continuous and each treatment
`cycle was 28 days. Subjects were treated across 4 dosing
`cohorts: cohort 1 (300 mg BID; n = 7); cohort 2 (600 mg
`BID; n = 4); cohort 3 (1200 mg BID; n = 7) and cohort 4
`(1500 mg BID; n = 9) (Fig. 1). The dosing schedule and
`starting dose were selected based on preclinical PK mod-
`eling and safety data, and on the results of the ongoing first-
`in-human phase I trial of BAY1436032 in subjects with
`mIDH1 solid tumors
`(NCT02746081;
`[23]). Baseline
`demographics and disease characteristics are provided in
`Table 1. Subjects had received a median of 2 (0–8) prior
`systemic therapies for AML and 4 had received no prior
`systemic therapies. The prevalence of individual IDH1-
`R132X mutations across the 27 treated subjects based on
`investigator-reported information was as follows: R132C
`(n = 15), R132H (n = 5), R132G (n = 3), R132L and
`
`R132S (n = 2 each). Consistent with previous reports
`[4, 24], R132C and R132H were the most prevalent IDH1
`mutations identified.
`
`Pharmacokinetics and Pharmacodynamics
`
`PK analysis was performed on C1-D1 after single-dose oral
`administration and on C1-D15 following continuous BID
`dosing. Following single oral administration, BAY1436032
`plasma concentrations were detectable 30 min after admin-
`istration. Maximum plasma concentrations were observed
`~3 to 4 h after single-dose and continuous BID adminis-
`tration (Supplementary Fig. S1). In the evaluated dose
`range, BAY1436032 exposure after single-dose adminis-
`tration generally increased in a dose-proportional manner
`(1.6–2.0-fold); however, dose-proportionality was not
`apparent after continuous BID administration. Minimal
`accumulation was evident at C1-D15 and inter-subject
`variability was high in all cohorts for the main PK para-
`meters evaluated (Supplementary Fig. S1 and Table S1).
`To evaluate potential effects of the study drug on target
`inhibition, R-2HG levels were measured in plasma samples
`obtained at baseline and at various time points during
`
`Rigel Exhibit 1041
`Page 4 of 11
`
`

`

`Safety and efficacy of BAY1436032 in IDH1-mutant AML: phase I study results
`
`2907
`
`Table 1 Baseline demographic and disease characteristicsa.
`
`Cohort 1:
`300 mg
`BID
`n = 7
`
`Cohort 2:
`600 mg
`BID
`n = 4
`
`Cohort 3:
`1200 mg
`BID
`n = 7
`
`Cohort 4:
`1500 mg
`BID
`n = 9
`
`Total
`
`n = 27
`
`67 (37–86)
`
`72 (51–76)
`
`70 (42–83)
`
`68 (27–79)
`
`69 (27–86)
`
`4 (57)
`3 (43)
`
`5 (71)
`2 (29)
`
`1 (14)
`4 (57)
`2 (29)
`9 (1–32)
`
`0
`1 (14)
`4 (57)
`2 (29)
`
`3 (43)
`3 (43)
`1 (14)
`0
`0
`1 (0–4)
`
`2 (50)
`2 (50)
`
`3 (75)
`1 (25)
`
`1 (25)
`2 (50)
`1 (25)
`10 (2–19)
`
`0
`1 (25)
`2 (50)
`1 (25)
`
`2 (50)
`0
`2 (50)
`0
`0
`1 (1–3)
`
`3 (43)
`4 (57)
`
`4 (51)
`3 (43)
`
`1 (14)
`6 (86)
`0
`9 (1–25)
`
`1 (14)
`3 (43)
`2 (29)
`1 (14)
`
`5 (71)
`0
`0
`2 (29)
`0
`2 (0–4)
`
`3 (33)
`6 (67)
`
`7 (78)
`2 (22)
`
`1 (11)
`7 (78)
`1 (11)
`15 (5–44)
`
`2 (22)
`2 (22)
`5 (56)
`0
`
`5 (56)
`2 (22)
`0
`0
`2 (22)
`3 (1–8)
`
`12 (44)
`15 (56)
`
`19 (70)
`8 (30)
`
`4 (15)
`19 (70)
`4 (15)
`11 (1–44)
`
`3 (11)
`7 (26)
`13 (48)
`4 (15)
`
`15 (56)
`5 (19)
`3 (11)
`2 (7)
`2 (7)
`2 (0–8)
`
`5 (71)
`
`4 (100)
`
`5 (71)
`
`9 (100)
`
`23 (85)
`
`6 (86)
`1 (14)
`
`4 (100)
`0
`
`7 (100)
`0
`
`7 (78)
`2 (22)
`
`24 (89)
`3 (11)
`
`Age [median (range), in years]
`Sex, n (%)
`Male
`Female
`AML classification, n (%)
`De novo AML
`Secondary AML
`ECOG performance status, n (%)
`0
`1
`2
`Time from initial diagnosis to 1st dose of
`study drug [median (range), in months]b
`ELN risk classification, n (%)c
`Favorable
`Intermediate
`Adverse
`Missing
`mIDH1 identified, n (%)d
`R132C
`R132H
`R132G
`R132L
`R132S
`Number of prior systemic antileukemic
`lines of therapies for AML, [median
`(range)]
`Subjects having received at least 1 prior
`systemic antileukemic therapy for AML, n
`(%)
`Prior allogeneic transplantation
`No
`Yes
`
`BID twice-daily, ECOG Eastern Cooperative Oncology Group, ELN European LeukemiaNet, n number of subjects.
`aPercentages are calculated including missing values.
`bFor the calculation of time from initial diagnosis, only subjects with complete date information (year, month, day) are included.
`cELN classification as per 2010 recommendations [20].
`dInvestigator-reported mIDH1 results used for subject enrollment are shown. Retrospective evaluation of baseline leukemic samples at a central
`laboratory via next-generation sequencing confirmed investigator-reported results in all evaluable cases.
`
`treatment. Baseline R-2HG levels were highly variable,
`with a median concentration of 1755 ng/mL (78–14,749)
`(Supplementary Table S2). Of the 27 subjects treated, 26
`had baseline R-2HG levels above those seen in wtIDH1
`cancers (61 ng/mL) and above the 97th percentile upper
`reference limit found in healthy individuals (138 ng/mL)
`[25, 26].
`
`All subjects achieved a lowering of baseline R-2HG
`levels during BAY1436032 treatment, with a median
`maximal decrease across all subjects of 66% (16–99)
`(Supplementary Table S2). However, only 5/26 subjects
`with an elevated baseline R-2HG level experienced a
`reduction to a normal level of ≤138 ng/mL. Maximal R-
`2HG decreases did not show a clear relationship with
`
`Rigel Exhibit 1041
`Page 5 of 11
`
`

`

`2908
`
`M. Heuser et al.
`
`Table 2 BAY1436032-related treatment-emergent adverse events occurring in ≥5% of treated subjects.
`
`TEAE
`
`Sinus tachycardia
`Diarrhea
`Nausea
`
`Vomiting
`Fatigue
`
`Decreased appetite
`
`Hypomagnesaemia
`Differentiation syndromea
`
`Dyspnea
`
`CTCAE
`grade
`
`Grade 1
`Grade 2
`Grade 1
`Grade 2
`Grade 1
`Grade 1
`Grade 2
`Grade 3
`Grade 1
`Grade 2
`Grade 1
`Grade 2
`Grade 3
`Grade 1
`Grade 2
`
`Cohort 1:
`300 mg
`BID
`n = 7
`
`Cohort 2:
`600 mg
`BID
`n = 4
`
`Cohort 3:
`1200 mg
`BID
`n = 7
`
`Cohort 4:
`1500 mg
`BID
`n = 9
`
`Total
`
`n = 27 (%)
`
`1
`0
`1
`1
`2
`0
`1
`0
`1
`1
`1
`0
`0
`1
`1
`
`0
`0
`1
`0
`1
`1
`0
`0
`0
`0
`0
`0
`0
`0
`0
`
`0
`2
`1
`0
`0
`0
`1
`0
`0
`0
`0
`0
`1
`1
`0
`
`1
`0
`1
`0
`1
`0
`0
`1
`0
`0
`1
`1
`0
`0
`0
`
`2 (7)
`2 (7)
`4 (15)
`1 (4)
`4 (15)
`1 (4)
`2 (7)
`1 (4)
`1 (4)
`1 (4)
`2 (7)
`1 (4)
`1 (4)
`2 (7)
`1 (4)
`
`BID twice-daily, CTCAE common terminology criteria for adverse events, n number of subjects, TEAE treatment-emergent adverse events (as per
`MedDRA PT).
`aSymptoms were not reported, only the term differentiation syndrome (DS). Three additional subjects presented with symptoms associated with DS
`and are not listed in the table since the individual symptoms occurred in <5% of treated subjects.
`
`BAY1436032 dose, plasma exposure, or the specific IDH1-
`R132X mutation detected at baseline.
`
`Safety
`
`Seventeen of 27 treated subjects experienced at least 1
`treatment-emergent adverse event (TEAE) of any grade
`related to BAY1436032, with events occurring in ≥5% of
`treated subjects as listed in Table 2. Seven of 27 subjects
`least 1 TEAE of ≥grade 3 related to
`experienced at
`BAY1436032, with each of the following events occurring
`in a single subject: amylase increase, DS, fatigue, febrile
`neutropenia, hyponatremia,
`lung infiltration, peripheral
`edema, pneumonitis, and decreased WBC count (grade 3);
`anemia, ileus, decreased neutrophil count, decreased platelet
`count, and sepsis (grade 4). Twenty-four subjects experi-
`enced at least 1 TEAE ≥ grade 3 irrespective of relationship
`to study drug (Supplementary Table S3).
`DS was considered an AE of special interest (AESI).
`Five of 27 (19%) treated subjects (2 each in cohorts 1 and 4,
`1 in cohort 3) presented with DS, or symptoms associated
`with DS, which occurred between 8 and 76 days after the
`start of treatment. For treatment of DS symptoms, 4 subjects
`received steroids, 2 received hydroxyurea, and 1 had an
`interruption of study drug. All cases eventually resolved and
`2 of the subjects experienced a clinical response (1 CRp,
`1 MLFS).
`
`No MTD was identified. A single DLT was reported:
`grade 4 ileus in subject 21 (cohort 4) who had a prior his-
`tory of
`intestinal pseudo-obstruction. The ileus, which
`started on the 4th day of treatment, resolved following dose
`interruption and returned upon reintroduction of study drug.
`There were no clinically significant ECG such as pro-
`longation of corrected QT interval related to BAY1436032
`treatment.
`Six of 27 subjects died either during BAY1436032
`treatment or within 30 days of permanent treatment dis-
`continuation, and none of these deaths were attributed to
`study drug. Causes of death were reported as PD or AE, in
`3 subjects each. AEs associated with these deaths included
`sepsis,
`lung infection,
`and general physical health
`deterioration.
`
`Efficacy
`
`The median treatment duration for all 27 subjects was
`3.0 months (0.49–8.5) (Supplementary Table S2), and there
`was a trend towards increased neutrophil counts occurring
`during treatment (Supplementary Fig. S2).
`The overall response rate to BAY1436032, based on
`investigator assessment, was 15% (4/27): subject 16 (1200
`mg BID) attained CRp at C7; subject 5 (300 mg BID)
`attained MLFS at C3; subject 24 (1500 mg BID) attained
`MLFS at C2 and went on to receive a 2nd stem cell
`
`Rigel Exhibit 1041
`Page 6 of 11
`
`

`

`Safety and efficacy of BAY1436032 in IDH1-mutant AML: phase I study results
`
`2909
`
`transplant; and subject 13 (1200 mg BID) attained PR at C5
`(Supplementary Table S2). The median treatment duration
`of these four responding subjects was 6.0 months (3.9–8.5).
`Blast counts and plasma R-2HG levels determined at var-
`ious times during the treatment of
`these subjects are
`depicted in Fig. 2. In subject 13, R-2HG levels rapidly
`decreased to a normal level found in healthy individuals,
`whereas the reduction of R-2HG to a normal level took a
`longer time in subjects 5 and 16.
`Best response of SD and PD was achieved by 18/27 and
`5/27 subjects, respectively (Supplementary Table S2). Of
`the 18 subjects with SD, 8 remained stable for ≥2 con-
`secutive monthly assessments, stayed on treatment
`for
`≥3 months and are considered to have achieved relevant SD.
`This group of 8 subjects showed a median treatment dura-
`tion of 5.5 months (3.1–7.0). Some decrease in leukemic
`blast counts and plasma R-2HG levels was evident in most
`of these subjects, but R-2HG levels generally did not reach
`normal levels (Supplementary Table S2, Figs. S3 and S4).
`As of November 20, 2019, 26/27 subjects were reported
`as deceased. Survival data by dosing cohort, as well as for
`the 27 subjects as a single group, is presented in Fig. 3.
`Median overall survival for the group of 27 treated subjects
`was 6.6 months (95% CI 4.6–9.4). The small subject
`numbers in each dosing cohort (n = 4–9) preclude relevant
`comparisons among cohorts.
`
`Retrospective mutational analysis
`
`Following enrollment, retrospective mutational analysis of
`>400 tumor-associated genes was successfully performed
`on baseline leukemic samples obtained from 25/27 treated
`subjects and the mIDH1 results from this analysis were
`concordant with those obtained locally via investigator
`assessment (Supplementary Tables S2 and S4).
`Baseline leukemic samples from 24/25 of the subjects for
`whom retrospective mutational analysis was successfully
`performed harbored alterations in additional genes classified
`as known or likely pathogenic, with a median of 2.5 (0–4)
`co-occurring alterations detected per subject. Co-occurring
`alterations in the following genes were found in >10% of
`evaluated subjects: DNMT3A (n = 11), ASXL1 (n = 7),
`NRAS (n = 5), NPM1 (n = 5), FLT3 (n = 4; 2 with an
`internal
`tandem duplication (ITD) and 2 with a kinase
`domain point mutation), TET2 and U2AF1 (n = 3 each).
`Co-occurring mutations in tyrosine kinase pathway genes
`such as NRAS, FLT3, and PTPN11 were absent
`in
`responding subjects.
`A co-occurring IDH2-R140Q mutation was detected in
`subjects 16 and 25, with variant allele frequencies (VAF) of
`13% for mIDH1/26% for mIDH2 in subject 16 and 5% for
`mIDH1/20% for mIDH2 in subject 25. Subject 16 experi-
`enced a significant
`reduction in R-2HG levels during
`
`BAY1436032 treatment, achieved a clinical response (CRp)
`and remained on treatment for 8.5 months (Supplementary
`Table S2, Fig. 2). Subject 25, who had received the mIDH2
`inhibitor enasidenib for the 10 months just prior to enroll-
`ment, experienced a modest reduction in R-2HG levels
`during BAY1436032 treatment, achieved a best response of
`SD and remained on treatment for 6.2 months (Supple-
`mentary Table S2, Fig. S4).
`An analysis of baseline samples showed the median
`VAF of mIDH1 to be 28% (5–49)
`(Supplementary
`Tables S2 and S4). The VAF of mIDH1 in comparison to
`the VAF of co-occurring mutations suggested that mIDH1
`was present
`in the founding clone in 18/25 evaluated
`subjects. There was no apparent correlation between the
`mIDH1 VAF and the specific IDH1-R132X mutation
`identified, or clinical outcome. Maximal R-2HG decreases
`detected during BAY1436032 treatment also did not cor-
`relate with baseline mIDH1 VAF.
`In addition to the analysis on baseline samples, for a
`limited number of subjects a retrospective mutational ana-
`lysis was also performed on bone marrow samples collected
`during BAY1436032 treatment. Results from three subjects
`who had been on treatment for at least three full cycles of
`treatment and had data available from baseline, during
`treatment and end-of-treatment (EOT) are shown in Sup-
`plementary Fig. S5. Although the IDH1 mutation present at
`baseline was always detected in samples collected during
`treatment and at the EOT, changes in other genes were
`evident: for subject 12 the EOT sample showed mutations
`in WT1, BCOR, and FLT3 (ITD) and a PDGFRB-TPM3
`gene fusion that were undetectable at baseline or at C3-D1;
`for subject 14 the EOT sample showed a MLL duplication/
`rearrangement that was not detected at baseline or C3-D1;
`and for subject 18 a mutation in STAG2 that was detected at
`baseline and at C3-D1 was not detected in the EOT sample,
`whereas the converse was true for mutations in PIK3C2G
`and CREBBP. Potential resistance mechanisms such as
`isoform switching and the acquisition of secondary altera-
`tions in mIDH1 were evaluated and not detected.
`
`Discussion
`
`the mIDH1 inhibitor BAY1436032 was
`In this study,
`evaluated at 4 different dose levels in a total of 27 AML
`subjects who harbored a mutation which altered the residue
`at position R132 of IDH1 to any one of 5 different amino
`acids, each of which is known to generate the R-2HG
`oncometabolite and to be inhibited by BAY1436032
`[16, 17].
`BAY1436032 was found to be generally safe, with just a
`single DLT (ileus) identified among the 27 treated subjects.
`The only other gastrointestinal-related TEAEs ≥grade 3
`
`Rigel Exhibit 1041
`Page 7 of 11
`
`

`

`2910
`
`M. Heuser et al.
`
`Fig. 2 Per-subject blast percentages and R-2HG levels from sub-
`jects who experienced clinical responses during BAY1436032
`treatment. Data from the four subjects who experienced clinical
`responses (1 CRp, 2 MLFS, 1 PR) is shown. Blast percentages are
`shown on the left Y-axis, plasma R-2HG levels on the right Y-axis and
`
`treatment cycle number on the X-axis. Bone marrow blasts are illu-
`strated with blue circles and peripheral blood blasts/leukocytes with
`red triangles. R-2HG levels are represented by gray bars and the R-
`2HG level associated with healthy individuals (138 ng/mL) is shown
`as black squares connected by a dotted black line.
`
`Fig. 3 Subject survival after
`start of BAY1436032
`treatment. Overall survival is
`depicted according to dosing
`cohort (a) and for all 27 treated
`subjects (b). Censored subjects
`are indicated with an asterisk.
`
`were two episodes of grade 3 nausea in cohort 4, which
`were not attributed to study drug. Each ≥grade 3 TEAE
`attributed to BAY1436032 occurred in a single subject. The
`most frequent ≥grade 3 TEAEs associated with ivosidenib,
`which is approved for a subset of subjects with mIDH1
`AML [27, 28], are QT prolongation (8%) and IDH DS (4%)
`[29]. Five subjects treated with BAY1436032 presented
`with DS, or symptoms associated with DS (all ≤grade 3),
`and all cases eventually resolved. There were no episodes of
`≥grade 3 QT prolongation with BAY1436032. The favor-
`able safety profile of BAY1436032 is consistent with pre-
`clinical studies demonstrating that BAY1436032 is a highly
`
`specific inhibitor of mIDH1 which exhibits little activity
`against wtIDH1 or wtIDH2 [16, 17].
`Modest clinical benefit from single-agent BAY1436032
`was identified in this study as evidenced by an overall
`response rate of 15% and a median treatment duration of
`6.0 months among responders. Eight additional subjects
`who achieved SD for ≥2 consecutive monthly assessments
`and stayed on treatment for ≥3 months showed a median
`treatment duration of 5.5 months. Most of these eight sub-
`jects experienced decreases in both bone marrow and per-
`ipheral blood blast counts during treatment, consistent with
`some degree of clinical benefit from BAY1436032. Clinical
`
`Rigel Exhibit 1041
`Page 8 of 11
`
`

`

`Safety and efficacy of BAY1436032 in IDH1-mutant AML: phase I study results
`
`2911
`
`relationship to
`
`outcome did not exhibit an obvious
`BAY1436032 dose.
`Although BAY1436032 demonstrated some degree of R-
`2HG inhibition in all subjects, only 5/26 subjects with an
`elevated baseline R-2HG level experienced a reduction to a
`normal level during BAY1436032 treatment. In support of a
`potential relationship between the degree of target inhibition
`and clinical benefit, the four subjects who achieved clinical
`responses showed a median maximal inhibition of baseline
`R-2HG levels of 95% (75–99), compared with 58% (16–93)
`among the 23 other subjects. A correlation between the
`degree of R-2HG suppression and clinical response was not
`found for ivosidenib [29], although in that study R-2HG
`levels were strongly suppressed in the majority of subjects
`regardless of clinical response, thereby making it difficult to
`directly address this question.
`It is unknown why a subset of subjects treated with
`BAY1436032 exhibit robust R-2HG suppression coupled
`with clinical benefit, while others do not. A dose-response
`relationship was not apparent when the degree of maximal
`percent R-2HG inhibition or the extent of clinical benefit
`was compared between cohorts, and subjects who achieved
`clinical benefit did not exhibit plasma concentrations
`markedly higher than those who did not.
`The overall response rate of 15% for BAY1436032 is
`lower than the 33–42% reported for other mIDH1 inhibitors
`in relapsed and refractory AML [29–32], and the median
`overall survival (6.6 months; 95% CI 4.6–9.4) is somewhat
`lower than that reported for ivosidenib in similar populations
`(8.8 months; 95% CI 6.7–10.2) [29]. It is likely that the
`incomplete target inhibition generated by BAY1436032

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket