scispace - formally typeset
Search or ask a question
Journal ArticleDOI

First-in-Human Phase I Study of the Oral Inhibitor of Indoleamine 2,3-Dioxygenase-1 Epacadostat (INCB024360) in Patients with Advanced Solid Malignancies.

TL;DR: Epacadostat was generally well tolerated, effectively normalized kynurenine levels, and produced maximal inhibition of IDO1 activity at doses of ≥100 mg BID.
Abstract: Purpose: Indoleamine 2,3-dioxygenase-1 (IDO1) catalyzes the degradation of tryptophan to N-formyl-kynurenine. Overexpressed in many solid malignancies, IDO1 can promote tumor escape from host immunosurveillance. This first-in-human phase I study investigated the maximum tolerated dose, safety, pharmacokinetics, pharmacodynamics, and antitumor activity of epacadostat (INCB024360), a potent and selective inhibitor of IDO1.Experimental Design: Fifty-two patients with advanced solid malignancies were treated with epacadostat [50 mg once daily or 50, 100, 300, 400, 500, 600, or 700 mg twice daily (BID)] in a dose-escalation 3 + 3 design and evaluated in 28-day cycles. Treatment was continued until disease progression or unacceptable toxicity.Results: One dose-limiting toxicity (DLT) occurred at the dose of 300 mg BID (grade 3, radiation pneumonitis); another DLT occurred at 400 mg BID (grade 3, fatigue). The most common adverse events in >20% of patients overall were fatigue, nausea, decreased appetite, vomiting, constipation, abdominal pain, diarrhea, dyspnea, back pain, and cough. Treatment produced significant dose-dependent reductions in plasma kynurenine levels and in the plasma kynurenine/tryptophan ratio at all doses and in all patients. Near maximal changes were observed at doses of ≥100 mg BID with >80% to 90% inhibition of IDO1 achieved throughout the dosing period. Although no objective responses were detected, stable disease lasting ≥16 weeks was observed in 7 of 52 patients.Conclusions: Epacadostat was generally well tolerated, effectively normalized kynurenine levels, and produced maximal inhibition of IDO1 activity at doses of ≥100 mg BID. Studies investigating epacadostat in combination with other immunomodulatory drugs are ongoing. Clin Cancer Res; 23(13); 3269-76. ©2017 AACR.

Content maybe subject to copyright    Report

Cancer Therapy: Clinical
First-in-Human Phase I Study of the Oral Inhibitor
of Indoleamine 2,3-Dioxygenase-1 Epacadostat
(INCB024360) in Patients with Advanced Solid
Malignancies
Gregory L. Beatty
1,2
, Peter J. O'Dwyer
1,2
, Jason Clark
3
, Jack G. Shi
3
, Kevin J. Bowman
3
,
Peggy A. Scherle
3
, Robert C. Newton
3
, Richard Schaub
3
, Janet Maleski
3
, Lance Leopold
3
,
and Thomas F. Gajewski
4
Abstract
Purpose: Indoleamine 2,3-dioxygenase-1 (IDO1) catalyzes the
degradation of tryptophan to N-formyl-kynurenine. Overex-
pressed in many solid malignancies, IDO1 can promote tumor
escape from host immunosurveillance. This rst-in-human phase
I study invest igated the maximum tolerated dose, safety, phar-
macokinetics, pharmacodynamics, and antitumor activity of epa-
cadostat (INCB024360), a potent and selective inhibitor of IDO1.
Experimental Design: Fifty-two patients with advanced solid
malignancies were treated with epacadostat [50 mg once daily or
50, 100, 300, 400, 500, 600, or 700 mg twice daily (BID)] in a
dose-escalation 3 þ 3 design and evaluated in 28-day cycles.
Treatment was continued until disease progression or unaccept-
able toxicity.
Results: One dose-limiting toxicity (DLT) occurred at the dose
of 300 mg BID (grade 3, radiation pneumonitis); another DLT
occurred at 400 mg BID (grade 3, fatigue). The most common
adverse events in >20% of patients overall were fatigue, nausea,
decreased appetite, vomiting, constipation, abdominal pain,
diarrhea, dyspnea, back pain, and cough. Treatment produced
signicant dose-dependent reductions in plasma kynurenine
levels and in the plasma kynurenine/tryptophan ratio at all doses
and in all patients. Near maximal changes were observed at doses
of 100 mg BID with >80% to 90% inhibition of IDO1 achieved
throughout the dosing period. Although no objective responses
were detected, stable disease lasting 16 weeks was observed in
7 of 52 patients.
Conclusions: Epacadostat was generally wel l tol erated,
effectively normalized kynurenine levels, and produced max-
imal inhibition of IDO1 activity at doses of 100 mg BID.
Studies investigating epacadostat in combination with othe r
immunomodulatory drugs are ongoing.
Clin Cancer Res; 23(13);
326976. 2017 AACR.
Introduction
Recently, immunotherapy with checkpoint inhibitors has pro-
duced potent long-lasting antitumor activity in patients with
advanced cancer (14). However, the majority of patients across
a wide range of malignancies do not respond (5). This observation
emphasizes the likely existence of additional immunoregulatory
pathways that control the effectiveness of immunosurveillance in
cancer (6, 7). Strategies designed to derail these pathways may be
critical for broadening the application of cancer immunotherapy.
Indoleamine 2,3-dioxygenase-1 (IDO1) is a heme-containing,
monomeric oxidoreductase rate-limiting enzyme that catalyzes
the degradation of the amino acid tryptophan to kynurenine (8).
IDO has been shown to play an important role in immune
regulation. During fetal development, IDO inhibition using the
competitive inhibitor 1-methyltryptophan leads to fetal rejection
by maternal T cells (9). In autoimmunity, IDO appears to be a key
regulator involved in suppressing T-cell responses toward apo-
ptotic cell-associated antigens (10) and in controlling T-cell
activity at sites of graft-versus-host disease (11). In cancer, IDO1
expression can block the development of a productive antitumor
immune response (1216).
Indoleamine 2,3-dioxygenase-1 is expressed by various cell
types, including malignant epithelial and myeloid cells such as
dendritic cells and macrophages that localize in tumor sites and
within tumor-draining lymph nodes. IDO1 expression by malig-
nant and nonmalignant cells can inhibit T-cell immune
responses, leading to immune evasion and tumor outgrowth
(1719). Tryptophan metabolites such as kynurenine, produced
by IDO1, inhibit immunosurveillance in cancer by arresting T
cells in the G
1
phase of the cell cycle (18), promoting T-cell and
1
Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylva-
nia.
2
Division of Hematology-Oncology, Department of Medicine, University of
Pennsylvania, Philadelphia, Pennsylvania.
3
Incyte Corporation, Wilmington,
Delaware.
4
Section of Hematology/Oncology, Department of Medicine, Univer-
sity of Chicago, Chicago, Illinois.
Note: Trial registration ID: NCT01195311.
Prior presentation: This article presents original results of a phase I study o f
epacadostat (INCB024360) in patients with advanced solid malignancies.
Results were presented in part at the Annual Meeting of the American Society
of Clinical Oncology (J Clin Oncol 30, 2012 suppl; abstr 2500; J Clin Oncol 31, 2013
suppl; abstr 3025).
Corresponding Author: Gregory L. Beatty, Abramson Cancer Center of the
University of Pennsylvania, Smilow Center for Translational Research, Room
8-112, 3400 Civic Cente r Boulevard, Building 421, Philadelphia, PA 19104-5156.
Phone: 215-746-7764; Fax: 215-573-8590; E-mail:
gregory.beatty@uphs.upenn.edu
doi: 10.1158/107 8-0432.CCR-16-2272
2017 American Association for Cancer Research.
Clinical
Cancer
Research
www.aacrjournals.org
3269
Downloaded from http://aacrjournals.org/clincancerres/article-pdf/23/13/3269/2300480/3269.pdf by guest on 26 August 2022

dendritic cell apoptosis, and supporting regulatory T-cell gener-
ation (12, 1922). In addition, tryptophan metabolites have been
found to negatively affect natural killer cell function (23, 24).
Indoleamine 2,3-dioxygenase-1 is increased in tumors that are
actively inltrated by effector T cells (13). This enhanced tumor
expression of IDO1 is thought to be driven by IFNg (12). In
patients with advanced cancer, IDO1 activity is increased
with elevated levels of tryptophan metabolites detected in
the serum and is associated with a poor prognosis (12, 2529).
Based on these ndings, IDO1 inhibition using epacadostat
(INCB024360), a selective small-molecule inhibitor of IDO1,
was initially evaluated in a mouse model of cancer, where it was
found to induce T-celldependent antitumor immunity (12).
This activity was related to the ability of epacadostat to block
regulatory T-cell activity and promote dendritic cell maturation
and function. Here, we report on the safety and activity of single-
agent epacadostat in patients with advanced solid malignancies.
The primary objective was to dene the safety, tolerability, and
maximum tolerated dose (MTD) of epacadostat. Secondary objec-
tives were to evaluate clinical activity, pharmacokinetics (PK),
pharmacodynamics, and the effect on blood biomarkers of
inammation.
Patients and Methods
Patients
Fifty-two patients with a neoplastic disease that was refractory
to currently available therapies or for which no effective treatment
was available were enrolled at the Abramson Cancer Center,
University of Pennsylvania (Philadelphia, PA), and the University
of Chicago (Chicago, IL). Inclusion criteria were age 18 years,
Eastern Cooperative Oncology Group (ECOG) performance sta-
tus of 0 to 2, life expectancy 12 weeks, and adequate end organ
function. Exclusion criteria included receipt of an investigational
study drug within 28 days (42 days for monoclonal antibodies)
before study entry and prior anticancer medication within 21 days
of the rst dose of study medication or 6 weeks for mitomycin-C
or nitrosoureas. Hormonal treatments were allowed and could be
continued, and there were no restrictions on the number of prior
lines of therapy. Additional exclusion criteria were active viral
hepatitis or other active chronic or acute systemic infection, active
autoimmune disorder, receipt of any compound known to be a
potent inducer or inhibitor of CYP3A4, prior bone marrow or
solid organ transplant, receipt of serotonin reuptake inhibitor
within 3 weeks before study entry, major surgery within 4 weeks of
study entry, active cardiovascular disease, history of gastrointes-
tinal condition causing malabsorption or obstruction, history of
brain metastases or spinal cord compression, and active infection.
All patients provided written informed consent. The study was
approved by the institutional review boards of the participating
institutions.
Study design and treatment
This was an open-label, multicenter, phase I dose-escalation
study using a 3 þ 3 design. The primary objective was to determine
safety, tolerability, and MTD of epacadostat. Secondary objectives
were to characterize PK, pharmacodynamics, biomarkers, and
tumor response rates.
Epacadostat was supplied by Incyte Corp oration (Wilming-
ton, DE ; 25-, 100-, and 300-mg tablets) and was administered
orally once or twic e daily after a 2-hour fast [50 m g once daily
or 50, 100 , 300, 400, 50 0, 6 00, or 700 mg twice daily (BID)] in
28-day cycles. The p redetermi ned doses of 50, 100, and 300 mg
BID were predicted bas ed on preclinical studies to achieve 50%,
75%, and 90 % inhibi tory concentrations (IC
50
,IC
75
,andIC
90
)
of IDO1 at trough serum levels in humans . The starting dose of
50 mg daily was proposed to minimize risk while stil l provid-
ing some inhibitory effect. Patients were instructed to fast for an
additional hour following dosing. Additionally, the foo d effect
of a standardized high-fat meal on steady-stat e PK was eva lu-
ated for the epacadostat 600 mg BID grou p on cy cle 2, day 1.
Patients continued treatment until disease progression or unac-
ceptable toxicity.
Safety and efcacy assessments
All patients who received at least 1 dose of epacadostat were
evaluated for safety and efcacy. Safety assessments included the
incidence of all adverse events (AE), irrespective of relationship to
study drug, according to the National Cancer Institute Common
Terminology Criteria for Adverse Events, version 4.0, and the
incidence of patients experiencing dose modications and/or
premature discontinuation of study drug. AEs were monitored
at screening; on days 1, 8, 15, and 22 during cycle 1; and on days 1
and 15 for all subsequent cycles. A dose-limiting toxicity (DLT)
was dened as a treatment-related grade 4 hematologic toxicity
of any duration except for grade 4 neutropenia without fever
lasting less than 7 days or any grade 3 nonhematologic toxicity
that occurred during the rst 28 days of treatment (cycle 1). The
MTD was estimated as the dose level below that at which one third
or more patients in a group (minimum 6) experienced DLTs
related to treatment with epacadostat. Tumor response was
assessed by computed tomography at baseline and every 8 weeks
according to Response Evaluation Criteria in Solid Tumors, ver-
sion 1.1.
Pharmacokinetics assessment
Plasma samples were collected on days 1 and 15 (steady state)
of cycle 1 before dosing and at dened time points after dosing.
Samples were analyzed for epacadostat by a validated liquid
chromatography with tandem mass spectrometry (LC/MS/MS)
Translational Relevance
Immunotherapy has emerged as a promising treatment
option for advanced cancer; however, many patients do not
receive adequate benet, possibly because of immune check-
point pathway redundancies. Here, we report results of a rst-
in-human study of epacadostat, an oral selective inhibitor of
indoleamine 2,3-dioxygenase (IDO1), in patients with treat-
ment-refractory advanced carcinoma. IDO1 is a rate-limiting
enzyme that catalyzes the degradation of the amino acid
tryptophan to kynurenine and, in doing so, can inhibit anti-
tumor immune activity. In this phase I study, epacadostat was
generally well tolerated and was associated with stable disease
lasting 16 weeks in 7 of 52 patients. Reductions in plasma
kynurenine levels and the kynurenine/tryptophan ratio were
found to be relevant blood-based biomarkers for monitoring
IDO1 inhibition in vivo. Our ndings support further evalu-
ation of epacadostat in combination with other treatment
approaches for improving antitumor immune activity in
patients with advanced cancer.
Beatty et al.
Clin Cancer Res; 23(13) July 1, 2017 Clinical Cancer Research3270
Downloaded from http://aacrjournals.org/clincancerres/article-pdf/23/13/3269/2300480/3269.pdf by guest on 26 August 2022

assay, with a lower limit of quantitation of 2 nmol/L (30).
Noncompartmental PK analysis was performed using Phoenix
WinNonlin (Certara USA, Inc.). Where available, the actual sam-
ple collection times were used for PK analysis. For a small number
of samples for which actual collection times were not recorded,
the scheduled collection times were used instead.
Pharmacodynamic assessments
Plasma levels of tryptophan and kynurenine at different time
points were evaluated by LC/MS/MS. In addition, whole blood
samples, collected at times correlating with PK assessments, were
evaluated for IDO1 inhibition by stimulating the blood with IFNg
(100 ng/mL) and lipopolysaccharide (LPS; 100 ng/mL) for 18
hours and then analyzing the plasma from the samples for
tryptophan and kynurenine levels by LC/MS/MS as previously
described (30). For each patient, the percentage inhibition of
IDO1, as determined by the decrease in kynurenine levels, was
calculated by comparing predose values with values obtained at
different times after dosing.
Changes in the plasma levels of proteins related to immunity or
inammation were monitored using Evidence Investigator Bio-
chip Array technology, a custom-designed multiplex biochip array
based on sandwich chemiluminescent immunoassays (Randox
Laboratories).
Statistical analysis
Descriptive statistical analyses were used to evaluate all appli-
cable outcomes.
Results
Patient characteristics
Fifty-two patients with advanced solid malignancies were
enrolled (Table 1). The predominant tumor sites were colorectal
(n ¼ 29, 55.8%) and melanoma (n ¼ 6, 11.5%), and the most
frequently reported locations of metastatic disease were lung,
liver, and lymph nodes. Each patient had received at least 1 prior
treatment for metastatic disease before study entry, and all
patients except 1 had ECOG performance status of 0 or 1.
Dose-limiting toxicities
Two patients experienced DLTs: grade 3 radiation pneumonitis
and grade 3 fatigue occurred in 1 patient each at the 300- and 400-
mg BID dose levels, respectively. In light of the projection from
preclinical animal modeling data (30) that all BID doses admin-
istered during dose escalation (50700 mg) would be efcacious,
no other DLT occurred with dosing up to 700 mg BID; thus, the
MTD was not established for epacadostat.
Safety and tolerability
The median (range) duration of epacadostat exposure was 51.5
(7284) days with a median (range) total daily dose of 800 (43.2
1400) mg. Irrespective of association with therapy, the most
common AEs (all grades) occurring throughout the study period
were fatigue (69.2%), nausea (65.4%), decreased appetite
(53.8%), and vomiting (42.3%; Table 2). These AEs were man-
aged by investigators using routine supportive care measures.
Seven patients (13.5%) discontinued therapy because of AEs
(50 mg once daily, n ¼ 1; 100 mg BID, n ¼ 1; 300 mg BID,
n ¼ 2; 400 mg BID, n ¼ 3), including pain, hepatic infection,
pneumonia, radiation recall pneumonitis (DLT), fatigue (DLT),
dyspnea and hypoxia, and nausea and vomiting. Only radiation
pneumonitis and fatigue were considered DLTs and possibly
related, but these dose levels were expanded and determined to
not exceed the MTD. Liver enzymes were monitored closely
throughout treatment in all patients. No grade 4 elevations were
observed in aspartate aminotransferase (AST) or alanine amino-
transferase (ALT) levels. Grade 3 AST/ALT elevation was observed
in 1 patient but was attributed to biliary duct obstruction con-
sistent with progressive disease. A second patient also experienced
grade 3 AST/ALT elevations, but this was determined to be most
likely related to acetaminophen ingestion over the maximum
recommended daily dose for tumor fevers.
Pharmacokinetics
Following oral d ose administratio n in t he fasted state, the PK
of epacadostat was characterized by a time of maximu m
observed concentration at approximately 2 hours and a bip has-
ic disposition with an apparent terminal-phase dispositio n
half-life of 2.9 hours, which appeared to be dose-i ndep endent.
Systemic accumulatio n followi ng BID dosi ng increased mea n
epacadostat maximum observed concentration (C
max
)andarea
under the concentration versu s time curve over 1 steady- state
dosing interval ( AUC
0t
) by 16% and 33 %, respe ctively , s ug-
gesting a relatively longer effective half-life of 4 to 6 ho urs.
Increases in epacadostat C
max
and AUC
0t
were slig htly less
Table 1. Patient demographics and disease characteristics at baseline
Total
Characteristic (N ¼ 52)
Age, y
Median 59.5
Min, max 38.0, 78.0
65 y, n (%) 36 (69.2)
Gender, n (%)
Male 31 (59.6)
Female 21 (40.4)
Race, n (%)
White 45 (86.5)
Black/African American 4 (7.7)
Asian 2 (3.8)
Native Hawaiian/Other Pacic Islander 1 (1.9)
ECOG status, n (%)
0 20 (38.5)
1 31 (59.6)
2 1 (1.9)
Tumor site, n (%)
Colorectal 29 (55.8)
Melanoma 6 (11.5)
Renal cell carcinoma 3 (5.8)
Other 14 (26.9)
Locally advanced disease, n (%)
Yes 33 (63.5)
No 19 (36.5)
Locations of metastatic disease, n (%)
Lung 38 (73.1)
Liver 30 (57.7)
Lymph nodes 27 (51.9)
Bone 6 (11.5)
Ascites 3 (5.8)
Skin or subcutaneous tissue 2 (3.8)
Pancreas 1 (1.9)
Pleural effusion 1 (1.9)
Other
a
22 (42.3)
Abbreviation: ECOG, Eastern Cooperati ve Oncology Group.
a
Includes lesions detected in or invading the spleen, adrenals, abdominal wall,
ovary, peritoneum, kidney, small bowel, mesentery, bladder, chest wall, medi-
astinum, pleura, pelvis, uterus, and ureters.
IDO1 Inhibitor in Advanced Solid Cancers
www.aacrjournals.org Clin Cancer Res; 23(13) July 1, 2017 3271
Downloaded from http://aacrjournals.org/clincancerres/article-pdf/23/13/3269/2300480/3269.pdf by guest on 26 August 2022

than proporti onal to dose within the r ange of 5 0 to 700 mg
BID. Moderate inter subject v ariabil ity was observed for ep aca-
dostat plasma e xposure s (Table 3). Administration of a high-fat
meal with e pacados tat 600 mg BID decreased t he geometric
mean C
max
by approximately 10% and increased the geometric
mean area under the curve from 0 to 12 hours (AUC
012h
)by
22%. The 90% C Is of the geometric mean rat io point estimates
for C
max
and AUC
012h
were 0.645 to 1.25 and 0.952 to 1.57,
respectively, bo th including the value of 1 and indica ting that
the effect on epacadostat plasma exposu res from a high-fat
meal was not s tatis tically signicant.
Pharmacodynamics
Treatment with epacadostat did not produce any signicant
changes in plasma proteins related to immune function, includ-
ing IL-4, IL-10, IL12, CCL22 (macrophage-derived chemokine), or
CCL5 (RANTES). In addition, no changes were observed with
treatment in plasma proteins related to inammation, including
C-reactive protein, intercellular adhesion molecule 1, IL-6, tumor
necrosis factor (TNF)-a, TNF receptor 2, and vascular cell adhe-
sion protein 1.
In the published literature, kynurenine levels are generally
elevated in patients with cancer (12), suggesting activation of the
Table 2. Treatment-emergent adverse events
50 mg 50 mg 100 mg 300 mg 400 mg 500 mg 600 mg 700 mg
QD BID BID BID BID BID BID BID Total
AE, n (%) (n ¼ 3) (n ¼ 4) (n ¼ 5) (n ¼ 6) (n ¼ 11) (n ¼ 5) (n ¼ 14) (n ¼ 4) (N ¼ 52)
All-grade AEs in 10% of patients
Fatigue 2 (66.7) 2 (50.0) 4 (80.0) 3 (50.0) 10 (90.9) 5 (100.0) 8 (57.1) 2 (50.0) 36 (69.2)
Nausea 3 (100.0) 4 (100.0) 3 (60.0) 4 (66.7) 5 (45.5) 4 (80.0) 8 (57.1) 3 (75.0) 3 4 (65.4)
Decreased appetite 0 2 (50.0) 1 (20.0) 3 (50.0) 7 (63.6) 4 (80.0) 8 (57.1) 3 (75.0) 28 (53.8)
Vomiting 2 (66.7) 2 (50.0) 0 3 (50.0) 3 (27.3) 2 (40.0) 8 (57.1) 2 (50.0) 22 (42.3)
Constipation 1 (33.3) 2 (50.0) 1 (20.0) 4 (66.7) 2 (18.2) 0 6 (42.9) 3 (75.0) 19 (36.5)
Abdominal pain 1 (33.3) 1 (25.0) 0 2 (33.3) 5 (45.5) 1 (20.0) 4 (28.6) 1 (25.0) 15 (28.8)
Diarrhea 1 (33.3) 2 (50.0) 1 (20.0) 1 (16.7) 4 (36.4) 1 (20.0) 3 (21.4) 1 (25.0) 14 (26.9)
Back pain 1 (33.3) 0 0 3 (50.0) 3 (27.3) 0 4 (28.6) 2 (50.0) 13 (25.0)
Dyspnea 1 (33.3) 1 (25.0) 1 (20.0) 1 (16.7) 2 (18.2) 1 (20.0) 5 (35.7) 1 (25.0) 13 (25.0)
Cough 1 (33.3) 0 2 (40.0) 2 (33.3) 1 (9.1) 2 (40.0) 3 (21.4) 0 11 (21.2)
Hypokalemia 0 2 (50.0) 2 (40.0) 1 (16.7) 2 (18.2) 0 2 (14.3) 1 (25.0) 10 (19.2)
Weight decreased 2 (66.7) 0 1 (20.0) 1 (16.7) 3 (27.3) 0 2 (14.3) 1 (25.0) 10 (19.2)
Pain 1 (33.3) 1 (25.0) 0 1 (16.7) 1 (9.1) 2 (40.0) 1 (7.1) 2 (50.0) 9 (17.3)
Pyrexia 1 (33.3) 1 (25.0) 0 1 (16.7) 3 (27.3) 0 2 (14.3) 0 8 (15.4)
Dehydration 0 1 (25.0) 0 1 (16.7) 3 (27.3) 0 2 (14.3) 0 7 (13.5)
Dizziness 1 (33.3) 0 0 0 1 (9.1) 0 2 (14.3) 2 (50.0) 6 (11.5)
Dysgeusia 0 0 1 (20.0) 1 (16.7) 1 (9.1) 0 2 (14.3) 1 (25.0) 6 (11.5)
Grade 3 or 4 AEs occurring in 2 patients
Fatigue 0 0 0 0 1 (9.1) 1 (20.0) 3 (21.4) 1 (25.0) 6 (11.5)
Abdominal pain 0 1 (25.0) 0 0 3 (27.3) 0 1 (7.1) 0 5 (9.6)
Hypokalemia 0 1 (25.0) 1 (20.0) 0 1 (9.1) 0 1 (7.1) 1 (25.0) 5 (9.6)
Nausea 0 1 (25.0) 0 1 (16.7) 1 (9.1) 0 2 (14.3) 0 5 (9.6)
Dehydration 0 1 (25.0) 0 1 (16.7) 1 (9.1) 0 0 0 3 (5.8)
Dyspnea 0 0 0 0 1 (9.1) 0 2 (14.3) 0 3 (5.8)
Hypotension 0 0 1 (20.0) 1 (16.7) 1 (9.1) 0 0 0 3 (5.8)
Vomiting 0 1 (25.0) 0 0 1 (9.1) 0 1 (7.1) 0 3 (5.8)
ALT increased 0 1 (25.0) 0 0 0 0 1 (7.1) 0 2 (3.8)
Anemia 0 0 1 (20.0) 0 1 (9.1) 0 0 0 2 (3.8)
Back pain 0 0 0 0 1 (9.1) 0 1 (7.1) 0 2 (3.8)
Bacterial infection 0 0 0 0 0 0 2 (14.3) 0 2 (3.8)
Hyponatremia 0 0 0 0 0 0 2 (14.3) 0 2 (3.8)
Pneumonia 0 0 0 1 (16.7) 0 0 1 (7.1) 0 2 (3.8)
Small intestinal obstruction 0 2 (50.0) 0 0 0 0 0 0 2 (3.8)
Abbreviations: AE, adverse event; ALT, alanine aminotransferase; BID, twice daily; QD, once daily.
Table 3. Summary of epacadostat steady-state (day 15) pharmacokinetic parameters
Mean SD
Dose group C
max
, mmol/L t
max
,h
a
t
1/2
, h AUC
0t
, mmol/L h
50 mg QD (n ¼ 3) 0.396 0.172 2.0 (1.04.0) 2.4 0.26 1.58 0.31
50 mg BID (n ¼ 4) 0.742 0.212 2.0 (1.03.9) 2.4 0.56 3.05 1.36
100 mg BID (n ¼ 5) 1.23 0.348 2.0 (1.02.2) 3.3 0.75 5.77 2.34
300 mg BID (n ¼ 5) 2.48 0.515 2.0 (1.02.0) 3.9 2.1 9.78 0.86
400 mg BID (n ¼ 8) 4.39 2.02 2.0 (1.06.0) 2.7 0.62 19.6 7.43
500 mg BID (n ¼ 5) 4.82 2.26 2.0 (2.02.4) 2.4 0.37 20.6 6.82
600 mg BID (n ¼ 12) 4.82 2.16 2.0 (1.02.1) 3.3 0.97 22.9 10.0
700 mg BID (n ¼ 4) 6.23 2.09 3.0 (2.04.5) 3.0 1.2 35.8 15.5
Abbreviations: AUC
0t
, area under the concentration versus time curve over 1 steady-state dosing interval; BID, twice daily; C
max
, maximum observed concentration;
QD, once daily; t
1/2
, half-life; t
max
, time of maximum observed concentration.
a
t
max
is reported as median (range).
Beatty et al.
Clin Cancer Res; 23(13) July 1, 2017 Clinical Cancer Research3272
Downloaded from http://aacrjournals.org/clincancerres/article-pdf/23/13/3269/2300480/3269.pdf by guest on 26 August 2022

IDO1 pathway. Treatment with epacadostat reduced plasma
kynurenine levels by 2 hours (data not shown) and, at doses
100 mg BID, was shown to reduce kynurenine levels to within
the range observed in normal, healthy volunteers (median value
of healthy subjects, 1499 nmol/L; Fig. 1A). In patients treated with
epacadostat, plasma kynurenine levels were found to decrease at
all doses, with near maximal inhibition achieved at doses 100
mg BID (Fig. 1B). Similar ndings were also observed for kynur-
enine to tryptophan ratios (data not shown).
Basal kynurenine levels in the plasma can be inuenced by both
IDO1 and tryptophan 2,3-dioxygenase (TDO), a liver enzyme that
controls dietary tryptophan catabolism (31). To determine the
ability of epacadostat to inhibit IDO1 activity in vivo and mini-
mize the TDO component, whole blood was stimulated for 18
hours with IFNg and LPS to induce IDO1 expression and then
assayed for kynurenine levels. Within 2 hours of dosing, a rapid
onset of IDO1 inhibition was seen with >90% inhibition achieved
at all dose levels. Maximal inhibition at trough was observed at
doses 100 mg BID (Fig. 1C). The 90% inhibitory concentration
was achievable at plasma concentrations greater than 500 nmol/L
(Fig. 1D), which is consistent with the in vitro potency of epacado-
stat (12).
Efcacy
The overall objective response rates are summarized in Table 4.
Stable disease, seen in 18 patients (34.6%), was the best overall
response. Eleven patients (21.2%), including 2 melanoma
patients who had progressed on ipilimumab immunotherapy
before study entry, showed prolonged stable disease compared
with their last prior therapy (Fig. 2). Stable disease lasting 16
weeks was seen in 7 patients (13.5%).
A
DC
B
Baseline
Day 15 (Predose)
Average Kyn, nmol/L
0
1,000
2,000
3,000
4,000
5,000
50 mg
BID
100 mg
BID
300 mg
BID
400 mg
BID
500 mg
BID
600 mg
BID
700 mg
BID
8007001000 200 300 400 500 600
Average percentage decrease in Kyn levels
from baseline at day 15
0
25
50
75
Dose, mg BID
210
Kyn, % Inhibition
0
10
20
30
40
50
60
70
80
90
100
Epacadostat plasma concentration, mmol/L
Average Kyn, % Inhibition
Time, h
0
20
2406
40
60
100
80
120
50 mg BID
100 mg BID
300 mg BID
400 mg BID
500 mg BID
600 mg BID
700 mg BID
Figure 1.
Pharmacodynamic effects of epacadostat BID dosing in vivo. A, Kyn levels by dose group at baseline and day 15. The dashed line represents the median Kyn
value observed in normal, healthy volunteers (1,499 nmol/L). B, Percentage decrease in Kyn levels from baseline by dose. C, Average Kyn inhibition over
time by dose on day 15 (relative to day 1) following IDO1 induction with IFNg and LPS. Time 0 represents the trough levels before the next dose of epacadostat
treatment. D, Epacadostat plasma concentrations relative to percentage Kyn inhibition. BID, twice daily; IDO1, indoleamine 2,3-dioxygenase-1; IFNg, interferon
gamma; Kyn, kynurenine; LPS, lipopolysaccharide.
Table 4. Best overall tumor response by RECIST, version 1.1
Total
n (%) (N ¼ 52)
Complete response 0
Partial response 0
Stable disease 18 (34.6)
Progressive disease 22 (42.3)
Not evaluable 12 (23.1)
Abbreviation: RECIST, Response Evaluation Criteria in Solid Tumors.
IDO1 Inhibitor in Advanced Solid Cancers
www.aacrjournals.org Clin Cancer Res; 23(13) July 1, 2017 3273
Downloaded from http://aacrjournals.org/clincancerres/article-pdf/23/13/3269/2300480/3269.pdf by guest on 26 August 2022

Citations
More filters
Journal ArticleDOI
TL;DR: Novel insights are discussed into the roles of Treg cells in cancer, which can hopefully be used to develop Treg cell-targeted therapies and facilitate immune precision medicine.
Abstract: Regulatory T (Treg) cells, an immunosuppressive subset of CD4+ T cells characterized by the expression of the master transcription factor forkhead box protein P3 (FOXP3), are a component of the immune system with essential roles in maintaining self-tolerance. In addition, Treg cells can suppress anticancer immunity, thereby hindering protective immunosurveillance of neoplasia and hampering effective antitumour immune responses in tumour-bearing hosts, thus promoting tumour development and progression. Identification of the factors that are specifically expressed in Treg cells and/or that influence Treg cell homeostasis and function is important to understanding cancer pathogenesis and to identifying therapeutic targets. Immune-checkpoint inhibitors (ICIs) have provided a paradigm shift in the treatment of cancer. Most immune-checkpoint molecules are expressed in Treg cells, but the effects of ICIs on Treg cells, and thus the contributions of these cells to treatment responses, remain unclear. Notably, evidence indicates that ICIs targeting programmed cell death 1 (PD-1) might enhance the immunosuppressive function of Treg cells, whereas cytotoxic T lymphocyte antigen 4 (CTLA-4) inhibitors might deplete these cells. Thus, although manipulation of Treg cells is a promising anticancer therapeutic strategy, approaches to controlling these cells require further research. Herein, we discuss novel insights into the roles of Treg cells in cancer, which can hopefully be used to develop Treg cell-targeted therapies and facilitate immune precision medicine. Regulatory T (Treg) cells are implicated in cancer immune evasion and escape and thus contribute to tumour development and progression. In this Review, the authors provide an overview of the phenotypes and roles of Treg cells in the context of cancer and outline potential strategies to target this cell type in anticancer immunotherapy.

717 citations

Journal ArticleDOI
TL;DR: An overview of the physiological and pathophysiological roles of tryptophan metabolism is provided, focusing on the clinical potential and challenges associated with targeting this pathway.
Abstract: L-Tryptophan (Trp) metabolism through the kynurenine pathway (KP) is involved in the regulation of immunity, neuronal function and intestinal homeostasis. Imbalances in Trp metabolism in disorders ranging from cancer to neurodegenerative disease have stimulated interest in therapeutically targeting the KP, particularly the main rate-limiting enzymes indoleamine-2,3-dioxygenase 1 (IDO1), IDO2 and tryptophan-2,3-dioxygenase (TDO) as well as kynurenine monooxygenase (KMO). However, although small-molecule IDO1 inhibitors showed promise in early-stage cancer immunotherapy clinical trials, a phase III trial was negative. This Review summarizes the physiological and pathophysiological roles of Trp metabolism, highlighting the vast opportunities and challenges for drug development in multiple diseases.

664 citations

Journal ArticleDOI
TL;DR: An overview of the field is provided and current immunotherapeutic approaches for solid tumours and haematological malignancies are summarized and the authors highlight the current immunotherapy trials that are targeting NK cells to treat patients with cancer.
Abstract: Immuno-oncology is an emerging field that has revolutionized cancer treatment Most immunomodulatory strategies focus on enhancing T cell responses, but there has been a recent surge of interest in harnessing the relatively underexplored natural killer (NK) cell compartment for therapeutic interventions NK cells show cytotoxic activity against diverse tumour cell types, and some of the clinical approaches originally developed to increase T cell cytotoxicity may also activate NK cells Moreover, increasing numbers of studies have identified novel methods for increasing NK cell antitumour immunity and expanding NK cell populations ex vivo, thereby paving the way for a new generation of anticancer immunotherapies The role of other innate lymphoid cells (group 1 innate lymphoid cell (ILC1), ILC2 and ILC3 subsets) in tumours is also being actively explored This Review provides an overview of the field and summarizes current immunotherapeutic approaches for solid tumours and haematological malignancies

628 citations

Journal ArticleDOI
TL;DR: A comprehensive review of immune checkpoint pathways involved in cancer immunotherapy is provided, and their mechanisms and the therapeutic interventions currently under investigation in phase I/II clinical trials are discussed.
Abstract: Immune checkpoints consist of inhibitory and stimulatory pathways that maintain self-tolerance and assist with immune response. In cancer, immune checkpoint pathways are often activated to inhibit the nascent anti-tumor immune response. Immune checkpoint therapies act by blocking or stimulating these pathways and enhance the body’s immunological activity against tumors. Cytotoxic T lymphocyte-associated molecule-4 (CTLA-4), programmed cell death receptor-1 (PD-1), and programmed cell death ligand-1(PD-L1) are the most widely studied and recognized inhibitory checkpoint pathways. Drugs blocking these pathways are currently utilized for a wide variety of malignancies and have demonstrated durable clinical activities in a subset of cancer patients. This approach is rapidly extending beyond CTLA-4 and PD-1/PD-L1. New inhibitory pathways are under investigation, and drugs blocking LAG-3, TIM-3, TIGIT, VISTA, or B7/H3 are being investigated. Furthermore, agonists of stimulatory checkpoint pathways such as OX40, ICOS, GITR, 4-1BB, CD40, or molecules targeting tumor microenvironment components like IDO or TLR are under investigation. In this article, we have provided a comprehensive review of immune checkpoint pathways involved in cancer immunotherapy, and discuss their mechanisms and the therapeutic interventions currently under investigation in phase I/II clinical trials. We also reviewed the limitations, toxicities, and challenges and outline the possible future research directions.

529 citations


Cites background from "First-in-Human Phase I Study of the..."

  • ...No OR was reported, but 7/52 patients achieved SD greater than 16 weeks [113]....

    [...]

Journal ArticleDOI
TL;DR: Epacadostat 100 mg twice daily plus pembrolizumab did not improve progression-free survival or overall survival compared with placebo plus pEmbrolizumsab in patients with unresectable or metastatic melanoma.
Abstract: Summary Background Immunotherapy combination treatments can improve patient outcomes. Epacadostat, an IDO1 selective inhibitor, and pembrolizumab, a PD-1 inhibitor, showed promising antitumour activity in the phase 1–2 ECHO-202/KEYNOTE-037 study in advanced melanoma. In this trial, we aimed to compare progression-free survival and overall survival in patients with unresectable stage III or IV melanoma receiving epacadostat plus pembrolizumab versus placebo plus pembrolizumab. Methods In this international, randomised, placebo-controlled, double-blind, parallel-group, phase 3 trial, eligible participants were aged 18 years or older, with unresectable stage III or IV melanoma previously untreated with PD-1 or PD-L1 checkpoint inhibitors, an ECOG performance status of 0 or 1, and had a known BRAFV600 mutant status or consented to BRAFV600 mutation testing during screening. Patients were stratified by PD-L1 expression and BRAFV600 mutation status and randomly assigned (1:1) through a central interactive voice and integrated web response system to receive epacadostat 100 mg orally twice daily plus pembrolizumab 200 mg intravenously every 3 weeks or placebo plus pembrolizumab for up to 2 years. We used block randomisation with a block size of four in each stratum. Primary endpoints were progression-free survival and overall survival in the intention-to-treat population. The safety analysis population included randomly assigned patients who received at least one dose of study treatment. The study was stopped after the second interim analysis; follow-up for safety is ongoing. This study is registered with ClinicalTrials.gov , number NCT02752074 . Findings Between June 21, 2016, and Aug 7, 2017, 928 patients were screened and 706 patients were randomly assigned to receive epacadostat plus pembrolizumab (n=354) or placebo plus pembrolizumab (n=352). Median follow-up was 12·4 months (IQR 10·3–14·5). No significant differences were found between the treatment groups for progression-free survival (median 4·7 months, 95% CI 2·9–6·8, for epacadostat plus pembrolizumab vs 4·9 months, 2·9–6·8, for placebo plus pembrolizumab; hazard ratio [HR] 1·00, 95% CI 0·83–1·21; one-sided p=0·52) or overall survival (median not reached in either group; epacadostat plus pembrolizumab vs placebo plus pembrolizumab: HR 1·13, 0·86–1·49; one-sided p=0·81). The most common grade 3 or worse treatment-related adverse event was lipase increase, which occurred in 14 (4%) of 353 patients receiving epacadostat plus pembrolizumab and 11 (3%) of 352 patients receiving placebo plus pembrolizumab. Treatment-related serious adverse events were reported in 37 (10%) of 353 patients receiving epacadostat plus pembrolizumab and 32 (9%) of 352 patients receiving placebo plus pembrolizumab. There were no treatment-related deaths in either treatment group. Interpretation Epacadostat 100 mg twice daily plus pembrolizumab did not improve progression-free survival or overall survival compared with placebo plus pembrolizumab in patients with unresectable or metastatic melanoma. The usefulness of IDO1 inhibition as a strategy to enhance anti-PD-1 therapy activity in cancer remains uncertain. Funding Incyte Corporation, in collaboration with Merck Sharp & Dohme.

528 citations

References
More filters
Journal ArticleDOI
TL;DR: Ipilimumab, with or without a gp100 peptide vaccine, as compared with gp100 alone, improved overall survival in patients with previously treated metastatic melanoma.
Abstract: Background An improvement in overall survival among patients with metastatic melanoma has been an elusive goal. In this phase 3 study, ipilimumab — which blocks cytotoxic T-lymphocyte–associated antigen 4 to potentiate an antitumor T-cell response — administered with or without a glycoprotein 100 (gp100) peptide vaccine was compared with gp100 alone in patients with previously treated metastatic melanoma. Methods A total of 676 HLA-A*0201–positive patients with unresectable stage III or IV melanoma, whose disease had progressed while they were receiving therapy for metastatic disease, were randomly assigned, in a 3:1:1 ratio, to receive ipilimumab plus gp100 (403 patients), ipilimumab alone (137), or gp100 alone (136). Ipilimumab, at a dose of 3 mg per kilogram of body weight, was administered with or without gp100 every 3 weeks for up to four treatments (induction). Eligible patients could receive reinduction therapy. The primary end point was overall survival. Results The median overall survival was 10.0 months among patients receiving ipilimumab plus gp100, as compared with 6.4 months among patients receiving gp100 alone (hazard ratio for death, 0.68; P<0.001). The median overall survival with ipilimumab alone was 10.1 months (hazard ratio for death in the comparison with gp100 alone, 0.66; P = 0.003). No difference in overall survival was detected between the ipilimumab groups (hazard ratio with ipilimumab plus gp100, 1.04; P = 0.76). Grade 3 or 4 immune-related adverse events occurred in 10 to 15% of patients treated with ipilimumab and in 3% treated with gp100 alone. There were 14 deaths related to the study drugs (2.1%), and 7 were associated with immune-related adverse events. Conclusions Ipilimumab, with or without a gp100 peptide vaccine, as compared with gp100 alone, improved overall survival in patients with previously treated metastatic melanoma. Adverse events can be severe, long-lasting, or both, but most are reversible with appropriate treatment. (Funded by Medarex and Bristol-Myers Squibb; ClinicalTrials.gov number, NCT00094653.)

13,081 citations

Journal ArticleDOI
TL;DR: This study showed that mismatch-repair status predicted clinical benefit of immune checkpoint blockade with pembrolizumab, and high somatic mutation loads were associated with prolonged progression-free survival.
Abstract: BackgroundSomatic mutations have the potential to encode “non-self” immunogenic antigens. We hypothesized that tumors with a large number of somatic mutations due to mismatch-repair defects may be susceptible to immune checkpoint blockade. MethodsWe conducted a phase 2 study to evaluate the clinical activity of pembrolizumab, an anti–programmed death 1 immune checkpoint inhibitor, in 41 patients with progressive metastatic carcinoma with or without mismatch-repair deficiency. Pembrolizumab was administered intravenously at a dose of 10 mg per kilogram of body weight every 14 days in patients with mismatch repair–deficient colorectal cancers, patients with mismatch repair–proficient colorectal cancers, and patients with mismatch repair–deficient cancers that were not colorectal. The coprimary end points were the immune-related objective response rate and the 20-week immune-related progression-free survival rate. ResultsThe immune-related objective response rate and immune-related progression-free survival ...

6,835 citations

Journal ArticleDOI
TL;DR: Nivolumab was associated with significant improvements in overall survival and progression-free survival, as compared with dacarbazine, among previously untreated patients who had metastatic melanoma without a BRAF mutation.
Abstract: Nivolumab was associated with higher rates of objective response than chemotherapy in a phase 3 study involving patients with ipilimumab-refractory metastatic melanoma. The use of nivolumab in previously untreated patients with advanced melanoma has not been tested in a phase 3 controlled study. METHODS We randomly assigned 418 previously untreated patients who had metastatic melanoma without a BRAF mutation to receive nivolumab (at a dose of 3 mg per kilogram of body weight every 2 weeks and dacarbazine-matched placebo every 3 weeks) or dacarbazine (at a dose of 1000 mg per square meter of body-surface area every 3 weeks and nivolumab-matched placebo every 2 weeks). The primary end point was overall survival. RESULTS At 1 year, the overall rate of survival was 72.9% (95% confidence interval [CI], 65.5 to 78.9) in the nivolumab group, as compared with 42.1% (95% CI, 33.0 to 50.9) in the dacarbazine group (hazard ratio for death, 0.42; 99.79% CI, 0.25 to 0.73; P<0.001). The median progression-free survival was 5.1 months in the nivolumab group versus 2.2 months in the dacarbazine group (hazard ratio for death or progression of disease, 0.43; 95% CI, 0.34 to 0.56; P<0.001). The objective response rate was 40.0% (95% CI, 33.3 to 47.0) in the nivolumab group versus 13.9% (95% CI, 9.5 to 19.4) in the dacarbazine group (odds ratio, 4.06; P<0.001). The survival benefit with nivolumab versus dacarbazine was observed across prespecified subgroups, including subgroups defined by status regarding the programmed death ligand 1 (PD-L1). Common adverse events associated with nivolumab included fatigue, pruritus, and nausea. Drugrelated adverse events of grade 3 or 4 occurred in 11.7% of the patients treated with nivolumab and 17.6% of those treated with dacarbazine. CONCLUSIONS Nivolumab was associated with significant improvements in overall survival and progression-free survival, as compared with dacarbazine, among previously untreated patients who had metastatic melanoma without a BRAF mutation. (Funded by Bristol-Myers Squibb; CheckMate 066 ClinicalTrials.gov number, NCT01721772.)

4,602 citations

Journal ArticleDOI
TL;DR: Ipilimumab (at a dose of 10 mg per kilogram) in combination with dacarbazine, as compared with dACarbazine plus placebo, improved overall survival in patients with previously untreated metastatic melanoma.
Abstract: A B S T R AC T Background Ipilimumab monotherapy (at a dose of 3 mg per kilogram of body weight), as compared with glycoprotein 100, improved overall survival in a phase 3 study involving patients with previously treated metastatic melanoma. We conducted a phase 3 study of ipilimumab (10 mg per kilogram) plus dacarbazine in patients with previously untreated metastatic melanoma. Methods We randomly assigned 502 patients with previously untreated metastatic melanoma, in a 1:1 ratio, to ipilimumab (10 mg per kilogram) plus dacarbazine (850 mg per square meter of body-surface area) or dacarbazine (850 mg per square meter) plus placebo, given at weeks 1, 4, 7, and 10, followed by dacarbazine alone every 3 weeks through week 22. Patients with stable disease or an objective response and no doselimiting toxic effects received ipilimumab or placebo every 12 weeks thereafter as maintenance therapy. The primary end point was overall survival. Results Overall survival was significantly longer in the group receiving ipilimumab plus dacarbazine than in the group receiving dacarbazine plus placebo (11.2 months vs. 9.1 months, with higher survival rates in the ipilimumab–dacarbazine group at 1 year (47.3% vs. 36.3%), 2 years (28.5% vs. 17.9%), and 3 years (20.8% vs. 12.2%) (hazard ratio for death, 0.72; P<0.001). Grade 3 or 4 adverse events occurred in 56.3% of patients treated with ipilimumab plus dacarbazine, as compared with 27.5% treated with dacarbazine and placebo (P<0.001). No drug-related deaths or gastrointestinal perforations occurred in the ipilimumab–dacarbazine group. Conclusions Ipilimumab (at a dose of 10 mg per kilogram) in combination with dacarbazine, as compared with dacarbazine plus placebo, improved overall survival in patients with previously untreated metastatic melanoma. The types of adverse events were consistent with those seen in prior studies of ipilimumab; however, the rates of elevated liver-function values were higher and the rates of gastrointestinal events were lower than expected on the basis of prior studies. (Funded by Bristol-Myers Squibb; ClinicalTrials.gov number, NCT00324155.)

4,069 citations

Journal ArticleDOI
03 Apr 2015-Science
TL;DR: The way forward for this class of novel agents lies in the ability to understand human immune responses in the tumor microenvironment, which will provide valuable information regarding the dynamic nature of the immune response and regulation of additional pathways that will need to be targeted through combination therapies to provide survival benefit for greater numbers of patients.
Abstract: Immune checkpoint therapy, which targets regulatory pathways in T cells to enhance antitumor immune responses, has led to important clinical advances and provided a new weapon against cancer. This therapy has elicited durable clinical responses and, in a fraction of patients, long-term remissions where patients exhibit no clinical signs of cancer for many years. The way forward for this class of novel agents lies in our ability to understand human immune responses in the tumor microenvironment. This will provide valuable information regarding the dynamic nature of the immune response and regulation of additional pathways that will need to be targeted through combination therapies to provide survival benefit for greater numbers of patients.

3,499 citations


"First-in-Human Phase I Study of the..." refers background in this paper

  • ..., PD-1/PD-L1 and CTLA-4) involved in limiting the antitumor potential of the adaptive immune system (32)....

    [...]

Related Papers (5)