scispace - formally typeset
Search or ask a question
Journal ArticleDOI

Ricolinostat, the First Selective Histone Deacetylase 6 Inhibitor, in Combination with Bortezomib and Dexamethasone for Relapsed or Refractory Multiple Myeloma

TL;DR: At the recommended phase II dose of ricolinostat of 160 mg daily, the combination with bortezomib and dexamethasone is safe, well-tolerated, and active, suggesting that selective inhibition of HDAC6 is a promising approach to multiple myeloma therapy.
Abstract: Purpose: Histone deacetylase (HDAC) inhibition improves the efficacy of proteasome inhibition for multiple myeloma but adds substantial toxicity. Preclinical models suggest that the observed synergy is due to the role of HDAC6 in mediating resistance to proteasome inhibition via the aggresome/autophagy pathway of protein degradation.Experimental Design: We conducted a phase I/II trial of the HDAC6-selective inhibitor ricolinostat to define the safety, preliminary efficacy, and recommended phase II dose in combination with standard proteasome inhibitor therapy. Patients with relapsed or refractory multiple myeloma received oral ricolinostat on days 1-5 and 8-12 of each 21-day cycle.Results: Single-agent ricolinostat therapy resulted in neither significant toxicity nor clinical responses. Combination therapy with bortezomib and dexamethasone was well-tolerated during dose escalation but led to dose-limiting diarrhea in an expansion cohort at a ricolinostat dose of 160 mg twice daily. Combination therapy at a ricolinostat dose of 160 mg daily in a second expansion cohort was well tolerated, with less severe hematologic, gastrointestinal, and constitutional toxicities compared with published data on nonselective HDAC inhibitors. The overall response rate in combination with daily ricolinostat at ≥160 mg was 37%. The response rate to combination therapy among bortezomib-refractory patients was 14%. Samples taken during therapy showed dose-dependent increases of acetylated tubulin in peripheral blood lymphocytes.Conclusions: At the recommended phase II dose of ricolinostat of 160 mg daily, the combination with bortezomib and dexamethasone is safe, well-tolerated, and active, suggesting that selective inhibition of HDAC6 is a promising approach to multiple myeloma therapy. Clin Cancer Res; 23(13); 3307-15. ©2017 AACR.

Content maybe subject to copyright    Report

Cancer Therapy: Clinical
Ricolinostat, the First Selective Histone
Deacetylase 6 Inhibitor, in Combination with
Bortezomib and Dexamethasone for Relapsed or
Refractory Multiple Myeloma
Dan T. Vogl
1
, Noopur Raje
2
, Sundar Jagannath
3
, Paul Richardson
4
, Parameswaran Hari
5
,
Robert Orlowski
6
, Jeffrey G. Supko
2
, David Tamang
7
, Min Yang
7
, Simon S. Jones
7
,
Catherine Wheeler
7
, Robert J. Markelewicz
7
, and Sagar Lonial
8
Abstract
Purpose: Histone deacetylase (HDAC) inhibition improves the
efcacy of proteasome inhibition for multiple myeloma but adds
substantial toxicity. Preclinical models suggest that the observed
synergy is due to the role of HDAC6 in mediating resistance to
proteasome inhibition via the aggresome/autophagy pathway of
protein degradation.
Experimental Design: We conducted a phase I/II trial of the
HDAC6-selective inhibitor ricolinostat to dene the safety, pre-
liminary efcacy, and recommended phase II dose in combina-
tion with standard proteasome inhibitor therapy. Patients with
relapsed or refractory multiple myeloma received oral ricolinostat
on days 15 and 812 of each 21-day cycle.
Results: Si ngle-ag ent ricol inostat therapy resulted in neit her
signicant toxicity nor clinical responses. Combinat ion therapy
with bortezomib and dex amethasone w as well- tolera ted duri ng
dose escal ation but led to dose-limiting diarrhea in an expan-
sion cohort at a ricolinostat dose of 16 0 mg twice daily.
Combination t herapy at a ricolinostat dose o f 160 mg daily in
a second expansion co hort was well t oler ated, with les s sever e
hematologic, gastrointestinal, and constitutional toxicities
compared with publ ished data on nonselective HDAC inhibi-
tors. The overall response rate in combination with daily rico-
linostat at 160 mg was 37%. The respo nse rate to combination
therapy among bo rtezomib -refract ory p atients was 14%. S am-
ples taken during therapy showed dose-dependent i ncreases of
acetylated tubulin in peri pheral blood lymphocytes .
Conclusions: At the recommended phase II dose of ricolinostat
of 160 mg daily, the combination with bortezomib and dexa-
methasone is safe, well-tolerated, and active, suggesting that
selective inhibition of HDAC6 is a promising approach to mul-
tiple myeloma therapy.
Clin Cancer Res; 23(13); 330715. 2017
AACR.
Introduction
Multiple myeloma is an incurable plasma cell malignancy with
a unique biology characterized by high levels of protein synthesis
and consequent endoplasmic reticulum (ER) stress and activation
of the unfolded protein response (UPR). Plasma cell differenti-
ation and survival depend on UPR activation, which results in
upregulation of protein degradation by the 26S proteasome. The
introduction of proteasome inhibitors into the multiple myeloma
therapeutic armamentarium has led to a dramatic improvement
in clinical outcomes (15). However, despite these advances,
multiple myeloma cells inevitably develop resistance to protea-
some inhibition, leading to disease progression.
The aggresome/autophagy pathway is a regulated degradative
process for cellular proteins (6) that is activated in response to
accumulation of cytosolic polyubiquitinated proteins in the set-
ting of proteasome inhibition, serving as an alternative route
for protein degradation (7) and thereby contributing to thera-
peutic resistance to proteasome inhibitor therapy. Histone dea-
cetylase 6 (HDAC6) is a cytosolic microtubule-associated deace-
tylase that mediates trafcking of ubiquitinated misfolded pro-
teins to the aggresome/autophagy pathway (8). Selective inhibi-
tion of HDAC6 increases a-tubulin acetylation and accumulation
of ubiquitinated proteins in multiple myeloma cells, with syner-
gistic cytotoxicity in combination with bortezomib (9). Clinical
trials with nonselective HDAC inhibitors in combination with
bortezomib and dexamethasone have shown improved outcomes
but also substantially increased toxicity (10, 11). The unique role
of HDAC6 in the aggresome/autophagy pathway raises the pos-
sibility that selective inhibition of HDAC6 may yield improved
efcacy and reduced toxicity when combined with proteasome
inhibition.
Ricolinostat (ACY-1215) is an orally available selective
HDAC6 inhibitor, with p reclinical data showing anti-myeloma
efcacy in combination with proteasome inhibitors, mediated
by inhibition of aut ophagic protein degradat ion an d increased
ER stress. (12, 13). We th erefore conducted a rst-in-human
dose-escalation study of ricolinostat as a single a gent and then
1
Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylva-
nia.
2
Massachusetts General Hospital, Boston, Massachusetts.
3
Mount Sinai
Medical Center, New York, New York.
4
Dana Farber Cancer Institute, Boston,
Massachusetts.
5
Medical College of Wisconsin, Milwaukee, Wisconsin.
6
MD
Anderson Cancer Center, Houston, Texas.
7
Acetylon Pharmaceuticals Inc.,
Boston, Massachusetts.
8
Emory University, Atlanta, Georgia.
Corresponding Author: Dan T. Vogl, Perelman Center for Advanced Medicine
South Tower 12-176, 3400 Civic Center Boulevard, Philadelphia, PA 19104.
Phone: 215-615-6508; Fax: 215-615-5887; E-mail: dan.vogl@uphs.upenn.edu
doi: 10.1158/1078-0432.CCR-16-2526
2017 American Associati on for Cancer Research.
Clinical
Cancer
Research
www.aacrjournals.org
3307
Downloaded from http://aacrjournals.org/clincancerres/article-pdf/23/13/3307/2299894/3307.pdf by guest on 26 August 2022

in combination with bortezomib and dexamethasone in
patients with relapsed or re fractory multiple myelom a. We
aimedtodene the dose-limiting toxicities (DLT), maximu m
tolerated dose (MTD), pharmacokinetics and pharmacodynam-
ics o f ricolinostat alone and in c ombina tion with bortezomib
and dexamethasone and t o dene the response rate and toxicity
prole of the combination regimen.
Materials and Methods
Study design
This study was designed as a 3-part, phase I/II, single-arm,
multicenter, open-label study in patients with relapsed or refrac-
tory multiple myeloma. Parts 1 and 2 of the study employed a
sequential group dose-escalation design of ricolinostat as mono-
therapy (part 1) and in combination with bortezomib and dexa-
methasone (part 2), with planned enrollment of up to 20 patients
in an expansion cohort at the MTD. Part 3 was intended to be a
Simon optimal 2-stage phase II trial at the MTD; however, on
the basis of the preliminary results of the part 2 expansion cohort,
we did not proceed with a formal phase II cohort and instead
enrolled an additional expansion cohort to explore a daily dose of
ricolinostat.
Population
Patients were eligible for enroll ment if they had mult iple
myeloma that was relapsed (progressed after the most recent
therapy) or refractory (progressed on or within 60 days after
completion of the most recent therapy ) after at least 2 prio r
lines of therapy. Patients had to have received a proteasome
inhibitor, an immunomodulatory drug, and an autologous
stem cell transplant as part of their prior therapy, unless they
were considered not to b e a candidate for these therapies by
their t reating physician. At enrollment, patients had to have
measurable disease parameters according to the International
Myeloma Working Group (IMWG) criteria (14). Patients wer e
at le ast 18 years old and had a Karnof sky Performance Sta tus of
70, adequate bone marrow reserve [absolute neutrophil
count 1.010
9
/L and platelet count 7510
9
/L (50
10
9
/L in patients in whom 50% of bone marrow nucleated cells
were plasma cells), calculated creatinine clearance 30 mL/min,
adequate hepa tic function (serum bilirubin < 2.0 mg/dL, serum
alanine transaminase (ALT ) and aspartate trans aminase (AST)
<3 t imes the upper li mit of normal (ULN)], and corrected
serum calcium ULN. Patients could not have received radio-
therapy or systemic anticancer therapy within 2 weeks of
starting therapy, aut ologou s stem cell transplant within 12
weeks, or any al logenei c stem cell transplant or HDAC inhib-
itor. Patient s were excluded i f they had gr ade 2 or higher
neuropathy, an active systemic infection, other active malig-
nancies, known human immunodecie ncy virus, active hepa-
titis B viru s, or a ctiv e hepat itis C virus infection. Patients wer e
also excluded if th ey had New York He art Associat ion Class 3 o r
4 congestive heart failure, unstable angina, cardiac arrhythmia,
QTcF > 480 msec, myoca rdial infarction or stroke within 6
months, or sev ere hypertension, diabetes mellit us, or chronic
obstructive pulmonary disease. All patients provided written
informed consent prior to study participation.
Study treatment
Ricolinostat was administered as an oral liquid formulation
on days 15and812 of ea ch 21-da y cycle. Ricolinostat was
initially prepared as a 20 mg /mL solu tion; mid- way d uring the
study a 12 mg/mL solut ion was introduced with improved
stability at room temperat ure. Prespecied ricolinostat doses
during single-agent d ose escalation (part 1) were 40, 8 0, 160,
240, and 360 mg daily in s uccessive cohorts. In the combina-
tion cohorts (part 2), patients received b ortezomi b immedi-
ately after ricolinostat on days 1, 4, 8, and 11, with dexameth-
asone 20 mg administered orally 30 minutes after ricol inostat
on days 1, 2, 4, 5, 8, 9, 11, and 12. During dose escalatio n,
bortezomib was administered intravenously; when data
became available reg arding su bcutaneo us admini strati on of
bortezomib, we permitted switching to subcutaneous dosing
after compl eti on of the rst 3-week cycle. In cohort 4 (d ose
expansion at ricolinosta t 160 mg daily), pa tients could start
therapy with either intravenous or subcutaneou s bortezomib
administration, at the investigator's discretion. Prespecied
dose levels for the combination therapy dose-escalatio n
cohorts are shown in Table 1, with ricolino stat doses r anging
from 40 mg daily (q.d.) to 160 mg twice daily (b. i.d.) and
bortezomib ad ministered at 1.0 mg/m
2
in the rst combined
dose cohort and the standard 1.3 mg/m
2
subsequently.
Endpoints
The primary endpoints in the dose-escalation cohorts were
identication of DLTs and the MTD. Dose escalation occurred
usingastandard3þ 3 design, with enrollment of 3 patients to
each dose cohort, enrollment ofasecondgroupof3patientsif
1oftherst 3 experienced an DLT, and enrollment to the next
dose level permitted if fewer than 1 o f 3 or 2 of 6 pa tients at a
given dose level e xperienced a DLT. Toxicities were assessed
using t he National Cancer Institute Common Terminology
Criteria fo r Adverse Events versio n 4.0. DLT was dened as
one of the following events occurring du ring the rst cycle of
therapy and considered t o be rela ted to ricolinostat: grade 4
neutropenia lasting >5 days; febrile n eutrope nia; grade 4 nau-
sea, vomiting, or diarrhea; grad e 3 nausea or vomiting persi st-
ing >72 hours; grade 3 diarrhea persisting >48 ho urs; or any
Translational Rele vance
Ricolinostat is the rst isoform-selective histone deacetylase
(HDAC) inhibitor in human clinical trials, an approach to
drug development that utilizes rational targeting to achieve an
improved therapeutic index. Our data show that the combi-
nation of HDAC6-selective inhibition using ricolinostat with
the proteasome inhibitor bortezomib overcomes bortezomib
resistance in relapsed multiple myeloma, with a favorable
safety prole that offers potential advantages compared with
nonselective HDAC inhibition. Our ndings validate preclin-
ical data showing that HDAC6-mediated trafcking of ubi-
quitinated proteins to the aggresome/autophagy pathway is a
relevant alternative mechanism of protein degradation for
cells exposed to proteasome inhibition and therefore a mech-
anism of therapeutic resistance to proteasome inhibition.
Incorporation of HDAC6-selective inhibition into the anti-
myeloma armamentarium therefore offers the prospect of
improved myeloma outcomes.
Vogl et al.
Clin Cancer Res; 23(13) July 1, 2017 Clinical Cancer Research3308
Downloaded from http://aacrjournals.org/clincancerres/article-pdf/23/13/3307/2299894/3307.pdf by guest on 26 August 2022

other grade 3 nonhematologic tox icity, with the ex ception of
hyperglycemia in diabetic patients. Thrombocytopenia was
considered a DLT for p atients r eceiving single-agent ricolinostat
if it was grade 3 with grade 2 bleeding or grade 4 and for
patients receiving comb ination t herapy if it wa s grade 4 on 2
separate occasi ons unre sponsiv e to transfusion support. The
MTD w as dened as the dose immediately below the dose level
at which 2 of up to 6 pa tients expe rienced a DLT.
Responses were assessed according to standard IMWG crite-
ria (15). The overall response rate (ORR) was dened as the
sumoftheratesofstringentcompleteresponse(sCR),com-
plete response (CR), very good parti al response, and part ial
response (PR). The clinica l benetrate(CBR)wasdened as the
sumoftheORRandtherateofminimalresponse.Response
assessments (serum and urine) were conducted on day 15 of
each cycl e, with a bone marrow aspirate and biopsy requi red to
conrm CR.
Patients receiving bortezomib completed a neurotoxicity-
directed questionnaire from the Functional Assessment of Can-
cer/Gynecology Oncology Group (FACT/GOG) survey of neuro-
toxicity (16, 17).
Pharmacokinetic studies
Serial blood samples for pharmacokinetic assessments were
collected during the rst cycle of therapy. In part 1, samples were
collected before and at 0.25, 0.5, 1, 2, 4, and 24 hours after the rst
dose of ricolinostat, on days 4 and 8 before, 0.25, and 1 hour after
the morning ricolinostat dose, on day 11 before and at 0.25, 0.5, 1,
2, and 4 hours after ricolinostat, and on day 15. In part 2, samples
were collected at the same time intervals after the administration
of ricolinostat and bortezomib, with additional samples obtain-
ed 6 hours after dosing on days 1 and 11. Blood (6 mL) was drawn
from a peripheral vein into plastic tubes containing freeze-dried
sodium heparin, mixed by inversion, and placed over ice until
centrifuged (1,300 g, 10 minutes, 4
C). Plasma was removed
and stored at 80
C until assayed. The concentrations of ricoli-
nostat and bortezomib were concurrently determined using an
analytical method involving high-performance liquid chroma-
tography with tandem mass spectrometric detection. The assay
was validated and applied to the analysis of study samples as
recommended by the FDA Guidance for Industry: Bioanalytical
Method Validation, May 2001 (https://www.fda.gov/Drugs/
GuidancecomplianceRegulatoryInformation/Guidances/default.
htm). The lower limit of quantitation for both compounds was
0.50 ng/mL. Pharmacokinetic parameters were estimated by
analysis of the ricolinostat and bortezomib plasma concentra-
tiontime curves for individual patients by standard noncompart-
mental methods using WinNonlin Professional 5.0 software
(Pharsight Corp.) and are reported as the geometric mean SD
at each dose level.
Pharmacodynamic studies
Blood samples for pharmacodynamic assessment of acetylated
tubulin and acetylated histone levels were collected at baseline
and (in part 1) 1 and 4 hours after the rst ricolinostat dose or (in
part 2) 0.25, 0.5, 1, 2, 4, 6, and 24 hours after the rst bortezomib
dose. Blood samples were cryopreserved in equal volume of
freezing solution (10% DMSO in PBS v/v) and stored at 80
C.
Upon thaw, samples were xed with 3.8% formaldehyde (2:1
formaldehyde/sample v/v) and stained with primary antibodies
for anti-tubulin (Sigma-Aldrich, #T7451) and anti-histone
H2BK5 (Cell Signaling Technologies, #2574) or isotype controls
(Rabbit IgG polyclonal Control antibody, Cell Signaling Tech-
nologies, #2729; mouse IgG2b, clone MOPC-141,Sigma-Aldrich,
#M5534). Samples were labeled with secondary DyLight488
antibodies (DyLight 488 goat anti-Mu-IgG, KPL #072-03-18-
06; DyLight 488 goat anti-Rb-IgG, KPL #072-03-15-16). The target
population was labeled with anti-CD3 (PE-Cy5 mouse anti-
human CD3, clone UCHT1, eBioscience # 15-0038). Data anal-
ysis was performed by gating on the CD3
þ
cells in FlowJo software
(Treestar, Inc.) and applying a mean uorescent intensity (MFI)
statistic. Fold change of tubulin or histone acetylation was cal-
culated by subtracting the control IgG MFI from the sample MFI
and dividing the MFI of treated groups by the MFI of the control
[Fold change ¼ (Postdose Acetyl-MFI Postdose IgG-MFI)/(Pre-
dose Acetyl-MFI Predose IgG-MFI)]. Horizontal bars represent
arithmetic means. Error bars represent the SEM.
Statistical analysis
The size of the dose-escalation cohort was based on the con-
ventional 3 þ 3 dose-escalation design for phase I trials; conse-
quently, a formal sample size estimation was not performed. All
patients who received at least one dose of ricolinostat were
included in the toxicity analysis. Patients who discontinued
the study before the rst response evaluation at the beginning
of cycle 2 were considered unevaluable for response. Data were
summarized using descriptive statistics. Time-to-event analysis,
including progression-free survival, was estimated using the
KaplanMeier method. The MFI of pharmacodynamic samples
used to calculate the fold change were assessed for statistical
difference by comparing the pre-dose signal to the 1-hour post-
dose signal of a given group using a paired, 2-tailed t test. Groups
where P 0.05 are indicated with an "
". Groups where P > 0.05
are indicated by "ns" (not signicant). Statistical analyses were
Table 1. Phase Ib dose-escalation schema
Phase Ib dose-escalation schema (21-d cycle)
Phase Ib cohorts n
Ricolinostat
(Days 15, 812)
Bortezomib
(Cycles 15: Days 1, 4, 8, 11
Cycles 6þ: Days 1, 8)
Dexamethasone
(Cycles 15: Days 1, 2, 4, 5, 8, 9, 11, 12
Cycles 6þ: Days 1, 2, 8, 9)
Cohort 1 7 40 mg q.d. 1.0 mg/m
2
20 mg
Cohort 2 3 40 mg q.d. 1.3 mg/m
2
20 mg
Cohort 3 3 80 mg q.d. 1.3 mg/m
2
20 mg
Cohort 4 3 160 mg q.d. 1.3 mg/m
2
20 mg
Cohort 5 3 240 mg q.d. 1.3 mg/m
2
20 mg
Cohort 6 3 160 mg b.i.d. 1.3 mg/m
2
20 mg
Cohort 6
0
expansion 21 160 mg b.i.d. 1.3 mg/m
2
20 mg
Cohort 4
0
expansion 14 160 mg q.d. 1.3 mg/m
2
20 mg
Ricolinostat, Bortezomib, and Dexamethasone for Myeloma
www.aacrjournals.org Clin Cancer Res; 23(13) July 1, 2017 3309
Downloaded from http://aacrjournals.org/clincancerres/article-pdf/23/13/3307/2299894/3307.pdf by guest on 26 August 2022

performed using SAS version 9.3 (SAS Institute, Inc.) and R
version 3.2.3 (R Foundation).
Results
Population
We enrolled 15 patients to 5 single-agent ricolinostat co horts
between August 2011 a nd Sept ember 2012 and 57 pa tients to
9 combina tion dose cohorts b etween May 2012 and September
2015. Pa tien t characteristics in the single-agent ricolinostat
cohorts (Table 2) included a median age o f 70 years and
median number of prior regimens of 4 (range, 211). Patient
characteristics in the combination dose cohorts (Table 2)
included a median age of 65 ye ars and median number of
prior regimens of 5 ( range, 212) . All p atients had received
prior bortezomib, a nd 33 % in the sin gle-ag ent ricol inostat
cohorts and 63% in the co mbinatio n dose cohorts were refrac-
tory to bortezomib, respectively.
Single-agent ricolinostat
DLTs and adverse events. We did not observe any DLTs during
single-agent dose escalation and therefore did not identify an
MTD. We did not explore doses higher than 360 mg q.d. because
pharmacokinetic analysis showed evidence of a plateau in expo-
sure at dose levels 160 mg, suggestive of saturable absorption of
the drug (see below).
The most common advers e events o bserved during therapy
(many attributed to dis ease or intercurrent illness) were renal
insufciency (33%), fatigue (27%), anemia (20%), and diar-
rhea (20%). Diarrhea occurred only at ricolinostat dose s 160
mg q.d. The only grade 3 or 4 adverse events assessed by the
investigator as possibly related to ricolinostat were seen at
doses 160 mg q.d. and were all hema tolog ic abnorma lities ,
including anemia (at 160 mg q.d.) and neutropenia and leu-
kopenia (at 360 mg q.d.). Two patients experienced a serious
adverse ev ent (SAE), including grade 5 (fatal ) cardiac ar rest at
40 mg q.d. (which occurred 27 days after study treatment
was discontinued for disease progression) and an exacerbation
of chronic pulmonary disease at 16 0 mg q.d.; neit her was
considered to be rela ted to ricolinostat. No patient d iscontin-
ued single-agent ricolinostat because of a treatment-emergent
adverse event. Serial triplicate electro cardiograms show ed no
evidence of QT interval prolongation.
Response. Of the 15 patients treated with single-agent ricolinostat,
6 had stable disease for a median of 11 weeks (range, 530), and
no patient had a minor response or PR (Fig. 1).
Combination dosing with ricolinostat, bortezomib, and
dexamethasone
DLTs and adverse events. The rst combination cohort was expand-
ed to 6 patients due to an asymptomatic increase in amylase. No
other DLTs were observed during dose escalation. Toxicities
observed during dose escalation (Table 3) were primarily low-
grade gastrointestinal toxicities, cytopenias, and fatigue. On the
basis of the combined pharmacokinetic and pharmacodynamic
data from the monotherapy and combination dose-escalation
cohorts, we enrolled an expansion cohort at the top dose of
ricolinostat (160 mg b.i.d.).
Of the 21 patients treated on the 160 mg b.i.d. expansion
cohort, 29% were hospitalized for reasons potentially related to
diarrhea and dehydration. Because of a concern that this repre-
sented excess toxicity with twice daily dosing, we enrolled a
second expansion cohort at a lower dose of ricolinostat (160
mg q.d.) to better understand the relationship between dose and
both toxicity and efcacy. Among the 24 patients who received
ricolinostat 160 mg b.i.d. (3 patients treated in the dose-escalation
cohort and 21 patients in the dose expansion cohort), the most
common treatment-emergent adverse events were thrombocyto-
penia (71%), diarrhea (67%), anemia (42%), fatigue (42%),
nausea (38%), hypokalemia (33%), vomiting (29%), peripheral
Table 2. Patient demographics
Characteristics Single-agent ricolinostat
Ricolinostat with
bortezomib and
dexamethasone
Combination therapy with
160 mg q.d. ricolinostat dosing
(cohorts 4, 4
0
,5)
Combination therapy with
160 mg b.i.d. ricolinostat dosing
(cohorts 6, 6
0
)
Patients enrolled N ¼ 15 N ¼ 57 N ¼ 20 N ¼ 24
Age, y
Median (range) 70 (5179) 65 (4784) 65 (4783) 67 (4884)
75 5 (33) 14 (25) 5 (25) 8 (33)
6574 7 (47) 15 (26) 5 (25) 6 (25)
64 3 (20) 28 (49) 10 (50) 10 (42)
Sex
Male 10 (67) 35 (61) 12 (60) 15 (63)
Female 5 (33) 22 (39) 8 (40) 9 (38)
Race
White 8 (53) 39 (68) 16 (80) 14 (58)
Black or African American 6 (40) 13 (23) 4 (20) 6 (25)
Other 0 (0) 3 (5) 0 (0) 3 (13)
Asian 1 (7) 2 (4) 0 (0) 1 (4)
Prior regimens
Median (range) 4 (211) 5 (213) 5 (29) 7 (313)
Refractory to
Lenalidomide 7 (47) 38 (67) 13 (65) 17 (71)
Bortezomib 5 (33) 36 (63) 13 (65) 15 (63)
Pomalidomide 0 (0) 18 (32) 5 (25) 12 (50)
Cyclophosphamide 1 (7) 19 (33) 5 (25) 10 (42)
Carlzomib 0 (0) 17 (30) 4 (20) 12 (50)
Thalidomide 0 (0) 12(21) 2 (10) 7 (29)
Vogl et al.
Clin Cancer Res; 23(13) July 1, 2017 Clinical Cancer Research3310
Downloaded from http://aacrjournals.org/clincancerres/article-pdf/23/13/3307/2299894/3307.pdf by guest on 26 August 2022

neuropathy (29%), hyperglycemia (25%), and renal insufciency
(21%; Table 3). Because of similar pharmacokinetic exposure at
doses of 240 mg daily and 160 mg daily (see below), we analyzed
patients treated at these 2 doses as a single cohort. Of the 20
patients receiving once daily ricolinostat at a dose of 160 mg or
greater (including 3 patients each treated at 160 mg q.d. and 240
mg q.d. during dose escalation and an additional 14 patients
treated at 160 mg q.d. during dose expansion), the most common
treatment-emergent adverse events were thrombocytopenia
(40%), anemia (35%), diarrhea (30%), hypertension (25%),
fatigue (25%), hyperglycemia (25%), renal insufciency
(25%), nausea (25%), hypophosphatemia (20%), and hypona-
tremia (20%; Table 3). Similar to the single-agent cohort, we did
not observe any signicant prolongation of the QT interval with
combination dosing.
Because the toxicity of bortezomib is greater with intravenous
dosing than with subcutaneous dosing (18), we analyzed toxicity
rates by route of bortezomib administration. Of the 20 subjects
treated with doses of ricolinostat 160 mg q.d. in combination
with bortezomib and dexamethasone, 10 received at least one full
Figure 1.
Treatment responses. A, Responses for the entire cohort of patients treated with (from left to right) single-agent ricolinostat, ricolinostat with bortezomib (Bz) and
dexamethasone (dex), ricolinostat 160 mg q.d. with Bz/dex, and ricolinostat 160 mg b.i.d. with Bz/dex. B, Responses among patients with bortezomib-
refractory myeloma treated with (from left to right) ricolinostat with Bz/dex, ricolinostat 160 mg q.d. with Bz/d ex, or ricolinostat 160 mg b.i.d. with Bz/dex. Numbers
indicate the percentage of patients in each category of response. MR, minimal response; PD, progressive disease; SD, stable disease; VGPR, very good partial
response.
Table 3. Treatment-emergent adverse events (in order of frequency of grade 3/4 events in patients receiving combined therapy with ricolinostat, bortezomib,and
dexamethasone)
Single-agent ricolinostat
Ricolinostat with
bortezomib and dexameth asone
Ricolinostat 160
mg q.d. with
bortezomib and
dexamethasone
(cohorts 4, 4
0
,5)
Ricolinostat 160 mg
b.i.d. with
bortezomib and
dexamethasone
(cohorts 6, 6
0
)
N ¼ 15 (%) N ¼ 57 (%) N ¼ 20 (%) N ¼ 24 (%)
Adverse event All grades Grade 3/4 All grades Grade 3/4 All grades Grade 3/4 All grades Grade 3/4
Thrombocytopenia 2 (13) 2 (13) 31 (54) 21 (37) 8 (40) 4 (20) 17 (71) 13 (54)
Anemia 3 (20) 1 (7) 23 (40) 11 (19) 7 (35) 4 (20) 10 (42) 5 (21)
Amylase elevation 0 (0) 0 (0) 8 (14) 6 (11) 1 (5) 1 (5) 3 (13) 1 (4)
Hypertension 0 (0) 0 (0) 8 (14) 6 (11) 5 (25) 4 (20) 2 (8) 1 (4)
Hypophosphatemia 2 (13) 1 (7) 10 (18) 5 (9) 4 (20) 2 (10) 2 (8) 1 (4)
Fatigue 4 (27) 0 (0) 20 (35) 4 (7) 5 (25) 1 (5) 10 (42) 2 (8)
Hypokalemia 1 (7) 0 (0) 15 (26) 4 (7) 3 (15) 1 (5) 8 (33) 2 (8)
Hyperglycemia 0 (0) 0 (0) 13 (23) 4 (7) 5 (25) 1 (5) 6 (25) 2 (8)
Pneumonia 0 (0) 0 (0) 6 (11) 4 (7) 0 (0) 0 (0) 4 (17) 3 (13)
Diarrhea 3 (20) 0 (0) 25 (44) 3 (5) 6 (30) 1 (5) 16 (67) 2 (8)
Peripheral neuropathy 0 (0) 0 (0) 15 (26) 3 (5) 3 (15) 0 (0) 7 (29) 1 (4)
Hyponatremia 1 (7) 0 (0) 9 (16) 3 (5) 4 (20) 1 (5) 3 (13) 1 (4)
Transaminase elevation 1 (7) 0 (0) 9 (16) 3 (5) 3 (15) 2 (10) 2 (8) 0 (0)
Febrile neutropenia 0 (0) 0 (0) 3 (5) 3 (5) 1 (5) 1 (5) 0 (0) 0 (0)
Blood creatinine elevation 5 (33) 0 (0) 16 (28) 2 (4) 5 (25) 2 (10) 5 (21) 0 (0)
Neutropenia 2 (13) 1 (7) 7 (12) 2 (4) 3 (15) 2 (10) 2 (8) 0 (0)
Bronchitis 0 (0) 0 (0) 2 (4) 2 (4) 1 (5) 1 (5) 1 (4) 1 (4)
Pulmonary embolism 0 (0) 0 (0) 2 (4) 2 (4) 0 (0) 0 (0) 2 (8) 2 (8)
Inuenza 0 (0) 0 (0) 2 (4) 2 (4) 0 (0) 0 (0) 1 (4) 1 (4)
Nausea 2 (13) 0 (0) 16 (28) 1 (2) 5 (25) 0 (0) 9 (38) 1 (4)
Vomiting 0 (0) 0 (0) 8 (14) 1 (2) 0 (0) 0 (0) 7 (29) 1 (4)
Dyspnea 1 (7) 0 (0) 8 (14) 1 (2) 2 (10) 0 (0) 3 (13) 0 (0)
Ricolinostat, Bortezomib, and Dexamethasone for Myeloma
www.aacrjournals.org Clin Cancer Res; 23(13) July 1, 2017 3311
Downloaded from http://aacrjournals.org/clincancerres/article-pdf/23/13/3307/2299894/3307.pdf by guest on 26 August 2022

Citations
More filters
Journal ArticleDOI
TL;DR: It is suggested that stringent genetic validation of the mechanism of action of cancer drugs in the preclinical setting may decrease the number of therapies tested in human patients that fail to provide any clinical benefit.
Abstract: Ninety-seven percent of drug-indication pairs that are tested in clinical trials in oncology never advance to receive U.S. Food and Drug Administration approval. While lack of efficacy and dose-limiting toxicities are the most common causes of trial failure, the reason(s) why so many new drugs encounter these problems is not well understood. Using CRISPR-Cas9 mutagenesis, we investigated a set of cancer drugs and drug targets in various stages of clinical testing. We show that-contrary to previous reports obtained predominantly with RNA interference and small-molecule inhibitors-the proteins ostensibly targeted by these drugs are nonessential for cancer cell proliferation. Moreover, the efficacy of each drug that we tested was unaffected by the loss of its putative target, indicating that these compounds kill cells via off-target effects. By applying a genetic target-deconvolution strategy, we found that the mischaracterized anticancer agent OTS964 is actually a potent inhibitor of the cyclin-dependent kinase CDK11 and that multiple cancer types are addicted to CDK11 expression. We suggest that stringent genetic validation of the mechanism of action of cancer drugs in the preclinical setting may decrease the number of therapies tested in human patients that fail to provide any clinical benefit.

382 citations

Journal ArticleDOI
TL;DR: This perspective reviews the biological and medicinal chemistry advances over the last three decades with an emphasis on the design of selective inhibitors that discriminate between the eleven human HDAC isoforms.
Abstract: It is now 30 years since the first report of a potent zinc-dependent histone deacetylase (HDAC) inhibitor appeared Since then, five HDAC inhibitors have received regulatory approval for cancer chemotherapy while many others are in clinical development for oncology as well as other therapeutic indications This Perspective reviews the biological and medicinal chemistry advances over the past 3 decades with an emphasis on the design of selective inhibitors that discriminate between the 11 human HDAC isoforms

279 citations

Journal ArticleDOI
01 Feb 2018-Leukemia
TL;DR: Clinical trial data of next generation proteasome inhibitors, next-generation immunomodulatory agent, and monoclonal antibodies approved for relapsed and refractory multiple myeloma are summarized, cautious interpretation of RCTs highlighted, and algorithm for salvage treatment of relapse/refractory MM proposed.
Abstract: Despite enormous advances, management of multiple myeloma (MM) remains challenging. Multiple factors impact the decision to treat or which regimen to use at MM relapse/progression. Recent major randomized controlled trials (RCTs) showed widely varying progression-free survivals (PFS), ranging from a median of 4 months (MM-003) to 23.6 months (ASPIRE). Based on these RCTs, next-generation proteasome inhibitors (carfilzomib and ixazomib), next-generation immunomodulatory agent (pomalidomide), and monoclonal antibodies (elotuzumab and daratumumab) were approved for relapsed and refractory MM. Daratumumab, targeting CD38, has multiple mechanisms of action including modulation of the immunosuppressive bone marrow micro-environment. In addition to the remarkable single agent activity in refractory MM, daratumumab produced deep responses and superior PFS in MM when combined with lenalidomide/dexamethasone, or bortezomib/dexamethasone. Other anti-CD38 antibodies, such as isatuximab and MOR202, are undergoing assessment. Elotuzumab, targeting SLAMF7, yielded superior response rates and PFS when combined with lenalidomide/dexamethasone. New combinations of these next generation novel agents and/or antibodies are undergoing clinical trials. Venetoclax, an oral BH3 mimetic inhibiting BCL2, showed single agent activity in MM with t(11;14), and is being studied in combination with bortezomib/dexamethasone. Selinexor, an Exportin-1 inhibitor, yielded promising results in quad- or penta-refractory MM including patients resistant to daratumumab. Pembrolizumab, an anti-PD1 check-point inhibitor, is being tested in combination with lenalidomide/dexamethasone or pomalidomide/dexamethasone. Chimeric antigen receptor-T cells targeting B-cell maturation antigen have yielded deep responses in RRMM. Finally, salvage autologous stem cell transplantation (ASCT) remains an important treatment in MM relapsing/progressing after a first ASCT. Herein, the clinical trial data of these agents are summarized, cautious interpretation of RCTs highlighted, and algorithm for salvage treatment of relapse/refractory MM proposed.

224 citations

Journal ArticleDOI
TL;DR: Proteasome inhibitor-based combination regimens have become established as a cornerstone of therapy throughout the myeloma treatment algorithm, incorporating agents from the other key classes of antimyeloma agents, including the immunomodulatory drugs, monoclonal antibodies, and histone deacetylase inhibitors.
Abstract: Proteasome inhibitors are one of the most important classes of agents to have emerged for the treatment of multiple myeloma in the past two decades, and now form one of the backbones of treatment. Three agents in this class have been approved by the United States Food and Drug Administration-the first-in-class compound bortezomib, the second-generation agent carfilzomib, and the first oral proteasome inhibitor, ixazomib. The success of this class of agents is due to the exquisite sensitivity of myeloma cells to the inhibition of the 26S proteasome, which plays a critical role in the pathogenesis and proliferation of the disease. Proteasome inhibition results in multiple downstream effects, including the inhibition of NF-κB signaling, the accumulation of misfolded and unfolded proteins, resulting in endoplasmic reticulum stress and leading to the unfolded protein response, the downregulation of growth factor receptors, suppression of adhesion molecule expression, and inhibition of angiogenesis; resistance to proteasome inhibition may arise through cellular responses mediating these downstream effects. These multiple biologic consequences of proteasome inhibition result in synergistic or additive activity with other chemotherapeutic and targeted agents for myeloma, and proteasome inhibitor-based combination regimens have become established as a cornerstone of therapy throughout the myeloma treatment algorithm, incorporating agents from the other key classes of antimyeloma agents, including the immunomodulatory drugs, monoclonal antibodies, and histone deacetylase inhibitors. This review gives an overview of the critical role of the proteasome in myeloma and the characteristics of the different proteasome inhibitors and provides a comprehensive summary of key clinical efficacy and safety data with the currently approved proteasome inhibitors.

207 citations

Journal ArticleDOI
26 Jul 2018-Cell
TL;DR: This study provides a concept in which a coordinated physico-chemical mechanism promotes platelet biogenesis and an innovative strategy for ex vivo platelet manufacturing is provided.

206 citations


Cites methods from "Ricolinostat, the First Selective H..."

  • ...A recent clinical trial for myeloma using a combination of chemotherapy and HDAC6 inhibitor Ricolinostat found a correlation between thrombocytopenia and the dosage of Ricolinostat independent of chemotherapy dose (Vogl et al., 2017), suggesting HDAC6 is an indispensable factor in human platelet biogenesis....

    [...]

References
More filters
Journal ArticleDOI
28 Feb 2008-Nature
TL;DR: Understanding autophagy may ultimately allow scientists and clinicians to harness this process for the purpose of improving human health, and to play a role in cell death.
Abstract: Autophagy, or cellular self-digestion, is a cellular pathway involved in protein and organelle degradation, with an astonishing number of connections to human disease and physiology. For example, autophagic dysfunction is associated with cancer, neurodegeneration, microbial infection and ageing. Paradoxically, although autophagy is primarily a protective process for the cell, it can also play a role in cell death. Understanding autophagy may ultimately allow scientists and clinicians to harness this process for the purpose of improving human health.

5,831 citations


"Ricolinostat, the First Selective H..." refers background in this paper

  • ...The aggresome/autophagy pathway is a regulated degradative process for cellular proteins (6) that is activated in response to accumulation of cytosolic polyubiquitinated proteins in the setting of proteasome inhibition, serving as an alternative route for protein degradation (7) and thereby contributing to therapeutic resistance to proteasome inhibitor therapy....

    [...]

  • ...When proteasomal degradation of misfolded and unneeded proteins is inhibited, those proteins are shuttled along the cytoskeleton in an HDAC6-dependent manner to perinuclear aggresomes, which subsequently fuse with degradative autophagic lysosomes (6, 7)....

    [...]

Journal ArticleDOI
TL;DR: The FACT-G meets or exceeds all requirements for use in oncology clinical trials, including ease of administration, brevity, reliability, validity, and responsiveness to clinical change.
Abstract: PURPOSEWe developed and validated a brief, yet sensitive, 33-item general cancer quality-of-life (QL) measure for evaluating patients receiving cancer treatment, called the Functional Assessment of Cancer Therapy (FACT) scale.METHODS AND RESULTSThe five-phase validation process involved 854 patients with cancer and 15 oncology specialists. The initial pool of 370 overlapping items for breast, lung, and colorectal cancer was generated by open-ended interview with patients experienced with the symptoms of cancer and oncology professionals. Using preselected criteria, items were reduced to a 38-item general version. Factor and scaling analyses of these 38 items on 545 patients with mixed cancer diagnoses resulted in the 28-item FACT-general (FACT-G, version 2). In addition to a total score, this version produces subscale scores for physical, functional, social, and emotional well-being, as well as satisfaction with the treatment relationship. Coefficients of reliability and validity were uniformly high. The ...

5,232 citations


"Ricolinostat, the First Selective H..." refers methods in this paper

  • ...Patients receiving bortezomib completed a neurotoxicitydirected questionnaire from the Functional Assessment of Cancer/Gynecology Oncology Group (FACT/GOG) survey of neurotoxicity (16, 17)....

    [...]

  • ...Transaminase elevation 1 (7) 0 (0) 9 (16) 3 (5) 3 (15) 2 (10) 2 (8) 0 (0)...

    [...]

  • ...Hyponatremia 1 (7) 0 (0) 9 (16) 3 (5) 4 (20) 1 (5) 3 (13) 1 (4)...

    [...]

Journal ArticleDOI
TL;DR: Bortezomib is superior to high-dose dexamethasone for the treatment of patients with multiple myeloma who have had a relapse after one to three previous therapies.
Abstract: background This study compared bortezomib with high-dose dexamethasone in patients with relapsed multiple myeloma who had received one to three previous therapies. methods We randomly assigned 669 patients with relapsed myeloma to receive either an intravenous bolus of bortezomib (1.3 mg per square meter of body-surface area) on days 1, 4, 8, and 11 for eight three-week cycles, followed by treatment on days 1, 8, 15, and 22 for three five-week cycles, or high-dose dexamethasone (40 mg orally) on days 1 through 4, 9 through 12, and 17 through 20 for four five-week cycles, followed by treatment on days 1 through 4 for five four-week cycles. Patients who were assigned to receive dexamethasone were permitted to cross over to receive bortezomib in a companion study after disease progression. results Patients treated with bortezomib had higher response rates, a longer time to progression (the primary end point), and a longer survival than patients treated with dexamethasone. The combined complete and partial response rates were 38 percent for bortezomib and 18 percent for dexamethasone (P<0.001), and the complete response rates were 6 percent and less than 1 percent, respectively (P<0.001). Median times to progression in the bortezomib and dexamethasone groups were 6.22 months (189 days) and 3.49 months (106 days), respectively (hazard ratio, 0.55; P<0.001). The oneyear survival rate was 80 percent among patients taking bortezomib and 66 percent among patients taking dexamethasone (P=0.003), and the hazard ratio for overall survival with bortezomib was 0.57 (P=0.001). Grade 3 or 4 adverse events were reported in 75 percent of patients treated with bortezomib and in 60 percent of those treated with dexamethasone. conclusions Bortezomib is superior to high-dose dexamethasone for the treatment of patients with multiple myeloma who have had a relapse after one to three previous therapies.

2,333 citations

Journal ArticleDOI
TL;DR: Bortezomib plus melphalan-prednisone alone in patients with newly diagnosed myeloma who were ineligible for high-dose therapy was superior to melphAlan-predisonsone alone.
Abstract: The time to progression among patients receiving bortezomib plus melphalan– prednisone (bortezomib group) was 24.0 months, as compared with 16.6 months among those receiving melphalan–prednisone alone (control group) (hazard ratio for the bortezomib group, 0.48; P<0.001). The proportions of patients with a partial response or better were 71% in the bortezomib group and 35% in the control group; complete-response rates were 30% and 4%, respectively (P<0.001). The median duration of the response was 19.9 months in the bortezomib group and 13.1 months in the control group. The hazard ratio for overall survival was 0.61 for the bortezomib group (P = 0.008). Adverse events were consistent with established profiles of toxic events associated with bortezomib and melphalan–prednisone. Grade 3 events occurred in a higher proportion of patients in the bortezomib group than in the control group (53% vs. 44%, P = 0.02), but there were no significant differences in grade 4 events (28% and 27%, respectively) or treatment-related deaths (1% and 2%). Conclusions Bortezomib plus melphalan–prednisone was superior to melphalan–prednisone alone in patients with newly diagnosed myeloma who were ineligible for high-dose therapy. (ClinicalTrials.gov number, NCT00111319.)

1,728 citations


Additional excerpts

  • ...Dyspnea 1 (7) 0 (0) 8 (14) 1 (2) 2 (10) 0 (0) 3 (13) 0 (0) Ricolinostat, Bortezomib, and Dexamethasone for Myeloma...

    [...]

  • ...Nausea 2 (13) 0 (0) 16 (28) 1 (2) 5 (25) 0 (0) 9 (38) 1 (4)...

    [...]

  • ...Vomiting 0 (0) 0 (0) 8 (14) 1 (2) 0 (0) 0 (0) 7 (29) 1 (4)...

    [...]

Journal ArticleDOI
12 Dec 2003-Cell
TL;DR: It is shown that cells deficient in HDAC6 fail to clear misfolded protein aggregates from the cytoplasm, cannot form aggresomes properly, and are hypersensitive to the accumulation of misfolding proteins.

1,414 citations


"Ricolinostat, the First Selective H..." refers background in this paper

  • ...Fatigue 4 (27) 0 (0) 20 (35) 4 (7) 5 (25) 1 (5) 10 (42) 2 (8)...

    [...]

  • ...Neutropenia 2 (13) 1 (7) 7 (12) 2 (4) 3 (15) 2 (10) 2 (8) 0 (0)...

    [...]

  • ...Hypophosphatemia 2 (13) 1 (7) 10 (18) 5 (9) 4 (20) 2 (10) 2 (8) 1 (4)...

    [...]

  • ...Hypokalemia 1 (7) 0 (0) 15 (26) 4 (7) 3 (15) 1 (5) 8 (33) 2 (8)...

    [...]

  • ...Pulmonary embolism 0 (0) 0 (0) 2 (4) 2 (4) 0 (0) 0 (0) 2 (8) 2 (8)...

    [...]

Related Papers (5)