ArticlePDF Available

Phase 2study of dovitinib in patients with relapsed or refractory multiple myeloma with or without t(4;14) translocation

Authors:

Abstract and Figures

Objectives: Approximately 15% of patients with multiple myeloma (MM) exhibit a t(4;14) translocation, which often results in constitutive activation of the receptor tyrosine kinase (RTK) fibroblast growth factor receptor 3 (FGFR3). This study evaluated the efficacy and safety of dovitinib, an RTK inhibitor with in vitro inhibitory activity against FGFR, in patients with relapsed or refractory MM with or without t(4;14) translocation. Methods: Adult patients with relapsed or refractory MM who had received ≥2 prior regimens were enrolled in this multicenter, 2-stage, phase 2 trial. Patients were grouped based on their t(4;14) status. Dovitinib (500 mg/day orally) was administered on a 5-days-on/2-days-off schedule. The primary endpoint was overall response rate by local investigator review (per International Myeloma Working Group criteria). In non-responding patients, treatment could continue with the addition of low-dose dexamethasone. Results: In total, 43 patients (median age, 63 years) were enrolled (13 t(4;14) positive, 26 t(4;14) negative, and 4 t(4;14) status non-interpretable). Patients had received a median of 5 prior regimens. Median duration of treatment was 8.7 weeks in the t(4;14)-positive group and 3.7 weeks in the t(4;14)-negative group. None of the patients on dovitinib had objective responses. The stable disease rate was 61.5% in the t(4;14)-positive group and 34.6% in the t(4;14)-negative group. Overall, 39 patients (90.7%) had adverse events suspected to be related to study drug, most commonly diarrhea (60.5%), nausea (58.1%), vomiting (46.5%), and fatigue (32.6%). Conclusion: Dovitinib showed no single-agent activity in relapsed or refractory MM but may stabilize disease in some t(4;14)-positive patients.
Content may be subject to copyright.
ORIGINAL ARTICLE
Phase 2 study of dovitinib in patients with relapsed or
refractory multiple myeloma with or without t(4;14)
translocation
Christof Scheid
1
, Donna Reece
2
, Meral Beksac
3
, Andrew Spencer
4
, Natalie Callander
5
,
Pieter Sonneveld
6
, Ghulam Kalimi
7
, Can Cai
7
, Michael Shi
7
, Jeffrey W. Scott
7
, A. Keith Stewart
8
1
University Hospital of Cologne, Cologne, Germany;
2
Princess Margaret Cancer Centre, Toronto, ON, Canada;
3
Ankara University School of
Medicine, Ankara, Turkey;
4
Alfred Health-Monash University, Melbourne, Vic., Australia;
5
University of Wisconsin, Madison, WI, USA;
6
Erasmus
Medical Center, Rotterdam, the Netherlands;
7
Novartis Pharmaceuticals, East Hanover, NJ;
8
Mayo Clinic, Scottsdale, AZ, USA
Abstract
Objectives: Approximately 15% of patients with multiple myeloma (MM) exhibit a t(4;14) translocation,
which often results in constitutive activation of the receptor tyrosine kinase (RTK) fibroblast growth factor
receptor 3 (FGFR3). This study evaluated the efficacy and safety of dovitinib, an RTK inhibitor with in vitro
inhibitory activity against FGFR, in patients with relapsed or refractory MM with or without t(4;14)
translocation. Methods: Adult patients with relapsed or refractory MM who had received 2 prior
regimens were enrolled in this multicenter, 2-stage, phase 2 trial. Patients were grouped based on their t
(4;14) status. Dovitinib (500 mg/day orally) was administered on a 5-days-on/2-days-off schedule. The
primary endpoint was overall response rate by local investigator review (per International Myeloma
Working Group criteria). In non-responding patients, treatment could continue with the addition of low-
dose dexamethasone. Results: In total, 43 patients (median age, 63 years) were enrolled (13 t(4;14)
positive, 26 t(4;14) negative, and 4 t(4;14) status non-interpretable). Patients had received a median of 5
prior regimens. Median duration of treatment was 8.7 weeks in the t(4;14)-positive group and 3.7 weeks
in the t(4;14)-negative group. None of the patients on dovitinib had objective responses. The stable
disease rate was 61.5% in the t(4;14)-positive group and 34.6% in the t(4;14)-negative group. Overall, 39
patients (90.7%) had adverse events suspected to be related to study drug, most commonly diarrhea
(60.5%), nausea (58.1%), vomiting (46.5%), and fatigue (32.6%). Conclusion: Dovitinib showed no single-
agent activity in relapsed or refractory MM but may stabilize disease in some t(4;14)-positive patients.
Key words phase 2; relapsed or refractory multiple myeloma; FGFR3; dovitinib; t(4;14) translocation
Correspondence Christof Scheid, MD, PhD, Department of Internal Medicine, University Hospital of Cologne, Kerpener Str. 62,
50924 Cologne, Germany. Tel: +49 221 478 6296; Fax +49 221 478 86712; e-mail: c.scheid@uni-koeln.de
Accepted for publication 13 November 2014 doi:10.1111/ejh.12491
Multiple myeloma (MM) is a neoplastic disorder of clonal B
cells characterized by uncontrolled and aberrant proliferation
of plasma cells within bone marrow, monoclonal protein in
the blood or urine, and associated organ dysfunction (1).
About 24 050 new cases of myeloma are estimated in 2014
in the United States (2), whereas the estimated number of
new MM cases in Europe was 33 000 in 2012 (3). Over the
past decade, the outcome of patients with MM has signi-
cantly improved with the development of immunomodula-
tory drugs (thalidomide, lenalidomide, and pomalidomide)
and proteasome inhibitors (bortezomib and carlzomib).
However, despite these advances, nearly all patients with
MM eventually relapse. Although patients can sometimes
achieve subsequent responses with additional treatment regi-
mens, these responses are often of shorter duration, necessi-
tating novel treatment options for MM (4, 5).
The broblast growth factor (FGF) receptor (FGFR) signal-
ing pathway is implicated in the pathogenesis of a variety of
cancers including MM (6); FGFR signaling promotes tumor
angiogenesis, and deregulated expression of FGFs leads to
316 ©2015 The Authors. European Journal of Haematology Published by John Wiley & Sons Ltd.
This is an open access article under the terms of the Creative Commons Attribution-NonCommercial-NoDerivs License, which permits use and
distribution in any medium, provided the original work is properly cited, the use is non-commercial and no modifications or adaptations are made.
European Journal of Haematology 95 (316–324)
tumor cell proliferation, survival, and chemoresistance (7).
Approximately 15% of patients with MM exhibit a t(4;14)
translocation (8), which often results in FGFR3 expression.
FGFR3 is receptor tyrosine kinase (RTK) of the FGF family,
consisting of 3 immunoglobulin-like extracellular domains, a
transmembrane domain, and a split cytoplasmic tyrosine
kinase domain. Following ligand stimulation, it signals pri-
marily through the extracellular signal-regulated kinases 1
and 2, phosphatidylinositol 3-kinase, and phospholipase C-ɣ
pathways (911). Furthermore, somatic mutation may occur
after the translocation, resulting in the constitutive activation
of FGFR3 (12). Patients with t(4;14) translocation appear to
have a worse prognosis and shorter survival compared with
patients without this translocation (13, 14). About 25% of
patients lose FGFR3 protein expression over time, and Ras
mutations common to almost 50% of patients with MM may
circumvent FGFR3 signaling dependency (15, 16).
Dovitinib, an RTK inhibitor, has demonstrated in vitro
inhibitory activity against FGFR, vascular endothelial growth
factor receptor (VEGFR), and platelet-derived growth factor
receptor, with half minimal inhibitory concentration values
of approximately 10 nM(17).
Tumor growth inhibition was observed with dovitinib
treatment in xenograft tumor models of MM with activating
FGFR3 mutations (8, 18), resulting in signicant improve-
ment in survival (8). Thus, the preclinical data established a
rationale for studying dovitinib in patients with MM. The
objective of this study was to evaluate the efcacy and
safety of dovitinib in patients with relapsed or refractory
MM with or without t(4;14) translocation.
Patients and methods
Patients
Adult patients (age 18 years), with cytopathologically or
histologically conrmed relapsed or refractory MM accord-
ing to International Myeloma Working Group (IMWG) cri-
teria (19), who had received 2 prior therapy regimens
including chemotherapy, autologous transplant, immunother-
apy, or other investigational agents, were included. Other
eligibility criteria were presence of measurable disease
(serum M-protein 1 g/dL and/or urine M-protein 200 mg/
24 h by protein electrophoresis) (20); World Health Organi-
zation (WHO) performance status of 2; and absolute neu-
trophil and platelet count 1000/mm
3
and 75 000/mm
3
,
respectively (or 750/mm
3
and 50 000/mm
3
, respectively,
if neutropenia and thrombocytopenia was clinically related
to progressive myeloma with bone marrow inltration),
hemoglobin 8 g/dL, aspartate aminotransferase (AST) and
alanine aminotransferase (ALT) 3.0 9upper limit of nor-
mal (ULN), serum bilirubin 1.5 9ULN, and serum creati-
nine 2.0 9ULN (or 24-hour creatinine clearance 50 mL/
min).
Patients were excluded if they had non-secretory or olig-
osecretory MM, symptomatic amyloidosis or plasma cell
leukemia, previous allogeneic stem cell transplant with evi-
dence of active graft-vs.-host disease requiring immunosup-
pressive therapy, history of another malignancy within
3 years prior to study entry (except cured basal cell carci-
noma of the skin or excised in situ carcinoma of the cervix),
impaired cardiac function or clinically signicant cardiac dis-
eases, uncontrolled hypertension dened as systolic blood
pressure 160 mmHg and/or diastolic blood pressure
100 mmHg with or without antihypertensive medication(s),
treatment with therapeutic doses of warfarin, cirrhosis,
chronic active hepatitis, or chronic persistent hepatitis, and
other concurrent severe and/or uncontrolled concomitant
medical conditions that could have caused unacceptable
safety risks. Patients could not have undergone a major sur-
gery 2 weeks prior to starting the study drug.
Study design and treatment
This was a multicenter, non-randomized, open-label, 2-stage,
phase 2 trial. Efcacy and safety of the treatment was sepa-
rately evaluated in the t(4;14)-positive or t(4;14)-negative
groups of patients. The study objectives did not include
direct comparisons of the two groups. At the baseline visit,
the patientsbone marrow aspirate samples were analyzed
for t(4;14) translocation status by cytoplasmic immunoglobu-
lin-enhanced interphase uorescent in situ hybridization
(FISH; Vysis; Abbott Laboratories, Abbott Park, IL, USA),
and they were assigned to either t(4;14)-positive or t(4;14)-
negative groups. Patients were considered assessable if at
least 100 clonal plasma cells from each slide could be
scored. Bone marrow aspirate samples were analyzed for
FGFR3 expression by ow cytometry, and the results were
qualitatively expressed as positive or negative. Patients were
assigned as t(4;14)-non-interpretable if there were too few
plasma cells in the samples to provide an accurate assess-
ment by FISH and ow cytometry and were not included in
the primary and secondary efcacy analyses.
All patients received dovitinib at a dose of 500 mg/day
orally on a 5-days-on/2-days-off schedule (dosing on days 1
to 5, 8 to 12, 15 to 19, and 22 to 26) every 28 days until pro-
gressive disease (PD), unacceptable toxicities, or patients/
investigators decision to discontinue. The 500 mg/day orally
on a 5-days-on/2-days-off dosing schedule was selected
based on the phase 1/2 study of dovitinib in metastatic renal
cell carcinoma (mRCC). In a Phase 1/2 dose escalation study,
dovitinib at the dose of 500 mg on a 5-days-on/2-days-off
schedule was generally well tolerated and showed antitumor
activity in heavily pretreated patients with mRCC. Also, the
pharmacodynamic analysis revealed VEGFR inhibition and
FGFR inhibition (21). Further, dovitinib (500 mg/day on a 5-
days-on/2-days-off schedule) was effective and tolerable in
patients with advanced or metastatic RCC enriched for
©2015 The Authors. European Journal of Haematology Published by John Wiley & Sons Ltd. 317
Scheid et al. Phase 2 dovitinib in multiple myeloma
patients previously treated with a VEGFR TKI and an mTOR
inhibitor (22).
As an exploratory objective, after disease progression on
dovitinib monotherapy, patients had the option to continue
treatment with the addition of low-dose dexamethasone
(40 mg every 7 days), with M-protein levels at the time of
progression considered as the new baseline.
Dose reductions [400 mg (rst reduction) and 300 mg
(second reduction)] were permitted for patients unable to tol-
erate the protocol-specied dosing schedule of dovitinib. No
dose escalation was allowed once a patient was dose
reduced. Dose reductions for dexamethasone were not per-
mitted in the dexamethasone-supplemented dovitinib regi-
men.
All patients provided written informed consent. The study
protocol and its amendments were approved by the indepen-
dent ethics committees at each center. The study conformed
to Good Clinical Practices guidelines and the ethical princi-
ples of the Declaration of Helsinki. The study was designed
by the academic investigators and by representatives of the
sponsor, Novartis Oncology.
Efficacy and safety assessments
The primary endpoint was extended overall response rate
(ORR) by local investigator review. Extended ORR was
dened as the rate of patients with ORR [best overall
response of complete response (CR), very good partial
response (VGPR), or partial response (PR)], plus patients
with best overall response of minor response (MR) accord-
ing to IMWG criteria (19). Secondary endpoints included
safety and ORR and progression-free survival (PFS) by local
investigator review. PFS was dened as the time from the
start of treatment to the date of the rst-documented PD or
death due to any cause, as assessed by the investigator. If a
patient had not progressed or died at the date of the analysis
cutoff or when he/she received any further antineoplastic
therapy, PFS was censored at the time of the last tumor
assessment before the cutoff date or the antineoplastic ther-
apy start date. Documentation of response in patients who
opted to continue on a dexamethasone-supplemented doviti-
nib regimen was an exploratory objective. Patients were
evaluated at the end of each cycle. Any response of MR or
better had to be conrmed by the next set of evaluations
4 weeks later. Any response of PD had to be conrmed as
soon as possible, preferably within 7 days after the observa-
tion.
Safety assessments consisted of collecting all adverse
events (AEs) and serious AEs (SAEs), including the regular
monitoring of laboratory tests, regular measurement of vital
signs, weight, performance status, physical examination, and
cardiac assessments (electrocardiogram, blood pressure, car-
diac enzymes, and multigated acquisition/echocardiogram).
All AEs were recorded using the National Cancer Institute
Common Terminology Criteria for Adverse Events version
4.0.
Statistical analysis
A 2-stage design (23) was used for each group to test the
null hypothesis that the extended ORR is 10% using a
1-sided test with 10% level of signicance and 90%
power at the alternative extended ORR of 30%. The null
hypothesis of extended ORR =0.10 and the alternative of
extended ORR =0.30 were chosen with reference to the
outcomes of single-agent phase 2 studies of lenalidomide
and bortezomib in patients with relapsed or refractory
MM (24, 25). For stage 1 enrollment, a total of 20
patients were planned in each group. If 3 patients had a
response, an additional 20 patients were planned in stage
2 of that group. The null hypothesis was rejected at the
10% level of signicance (1-sided) if there were 7
responders among all 40 patients at the end of stage 2.
KaplanMeier product-limit method was performed to esti-
mate median PFS.
Results
Patient characteristics
A total of 43 patients with relapsed or refractory MM were
enrolled between May 28, 2010, and December 29, 2011. As
of the latter date, only 13 patients were enrolled in the t(4;14)-
positive group. Enrollment in the t(4;14)-positive group was
closed due to lack of objective responses and slow rate of
enrollment. Twenty-six patients were enrolled in the t(4;14)-
negative group. The t(4;14) translocation status could not be
determined in 4 patients; these patients were summarized in a
non-interpretable group.
A majority of the patients were white (90.7%), had a
WHO performance status of 1 (58.1%), and were refractory
to last treatment (60.5%) (Table 1). Independent of translo-
cation status, 12 patients (27.9%) had FGFR3 expression (9
in the t(4;14)-positive group, 2 in the t(4;14)-negative group,
and 1 in t(4;14)non-interpretable group). Patients had
received a median of 5 (range, 210) prior antineoplastic
regimens.
Patient disposition
All 43 patients received dovitinib monotherapy (Table 2).
The median duration of exposure to the study drug was
8.7 weeks and 3.7 weeks for the patients enrolled in t
(4;14)-positive and t(4;14)-negative groups, respectively.
Of the 27 patients with PD on dovitinib monotherapy, 6
patients continued on dovitinib with dexamethasone added
[3 each in the t(4;14)-positive and t(4;14)-negative
groups].
318 ©2015 The Authors. European Journal of Haematology Published by John Wiley & Sons Ltd.
Phase 2 dovitinib in multiple myeloma Scheid et al.
Table 1 Patient characteristics at baseline dovitinib monotherapy
t(4;14)-Positive
n=13
t(4;14)-Negative
n=26
t(4;14)-
Non-interpretable
n=4
All patients
N=43
Median age (range), years 61 (4769) 67 (2984) 55 (4066) 63 (2984)
Sex, n(%)
Female 6 (46.2) 10 (38.5) 4 (100) 20 (46.5)
Male 7 (53.8) 16 (61.5) 0 23 (53.5)
Race, n(%)
White 12 (92.3) 23 (88.5) 4 (100) 39 (90.7)
Black 1 (7.7) 2 (7.7) 0 3 (7.0)
Asian 0 1 (3.8) 0 1 (2.3)
World Health Organization performance status, n(%)
0 4 (30.8) 9 (34.6) 0 13 (30.2)
1 8 (61.5) 15 (57.7) 2 (50.0) 25 (58.1)
2 1 (7.7) 2 (7.7) 2 (50.0) 5 (11.6)
Median time since diagnosis (range), months 49.3 (24.984.3) 64.3 (15.8246.0) 62.1 (58.8140.1) 58.8 (15.8246.0)
Number of prior lines of therapy, n(%)
2 4 (30.8) 2 (7.7) 0 6 (14.0)
3 1 (7.7) 4 (15.4) 0 5 (11.6)
4 2 (15.4) 5 (19.2) 0 7 (16.3)
>4 6 (46.2) 15 (57.7) 4 (100) 25 (58.1)
Median prior lines of therapy (range) 4.0 (210) 5.0 (29) 7.0 (59) 5.0 (210)
Prior therapies, n(%)
Corticosteroids 13 (100) 26 (100) 4 (100) 43 (100)
Bevacizumab 1 (7.7) 1 (3.8) 0 2 (4.7)
Bortezomib 13 (100) 24 (92.3) 4 (100) 41 (95.3)
Vorinostat 1 (7.7) 2 (7.7) 0 3 (7.0)
Lenalidomide 11 (84.6) 21 (80.8) 4 (100.0) 36 (83.7)
Thalidomide 6 (46.2) 16 (61.5) 4 (100.0) 26 (60.5)
Patients relapsed or refractory from last treatment, n(%)
Refractory 7 (53.8) 16 (61.5) 3 (75.0) 26 (60.5)
Relapsed 6 (46.2) 10 (38.5) 1 (25.0) 17 (39.5)
FGFR3 expression, n(%)
Yes 9 (69.2) 2 (7.7) 1 (25.0) 12 (27.9)
No 0 19 (73.1) 2 (50.0) 21 (48.8)
Unknown 4 (30.8) 5 (19.2) 1 (25.0) 10 (23.3)
FGFR3, fibroblast growth factor receptor 3.
Table 2 Patient disposition
t(4;14)-Positive
n=13
t(4;14)-Negative
n=26
t(4;14)-Non-interpretable
n=4
All patients
N=43
Dovitinib monotherapy
EOT, n(%) 13 (100) 26 (100) 4 (100) 43 (100)
Primary reason for EOT, n(%)
Disease progression 7 (53.8) 20 (76.9) 0 27 (62.8)
Adverse event (s) 4 (30.8) 3 (11.5) 3 (75.0) 10 (23.3)
Subject withdrew consent 2 (15.4) 3 (11.5) 1 (25.0) 6 (14.0)
Dovitinib plus dexamethasone
EOT, n(%) 3 (23.1) 3 (11.5) 0 6 (14.0)
Primary reason for EOT, n(%)
Disease progression 2 (15.4) 3 (11.5) 0 5 (11.6)
Adverse event(s) 1 (7.7) 0 0 1 (2.3)
EOT, end of treatment.
©2015 The Authors. European Journal of Haematology Published by John Wiley & Sons Ltd. 319
Scheid et al. Phase 2 dovitinib in multiple myeloma
As of March 22, 2013 (nal database lock), all patients
discontinued the study drug (including those receiving the
dovitinib/dexamethasone combination regimen). The most
frequently reported reasons for discontinuing dovitinib
monotherapy were disease progression in 27 patients
(62.8%) and AEs in 10 patients (23.3%). The reasons to stop
the dovitinib/dexamethasone regimen were disease progres-
sion for 5 patients and AEs for 1 patient.
Efficacy
None of the patients on dovitinib monotherapy had MR,
PR, VGPR, or CR (Table 3); hence, the ORR (PR) and
extended ORR (MR) were 0%. Overall, 20 patients
(46.5%) had stable disease (SD).
Eight patients (61.5%) in the t(4;14)-positive group had
SD. The median PFS per local investigator assessment
was 2.6 months [95% condence interval (CI): 0.93.9] in
the t(4;14)-positive group in patients treated with dovitinib
monotherapy (Fig. 1).Of the 3 patients in the t(4;14)-positive
group who continued on the dexamethasone-supplemented
treatment regimen, 1 patient had SD, 1 patient had PD, and
the response was not evaluable for the remaining 1 patient.
Nine patients in the t(4;14)-negative group (34.6%) had
SD. The median PFS per local investigator assessment was
0.9 months (95% CI: 0.81.7) in the t(4;14)-negative group
in patients treated with dovitinib monotherapy (Fig. 1A). Of
the 3 patients in the t(4;14)-negative group, 1 patient had
PR and 2 patients had SD.
As the number of responders in each group failed to meet
the criteria for study continuation to stage 2 (3 responders
from stage 1), the study was terminated. Among the 12
patients with detectable FGFR3 expression, 58.3% had SD,
whereas the SD rate in the 21 patients without FGFR3
expression (excluding patients with unknown expression)
was 42.9%. The median PFS was 1.77 months [95% CI:
0.393.88]) in patients who had FGFR3 expression and
0.9 months [95% CI: 0.821.77] in patients who did not
have FGFR3 expression (Fig. 1B).
Safety
Overall, 39 patients (90.7%) treated with dovitinib mono-
therapy had AEs suspected to be related to study drug
(Table 4). The safety proles were similar for the t(4;14)-
positive and t(4;14)-negative groups. The most frequently
reported AEs suspected to be related to study drug were
diarrhea (60.5%), nausea (58.1%), vomiting (46.5%), fati-
gue (32.6%), and thrombocytopenia (20.9%). The most
frequently reported grade 3 or 4 treatment-related AEs
were diarrhea (20.9%), thrombocytopenia (16.3%), and
fatigue (14.0%). Diarrhea was the most commonly
reported grade 3 or 4 SAE occurring in 4 of the 8
patients (9.3%) with SAEs suspected to be related to
study drug. For the 6 patients who continued on the dovi-
tinib and dexamethasone combination, AEs and SAEs
were primarily gastrointestinal related.
In the overall population, the most common AEs leading
to discontinuation of dovitinib monotherapy were diarrhea
(11.6%), vomiting (7.0%), and fatigue (4.7%). Overall, 26
patients treated with dovitinib monotherapy had AEs requir-
ing a dose adjustment or study drug interruption, most com-
monly diarrhea (23.3%), nausea (9.3%), and vomiting
(9.3%). A total of 12 patients (27.9%) on dovitinib mono-
therapy had a dose change: 6 patients (14.0%) required 1
dose change, and 6 patients (14.0%) required 2 dose
changes. The median daily dose remained 500 mg/day in all
the groups.
In total, 4 patients died within 30 days following the last
treatment dose on dovitinib monotherapy; the reasons for
deaths were disease progression (3 patients) and cardiopul-
monary arrest (1 patient). The patient who died due to car-
diopulmonary arrest had discontinued study drug because of
disease progression on day 25 and started melphalan 2 days
later. The patients condition deteriorated; under mechanical
ventilation, cardiogenic shock was experienced 10 days after
last dose of the study drug, which was followed by cardio-
pulmonary arrest 24 days after the last dose of the study
drug. A total of 2 patients died within 30 days following the
Table 3 Overall response to dovitinib monotherapy, assessed by local review
Overall response, n(%)
t(4;14)-Positive
n=13
t(4;14)-Negative
n=26
t(4;14)-Non-interpretable
n=4
All patients
N=43
Complete response 0 0 0 0
Very good partial response 0 0 0 0
Partial response 0 0 0 0
Minor response 0 0 0 0
Stable disease 8 (61.5) 9 (34.6) 3 (75.0) 20 (46.5)
Progressive disease 4 (30.8) 13 (50.0) 1 (25.0) 18 (41.9)
Not determined 1 (7.7) 4 (15.4) 0 5 (11.6)
Extended overall response rate 0 0 0 0
Overall response rate 0 0 0 0
320 ©2015 The Authors. European Journal of Haematology Published by John Wiley & Sons Ltd.
Phase 2 dovitinib in multiple myeloma Scheid et al.
last dose on dovitinib plus dexamethasone combination ther-
apy; 1 of these patients died due to disease progression, and
the other patient died due to intestinal perforation secondary
to disease progression. None of these 6 deaths were sus-
pected to be related to study drug.
There was no evidence of QTcF increase from baseline of
>60 ms. One patient experienced a >20% decrease in left
ventricular ejection fraction.
Hematologic abnormality shifts were mostly grade 1 or 2.
Eleven patients (25.6%) had grade 3 decreased neutrophils,
A
B
Figure 1 KaplanMeier median progression-free
survival by local investigator review dovitinib
monotherapy by (A) t(4;14) translocation status
and (B) FGFR3 expression status.
Table 4 Adverse events suspected to be related to study drug Dovitinib monotherapy (10% patients)
Preferred term
t(4;14)-Positive
n=13
t(4;14)-Negative
n=26
t(4;14)-Non-interpretable
n=4
All patients
N=43
Any grade
n(%)
Grade 3/4
n(%)
Any grade
n(%)
Grade 3/4
n(%)
Any grade
n(%)
Grade 3/4
n(%)
Any grade
n(%)
Grade 3/4
n(%)
Total 12 (92.3) 8 (61.5) 23 (88.5) 12 (46.2) 4 (100) 2 (50.0) 39 (90.7) 22 (51.2)
Diarrhea 7 (53.8) 3 (23.1) 16 (61.5) 6 (23.1) 3 (75.0) 0 26 (60.5) 9 (20.9)
Nausea 7 (53.8) 1 (7.7) 14 (53.8) 2 (7.7) 4 (100) 0 25 (58.1) 3 (7.0)
Vomiting 6 (46.2) 1 (7.7) 11 (42.3) 2 (7.7) 3 (75.0) 0 20 (46.5) 3 (7.0)
Fatigue 4 (30.8) 3 (23.1) 10 (38.5) 3 (11.5) 0 0 14 (32.6) 6 (14.0)
Thrombocytopenia 4 (30.8) 3 (23.1) 3 (11.5) 2 (7.7) 2 (50.0) 2 (50.0) 9 (20.9) 7 (16.3)
Decreased appetite 2 (15.4) 1 (7.7) 5 (19.2) 1 (3.9) 0 0 7 (16.3) 2 (4.7)
Dehydration 1 (7.7) 0 5 (19.2) 4 (15.4) 0 0 6 (14.0) 4 (9.3)
Anemia 2 (15.4) 2 (15.4) 2 (7.7) 1 (3.8) 1 (25.0) 0 5 (11.6) 3 (7.0)
Neutropenia 3 (23.1) 3 (23.1) 2 (7.7) 1 (3.8) 0 0 5 (11.6) 4 (9.3)
Dyspepsia 0 0 2 (7.7) 0 2 (50.0) 0 4 (9.3) 0
Asthenia 0 0 3 (11.5) 0 0 0 3 (7.0) 0
Rash 2 (15.4) 0 1 (3.9) 1 (3.8) 0 0 3 (7.0) 1 (2.3)
Weight decreased 0 0 3 (11.5) 0 0 0 3 (7.0) 0
©2015 The Authors. European Journal of Haematology Published by John Wiley & Sons Ltd. 321
Scheid et al. Phase 2 dovitinib in multiple myeloma
12 patients (27.9%) had grade 3 decreased hemoglobin, and
12 patients (27.9%) had grade 3 decreased white blood cells.
A total of 6 patients (14.0%) experienced grade 3 decreased
lymphocytes and 1 patient (2.3%) experienced grade 4
decreased lymphocytes. Furthermore, grade 3 and 4
decreased platelet count was reported in 9 patients (20.9%)
and 10 patients (23.3%), respectively.
Grade 3 increases in ALT and AST were reported in 3
patients (7.3%) and 2 patients (4.7%), respectively, while for
bilirubin, no increase of more than grade 1 was found.
Discussion
Approximately 15% of patients with MM exhibit a t(4;14)
translocation (8), which often results in FGFR3 expression.
Loss of FGFR3 expression is, however, evidenced during
disease progression in approximately 25% of patients, lead-
ing some to believe that the reciprocal of the t(4;14) translo-
cation that results in MMSET protein activation may be the
dominant molecular event (15, 16). Treatment of patients
with MM bearing the t(4;14) translocation (one of the high-
riskcytogenetic factors) is challenging, as early and aggres-
sive relapse is common (26). Although development of
immunomodulatory agents and proteasome inhibitors has
dramatically improved the outlook for patients with MM,
including t(4;14), prognosis in this MM subset remains poor
overall (27). Dovitinib, an RTK inhibitor, demonstrated in
vitro cytotoxicity and tumor growth inhibition in xenograft
tumor models of MM with activating FGFR3 mutations (8,
18). Based on these preclinical ndings, we evaluated the
efcacy and safety of dovitinib in patients with relapsed or
refractory MM with and without t(4;14) translocation.
In the present study, although none of the patients on
dovitinib monotherapy had ORR (PR) or extended ORR
(MR), 20 patients (46.5%) had SD. Although the t(4;14)
translocation predicts poor prognosis in patients with MM
(28), it is of interest that the proportion of patients
with SD during study treatment was numerically higher in
the t(4;14)-positive group (61.5%) than the t(4;14)-negative
group (34.6%). These results need to be interpreted cau-
tiously as the sample size was small, and the study objec-
tives did not include direct comparison the t(4;14)-positive
or t(4;14)-negative groups of patients. Furthermore, the
median PFS in the t(4;14)-positive and t(4;14)-negative
group was 2.6 months and 0.9 months, respectively. These
results raise the possibility that dovitinib has some biologic
activity in relapsed or refractory MM patients with t(4;14)
translocation, a hypothesis that could be explored in com-
bination studies with other agents, such as proteasome
inhibitors or by studying similar agents earlier in the dis-
ease course. Although patient populations from different
studies cannot be compared, it is interesting to note that
carlzomib as a single agent produced a median PFS of
3.5 months in patients with relapsed and refractory MM
with high-risk cytogenetics (29). In a randomized phase 2
study comparing pomalidomide plus low-dose dexametha-
sone vs. pomalidomide alone in patients with relapsed and
refractory MM, the median PFS was 2.7 months in patients
treated with pomalidomide alone (30), which is similar to
the median PFS in the t(4;14)-positive group in our study.
Monotherapy has generally not proven to be effective in
patients with MM; thus, combination regimens have been
the mainstay of effective therapy in MM. In this study,
results were perhaps more promising in patients with t(4;14)
or FGFR3 overexpression, and further in vitro and xenograft
studies may suggest a role for using dovitinib in combina-
tion regimens for t(4;14)-positive patients.
The safety results in our study were in line with the
known safety prole of dovitinib or were not unexpected in
patients with relapsed or refractory MM. The most com-
monly affected primary system organ class (10%) for AEs
suspected to be related to study drug was gastrointestinal
disorders. Gastrointestinal toxicity remains a challenge, and
timely monitoring and supportive care during dovitinib treat-
ment may alleviate these symptoms.
In summary, dovitinib was found to have no or minimal
single-agent activity in relapsed or refractory MM irrespec-
tive of t(4;14) status. However, the higher rate of disease
stabilization, the longer exposure duration, and longer PFS
in the t(4;14)-positive group may provide support for explor-
ing the inclusion of dovitinib in a combination therapy regi-
men in this high-risk patient population, particularly those
patients with earlier-stage disease who retain expression of
FGFR3 and lack Ras or other genomic mutations that render
FGFR3 redundant. Success is only likely in patients meeting
these criteria who are still at least partially dependent on
FGFR3 signaling.
Acknowledgements
We thank Peter J. Simon, PhD, Articulate Science (funded
by Novartis Pharmaceuticals Corporation) and Pushkar Nar-
vilkar, Novartis Healthcare Pvt. Ltd. for providing medical
editorial assistance with this manuscript.
Conflict of interest and sources of funding
CS has received honoraria from Novartis. DR has received
research funding from Novartis, Millennium, Janssen, and
Celgene; honoraria from Novartis, Janssen, and Celgene;
and consultancy from Janssen and Celgene. MB has served
on advisory boards for Novartis, Janssen-Cilag, Celgene,
Amgen, and Bristol-Myers Squibb; and speakersbureau for
Janssen-Cilag, Celgene, and Amgen. AS has received per-
sonal fees from Novartis. NC has nothing to disclose. PS
has received grants and personal fees from Celgene, Janssen,
Millennium, and Onyx; and has served on advisory boards
for Novartis. GK, CC, MC, and JC are employees of
322 ©2015 The Authors. European Journal of Haematology Published by John Wiley & Sons Ltd.
Phase 2 dovitinib in multiple myeloma Scheid et al.
Novartis Pharmaceuticals Corporation. AKS has served as
consultant for Novartis. This study was funded by Novartis
Pharmaceuticals Corporation.
References
1. Kyle RA, Rajkumar SV. Multiple myeloma. N Engl J Med
2004;351:186073.
2. Siegel R, Naishadham D, Jemal A. Cancer statistics, 2014. CA
Cancer J Clin 2014;64:929.
3. Ferlay J, Steliarova-Foucher E, Lortet-Tieulent J, Rosso S,
Coebergh JW, Comber H, Forman D, Bray F. Cancer inci-
dence and mortality patterns in Europe: estimates for 40 coun-
tries in 2012. Eur J Cancer 2013;49:1374403.
4. Saini N, Mahindra A. Therapeutic strategies for the treatment
of multiple myeloma. Discov Med 2013;15:2518.
5. Anderson KC, Kyle RA, Rajkumar SV, Stewart AK, Weber
D, Richardson P. Clinically relevant end points and new drug
approvals for myeloma. Leukemia 2008;22:2319.
6. Chell V, Balmanno K, Little AS, Wilson M, Andrews S,
Blockley L, Hampson M, Gavine PR, Cook SJ. Tumour cell
responses to new broblast growth factor receptor tyrosine
kinase inhibitors and identication of a gatekeeper mutation in
FGFR3 as a mechanism of acquired resistance. Oncogene
2013;32:305970.
7. Pardo OE, Arcaro A, Salerno G, Raguz S, Downward J, Seckl
MJ. Fibroblast growth factor-2 induces translational regulation
of Bcl-XL and Bcl-2 via a MEK-dependent pathway: correla-
tion with resistance to etoposide-induced apoptosis. J Biol
Chem 2002;277:120406.
8. Xin X, Abrams TJ, Hollenbach PW, et al. CHIR-258 is efca-
cious in a newly developed broblast growth factor receptor
3-expressing orthotopic multiple myeloma model in mice. Clin
Cancer Res 2006;12:490815.
9. Kanai M, Goke M, Tsunekawa S, Podolsky DK. Signal trans-
duction pathway of human broblast growth factor receptor 3.
Identication of a novel 66-kDa phosphoprotein. J Biol Chem
1997;272:66218.
10. Hart KC, Robertson SC, Kanemitsu MY, Meyer AN, Tynan
JA, Donoghue DJ. Transformation and Stat activation by
derivatives of FGFR1, FGFR3, and FGFR4. Oncogene
2000;19:330920.
11. Hart KC, Robertson SC, Donoghue DJ. Identication of tyro-
sine residues in constitutively activated broblast growth fac-
tor receptor 3 involved in mitogenesis, Stat activation, and
phosphatidylinositol 3-kinase activation. Mol Biol Cell
2001;12:93142.
12. Chesi M, Brents LA, Ely SA, Bais C, Robbiani DF, Mesri
EA, Kuehl WM, Bergsagel PL. Activated broblast growth
factor receptor 3 is an oncogene that contributes to
tumor progression in multiple myeloma. Blood
2001;97:72936.
13. Moreau P, Facon T, Leleu X, Morineau N, Huyghe P,
Harousseau JL, Bataille R, Avet-Loiseau H. Recurrent 14q32
translocations determine the prognosis of multiple myeloma,
especially in patients receiving intensive chemotherapy. Blood
2002;100:157983.
14. Moreau P, Attal M, Garban F, et al. Heterogeneity of t(4;14)
in multiple myeloma. Long-term follow-up of 100 cases trea-
ted with tandem transplantation in IFM99 trials. Leukemia
2007;21:20204.
15. Santra M, Zhan F, Tian E, Barlogie B, Shaughnessy J Jr.
A subset of multiple myeloma harboring the t(4;14)(p16;
q32) translocation lacks FGFR3 expression but maintains an
IGH/MMSET fusion transcript. Blood 2003;101:2374
6.
16. Chesi M, Nardini E, Brents LA, Schr
ock E, Ried T, Kuehl
WM, Bergsagel PL. Frequent translocation t(4;14)(p16.3;
q32.3) in multiple myeloma is associated with increased
expression and activating mutations of broblast growth factor
receptor 3. Nat Genet 1997;16:2604.
17. Lee SH, de Lopes MD, Vora J, et al. In vivo target modula-
tion and biological activity of CHIR-258, a multitargeted
growth factor receptor kinase inhibitor, in colon cancer mod-
els. Clin Cancer Res 2005;11:363341.
18. Trudel S, Li ZH, Wei E, Wiesmann M, Chang H, Chen C,
Reece D, Heise C, Stewart AK. CHIR-258, a novel, multitar-
geted tyrosine kinase inhibitor for the potential treatment of t
(4;14) multiple myeloma. Blood 2005;105:29418.
19. Rajkumar SV, Harousseau JL, Durie B, et al. Consensus
recommendations for the uniform reporting of clinical trials:
report of the International Myeloma Workshop Consensus
Panel 1. Blood 2011;117:46915.
20. Durie BG, Harousseau JL, Miguel JS, et al. International
uniform response criteria for multiple myeloma. Leukemia
2006;20:146773.
21. Angevin E, Lopez-Martin JA, Lin CC, et al. Phase I study of
dovitinib (TKI258), an oral FGFR, VEGFR, and PDGFR
inhibitor, in advanced or metastatic renal cell carcinoma. Clin
Cancer Res 2013;19:125768.
22. Escudier B, Gr
unwald V, Ravaud A, et al. Phase II results of
Dovitinib (TKI258) in patients with metastatic renal cell
cancer. Clin Cancer Res 2014;20:301222.
23. Simon R. Optimal two-stage designs for phase II clinical
trials. Control Clin Trials 1989;10:110.
24. Richardson PG, Barlogie B, Berenson J, et al. A phase 2
study of bortezomib in relapsed, refractory myeloma. N Engl
J Med 2003;348:260917.
25. Richardson PG, Blood E, Mitsiades CS, et al. A randomized
phase 2 study of lenalidomide therapy for patients with
relapsed or relapsed and refractory multiple myeloma. Blood
2006;108:345864.
26. Kalff A, Spencer A. The t(4;14) translocation and FGFR3
overexpression in multiple myeloma: prognostic implications
and current clinical strategies. Blood Cancer J 2012;2:
e89.
27. Bergsagel PL, Mateos MV, Gutierrez NC, Rajkumar SV, San
Miguel JF. Improving overall survival and overcoming
adverse prognosis in the treatment of cytogenetically high-risk
multiple myeloma. Blood 2013;121:88492.
©2015 The Authors. European Journal of Haematology Published by John Wiley & Sons Ltd. 323
Scheid et al. Phase 2 dovitinib in multiple myeloma
28. Segges P, Braggio E. Genetic markers used for risk stratica-
tion in multiple myeloma. Genet Res Int 2011;2011:
798089.
29. Jakubowiak AJ, Siegel DS, Martin T, et al. Treatment out-
comes in patients with relapsed and refractory multiple mye-
loma and high-risk cytogenetics receiving single-agent
carlzomib in the PX-171-003-A1 study. Leukemia
2013;27:23516.
30. Richardson PG, Siegel DS, Vij R, et al. Pomalidomide alone
or in combination with low-dose dexamethasone in relapsed
and refractory multiple myeloma: a randomized phase 2 study.
Blood 2014;123:182632.
324 ©2015 The Authors. European Journal of Haematology Published by John Wiley & Sons Ltd.
Phase 2 dovitinib in multiple myeloma Scheid et al.
... In the study carried out by Scheid et al. [101], the activity of dovitinib, which acts on the fibroblast growth factor receptor 3 (FGFR3), was evaluated in MM patients. FGFR3 is an RTK of the FGFR family that is responsible for cell growth, differentiation, and migration in a wide variety of cell types and is present in MM and a variety of cancers. ...
... The other studies addressed treatments with other kinase inhibitors, such as Ruxolitinib, Cabozantinib, INCB052793, Tivantinib, Trametinib, Afuresertibe, Sorafenib, and Dovitinib. In total, 36.4% (4) of the articles described in the table are phase I clinical trials, while the other 63.6%(7)are phase II clinical trials[92][93][94][95][96][97][98][99][100][101]. ...
Article
Full-text available
Multiple myeloma (MM) is a blood cell neoplasm characterized by excessive production of malignant monoclonal plasma cells (activated B lymphocytes) by the bone marrow, which end up synthesizing antibodies or antibody fragments, called M proteins, in excess. The accumulation of this production, both cells themselves and of the immunoglobulins, causes a series of problems for the patient, of a systemic and local nature, such as blood hyperviscosity, renal failure, anemia, bone lesions, and infections due to compromised immunity. MM is the third most common hematological neoplasm, constituting 1% of all cancer cases, and is a disease that is difficult to treat, still being considered an incurable disease. The treatments currently available cannot cure the patient, but only extend their lifespan, and the main and most effective alternative is autologous hematopoietic stem cell transplantation, but not every patient is eligible, often due to age and pre-existing comorbidities. In this context, the search for new therapies that can bring better results to patients is of utmost importance. Protein tyrosine kinases (PTKs) are involved in several biological processes, such as cell growth regulation and proliferation, thus, mutations that affect their functionality can have a great impact on crucial molecular pathways in the cells, leading to tumorigenesis. In the past couple of decades, the use of small-molecule inhibitors, which include tyrosine kinase inhibitors (TKIs), has been a hallmark in the treatment of hematological malignancies, and MM patients may also benefit from TKI-based treatment strategies. In this review, we seek to understand the applicability of TKIs used in MM clinical trials in the last 10 years.
... In about 80% of patients with t(4; 14), there is also the reciprocal translocation that juxtaposes the powerful 3 0 IgH enhancer on der14 to FGFR3, a receptor tyrosine kinase expressed on the cell surface. A quarter of the patients that ectopically express FGFR3 also have an activating mutation indicating a critical role for the tyrosine kinase activity of FGFR3 in MM progression 39 Clinical trials using FGFR3 tyrosine kinase inhibitors dovitinib 40 and erdafitinib (NCT02952573) have not reported clinical responses in patients with t(4;14) MM. Preclinical studies suggest that kinase inhibition will only be effective in the presence of FGFR3 activating mutations, which has not been an eligibility criteria in the clinical trials. ...
... In about 80% of patients with t(4; 14), there is also the reciprocal translocation that juxtaposes the powerful 3 0 IgH enhancer on der14 to FGFR3, a receptor tyrosine kinase expressed on the cell surface. A quarter of the patients that ectopically express FGFR3 also have an activating mutation indicating a critical role for the tyrosine kinase activity of FGFR3 in MM progression 39 Clinical trials using FGFR3 tyrosine kinase inhibitors dovitinib 40 and erdafitinib (NCT02952573) have not reported clinical responses in patients with t(4;14) MM. Preclinical studies suggest that kinase inhibition will only be effective in the presence of FGFR3 activating mutations, which has not been an eligibility criteria in the clinical trials. ...
... Thus, although this patient succumbed to clinical disease progression, a dramatic molecular response was achieved with erdafitinib as evidenced by elimination of the intended FGFR3-mutant subclone. This is particularly important in light of several FGFR3-targeted clinical trials in MM showing best responses of stable disease, leading some to question whether this class of agents has a role in myeloma therapy (Arnulf et al. 2007;Scheid et al. 2015;Trudel et al. 2012). However, our study shows that measuring molecular responses at the single-cell level may more accurately reflect the effectiveness of targeted agents such as erdafitinib. ...
Article
Full-text available
Genomic characterization of cancer has enabled identification of numerous molecular targets, which has led to significant advances in personalized medicine. However, with few exceptions, precision medicine approaches in the plasma cell malignancy multiple myeloma (MM) have had limited success, likely owing to the subclonal nature of molecular targets in this disease. Targeted therapies against FGFR3 have been under development for the past decade in the hopes of targeting aberrant FGFR3 activity in MM. FGFR3 activation results from the recurrent transforming event of t(4;14) found in approximately 15% of MM patients, as well as secondary FGFR3 mutations in this subgroup. To evaluate the effectiveness of targeting FGFR3 in MM, we undertook a phase 2 clinical trial evaluating the small molecule FGFR1-4 inhibitor, erdafitinib, in relapsed/refractory myeloma patients with or without FGFR3 mutations ( NCT02952573 ). Herein, we report on a single t(4;14) patient enrolled on this study who was identified to have a subclonal FGFR3 stop-loss deletion. Although this individual eventually progressed on study and succumbed to their disease, the intended molecular response was revealed through an extensive molecular characterization of the patient's tumour at baseline and on treatment using single-cell genomics. We identified elimination of the FGFR3-mutant subclone after treatment and expansion of a pre-existing clone with loss of chromosome 17p. Altogether, our study highlights the utility of single cell genomics in targeted trials as they can reveal molecular mechanisms that underlie sensitivity and resistance. This in turn can guide more personalized and targeted therapeutic approaches, including those that involve FGFR3-targeting therapies.
... Clinically, FGFR3 has been targeted by both non-selective multikinase inhibitors and highly selective FGFR inhibitors. In multiple myeloma patients, dovitinib and nintedanib, two multikinase inhibitors which inhibit FGFR, VEGFR, and PDGFR, have demonstrated little more than a tolerable safety profile and the potential to stabilize myelomatous disease (90,91). Dasatinib, a multikinase inhibitor effective in CML, was investigated in a phase II study of RRMM patients produced only 1 partial response out of 21 treated patients (NCT00429949). ...
Article
Full-text available
Multiple myeloma remains an incurable disease despite numerous novel agents being approved in the last decade. Furthermore, disease behavior and susceptibility to current treatments often vary drastically from patient to patient. To date there are no approved therapies in myeloma that are targeted to specific patient populations based on genomic or immunologic findings. Precision medicine, using biomarkers descriptive of a specific tumor’s biology and predictive of response to appropriate agents, may continue to push the field forward by expanding our treatment arsenal while refining our ability to expose patients to only those treatments likely to be efficacious. Extensive research efforts have been carried out in this endeavor including the use of agents targeting Bcl2 and the RAS/MAPK and PI3K/AKT/mTOR pathways. Thus far, clinical trials have yielded occasional successes intermixed with disappointments, reflecting significant hurdles which still remain including the complex crosstalk between oncogenic pathways and the nonlinear genetic development of myeloma, prone to cultivating sub-clones with distinctive mutations. In this review, we explore the landscape of precision therapeutics in multiple myeloma and underscore the degree to which research efforts have produced tangible clinical results.
... Other targets have been identified. FGFR3, together with MMSET/WHSC1, is overexpressed in 10-15% of MM patients due to the t(4;14) translocation; however, first generation FGFR inhibitors have demonstrated limited efficacy [134]. The recent discovery of activating FGFR3 mutations in a third of t(4;14) cases has renewed interest in novel FGFR inhibitors targeting FGFR3mutated MM [135,136]. ...
Article
Full-text available
New approaches to stratify multiple myeloma patients based on prognosis and therapeutic decision-making, or prediction, are needed since patients are currently managed in a similar manner regardless of individual risk factors or disease characteristics. However, despite new and improved biomarkers for determining the prognosis of patients, there is currently insufficient information to utilise biomarkers to intensify, reduce or altogether change treatment, nor to target patient-specific biology in a so-called predictive manner. The ever-increasing number and complexity of drug classes to treat multiple myeloma have improved response rates and so clinically useful biomarkers will need to be relevant in the era of such novel therapies. Therefore, the field of multiple myeloma biomarker development is rapidly progressing, spurred on by new technologies and therapeutic approaches, and underpinned by a deeper understanding of tumour biology with individualised patient management the goal. In this review, we describe the main biomarker categories in multiple myeloma and relate these to diagnostic, prognostic and predictive applications.
... Similarly, over half of the patients with RAS-mutated MM treated with the MEK inhibitor trametinib experienced at least a 25% reduction in paraprotein although many responses were short lived, especially when the drugs were used as single agents121 . FGFR3, together with MMSET/WHSC1, is overexpressed in 10-15% of MM patients due to the t(4;14) translocation, however rst generation FGFR inhibitors have demonstrated limited e cacy122 . The recent discovery of activating FGFR3 mutations in a third of t(4;14)cases has, however, renewed interest in novel FGFR inhibitors targeting FGFR3-mutated MM123,124 . ...
Preprint
Full-text available
New approaches to stratify multiple myeloma patients for informing on prognosis and therapy selection are needed since patients are currently managed in a similar manner regardless of individual risk factors. However, despite new and improved biomarkers for determining the prognosis of patients, there is currently insufficient information to utilise biomarkers to intensify, reduce or altogether change treatment, nor to target patient-specific biology. The ever-increasing number and complexity of drug classes to treat multiple myeloma have improved response rates and so clinically useful biomarkers will need to be relevant in the era of such novel therapies. Therefore, the field of multiple myeloma biomarker development is rapidly progressing, spurred on by new technologies and therapeutic approaches, and underpinned by a deeper understanding of tumour biology with individualised patient management the goal. In this review we describe the main biomarker categories in multiple myeloma and relate these to prognostic and therapeutic applications.
Article
Although the aberrant activity of fibroblast growth factor receptor 3 (FGFR3) is implicated in various cancers, the reported kinase inhibitors of FGFR3 tend to cause side effects resulting from the inhibitory activity on vascular endothelial growth factor receptor 2 (VEGFR2). Therefore, it is necessary to find a novel high‐selective inhibitor of FGFR3 over VEGFR2 from the small‐molecule compound database. In this study, integrated virtual screening protocols were established to screen for selective inhibitors of FGFR3 over VEGFR2 in Drugbank and Asinex databases by combining three‐dimensional pharmacophore model, molecular docking, molecular dynamics (MD) simulation, and molecular mechanics Poisson–Boltzmann surface area (MMPBSA) calculations. Finally, it is found that Asinex‐5082, as an octahydropyrrolo[3,2‐b] pyridin derivative, has larger binding free energy with FGFR3 (−39.3 kcal/mol) than reference drug Erdafitinib (−29.9 kcal/mol), while cannot bind with VEGFR2, resulting in considerable inhibitory selectivity. This is because Asinex‐5082, unlike Erdafitinib, has not m‐dimethoxybenzene with large steric hindrance, thus can enter the larger ATP‐binding pocket of FGFR3 with DFG‐in conformation to form hydrophobic interaction with residues Met529, Ile539, and Tyr557 as well as hydrogen bond with Ala558. On the other hand, due to the fact that the benzodioxane and N‐heterocyclic rings are connected by carbonyl (C=O), Asinex‐5082 cannot rotate freely so as to enter the smaller ATP binding pocket of VEGFR2 on the DFG‐out conformation. The lead molecule Asinex‐5082 may facilitate the rational design and development of novel selective inhibitors of FGFR3 over VEGFR2 as anticancer drugs.
Article
Multiple myeloma (MM) is a clonal plasma cell malignancy with a heterogeneous disease course. Insights into the genetics of the disease have identified certain high-risk cytogenetic features that are associated with adverse outcomes. While the advances in therapy have translated into dramatic improvements in the outcome of patients with MM, those with high-risk genetic features continue to perform poorly. This has resulted in a need for clinical trials focusing on the high-risk subgroup of MM as they search for additional biomarkers and therapeutic targets continue. In this review, we discuss the currently existing data on prognostic and predictive biomarkers in MM and speculate the role of treatment stratification based on the genetic features of the disease.
Article
Full-text available
Fibroblast growth factor (FGF) signaling regulates tumor growth and vascularization and partly mediates antiangiogenic escape from vascular endothelial growth factor receptor (VEGFR) inhibitors. Dovitinib (TKI258) is a tyrosine kinase inhibitor (TKI) that inhibits FGF receptor (FGFR), VEGFR, and platelet-derived growth factor receptor, which are known drivers of antiangiogenic escape, angiogenesis, and tumor growth in renal cell carcinoma (RCC). Patients with advanced or metastatic RCC were treated with oral dovitinib 500 mg/day (5-days-on/2-days-off schedule). The study population was enriched for patients previously treated with a VEGFR TKI and a mammalian target of rapamycin (mTOR) inhibitor. Of 67 patients enrolled, 55 patients (82.1%) were previously treated with ≥ 1 VEGFR TKI and ≥ 1 mTOR inhibitor (per-protocol efficacy set). The 8-week overall response rate and disease control rate in this population were 1.8% and 52.7%, respectively. Disease control rate during the entire study period was 56.4% (50.9% ≥ 4 months). Median progression-free survival and overall survival in the entire population were 3.7 and 11.8 months, respectively. Pharmacodynamic analyses demonstrated dovitinib-induced inhibition of VEGFR (as determined by increased levels of placental growth factor and decreased levels of soluble VEGFR2) and FGFR (as determined by increased FGF23 serum measures). The most frequently reported treatment-related adverse events of all grades included nausea (65.7%), diarrhea (62.7%), vomiting (61.2%), decreased appetite (47.8%), and fatigue (32.8%). Dovitinib was shown to be an effective and tolerable therapy for patients with metastatic RCC who had progressed following treatment with VEGFR TKIs and mTOR inhibitors.
Article
Full-text available
Key Points Pomalidomide plus low-dose dexamethasone significantly improved PFS vs pomalidomide alone in relapsed and refractory multiple myeloma. Pomalidomide plus low-dose dexamethasone is an important new treatment option for RRMM patients who have received multiple prior therapies.
Article
Full-text available
Several cytogenetic abnormalities are associated with poor outcomes in multiple myeloma (MM). We prospectively analyzed the impact of cytogenetic abnormalities on outcomes during the phase 2 PX-171-003-A1 study of single-agent carfilzomib for relapsed and refractory MM. In the response-evaluable population (257/266), fluorescence in situ hybridization (FISH)/conventional cytogenetic profiles were available for 229 patients; 62 (27.1%) had high-risk cytogenetics-del 17p13, t(4;14), or t(14;16) by interphase FISH, or deletion 13 or hypodiploidy by metaphase cytogenetics-and 167 (72.9%) had standard-risk profiles. Generally, baseline characteristics were similar between subgroups, but ISS stage III disease was more common in high- versus standard-risk patients (41.9 vs 27.5%) as was ECOG performance status 1/2 (85.5 vs 68.3%). Overall response was comparable between subgroups (25.8 vs 24.6%, respectively; P=0.85), while time-to-event endpoints showed a trend of shorter duration in high-risk patients, including median duration of response (5.6 months [95% CI 3.7-7.8] vs 8.3 months [95% CI 5.6-12.3]) and overall survival (9.3 [95% CI 6.5-13.0] vs 19.0 months [95% CI 15.4-NE]; P=0.0003). Taken together, these findings demonstrate that single-agent carfilzomib is efficacious and has the potential to at least partially overcome the impact of high-risk cytogenetics in heavily pre-treated patients with MM.Leukemia accepted article preview online, 14 May 2013; doi:10.1038/leu.2013.152.
Article
Full-text available
The outcome of patients with myeloma has improved significantly in the past decade with the incorporation of the immunomodulatory drugs thalidomide and lenalidomide and the proteasome inhibitor, bortezomib. Considering nearly all patients relapse, myeloma remains an active area of investigation. There are several promising classes of agents including next generation immunomodulatory agents, proteasome inhibitors, antibody and antitumor immunotherapy approaches that are being evaluated. This article provides an overview on the therapeutic strategies in the treatment of multiple myeloma.
Article
Full-text available
Cancer incidence and mortality estimates for 25 cancers are presented for the 40 countries in the four United Nations-defined areas of Europe and for the European Union (EU-27) for 2012. We used statistical models to estimate national incidence and mortality rates in 2012 from recently-published data, predicting incidence and mortality rates for the year 2012 from recent trends, wherever possible. The estimated rates in 2012 were applied to the corresponding population estimates to obtain the estimated numbers of new cancer cases and deaths in Europe in 2012. There were an estimated 3.45 million new cases of cancer (excluding non-melanoma skin cancer) and 1.75 million deaths from cancer in Europe in 2012. The most common cancer sites were cancers of the female breast (464,000 cases), followed by colorectal (447,000), prostate (417,000) and lung (410,000). These four cancers represent half of the overall burden of cancer in Europe. The most common causes of death from cancer were cancers of the lung (353,000 deaths), colorectal (215,000), breast (131,000) and stomach (107,000). In the European Union, the estimated numbers of new cases of cancer were approximately 1.4 million in males and 1.2 million in females, and around 707,000 men and 555,000 women died from cancer in the same year. These up-to-date estimates of the cancer burden in Europe alongside the description of the varying distribution of common cancers at both the regional and country level provide a basis for establishing priorities to cancer control actions in Europe. The important role of cancer registries in disease surveillance and in planning and evaluating national cancer plans is becoming increasingly recognised, but needs to be further advocated. The estimates and software tools for further analysis (EUCAN 2012) are available online as part of the European Cancer Observatory (ECO) (http://eco.iarc.fr). Copyright © 2013 Elsevier Ltd. All rights reserved.
Article
The involvement of fibroblast growth factor-2 (FGF-2) in the biology of small cell lung cancer (SCLC) has not previously been investigated. Here we report that FGF-2 prevented etoposide-induced apoptosis in H-510 SCLC cells. Phosphatidylinositol 3-kinase/protein kinase B signaling did not mediate this effect because FGF-2 failed to activate phosphatidylinositol 3-kinase or protein kinase B. In contrast, the mitogen-activated extracellularly regulated kinase kinase (MEK) was crucial for this response because its inhibition abolished the prosurvival properties of FGF-2. Moreover, in H-69 SCLC cells, the failure of FGF-2 to prevent etoposide-induced apoptosis correlated with uncoupling from MEK activation. However, the introduction of an activated MEK rendered these cells resistant to etoposide killing. Cell rescue relied on de novo protein synthesis, and the anti-apoptotic proteins Bcl-XL and Bcl-2 were up-regulated in a MEK-dependent fashion within 4 h of FGF-2 treatment. Contrary to previous reports, we found that this up-regulation occurred at the translational rather than the transcriptional level. Indeed, actinomycin D failed to prevent up-regulation of Bcl-XL and Bcl-2, and FGF-2 did not increase the mRNA levels or the stability of these proteins. The induction of the pro-apoptotic protein Bad by etoposide was also blocked by FGF-2 in a MEK-dependent fashion. Thus, MEK/extracellularly regulated kinase signaling is critical in the coordinate modulation of both pro- and anti-apoptotic Bcl-2 family members by FGF-2.
Article
Each year, the American Cancer Society estimates the numbers of new cancer cases and deaths that will occur in the United States in the current year and compiles the most recent data on cancer incidence, mortality, and survival. Incidence data were collected by the National Cancer Institute, the Centers for Disease Control and Prevention, and the North American Association of Central Cancer Registries and mortality data were collected by the National Center for Health Statistics. A total of 1,665,540 new cancer cases and 585,720 cancer deaths are projected to occur in the United States in 2014. During the most recent 5 years for which there are data (2006-2010), delay-adjusted cancer incidence rates declined slightly in men (by 0.6% per year) and were stable in women, while cancer death rates decreased by 1.8% per year in men and by 1.4% per year in women. The combined cancer death rate (deaths per 100,000 population) has been continuously declining for 2 decades, from a peak of 215.1 in 1991 to 171.8 in 2010. This 20% decline translates to the avoidance of approximately 1,340,400 cancer deaths (952,700 among men and 387,700 among women) during this time period. The magnitude of the decline in cancer death rates from 1991 to 2010 varies substantially by age, race, and sex, ranging from no decline among white women aged 80 years and older to a 55% decline among black men aged 40 years to 49 years. Notably, black men experienced the largest drop within every 10-year age group. Further progress can be accelerated by applying existing cancer control knowledge across all segments of the population. CA Cancer J Clin 2014. (©) 2014 American Cancer Society, Inc.