ArticlePDF Available

Antitumor activity of lurbinectedin (PM01183) and doxorubicin in relapsed small-cell lung cancer: Results from a phase I study

Authors:
  • START Madrid - CIOCC

Abstract and Figures

Background: Lurbinectedin (PM01183) has synergistic antitumor activity when combined with doxorubicin in mice with xenografted tumors. This phase I trial determined the recommended dose (RD) of doxorubicin (bolus) and PM01183 (1-h intravenous infusion) on day 1 every 3 weeks (q3wk), and obtained preliminary evidence of antitumor activity for this combination in small-cell lung cancer (SCLC). Patients and methods: Patients with advanced solid tumors received doxorubicin and PM01183 following a standard dose escalation design and expansion at the RD. Twenty-seven patients had relapsed SCLC: 12 with sensitive disease (platinum-free interval ≥90 days) and 15 with resistant disease (platinum-free interval <90 days). Results: Doxorubicin 50 mg/m2 and PM01183 4.0 mg flat dose was the RD. In relapsed SCLC, treatment tolerance at the RD was manageable. Transient and reversible myelosuppression (including neutropenia, thrombocytopenia, and febrile neutropenia) was the main toxicity, managed with dose adjustment and colony-stimulating factors. Fatigue (79%), nausea/vomiting (58%), decreased appetite (53%), mucositis (53%), alopecia (42%), diarrhea/constipation (42%), and asymptomatic creatinine (68%) and transaminase increases (alanine aminotransferase 42%; aspartate aminotransferase 32%) were common, and mostly mild or moderate. Complete (n = 2, 8%) and partial response (n = 13, 50%) occurred in relapsed SCLC, mostly at the RD. Response rates at second line were 91.7% in sensitive disease [median progression-free survival (PFS)=5.8 months] and 33.3% in resistant disease (median PFS = 3.5 months). At third line, response rate was 20.0% (median PFS = 1.2 months), all in resistant disease. Conclusion: Doxorubicin 50 mg/m2 and PM01183 4.0 mg flat dose on day 1 q3wk has shown remarkable activity, mainly in second line, with manageable tolerance in relapsed SCLC, leading to further evaluation of this combination within an ongoing phase III trial.
Content may be subject to copyright.
ORIGINAL ARTICLE
Antitumor activity of lurbinectedin (PM01183) and
doxorubicin in relapsed small-cell lung cancer:
results from a phase I study
E. Calvo
1
, V. Moreno
2
, M. Flynn
3
, E. Holgado
1
, M. E. Olmedo
4
, M. P. Lopez Criado
5
, C. Kahatt
6
,
J. A. Lopez-Vilari~
no
6
, M. Siguero
6
, C. Fernandez-Teruel
6
, M. Cullell-Young
6
, A. Soto Matos-Pita
6
&
M. Forster
3
*
1
START Madrid – Oncology, HM CIOCC, Hospital Madrid Norte Sanchinarro, Madrid;
2
START Madrid – Oncology, FJD (Hospital Fundaci
on Jime´nez D
ıaz), Madrid,
Spain;
3
Department of Oncology, University College of London Hospital and UCL Cancer Institute, London, UK;
4
Department of Oncology, Hospital Ramon y Cajal,
Madrid;
5
Department of Oncology, M.D. Anderson Cancer Center, Madrid;
6
Clinical R&D, Pharma Mar, S.A., Colmenar Viejo, Madrid, Spain
*Correspondence to: Dr Martin Forster, Department of Oncology, University College of London Hospital, 235 Euston Road, London NW1 2BU, UK. Tel: þ44-020-3447-5085;
Fax: þ44-020-3447-9055; E-mail: m.forster@ucl.ac.uk
Background: Lurbinectedin (PM01183) has synergistic antitumor activity when combined with doxorubicin in mice with
xenografted tumors. This phase I trial determined the recommended dose (RD) of doxorubicin (bolus) and PM01183 (1-h
intravenous infusion) on day 1 every 3 weeks (q3wk), and obtained preliminary evidence of antitumor activity for this
combination in small-cell lung cancer (SCLC).
Patients and methods: Patients with advanced solid tumors received doxorubicin and PM01183 following a standard dose
escalation design and expansion at the RD. Twenty-seven patients had relapsed SCLC: 12 with sensitive disease (platinum-free
interval 90 days) and 15 with resistant disease (platinum-free interval <90 days).
Results: Doxorubicin 50 mg/m
2
and PM01183 4.0mg flat dose was the RD. In relapsed SCLC, treatment tolerance at the RD was
manageable. Transient and reversible myelosuppression (including neutropenia, thrombocytopenia, and febrile neutropenia)
was the main toxicity, managed with dose adjustment and colony-stimulating factors. Fatigue (79%), nausea/vomiting (58%),
decreased appetite (53%), mucositis (53%), alopecia (42%), diarrhea/constipation (42%), and asymptomatic creatinine (68%) and
transaminase increases (alanine aminotransferase 42%; aspartate aminotransferase 32%) were common, and mostly mild or
moderate. Complete (n¼2, 8%) and partial response (n¼13, 50%) occurred in relapsed SCLC, mostly at the RD. Response rates
at second line were 91.7% in sensitive disease [median progression-free survival (PFS)¼5.8 months] and 33.3% in resistant
disease (median PFS ¼3.5 months). At third line, response rate was 20.0% (median PFS ¼1.2 months), all in resistant disease.
Conclusion: Doxorubicin 50 mg/m
2
and PM01183 4.0 mg flat dose on day 1 q3wk has shown remarkable activity, mainly in
second line, with manageable tolerance in relapsed SCLC, leading to further evaluation of this combination within an ongoing
phase III trial.
Key words:lurbinectedin, PM01183, small-cell lung cancer, phase I study
Introduction
The synthetic tetrahydroisoquinoline lurbinectedin (PM01183;
Pharma Mar S.A., Colmenar Viejo, Madrid, Spain) has broad
in vitro activity in the low nanomolar range [1]. PM01183 inhibits
active transcription in tumor cells through binding to CG-rich
sequences, irreversible stalling and degradation of elongating RNA
polymerase II on the DNA template, generation of XPF-dependent
single- and double-strand DNA breaks, and subsequent apoptosis
[2]. PM01183 also has a selective apoptotic-inducing effect on
mononuclear phagocytes, and inhibits the production of inflamma-
tory cytokines by these cells [3].
A study in mice bearing xenografted human tumors found
strong dose-dependent antitumor activity for the combination of
V
CThe Author 2017. Published by Oxford University Press on behalf of the European Society for Medical Oncology.
This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0/), which permits non-commercial re-use,
distribution, and reproduction in any medium, provided the original work is properly cited. For commercial re-use, please contact journals.permissions@oup.com
Annals of Oncology 28: 2559–2566, 2017
doi:10.1093/annonc/mdx357
Published online 14 July 2017
Downloaded from https://academic.oup.com/annonc/article-abstract/28/10/2559/3965270/Antitumor-activity-of-lurbinectedin-PM01183-and
by guest
on 02 October 2017
PM01183 and doxorubicin, with a synergistic combination index
1. Doxorubicin has a different mechanism of action and its clin-
ical toxicity profile does not completely overlap that of PM01183
[4]. A prior phase I trial found antitumor activity for single-agent
PM01183 in patients with advanced solid tumors [5].
Based on these findings, this phase I trial was designed to deter-
mine the recommended dose (RD), and evaluate the safety and
antitumor activity of doxorubicin combined with PM01183 ad-
ministered every three weeks (q3wk) in selected advanced solid
tumors. Tumor types for this study were selected based on pre-
clinical evidence of potential activity of PM01183 and on the
standard clinical use of doxorubicin in solid tumor patients. Of
note, small-cell lung cancer (SCLC) cells are addicted to lineage-
specific and proto-oncogenic transcription factors that support
cell growth, hence further suggesting that PM01183 could play a
role in the treatment of this tumor type. Due to the relevance of
the antitumor activity observed during escalation, the results
shown here are focused on relapsed SCLC patients.
Materials and methods
Patients were recruited in Spain and the UK. The study followed ICH
Good Clinical Practice guidelines, and was approved by the respective
Research Ethics Committees. Written informed consent was obtained
from all patients before study-specific procedures. The study is registered
at http://www.clinicaltrials.gov as NCT01970540.
Eligibility criteria
Eligible patients were aged 18–75 years with confirmed advanced solid
tumors pre-treated with 1–2 cytotoxic-containing chemotherapy regimens
for advanced disease (not anthracyclines); who had recovered from previous
toxicities; 3 weeks since last anticancer therapy and 6weeks since sys-
temic nitrosoureas and mitomycin C; life expectancy 3 months; Eastern
Cooperative Oncology Group performance status 1; normal left ventricu-
lar ejection fraction (LVEF); and adequate bone marrow, hepatic and renal
function.
Patients were excluded if they had symptomatic progressive or
corticosteroid-requiring brain metastases or leptomeningeal involve-
ment; were pregnant or lactating women, or not using effective contra-
ception; had prior radiation therapy (>35% of bone marrow), prior
bone marrow/stem cell transplantation, relevant cardiac disease, alcohol
consumption or cirrhosis, active uncontrolled infection, or any disease
interfering with study outcome.
Study treatment
Treatment consisted of a fixed dose of doxorubicin 50mg/m
2
as bolus fol-
lowed by escalating doses of PM01183 intravenously (i.v.) over 1 h on day
1 q3wk. Doxorubicin was administered to a maximum cumulative dose of
450 mg/m
2
. After reaching this, patients were switched to PM01183 alone
at its single-agent RD of 7.0 mg flat dose (FD) on day 1 q3wk to prevent
doxorubicin-induced cardiomyopathy [6]. Commercially available doxo-
rubicin was provided. PM01183 was supplied as a lyophilized powder con-
centrate, reconstituted in sterile water for injection, and diluted with
glucose 5% or sodium chloride 0.9% solution. All patients received stand-
ard antiemetic prophylaxis before each infusion. Treatment was given until
disease progression, unacceptable toxicity, intercurrent illness precluding
study continuation, patient refusal and/or non-compliance with study re-
quirements, treatment delay >15 days (except if clear clinical benefit), and
requirement of >2dosereductions.
Dose escalation
Dose escalation followed a standard 3 þ3 design. Doxorubicin dose was
standard for combination schedules. The starting PM01183 dose (3.5 mg
FD daily) was 50% the RD defined for PM01183 alone on day 1 q3wk [5].
Dose-limiting toxicities were evaluated during cycle 1 and comprised
grade 4 neutropenia >7 days, grade 3 febrile neutropenia, grade 4
thrombocytopenia (or grade 3 requiring transfusion), grade 4 transamin-
ase increase (or grade 3 >14 days), grade 3 creatinine phosphokinase
increase, and clinically relevant grade 3 non-hematological toxicities.
Study assessments
Hematology and biochemistry tests were done at baseline, weekly during
cycle 1, and before each PM01183 infusion and on day 10 during subse-
quent cycles. Electrocardiograms and LVEF assessments were done at
baseline and repeated at doxorubicin discontinuation or if clinically
indicated.
Antitumor activity was evaluated every two cycles according to the
Response Evaluation Criteria In Solid Tumors (RECIST) v.1.1 [7]. Overall
response rate (ORR) was the percentage of patients with complete (CR) or
partial response (PR), and disease control rate (DCR) was the percentage
of patients with response or stable disease (SD). Time-to-event parameters
were progression-free survival (PFS) and duration of response.
Adverse events (AEs) and laboratory abnormalities were graded with
the National Cancer Institute Common Terminology Criteria for
Adverse Events (NCI-CTCAE) v.4 [8] and coded using the Medical
Dictionary for Regulatory Activities (MedDRA) v.14.1.
Statistical analysis
Continuous variables were presented with summary statistics and cat-
egorical variables in frequency tables. Time-to-event variables were calcu-
lated using Kaplan–Meier approach. Binomial exact distribution was
used to calculate 95% confidence intervals (95% CIs) for categorical
variables.
Results
Dose escalation
Seventy-four patients were included during dose escalation. Most
common tumor types were SCLC (n¼28, 38%), endometrial can-
cer (n¼15, 20%), neuroendocrine tumors (n¼9, 12%) and soft
tissue sarcoma (n¼8, 11%). Four dose levels were evaluated.Most
DLTs were hematological and mostly occurred at the highest dose
level (doxorubicin 50 mg/m
2
and PM01183 5.0 mg FD), which was
defined as the maximum tolerated dose. The RD was determined
at doxorubicin 50 mg/m
2
and PM01183 4.0 mg FD [9].
Characteristics of SCLC patients and treatment
All 28 SCLC patients were treated with doxorubicin/PM01183,
and 27 of them were included in the present analysis (Table 1);
one patient was considered not evaluable because he had lepto-
meningeal carcinomatosis in cycle 1 that had not been evaluated
at baseline. Median age was 62 years (range, 48–73 years) and
many were male (n¼17, 63%). At baseline, 18 (67%) had bulky
disease (target lesion >50 mm), 6 (22%) had brain metastases,
and 13 (48%) had received prophylactic cranial irradiation.
Twelve patients (44%) had sensitive disease [defined as
platinum-free interval (PFI) 90 days] and received doxorubicin/
PM01183 as second-line therapy. The other 15 (56%) had resistant
Original article Annals of Oncology
2560 | Calvo et al. Volume 28 | Issue 10 | 2017
Downloaded from https://academic.oup.com/annonc/article-abstract/28/10/2559/3965270/Antitumor-activity-of-lurbinectedin-PM01183-and
by guest
on 02 October 2017
disease (defined as PFI <90 days) and received it as second line
(n¼9), third line (n¼5) or fourth line (n¼1).
Six patients were treated below the RD, 19 at the RD (doxorubi-
cin 50 mg/m
2
þPM011834.0 mg FD), and 2 above the RD. A total
of 126 cycles of doxorubicin/PM01183 were administered at all
dose levels (101 cycles at the RD). Median relative dose intensity at
the RD was 95.8% (range, 78.6%–103.1%) for doxorubicin, and
94.5% (range, 70.1%–101.2%) for PM01183. Eight patients
(29.6%) received 45 cycles of single-agent PM01183 after doxo-
rubicin discontinuation [median relative dose intensity¼87.4%
(range, 76.5%–100.0%), including seven patients previously
treated with the combination at the RD]. Total median of cycles
(combination and single-agent PM01183) per patient was 4 (range,
1–33 cycles) at all dose levels and 6 (range, 1–33 cycles) at the RD.
Table 1. Baseline characteristics of patients with relapsed SCLC
Doxorubicin1PM01183
Second line Third or fourth line
a
Sensitive (n512) Resistant (n59) Resistant (n56)
n%n%n%
Gender
Male 10 83 6 67 1 17
Female 2 17 3 33 5 83
Median age (range) (years) 62.5 (48–73) 62.0 (50–70) 58.5 (50–63)
ECOG performance status
0 8 67 1 11 3 50
1 4 33 8 89 3 50
Median BSA (range) (m
2
) 1.9 (1.3–2.0) 1.9 (1.7–2.3) 1.7 (1.5–1.8)
Median albumin (range) (g/dl) 4.1 (3.2–4.8) 4.2 (2.5–4.5) 4.0 (3.6–4.5)
Median hemoglobin (range) (g/dl) 13.5 (10.5–15.2) 12.1 (9.2–17.7) 11.6 (9.6–13.2)
Median LDH (range) (x ULN) 0.6 (0.3–1.6) 1.1 (0.5–3.4) 1.7 (0.7–2.8)
Smoker
Current 2 17 4 44 2 33
Former
b
10 83 5 56 4 67
Median number of sites of disease involvement (range) 2.5 (1–4) 3 (1–5) 2.5 (2–7)
Metastasis at baseline
Visceral 6 50 8 89 3 50
Liver 6 50 3 33 2 33
Bone 3 25 4 44 2 33
CNS 4 44 2 33
Bulky disease (any target lesion >50 mm) 5 42 8 89 5 83
PCI 10 83 1 11 2 33
Prior therapy
Platinum compound 12 100 9 100 6 100
Etoposide 12 100 9 100 6 100
Topotecan – – – 3 50
Irinotecan – – – 2 33
Other (investigational drugs) 1
c
81
d
11 –
PFI
e
<90 days 9 100 6 100
90–179 days 7 58
180 days 5 42
TTP to prior therapy (months) 7.7 (2.1–13.6) 4.5 (1.2–6.0) 3.3 (1.6–6.4)
a
Only one patient with resistant disease received the combination as fourth-line therapy.
b
Defined as an adult patient who used to smoke but who had quit smoking by the time of registration into this trial.
c
LY2940680.
d
Nivolumab.
e
Time from the last prior platinum therapy before inclusion in the study.
BSA, body surface area; CNS, central nervous system; ECOG, Eastern Cooperative Oncology Group; LDH, lactate dehydrogenase; PCI, prophylactic cranial
irradiation; PFI, platinum-free interval; SCLC, small-cell lung cancer; TTP, time to progression; ULN, upper limit of normal.
Annals of Oncology Original article
Volume 28 | Issue 10 | 2017 doi:10.1093/annonc/mdx357 | 2561
Downloaded from https://academic.oup.com/annonc/article-abstract/28/10/2559/3965270/Antitumor-activity-of-lurbinectedin-PM01183-and
by guest
on 02 October 2017
Efficacy
Twenty-six patients were evaluable for efficacy. Overall, ORR was
57.7% (95% CI, 36.9%–76.6%) and DCR was 69.2% (95% CI,
48.2%–85.6%) (Table 2).
As second line, 14 of 21 patients (ORR ¼66.7%; 95% CI,
43.0%–85.4%) had response. ORR was 91.7% (95% CI, 61.5%–
99.8%) in sensitive disease [two CRs (16.7%) and nine PRs
(75.0%) among 12 patients], and 33.3% (95% CI, 7.5%–70.1%)
in resistant disease (three PRs among nine patients). Most re-
sponses were confirmed (14 of 15). One patient with sensitive dis-
ease and two with resistant disease had SD; DCR was 100.0%
(95% CI, 73.5%–100.0%) in sensitive disease and 55.6% (95%
CI, 21.2%–86.3%) in resistant disease (Table 2).
As third line (all resistant disease), ORR/DCR was 20.0% (95%
CI, 0.5%–71.6%) (one PR among five patients) (Table 2).
Tumor shrinkage occurred in 20 of 26 patients (77%) (Figure
1): all 12 with sensitive disease, and 7 of 9 (78%) with resistant
disease as second line; and 1 of 5 (20%) with resistant disease as
third line. Target lesion size decrease was maintained in seven of
eight (88%) patients treated with single-agent PM01183.
Median PFS was 4.1 months (95% CI, 1.4–5.8 months) in all
patients: 4.7 months (95% CI, 3.5–8.4 months) as second line
[sensitive disease: 5.8 months (95% CI, 3.6–10.9 months); resist-
ant disease: 3.5 months (95% CI, 1.1–8.0 months)], and
1.2 months (95% CI, 0.6–4.1 months) as third line (Figure 2).
Seven patients achieved PFS >6 months (Figure 1), including one
with sensitive disease who received 33 cycles as second line and
had CR and a PFS of 23.6 months before discontinuing due to
disease progression (time-to-progression after first-line therapy
had been 2.1 months).
Median duration of response was 4.5 months (95% CI, 2.3–
7.8 months) in all patients and as second line [sensitive disease:
4.5 months (95% CI, 2.1–9.1 months); resistant disease: 6.7 months
(95% CI, 2.3–7.2 months)]; and 2.8 months in one patient as third
line.
Twelve responses (including both CRs) occurred at the RD.
ORR was 64.7% (95% CI, 38.3%–85.7%) as second line [sensitive
disease: 88.9% (95% CI, 51.7%–99.7%); resistant disease: 37.5%
(95% CI, 8.5%–75.5%)]; and 50.0% (95% CI, 1.2%–98.7%) as
third line. Median PFS was 4.1 months (95% CI, 1.4–9.0 months)
as second line [sensitive disease: 9.0 months (95% CI, 3.3–
11.7 months); resistant disease: 2.4 months (95% CI, 1.1–
8.0 months)]; and 2.4 months (95% CI, 0.6–4.1 months) as third
line. Median duration of response was 6.7 months (95% CI, 2.1–
9.1 months) as second line [sensitive disease: 5.6 months (95%
CI, 1.0–9.8 months); resistant disease: 6.7 months (95% CI, 2.3–
7.2 months)].
Six patients at all dose levels had brain metastases at baseline
that were considered non-target lesions for the evaluation of the
disease. Of these six patients, four received the doxorubicin/
PM01183 combination as second-line therapy and two as third-
line therapy. All six patients discontinued treatment due to dis-
ease progression. In three of these patients, disease progression
consisted of the appearance of new tumor lesions after two to six
cycles while the brain lesions at baseline remained apparently sta-
ble (n¼2) or were not evaluated again while on treatment
(n¼1). Two patients showed progression in baseline brain le-
sions after two and five cycles. Finally, one patient had clinical de-
terioration after three cycles while the baseline brain lesions
remained apparently stable.
Toxicity
All patients were evaluable for safety. At the RD, the most com-
mon combination-related non-hematological AEs were fatigue
(n¼15; 79%), nausea/vomiting (n¼11; 58%), decreased
Table 2. Best tumor response according to Response Evaluation Criteria In Solid Tumors (RECIST)
Doxorubicin 1PM01183 Total (n526)
a
Second line Third line
Sensitive (n512) Resistant (n59) Resistant (n55)
n%n%n%n%
CR 2 16.7 – 2 7.7
PR 9
b
75.0 3 33.3 1 20.0 13 50.0
SD 1 8.3 2 22.2 – 3 11.5
PD 4 44.4 4 80.0 8 30.8
ORR (95% CI) 91.7% (61.5%–99.8%) 33.3% (7.5%–70.1%) 20.0% (–) 57.7% (36.9%–76.6%)
DCR (95% CI) 100.0% (73.5%–100.0%) 55.6% (21.2%–86.3%) 20.0% (–) 69.2% (48.2%–85.6%)
Median duration of response
(months) (95% CI)
4.5 (2.1–9.1) 6.7 (2.3–7.2) 2.8 (–) 4.5 (2.3–7.8)
a
One patient, who was the only one in this study who received the combination as fourth-line therapy, was not evaluable for efficacy and has been
excluded.
b
Partial response could not be confirmed in one patient.
CI, confidence interval; CR, complete response; DCR, disease control rate; ORR, overall response rate; PD, progressive disease; PR, partial response; SD, stable
disease.
Original article Annals of Oncology
2562 | Calvo et al. Volume 28 | Issue 10 | 2017
Downloaded from https://academic.oup.com/annonc/article-abstract/28/10/2559/3965270/Antitumor-activity-of-lurbinectedin-PM01183-and
by guest
on 02 October 2017
100
80
60
40
20
1.2 1.2 1.2 1.2 1.4
0.6 1.1 3.8
1.2
9.6
4.8
3.5 3.3 4.1
8.0
5.8
4.13.6
10.9
8.4 9.0 11.7
23.6
4.6
2.7
4.7*
**
*****
–20
–40
–60
–80
0
3
6
9
12
15
Progression free survival (months)
18
21
24
27
– 100
2nd/R
2nd/R
2nd/R
2nd/R
2nd/R
2nd/R
2nd/R
3rd/R
2nd/R
2nd/R
2nd/R
2nd/S
2nd/S
2nd/S
2nd/S
2nd/S
2nd/S
2nd/S
2nd/S
2nd/S
2nd/S
2nd/S
2nd/S
2nd/S
3rd/R
3rd/R
3rd/R
3rd/R
3rd/R
0RD
RD RD RD RD RD
RD
RD RD RD RD <RD <RD RD RD RD RD RD RD RD RD
<RD<RD <RD
100
80
60
40
20
Chan
g
e from baseline (%)
–20
–40
–60
–80
– 100
CR
PR
SD
PD
0
<RD
>RD
Figure 1. Waterfall plot showing maximum variation of target lesions and progression-free survival in patients with at least one radiological
tumor assessment (n¼26). Sixteen patients had target lesion decrease >30%: 15 with CR or PR, and 1 with PD who had response in extracra-
nial lesions and disease progression in the brain. Red stars ¼treatment switch to PM01183 alone. 2nd/R, second-line therapy and resistant
disease; 2nd/S, second-line therapy and sensitive disease; 3rd/R, third-line therapy and resistant disease; CR, complete response; PD, progres-
sive disease; PR, partial response; SD, stable disease.
1.0
0.9
0.8
2nd/S
2nd/R
3rd/R
0.7
0.6
0.5
0.4
Cumulative probability
0.3
0.2
0.1
0.0
036912
Time (months)
Global (N=26 C=1) Median PFS: 4.1 months (95%CI: 1.4-5.8)
2nd/S (N=12 C=1) Median PFS: 5.8 months (95%CI: 3.6-10.9)
2nd/R (N=9 C=0) Median PFS: 3.5 months (95%CI: 1.1-8.0)
Censored
3rd/R (N=5 C=0) Median PFS: 1.2 months (95%CI: 0.6-4.1)
15 18 21 24
Figure 2. Kaplan–Meier plot of progression-free survival with doxorubicin/PM01183. 2nd/R, second-line therapy and resistant disease; 2nd/S, second-
line therapy and sensitive disease; 3rd/R, third-line therapy and resistant disease; C, censored; CI, confidence interval; PFS, progression-free survival.
Annals of Oncology Original article
Volume 28 | Issue 10 | 2017 doi:10.1093/annonc/mdx357 | 2563
Downloaded from https://academic.oup.com/annonc/article-abstract/28/10/2559/3965270/Antitumor-activity-of-lurbinectedin-PM01183-and
by guest
on 02 October 2017
appetite, mucositis (n¼10 each; 53%), alopecia and diarrhea/con-
stipation (n¼8 each; 42%) (Table 3). Most AEs were grade 1/2.
Grade 3 related AEs comprised febrile neutropenia, fatigue,
mucositis and pneumonia. The only related grade 4 AE was one fe-
brile neutropenia episode. Regardless of relationship, hematolo-
gical abnormalities were common and severe cases comprised
neutropenia (n¼18; 95%) [grade 4 lasting a median of 4 days
(range, 2–13 days)], leukopenia (n¼15; 79%), anemia (n¼9;
47%), and thrombocytopenia (n¼5; 26%) (Table 3). Most bio-
chemical abnormalities were grade 1/2; the most frequent were
creatinine (n¼13; 68%), alanine aminotransferase (n¼8; 42%)
and aspartate aminotransferase (n¼6; 32%) increases. Grade 3
increases in transaminases and bilirubin occurred in one patient
and were concomitant with bile duct obstruction unrelated to
treatment.
At the RD, nine patients required red blood cell transfusions,
one had platelets transfusions, and seven required granulocyte
colony-stimulating factor support. Treatment-related AEs re-
sulted in 12 delays, two dose reductions, and one discontinu-
ation. No toxic deaths occurred.
Most common AEs related to single-agent PM01183 were fa-
tigue (all eight patients), decreased appetite (n¼4; 50%) and alo-
pecia (n¼3; 38%). Most were grade 1/2; single episodes of grade
3 febrile neutropenia, grade 3 hypomagnesemia, and grade 4
Table 3. Treatment-related adverse events and laboratory abnormalities regardless of relationship at the RD (10%of patients or grade 3/4)
Doxorubicin 50.0 mg/m
2
1PM01183 4.0 mg FD (n519)
NCI-CTCAE grade
3 4 All
n%n%n%
Treatment-related AEs
Alopecia – – – 8 42
Conjunctivitis – – – 2 11
Decreased appetite – – – 10 53
Diarrhea/constipation – – – 8 42
Dizziness – – – 2 11
Dry mouth – – – 2 11
Dry skin – – – 2 11
Dysgeusia – – – 7 37
Esophageal candidiasis 1 5 1 5
Fatigue 2 11 – 15 79
Hypokalemia 1 5 – – 1 5
Hypomagnesemia 1 5 – – 1 5
Mucositis 2 11 – 10 53
Myalgia – – – 3 16
Nail disorder – – – 2 11
Nausea/vomiting – – – 11 58
Neutropenic infection 1 5 1 5
Pneumonia 2 11 – 2 11
Pyrexia – – – 2 11
Somnolence – – – 2 11
Hematological abnormalities
Anemia 9 47 – 18 95
Febrile neutropenia 4 21 1 5 5 26
Leukopenia 9 47 6 32 19 100
Neutropenia 3 16 15 79 19 100
Thrombocytopenia 2 11 3 16 17 90
Biochemical abnormalities
ALP increased – – – 5 26
ALT increased 1 5 8 42
AST increased 1 5 6 32
Bilirubin increased 1 5 5 26
Creatinine increased – – – 13 68
AE, adverse event; ALP, alkaline phosphatase; ALT, alanine aminotransferase; AST, aspartate aminotransferase; FD, flat dose; NCI-CTCAE, National Cancer
Institute Common Terminology Criteria for Adverse Events; RD, recommended dose.
Original article Annals of Oncology
2564 | Calvo et al. Volume 28 | Issue 10 | 2017
Downloaded from https://academic.oup.com/annonc/article-abstract/28/10/2559/3965270/Antitumor-activity-of-lurbinectedin-PM01183-and
by guest
on 02 October 2017
hypokalemia occurred. Severe hematological abnormalities com-
prised neutropenia (75% of patients), anemia (63%), leukopenia
and thrombocytopenia (50% each). All biochemical abnormal-
ities were grade 1/2.
Discussion
Remarkable antitumor activity was found for the doxorubicin/
PM01183 combination in relapsed SCLC. At the RD, ORR was
65% as second line (sensitive disease: 89%; resistant disease:
38%); and 50% as third line. Most responses occurred at the RD
and were durable. Median PFS was 5.8 months (sensitive disease)
and 3.5 months (resistant disease) as second line, and 1.2 months
as third line. Median duration of response was 4.5, 6.7 and
2.8 months, respectively. One CR lasted almost two years in a
patient with sensitive disease. Tumor shrinkage was maintained
in 88% of patients who switched to PM01183 alone.
Therapeutic options for SCLC relapsing after first-line therapy
are few. Currently approved second-line therapies resulted in
ORRs of 7%–44% (sensitive disease: 25%–53%; resistant disease:
10%–18%) and median PFS of 2.2–4.5 months in patients with
PFIs of 45–90 days or no brain metastases [1012].
Immunotherapy drugs currently under evaluation for relapsed
SCLC have resulted in ORRs of 10%–33% for the monoclonal
antibody nivolumab alone or with ipilimumab [13] and 33%
for the monoclonal antibody pembrolizumab in selected patients
with PDL1-positive disease [14]. The antibody-drug conjugate
rovalpituzumab tesirine has been described to have an ORR of
18% in relapsed SCLC, with an ORR of 38% in highly selected pa-
tients with high DLL3 expression [15]. In contrast, in the present
study second-line doxorubicin/PM01183 combination resulted
in higher ORRs compared to currently approved therapies in un-
selected patients with sensitive (91.7%) and resistant (33.3%) dis-
ease. Median PFS was also longer. Of note, the 5.8 months
achieved with second-line doxorubicin/PM01183 in sensitive dis-
ease was similar to the median PFS of 5.5 months reported with
platinum/etoposide rechallenge in a retrospective analysis [16].
Remarkably, one-third of patients in this current study showed
longer PFS with second-line doxorubicin/PM01183 than with
first-line therapy.
Reversible myelosuppression was the most common toxicity
with the doxorubicin/PM01183 combination. Episodes of severe
neutropenia, thrombocytopenia and febrile neutropenia were
transient and successfully managed with cycle delays, dose reduc-
tions and colony-stimulating factors. Most other toxicities were
mild or moderate. Median dose intensities at the RD were 96%
for doxorubicin and 95% for PM01183. No toxic deaths
occurred.
Toxicities with doxorubicin/PM01183 were more frequent
than with single-agent PM01183 in a previous phase I study [5];
e.g. nausea/vomiting (58% versus 47%), mucositis (53% ver-
sus <10%) and alopecia (42% versus <10%). Severe hematolo-
gical toxicity was generally more frequent with doxorubicin/
PM01183 than with doxorubicin alone at 60 or 70 mg/m
2
q3wk
in other tumors [17,18] and similar than with approved second-
line therapies [19]. Some non-hematological toxicities were more
common with doxorubicin/PM01183 (anorexia, 53% versus
30%) than with doxorubicin alone; others occurred at similar
frequencies (nausea/vomiting, 58% versus 30%–75%; stomatitis/
mucositis, 53% versus 62%) or were less common (alopecia, 42%
versus 97%) [17,18]. The safety profile of single-agent PM01183
in this study generally agrees with that reported previously [5].
In summary, this doxorubicin/PM01183 q3wk regimen has re-
markable antitumor activity in relapsed SCLC that appears to be
meaningfully higher compared to standard second-line therapies,
particularly in sensitive disease. Tolerance suggested a positive
risk-benefit profile for doxorubicin/PM01183 in relapsed SCLC.
This trial is currently evaluating the combination in an expanded
cohort of SCLC patients at a reduced doxorubicin dose (40 mg/
m
2
) and with PM01183 transformed to a dose of 2.0 mg/m
2
to
improve safety over the profile described herein. An ongoing
randomized phase III trial is evaluating doxorubicin/PM01183
versus cyclophosphamide, doxorubicin and vincristine (CAV) or
topotecan, with primary colony-stimulating factor support as
second-line treatment of SCLC.
Funding
Pharma Mar S.A. (no grant numbers apply).
Disclosure
EC has been consultant for Novartis, Pierre Fabre, Boehringer
Ingelheim, EUSA and Seattle Genetics, and has participated in a
speaker’s bureau for Novartis. MPLC has been paid travel accom-
modation and expenses by Bristol. MF has been consultant for
Eli-Lilly, Pfizer, Clovis, Boehringer Ingelheim, Novartis, Merck,
AstraZeneca, BMS and MSD; has received research funding from
AstraZeneca and Boehringer Ingelheim; and has been paid travel
accommodation and expenses by Boehringer Ingelheim, BMS,
Celgene, Eli Lilly, Merck, MSD and Roche. In addition, MF is
supported by the UCL/UCLH NIHR Biomedical Research Centre
and runs early phase studies in the NIHR UCLH Clinical
Research Facility supported by the UCL ECMC. CK, MS, CFT,
MCY, and ASM-P are employees and stock owners of Pharma
Mar S.A. JAL-V is an employee of Pharma Mar S.A. All remaining
authors have declared no conflicts of interest.
References
1. Leal JF, Martinez-Diez M, Garcia-Hernandez V et al. PM01183, a new
DNA minor groove covalent binder with potent in vitro and in vivo anti-
tumour activity. Br J Pharmacol 2010; 161: 1099–1110.
2. Santamaria Nunez G, Robles CM, Giraudon C et al. Lurbinectedin spe-
cifically triggers the degradation of phosphorylated RNA polymerase II
and the formation of DNA breaks in cancer cells. Mol Cancer Ther 2016;
15: 1–14.
3. Belgiovine C, Bello E, Liguori M et al. Lurbinectedin reduces tumour-
associated macrophages and the inflammatory tumour microenviron-
ment in preclinical models. Br J Cancer 2017; doi: 10.1038/bjc.2017.205.
4. Malone H, Atassi G. DNA topoisomerasa targeting drugs: mechanisms
of actions and perspectives. Anticancer Drugs 1997; 8: 811–822.
5. Elez ME, Tabernero J, Geary D et al. First-in-human phase I study of
Lurbinectedin (PM01183) in patients with advanced solid tumors. Clin
Cancer Res 2014; 20: 2205–2214.
6. Von Hoff DD, Layard MW, Basa P et al. Risk factors for doxorubicin-
induced congestive heart failure. Ann Intern Med 1979; 91: 710–717.
Annals of Oncology Original article
Volume 28 | Issue 10 | 2017 doi:10.1093/annonc/mdx357 | 2565
Downloaded from https://academic.oup.com/annonc/article-abstract/28/10/2559/3965270/Antitumor-activity-of-lurbinectedin-PM01183-and
by guest
on 02 October 2017
7. Eisenhauer EA, Therasse P, Bogaerts J et al. New response evaluation cri-
teria in solid tumours: revised RECIST guideline (version 1.1). Eur J
Cancer 2009; 45: 228–247.
8. National Cancer Institute Common Terminology Criteria for Adverse
Events v.4.0 (CTCAE). At: https://evs.nci.nih.gov/ftp1/CTCAE/CTCAE_4.
03_2010-06-14_QuickReference_5x7.pdf (20 July 2017, date last accessed).
9. Calvo E, Foster M, Szyldergemajn S et al. Lurbinectedin (PM01183) in
combination with doxorubicin (DOX): preliminary results of a phase Ib
study. Eur J Cancer 2013; 49: 178–179.
10. Alvarado-Luna G, Morales-Espinosa D. Treatment for small cell lung can-
cer, where are we now?-a review. Transl Lung Cancer Res 2016; 5: 26–38.
11. Inoue A, Sugawara S, Yamazaki K et al. Randomized phase II trial com-
paring amrubicin with topotecan in patients with previously treated
small-cell lung cancer: North Japan Lung Cancer Study Group Trial
0402. J Clin Oncol 2008; 26: 5401–5406.
12. Jotte R, Conkling P, Reynolds C et al. Randomized phase II trial of
single-agent amrubicin or topotecan as second-line treatment in patients
with small-cell lung cancer sensitive to first-line platinum-based chemo-
therapy. J Clin Oncol 2011; 29: 287–293.
13. Antonia SJ, Lopez-Martin JA, Bendell J et al. Nivolumab alone and nivolu-
mab plus ipilimumab in recurrent small-cell lung cancer (CheckMate 032):
a multicentre, open-label, phase 1/2 trial. Lancet Oncol 2016; 17: 883–895.
14. Ott PA, Felip E, Hiret S et al. Pembrolizumab in patients with extensive-
stage small cell lung cancer: updated survival results from KEYNOTE-
028. J Thorac Oncol 2017; 12: S259.
15. Rudin CM, Pietanza MC, Bauer TM et al. Rovalpituzumab tesirine, a
DLL3-targeted antibody-drug conjugate, in recurrent small-cell lung
cancer: a first-in-human, first-in-class, open-label, phase 1 study. Lancet
Oncol 2017; 18: 42–51.
16. Genestreti G, Tiseo M, Kenmotsu H et al. Outcomes of platinum-
sensitive small-cell lung cancer patients treated with platinum/etoposide
rechallenge: a multi-institutional retrospective analysis. Clin Lung
Cancer 2015; 16: e223–e228.
17. Norris B, Pritchard KI, James K et al. Phase III comparative study of
vinorelbine combined with doxorubicin versus doxorubicin alone in dis-
seminated metastatic/recurrent breast cancer: National Cancer Institute
of Canada Clinical Trials Group Study MA8. J Clin Oncol 2000; 18:
2385–2394.
18. Thigpen JT, Brady MF, Homesley HD et al. Phase III trial of doxorubicin
with or without cisplatin in advanced endometrial carcinoma: a gyneco-
logic oncology group study. J Clin Oncol 2004; 22: 3902–3908.
19. von Pawel J, Schiller JH, Shepherd FA et al. Topotecan versus cyclophos-
phamide, doxorubicin, and vincristine for the treatment of recurrent
small-cell lung cancer. J Clin Oncol 1999; 17: 658–667.
Original article Annals of Oncology
2566 | Calvo et al. Volume 28 | Issue 10 | 2017
Downloaded from https://academic.oup.com/annonc/article-abstract/28/10/2559/3965270/Antitumor-activity-of-lurbinectedin-PM01183-and
by guest
on 02 October 2017
... A phase I study evaluated initially lurbinectedin in combination with DOX in patients with pretreated selected advanced solid tumors [15,16]. Lurbinectedin, a trabectedin analog with improved hepatic toxicity, showed promising results in early trials for STS. ...
... The two lurbinectedin plus DOX schedules evaluated during the phase IIb stage have previously shown promising antitumor activity in patients with metastatic LMS [17,19]. The lurbinectedin dose of 2.2 mg/m 2 used in the Experimental arm A is close to the 2.0 mg/m 2 dose used in a previous phase II study [17], and is similar to the 4.0 mg flat dose (FD) defined as RD in an early phase I study, as 4.0 mg FD corrected by the median patient BSA observed in that study (1.8 m 2 ) results in a BSAbased dose of 2.2 mg/m 2 [15]. In these two previous trials, the DOX dose was 50 mg/m 2 . ...
Article
Full-text available
Previous phase I/II trials indicate promising activity of lurbinectedin plus doxorubicin (DOX) in leiomyosarcoma (LMS). We describe here the rationale and design of SaLuDo, an open label, randomized, multicenter, seamless phase IIb/III study to evaluate the antitumor activity and safety of lurbinectedin plus DOX versus DOX alone in the first-line setting of metastatic LMS. The phase IIb stage will evaluate two schedules of the combination for the phase III stage given every 3 weeks (q3wk): DOX 50 mg/m2 plus lurbinectedin 2.2 mg/m2, and DOX 25 mg/m2 plus lurbinectedin 3.2 mg/m2. The control arm will be DOX 75 mg/m2 q3wk. The primary endpoint is progression-free survival by independent review; overall survival is the key secondary endpoint. Clinical trial registration: www.clinicaltrials.gov identifier is NCT06088290.
... Calvo et al. reported the first clinical data in a phase I study where lurbinectedin with a flat dose of 4.0 mg showed a remarkable activity in combination with doxorubicin at the dose of 50 mg/m 2 in patients with relapsed SCLC, particularly in patients with platinum sensitive disease. The overall response rate (ORR) reached 57.7% while the median progression-free survival (PFS) was 4.1 and 7.9 months, respectively [29]. These data were also supported by the results reported by Trigo et al. in a phase II, single arm, open-label basket trial investigating the safety and efficacy of single-agent lurbinectedin (3.2 mg/m 2 every 3 weeks) in 105 SCLC patients relapsing following first-line chemotherapy. ...
... Moreover, another phase Ib study on the efficacy of lurbinectedin in combination with doxorubicin (50 mg/m 2 ) included eight patients with STS. Out of these eight patients, four STS patients exhibited a PFS above 3 months with one patient with synovial sarcoma actually reaching a complete response [29]. ...
Article
Full-text available
Opinion statement Soft tissue sarcoma (STS), a substantial group of aggressive and rare tumors with tissue heterogeneity, is infrequently represented in clinical trials with an urgent necessity for newer treatment options. Lurbinectedin, an analog of trabectedin, is currently approved, in various countries, as a single agent, for the treatment of patients with relapsed small cell lung cancer (SCLC). However, preclinical and phase I and phase II trials have demonstrated the efficacy of lurbinectedin in different tumor types, including STS. The better understanding of the pathophysiology and evolution of STS as well as the mechanism of action of lurbinectedin in addition to the available data regarding the activity of this drug in this subset of patients will pave the way to newer therapeutic options and strategies.
... A phase-II clinical study showed that the response rate of temozolomide in patients with SCLC could reach 23% [3]. A Phase-I clinical study showed that lubitidine combined with doxorubicin achieved an objective response rate of 33.3% in patients with recurrent SCLC [32]. Based on these reports, we evaluated the effects of cisplatin, doxorubicin, and temozolomide on the proliferation and metastasis of H1048-ER cells in vitro and in vivo. ...
Article
Full-text available
Small cell lung cancer (SCLC) is a type of high-grade neuroendocrine malignancy with low gene mutation. Chemotherapy is the major treatment strategy, but long-term clinical application often leads to drug resistance. Etoposide is a first-line drug approved by the US Food and Drug Administration for SCLC treatment, but etoposide-resistance is a problem. In this study, a SCLC cell line with etoposide-acquired resistance, H1048-ER, was constructed through a concentration gradient increasing method, and its resistance to etoposide was investigated in vitro and in a zebrafish model. Through transcriptome sequencing, real-time reverse transcription-quantitative polymerase chain reaction, and bioinformatic analyses of H1048-ER vs. H1048 cells, 51 differentially expressed genes were found to be significantly enriched in “collagen degradation” and “MET/FAK signaling activation in ECM”. Among them, six genes (COL11A1, COL26A1, COL4A3, COL4A4, LAMA4, and LAMC1) had strong correlations with the prognosis of lung cancer. They may be key factors in the acquired etoposide resistance of H1048-ER cells. H1048-ER cells showed cross-resistance to cisplatin but were sensitive to doxorubicin and temozolomide. Our study provides novel insights into etoposide resistance in SCLC and affords the potential treatment options after etoposide resistance.
... It is a different-patterned derivative of ecteinascidin that was isolated from tunicate E. turbinata extracts. According to Calvo et al. (2017), lurbinectedin causes DNA alkylation, inhibits active transcription, causes DNA breakage, and ultimately induces apoptosis. ...
Article
Full-text available
In terms of focus, promising results obtained from extensive investigations showed that the biodiversity of marine invertebrates has been documented. This poses the marine ecosystem as a hopeful resource in the discovery of novel compounds, with an ideal starting point in scaffolding additional screening of marine natural products (MNPs), especially to face the rise in the burden of diseases and treatment failure. A wide range of antimicrobial, antiviral, anticancer, anti-inflammatory, antiparasitic, neuroprotective, analgesic, antimalarial and other agents of marine origin have been pursued in the control and management of diseases. In the current review, we will focus on figuring out drugs from well-known marine species within invertebrates (e.g., sea stars, sea sponges, sea cucumbers, sea urchins, sea worms, soft corals, sea hares, and sea worms). Moreover, we will discuss the current status and challenges being faced in the marine drug discovery field, the investment in the marine bio-drug sector, and how it provides the market with novel products for treating numerous human diseases.
... It is a different-patterned derivative of ecteinascidin that was isolated from tunicate E. turbinata extracts. According to Calvo et al. (2017), lurbinectedin causes DNA alkylation, inhibits active transcription, causes DNA breakage, and ultimately induces apoptosis. ...
Article
Full-text available
Small-cell lung cancer (SCLC) is a highly aggressive and rapidly proliferative malignancy that has historically had limited therapeutic advancements. Recent advancements in the understanding of SCLC have led to attempts at subtyping the disease based on transcription factor characteristics, offering new insights into its biology and potential therapeutic targets. In addition, significant progress has been made in developing treatment regimens, providing new hope for improved patient outcomes. The introduction of immune checkpoint inhibitors, such as atezolizumab and durvalumab, in combination with traditional chemotherapy, has marked a significant advancement, demonstrating improved overall survival and progression-free survival compared to chemotherapy alone. Despite these advancements, the prognosis for extensive-stage SCLC (ES-SCLC), the more advanced form of SCLC, remains poor, highlighting the critical need for ongoing research and the development of novel therapeutic strategies. New treatment modalities, such as lurbinectedin and anti-Delta-like Canonical Notch Ligand 3 antibodies, are now included in the treatment options for refractory SCLC, and many more treatment strategies involving combination therapies are being studied. Advances in molecular profiling and the identification of biomarkers are aiding in the development of personalized treatment approaches. This review focuses on these recent advancements and emerging strategies in the treatment of ES-SCLC.
Article
Full-text available
Objective This study aimed to systematically analyze the efficacy and toxicity of lurbinectedin as a second-line or subsequent treatment for extensive-stage small cell lung cancer (ES-SCLC). Methods Candidate studies were identified in PubMed, Embase, Cochrane Library, ClinicalTrials.gov, CNKI, and Wanfang databases up to 1 May 2024. Objective remission rate (ORR), disease control rate (DCR), duration of response (DOR), progression-free survival (PFS), overall survival (OS), and adverse events (AEs) were extracted, respectively. The efficacy and toxicity of lurbinectedin in ES-SCLC were analyzed by meta-analysis. Results Six eligible prospective studies were included in this meta-analysis, including 536 patients with ES-SCLC who received second-line or subsequent treatment. In pooled analysis, the ORR of lurbinectedin was 35% (95% confidence interval [CI] 29–41), DCR was 67% (95%CI 58–76), DOR was 5.33 months (95%CI 4.51-6.16), PFS was 3.38 months (95%CI 2.59-4.17), and OS was 7.49 months (95%CI 5.11–9.87). The incidence of AEs and severe adverse events (SAEs) was 92% (95%CI 78–100) and 37% (95%CI 19–57), respectively. The most common AEs were leukopenia, neutropenia, anemia, and thrombocytopenia, with incidences of 81% (68–91), 74% (57–88), 73% (35–98) and 57% (46–68), respectively. Conclusion As a promising alternative for second-line treatment for ES-SCLC, lurbinectedin has a certain level of efficacy and a favorable safety profile. The integration of lurbinectedin with other therapeutic modalities presents an emerging area warranting further investigation.
Article
Background and Aims Lurbinectedin is a novel oncogenic transcription inhibitor active in several cancers, including small cell lung cancer (SCLC). We aimed to describe the first Australian experience of the clinical efficacy and tolerability of lurbinectedin for the treatment of SCLC after progression on platinum‐containing therapy. Methods Multicentre real‐world study of individuals with SCLC initiating lurbinectedin monotherapy (3.2 mg/m ² three‐weekly) on an early access programme between May 2020 and December 2021. Key outcomes were clinical utilisation, efficacy and tolerability. Progression‐free survival (PFS) and overall survival (OS) were calculated using the Kaplan–Meier method. Outcome data were collected within the AUstralian Registry and biObank of thoRacic cAncers (AURORA). Results Data were analysed for 46 individuals across seven sites. Lurbinectedin was given as second‐ (83%, 38/46) or subsequent‐ (17%, 8/46) line therapy, mostly with prior chemoimmunotherapy (87%, 40/46). We report dose modifications (17%, 8/46), interruptions/delays (24%, 11/46), high‐grade toxicities (28%, 13/46) and hospitalisations (54%, 25/46) during active treatment. The overall response rate was 33% and the disease control rate was 50%. Six‐month OS was 44% (95% confidence interval (CI): 29.0–57.1). Twelve‐month OS was 15% (95% CI: 6.5–26.8). From lurbinectedin first dose, the median PFS was 2.5 months (95% CI: 1.8–2.9) and OS was 4.5 months (95% CI: 3.5–7.2). From SCLC diagnosis, the median OS was 12.9 months (95% CI: 11.0–17.2). Individuals with a longer chemotherapy‐free interval prior to lurbinectedin had longer PFS and OS. Conclusion This real‐world national experience of lurbinectedin post‐platinum chemotherapy and immunotherapy for individuals with SCLC was similar to that reported in clinical trials.
Article
Objective To investigate the mechanism of Ling-Gui-Zhu-Gan decoction (LGZGD) protects against doxorubicin (DOX)-induced myocardial injury. Methods In vivo experiment, rats were divided into six groups: normal group, model group (15 mg/kg, DOX), Dex group(150 mg/kg, Dex), LGZGD-L group (2.1 g/kg), LGZGD-M group (4.2 g/kg), and LGZGD-H group (8.4 g/kg). We used HE and Masson staining to observe the histopathological changes, echocardiography to assess the cardiac function, and western blot and RT-qPCR to detect the expressions of Nrf2, GPX4, Fpn1, and Ptgs2. In vitro experiment, we used immunofluorescence to detect ROS production, and RT-qPCR to detect gene expression of GPX4, Fpn1, and Ptgs2. Key findings In vivo, LGZGD improved cardiac systolic function. LGZGD significantly reduced MDA, LDH, and CK levels, increased SOD activity, enhanced the protein expression of Nrf2, GPX4, and Fpn1, and decreased Ptgs2 levels. In vitro, LGZGD-containing serum significantly reduced ROS, increased the gene expression of GPX4 and Fpn1, and decreased the gene expression of Ptgs2. Furthermore, compared with the LGZGD (si-NC) group, the LGZGD (si-Nrf2) group had decreased gene expression of Nrf2, GPX4, and Fpn1 and increased gene expression of Ptgs2. Conclusions LGZGD can ameliorate DOX-cardiotoxicity by activating the Nrf2 signaling pathway and inhibiting ferroptosis in cardiomyocytes.
Chapter
This reference highlights the significance of marine ecosystems, encompassing seaweed beds, seagrasses, coral reefs, mangroves, estuaries, and protected areas, as a remarkable gateway to overcoming healthcare challenges and unlocking a rich trove of bioactive compounds for drug discovery. One of the key highlights of this book is its exploration of the development of marine bio-drugs, a field that demands collaboration among scientists from both academic and industrial fronts. The editors also include a prospective review on marine environments, emphasizing the necessity for big data, collective knowledge sharing, financial support, and streamlined administrative processes, all of which contribute to enhancing innovation in the drug discovery process. Another feature includes reference lists that allow researchers to explore topics of interest in depth. With twelve comprehensive chapters, this book extensively covers marine ecosystem biodiversity, productivity, protected areas, and the intricate interplay of biotic and abiotic factors that shape these ecosystems. Readers will learn about important bioactive compounds within marine organisms and how to use this knowledge to outline a strategy for bio-drug discovery. The book caters to a diverse audience of researchers, students, ecologists, microbiologists, pharmacologists, and biotechnologists who are engaged in studying the dynamic components of marine environments. By providing the latest insights and strategies in the realm of bio-drug discovery from marine resources, this book serves as an invaluable resource for scholars and professionals seeking to tap into the potential of these unique ecosystems.
Article
Full-text available
Background: Lurbinectedin is a novel anticancer agent currently undergoing late-stage (Phase II /III) clinical evaluation in platinum-resistant ovarian, BRCA1/2-mutated breast and small-cell lung cancer. Lurbinectedin is structurally related to trabectedin and it inhibits active transcription and the DNA repair machinery in tumour cells. Methods: In this study we investigated whether lurbinectedin has the ability to modulate the inflammatory microenvironment and the viability of myeloid cells in tumour-bearing mice. Results: Administration of lurbinectedin significantly and selectively decreased the number of circulating monocytes and, in tumour tissues, that of macrophages and vessels. Similar findings were observed when a lurbinectedin-resistant tumour variant was used, indicating a direct effect of lurbinectedin on the tumour microenviroment. In vitro, lurbinectedin induced caspase-8-dependent apoptosis of human purified monocytes, whereas at low doses it significantly inhibited the production of inflammatory/growth factors (CCL2, CXCL8 and VEGF) and dramatically impaired monocyte adhesion and migration ability. These findings were supported by the strong inhibition of genes of the Rho-GTPase family in lurbinectedin-treated monocytes. Conclusions: The results illustrate that lurbinectedin affects at multiple levels the inflammatory microenvironment by acting on the viability and functional activity of mononuclear phagocytes. These peculiar effects, combined with its intrinsic activity against cancer cells, make lurbinectedin a compound of particular interest in oncology.British Journal of Cancer advance online publication, 6 July 2017; doi:10.1038/bjc.2017.205 www.bjcancer.com.
Article
Full-text available
We have defined the mechanism of action of lurbinectedin, a marine-derived drug exhibiting a potent antitumor activity across several cancer cell lines and tumor xenografts. This drug, currently undergoing clinical evaluation in ovarian, breast, and small cell lung cancer patients, inhibits the transcription process through (i) its binding to CG-rich sequences, mainly located around promoters of protein-coding genes; (ii) the irreversible stalling of elongating RNA polymerase II (Pol II) on the DNA template and its specific degradation by the ubiquitin/proteasome machinery; and (iii) the generation of DNA breaks and subsequent apoptosis. The finding that inhibition of Pol II phosphorylation prevents its degradation and the formation of DNA breaks after drug treatment underscores the connection between transcription elongation and DNA repair. Our results not only help to better understand the high specificity of this drug in cancer therapy but also improve our understanding of an important transcription regulation mechanism. Mol Cancer Ther; 15(10); 1-14. ©2016 AACR.
Article
Background: Rovalpituzumab tesirine is a first-in-class antibody-drug conjugate directed against delta-like protein 3 (DLL3), a novel target identified in tumour-initiating cells and expressed in more than 80% of patients with small-cell lung cancer. We aimed to assess the safety and activity of rovalpituzumab tesirine in patients who progressed after one or more previous regimen. Methods: We conducted a phase 1 open-label study at ten cancer centres in the USA. Eligible patients were aged 18 years or older and had histologically or cytologically confirmed small-cell lung cancer or large-cell neuroendocrine tumours with progressive measurable disease (according to Response Evaluation Criteria in Solid Tumors [RECIST], version 1.1) previously treated with one or two chemotherapeutic regimens, including a platinum-based regimen. We assigned patients to dose-escalation or expansion cohorts, ranging from 0·05 mg/kg to 0·8 mg/kg rovalpituzumab tesirine intravenously every 3 weeks or every 6 weeks, followed by investigation of the dose schedules 0·3 mg/kg and 0·4 mg/kg every 6 weeks and 0·2 mg/kg every 3 weeks. Primary objectives were to assess the safety of rovalpituzumab tesirine, including the maximum tolerated dose and dose-limiting toxic effects. The primary activity endpoint was objective response by intention-to-treat analysis. This study is registered with ClinicalTrials.gov, number NCT01901653. The study is closed to enrolment; this report focuses on the cohort with small-cell lung cancer. Findings: Between July 22, 2013, and Aug 10, 2015, 82 patients were enrolled, including 74 patients with small-cell lung cancer and eight with large-cell neuroendocrine carcinoma, all of whom received at least one dose of rovalpituzumab tesirine. Dose-limiting toxic effects of rovalpituzumab tesirine occurred at a dose of 0·8 mg/kg every 3 weeks, including grade 4 thrombocytopenia (in two of two patients at that dose level) and grade 4 liver function test abnormalities (in one patient). The most frequent grade 3 or worse treatment-related adverse events in 74 patients with small-cell lung cancer were thrombocytopenia (eight [11%]), pleural effusion (six [8%]), and increased lipase (five [7%]). Drug-related serious adverse events occurred in 28 (38%) of 74 patients. The maximum tolerated dose of rovalpituzumab tesirine was 0·4 mg/kg every 3 weeks; the recommended phase 2 dose and schedule is 0·3 mg/kg every 6 weeks. At active doses of rovalpituzumab tesirine (0·2 mg/kg or 0·4 mg/kg every 3 weeks or 0·3 mg/kg or 0·4 mg/kg every 6 weeks), 11 (18%) of 60 assessable patients had a confirmed objective response. 11 (18%) of 60 assessable patients had a confirmed objective response, including ten (38%) of 26 patients confirmed to have high DLL3 expression (expression in 50% or more of tumour cells). Interpretation: Rovalpituzumab tesirine shows encouraging single-agent antitumour activity with a manageable safety profile. Further development of rovalpituzumab tesirine in DLL3-expressing malignant diseases is warranted. Funding: Stemcentrx Inc.
Article
Background: Treatments for small-cell lung cancer (SCLC) after failure of platinum-based chemotherapy are limited. We assessed safety and activity of nivolumab and nivolumab plus ipilimumab in patients with SCLC who progressed after one or more previous regimens. Methods: The SCLC cohort of this phase 1/2 multicentre, multi-arm, open-label trial was conducted at 23 sites (academic centres and hospitals) in six countries. Eligible patients were 18 years of age or older, had limited-stage or extensive-stage SCLC, and had disease progression after at least one previous platinum-containing regimen. Patients received nivolumab (3 mg/kg bodyweight intravenously) every 2 weeks (given until disease progression or unacceptable toxicity), or nivolumab plus ipilimumab (1 mg/kg plus 1 mg/kg, 1 mg/kg plus 3 mg/kg, or 3 mg/kg plus 1 mg/kg, intravenously) every 3 weeks for four cycles, followed by nivolumab 3 mg/kg every 2 weeks. Patients were either assigned to nivolumab monotherapy or assessed in a dose-escalating safety phase for the nivolumab/ipilimumab combination beginning at nivolumab 1 mg/kg plus ipilimumab 1 mg/kg. Depending on tolerability, patients were then assigned to nivolumab 1 mg/kg plus ipilimumab 3 mg/kg or nivolumab 3 mg/kg plus ipilimumab 1 mg/kg. The primary endpoint was objective response by investigator assessment. All analyses included patients who were enrolled at least 90 days before database lock. This trial is ongoing; here, we report an interim analysis of the SCLC cohort. This study is registered with ClinicalTrials.gov, number NCT01928394. Findings: Between Nov 18, 2013, and July 28, 2015, 216 patients were enrolled and treated (98 with nivolumab 3 mg/kg, three with nivolumab 1 mg/kg plus ipilimumab 1 mg/kg, 61 with nivolumab 1 mg/kg plus ipilimumab 3 mg/kg, and 54 with nivolumab 3 mg/kg plus ipilimumab 1 mg/kg). At database lock on Nov 6, 2015, median follow-up for patients continuing in the study (including those who had died or discontinued treatment) was 198·5 days (IQR 163·0-464·0) for nivolumab 3 mg/kg, 302 days (IQR not calculable) for nivolumab 1 mg/kg plus ipilimumab 1 mg/kg, 361·0 days (273·0-470·0) for nivolumab 1 mg/kg plus ipilimumab 3 mg/kg, and 260·5 days (248·0-288·0) for nivolumab 3 mg/kg plus ipilimumab 1 mg/kg. An objective response was achieved in ten (10%) of 98 patients receiving nivolumab 3 mg/kg, one (33%) of three patients receiving nivolumab 1 mg/kg plus ipilimumab 1 mg/kg, 14 (23%) of 61 receiving nivolumab 1 mg/kg plus ipilimumab 3 mg/kg, and ten (19%) of 54 receiving nivolumab 3 mg/kg plus ipilimumab 1 mg/kg. Grade 3 or 4 treatment-related adverse events occurred in 13 (13%) patients in the nivolumab 3 mg/kg cohort, 18 (30%) in the nivolumab 1 mg/kg plus ipilimumab 3 mg/kg cohort, and ten (19%) in the nivolumab 3 mg/kg plus ipilimumab 1 mg/kg cohort; the most commonly reported grade 3 or 4 treatment-related adverse events were increased lipase (none vs 5 [8%] vs none) and diarrhoea (none vs 3 [5%] vs 1 [2%]). No patients in the nivolumab 1 mg/kg plus ipilimumab 1 mg/kg cohort had a grade 3 or 4 treatment-related adverse event. Six (6%) patients in the nivolumab 3 mg/kg group, seven (11%) in the nivolumab 1 mg/kg plus ipilimumab 3 mg/kg group, and four (7%) in the nivolumab 3 mg/kg plus ipilimumab 1 mg/kg group discontinued treatment due to treatment-related adverse events. Two patients who received nivolumab 1 mg/kg plus ipilimumab 3 mg/kg died from treatment-related adverse events (myasthenia gravis and worsening of renal failure), and one patient who received nivolumab 3 mg/kg plus ipilimumab 1 mg/kg died from treatment-related pneumonitis. Interpretation: Nivolumab monotherapy and nivolumab plus ipilimumab showed antitumour activity with durable responses and manageable safety profiles in previously treated patients with SCLC. These data suggest a potential new treatment approach for a population of patients with limited treatment options and support the evaluation of nivolumab and nivolumab plus ipilimumab in phase 3 randomised controlled trials in SCLC. Funding: Bristol-Myers Squibb.
Article
Small cell lung cancer (SCLC) represents between 13% and 15% of all diagnosed lung cancers worldwide. It is an aggressive neoplasia, with a 5-year mortality of 90% or more. It has historically been classified as limited disease (LD) and extensive disease (ED) in most study protocols. The cornerstone of treatment for any stage of SCLC is etoposide-platinum based chemotherapy; in limited stage (LS), concomitant radiotherapy to thorax and mediastinum. Prophylactic radiotherapy to the central nervous system (CNS) [prophylactic cerebral irradiation (PCI)] has diminished the incidence of brain metastasis as the site for relapse in LD and ED patients, therefore it should be offered to patients with complete response to induction first-line treatment. Regarding second-line treatment, results are more modest and topotecan is accepted as treatment for this scenario offering a modest benefit.
Article
Patients with small-cell lung cancer (SCLC) that progresses after first-line (FL) chemotherapy have a poor prognosis and second-line (SL) chemotherapy has limited efficacy. Patients whose disease relapses/progresses > 90 days after FL platinum-based treatment are considered platinum-sensitive and could be rechallenged with a similar regimen. We conducted a multicenter retrospective analysis to evaluate outcomes of SCLC patients rechallenged with platinum/etoposide. Records of all SCLC patients treated in 7 institutions between January 2007 and December 2011 were reviewed. The primary end point was overall survival from the time of rechallenge (OS-R); secondary end points were progression-free survival (PFS) and overall survival from the time of diagnosis (OS-D). Survival curves were calculated using the Kaplan-Meier method. Of the 2000 SCLC patients identified, 112 (5.6%) had sensitive disease treated with rechallenge platinum/etoposide; 65% were men with a median age of 64 years. At the time of diagnosis, 44% of patients had limited disease, 82% had an Eastern Cooperative Oncology Group performance status of 0 to 1. A median of 4 cycles of rechallenge was administered. Tumor response was 3% for complete response and 42% for partial response, 19% of patients maintained stable disease, 27% progressive disease, and 9% were not evaluable. Median PFS from the time of rechallenge was 5.5 months (95% confidence interval [CI], 4.4-6.3). Median OS-R and OS-D were 7.9 months (95% CI, 6.9-9.7) and 21.4 months (95% CI, 19.8-24.1), respectively. Subgroup analysis according to relapse-free interval (90-119 vs. 120-149 vs. > 150 days) did not show any statistically significant difference in PFS or OS-R. The outcome for SL chemotherapy for SCLC is poor. Rechallenge platinum/etoposide is a reasonable option with potentially better outcomes than standard chemotherapy. Copyright © 2015 Elsevier Inc. All rights reserved.
Article
PM01183 is a new synthetic tetrahydroisoquinoline alkaloid that is currently in phase I clinical development for the treatment of solid tumours. In this study we have characterized the interactions of PM01183 with selected DNA molecules of defined sequence and its in vitro and in vivo cytotoxicity. DNA binding characteristics of PM01183 were studied using electrophoretic mobility shift assays, fluorescence-based melting kinetic experiments and computational modelling methods. Its mechanism of action was investigated using flow cytometry, Western blot analysis and fluorescent microscopy. In vitro anti-tumour activity was determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay and the in vivo activity utilized several human cancer models. Electrophoretic mobility shift assays demonstrated that PM01183 bound to DNA. Fluorescence-based thermal denaturation experiments showed that the most favourable DNA triplets providing a central guanine for covalent adduct formation are AGC, CGG, AGG and TGG. These binding preferences could be rationalized using molecular modelling. PM01183-DNA adducts in living cells give rise to double-strand breaks, triggering S-phase accumulation and apoptosis. The potent cytotoxic activity of PM01183 was ascertained in a 23-cell line panel with a mean GI(50) value of 2.7 nM. In four murine xenograft models of human cancer, PM01183 inhibited tumour growth significantly with no weight loss of treated animals. PM01183 is shown to bind to selected DNA sequences and promoted apoptosis by inducing double-strand breaks at nanomolar concentrations. The potent anti-tumour activity of PM01183 in several murine models of human cancer supports its development as a novel anti-neoplastic agent.
Article
Lurbinectedin (PM01183) binds covalently to DNA and has broad activity against tumor cell lines. This first-in-human phase I study evaluated dose-limiting toxicities (DLTs) and defined a phase II recommended dose (RD) for PM01183 as a 1-hour intravenous (i.v.) infusion every 3 weeks (q3wk). Experimental design: Thirty-one patients with advanced solid tumors received escalating doses of PM01183 following an accelerated titration design. PM01183 was safely escalated over 200-fold, from 0.02 mg/m(2) to 5.0 mg/m(2). Dose-doubling was utilized, requiring 15 patients and 9 dose levels to identify DLT. The RD was 4.0 mg/m(2), with one of 15 patients having DLT (grade 4 thrombocytopenia). Clearance was independent of body surface area; thus, a flat dose (FD) of 7.0 mg was used during expansion. Myelosuppression, mostly grade 4 neutropenia, occurred in 40% of patients but was transient and manageable, and none was febrile. All other toxicity was mild, and fatigue, nausea and vomiting were the most common at the RD. Pharmacokinetic parameters showed high interindividual variation, though linearity was observed. At or above the RD, the myelosuppressive effect was significantly associated with the area under the concentration-time curve (white blood cells, p=0.0007; absolute neutrophil count, p=0.016). A partial response was observed in one pancreatic adenocarcinoma patient at the RD. A FD of 7.0 mg is the RD for PM01183 as a 1-hour infusion q3wk. This dose is tolerated and active. Severe neutropenia occurred at this dose, although it was transient and with no clinical consequences in this study.