ArticlePDF Available

Inter- and Intra-Patient Heterogeneity of Response and Progression to Targeted Therapy in Metastatic Melanoma

PLOS
PLOS ONE
Authors:

Abstract and Figures

MAPK inhibitors (MAPKi) are active in BRAF-mutant metastatic melanoma patients, but the extent of response and progression-free survival (PFS) is variable, and complete responses are rare. We sought to examine the patterns of response and progression in patients treated with targeted therapy. MAPKi-naïve patients treated with combined dabrafenib and trametinib had all metastases ≥5 mm (lymph nodes ≥15 mm in short axis) visible on computed tomography measured at baseline and throughout treatment. 24 patients had 135 measured metastases (median 4.5/patient, median diameter 16 mm). Time to best response (median 5.5 mo, range 1.7-20.1 mo), and the degree of best response (median -70%, range +9 to -100%) varied amongst patients. 17% of patients achieved complete response (CR), whereas 53% of metastases underwent CR, including 42% ≥10 mm. Metastases that underwent CR were smaller than non-CR metastases (median 11 vs 20 mm, P<0.001). PFS was variable among patients (median 8.2 mo, range 2.6-18.3 mo), and 50% of patients had disease progression in new metastases only. Only 1% (1/71) of CR-metastases subsequently progressed. Twelve-month overall survival was poorer in those with a more heterogeneous initial response to therapy than less heterogeneous (67% vs 93%, P = 0.009). Melanoma response and progression with MAPKi displays marked inter- and intra-patient heterogeneity. Most metastases undergo complete response, yet only a small proportion of patients achieve an overall complete response. Similarly, disease progression often occurs only in a subset of the tumor burden, and often in new metastases alone. Clinical heterogeneity, likely reflecting molecular heterogeneity, remains a barrier to the effective treatment of melanoma patients.
Content may be subject to copyright.
Inter- and Intra-Patient Heterogeneity of Response and
Progression to Targeted Therapy in Metastatic
Melanoma
Alexander M. Menzies
1,2
*, Lauren E. Haydu
1,2
, Matteo S. Carlino
1,2,3,5
, Mary W. F. Azer
3
, Peter J. A. Carr
2,4
,
Richard F. Kefford
1,2,3,5
, Georgina V. Long
1,2
1Melanoma Institute Australia, Sydney, Australia, 2The University of Sydney, Sydney, Australia, 3Westmead Hospital, Crown Princess Mary Cancer Centre, Sydney,
Australia, 4Westmead Hospital, Department of Radiology, Sydney, Australia, 5Westmead Institute for Cancer Research, Westmead, Australia
Abstract
Background:
MAPK inhibitors (MAPKi) are active in BRAF-mutant metastatic melanoma patients, but the extent of response
and progression-free survival (PFS) is variable, and complete responses are rare. We sought to examine the patterns of
response and progression in patients treated with targeted therapy.
Methods:
MAPKi-naı
¨ve patients treated with combined dabrafenib and trametinib had all metastases $5 mm (lymph nodes
$15 mm in short axis) visible on computed tomography measured at baseline and throughout treatment.
Results:
24 patients had 135 measured metastases (median 4.5/patient, median diameter 16 mm). Time to best response
(median 5.5 mo, range 1.7–20.1 mo), and the degree of best response (median 270%, range +9to2100%) varied amongst
patients. 17% of patients achieved complete response (CR), whereas 53% of metastases underwent CR, including 42%
$10 mm. Metastases that underwent CR were smaller than non-CR metastases (median 11 vs 20 mm, P,0.001). PFS was
variable among patients (median 8.2 mo, range 2.6–18.3 mo), and 50% of patients had disease progression in new
metastases only. Only 1% (1/71) of CR-metastases subsequently progressed. Twelve-month overall survival was poorer in
those with a more heterogeneous initial response to therapy than less heterogeneous (67% vs 93%, P=0.009).
Conclusion:
Melanoma response and progression with MAPKi displays marked inter- and intra-patient heterogeneity. Most
metastases undergo complete response, yet only a small proportion of patients achieve an overall complete response.
Similarly, disease progression often occurs only in a subset of the tumor burden, and often in new metastases alone. Clinical
heterogeneity, likely reflecting molecular heterogeneity, remains a barrier to the effective treatment of melanoma patients.
Citation: Menzies AM, Haydu LE, Carlino MS, Azer MWF, Carr PJA, et al. (2014) Inter- and Intra-Patient Heterogeneity of Response and Progression to Targeted
Therapy in Metastatic Melanoma. PLoS ONE 9(1): e85004. doi:10.1371/journal.pone.0085004
Editor: Keiran Smalley, The Moffitt Cancer Center & Research Institute, United States of America
Received October 30, 2013; Accepted November 27, 2013; Published January 6, 2014
Copyright: ß2014 Menzies et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits
unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Funding: This work was supported by Program Grants from the National Health and Medical Research Council of Australia (NHMRC), Cancer Institute NSW,
Australian Cancer Research Foundation, the Melanoma Foundation of the University of Sydney and Melanoma Institute Australia. GlaxoSmithKline funded the
clinical trial from which these data were obtained. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the
manuscript.
Competing Interests: The authors have read the journal’s policy and have the following conflicts: AMM Roche (H & T), GlaxoSmithKline (T). RFK Roche (C),
GlaxoSmithKline (C), Novartis (C). GVL Roche (C, T & H), GlaxoSmithKline (C), Novartis (C). None to declare for the remaining authors. There are no employment or
leadership positions, no stock ownership, no expert testimony. Note: (C) = Consultant advisor, (H) Honoraria and (T) Travel support for conference attendance. This
does not alter the authors’ adherence to all the PLOS ONE policies on sharing data and materials.
* E-mail: alexander.menzies@sydney.edu.au
Introduction
Molecular heterogeneity exists in all cancers [1,2], particularly
melanoma [3–5]. Genetic divergence occurs during clonal
evolution, resulting in inter- and intra-tumoral molecular hetero-
geneity within patients [3,6,7]. Certain driver genetic aberrations
exist in all tumor cells within an individual, but several others exist
in subclones, conferring varying degrees of drug resistance [2].
Intrinsic resistance mechanisms present in subclones of the overall
tumor burden diminish the initial response to systemic treatment,
and these and acquired mechanisms result in disease progression.
Ultimately the presence or development of these mechanisms
influence the initial response to systemic treatment, time to
progression, and overall survival. The influence and heterogeneity
of the tumor micro-environment is also increasingly understood to
play a role in tumor cell heterogeneity and treatment outcome [8].
Clinically, inter- and intra-patient molecular heterogeneity is
manifest by the variable responses observed between and within
patients treated with targeted therapies. BRAF inhibitors, used as
single agents or in combination with MEK inhibitors, are active in
most patients with metastatic melanoma, but the extent of response
and time to progression are variable between patients, and complete
responses are uncommon [9–11]. Patterns of disease progression are
also variable, with existing metastases progressing or new metastases
developing at the same time as ongoing response in other metastases
[12,13]. The terms ‘‘mixed response’’ and ‘‘isolated progression’’
are now used commonly, however these terms have not yet been
PLOS ONE | www.plosone.org 1 January 2014 | Volume 9 | Issue 1 | e85004
accurately defined, and there is little known as to the prevalence or
predictors of these phenomena, nor the clinical outcomes of patients
with these patterns of response and progression.
We therefore sought to examine the patterns of response and
progression to targeted therapy by measuring every metastasis
$5 mm via computed tomography (CT) in a cohort of patients
with metastatic melanoma treated with combined BRAF and
MEK inhibitors.
Patients and Methods
Patients and Treatment
All MAPK inhibitor naı
¨ve BRAF-mutant metastatic melanoma
patients treated with dabrafenib and trametinib (CombiDT) on
parts B–D of the BRF113220 Phase 1/2 [11] trial
(NCT01072175) at Westmead Hospital in association with
Melanoma Institute Australia were included for analysis. The
collection and analysis of clinical data was approved by the
Westmead and Royal Prince Alfred Hospitals Human Research
Ethics Committees (Protocol No. X11-0023 and HREC/11/
RPAH/32) and written informed consent was obtained from each
patient. Patients received a range of doses of dabrafenib and
trametinib. Patient demographic and disease characteristic data at
trial entry were collected.
Disease Assessments
CT scans of 3 mm slice thickness were performed at baseline
and then every 8 weeks as per the clinical trial protocol. In
Table 1. Patient demographics and clinical characteristics.
Feature All patients
Uniform Response
at First Scan*
Mixed Response
at First Scan*
P
-value
#
N%N%N%
Number of patients 24 100 15 62 9 38
Age (years)
Median 51 57 42 0.290
Range 29–78 28–77 38–74
Sex
Male 13 54 8 53 5 56 0.625
Female 11 46 7 47 4 44
BRAF genotype
V600E 20 85 13 87 7 78 0.486
V600K 4 15 2 13 2 22
ECOG PS
0 19 79 11 73 8 89 0.360
1521427111
AJCC Stage
M1a 5 21 3 20 2 22 0.418
M1b 5 21 4 27 1 11
M1c 14 58 8 53 6 67
Baseline LDH
,16ULN 19 79 13 87 6 67 0.255
.16ULN 5 21 2 13 3 33
Drug doses (Dab/Tra)
300/2 8 33 6 40 2 22 Not Tested
300/1.5 1 4 0 0 1 11
300/1 4 17 2 13 2 22
300/0 then 300/2 at PD 2 8 2 13 0 0
150
,
/2 8 33 4 27 4 44
300
,
/2 141700
Dab with 2 mg Tra 17 71 11 73 6 67 0.539
Dab with ,2 mg Tra 7 29 4 27 3 33
Abbreviations: ECOG PS, Eastern Cooperative Oncology Group Performance Status; AJCC, American Joint Committee on Cancer; LDH, lactate dehydrogenase; ULN,
upper limit of normal; Dab, dabrafenib total daily dose; Tra, trametinib daily dose; PD, progressive disease.
,
hydroxymethylcellulose dabrafenib preparation.
testing M1a & M1b versus M1c.
*Uniform response: $80% of metastases with a complete or partial response and no progressing or new metastases. Mixed response: ,80% of metastases with a
complete or partial response, or the presence of any progressing or new metastases.
#
testing uniform versus mixed response cohorts.
doi:10.1371/journal.pone.0085004.t001
Heterogeneity of Tumour Response in Melanoma
PLOS ONE | www.plosone.org 2 January 2014 | Volume 9 | Issue 1 | e85004
addition to the RECIST v1.1 assessments [14] conducted
prospectively as part of the clinical trial, a more detailed radiologic
assessment of every metastasis $5 mm diameter in long axis
(lymph nodes $15 mm in short axis) visible on CT was performed
on every scan. This was referred to as the ‘‘ALL metastasis’’
assessment, and was conducted retrospectively, blinded to the
RECIST assessment and clinical data. Measurements were made
on each scan to the nearest millimeter using the IntelePACSß
computer software program.
RECIST data were used only as a comparison to the ALL
metastasis assessment data to assess for concordance of these
measures for best overall response, time to best response (TTBR),
and progression-free survival (PFS) (see supplementary methods).
The patient’s overall response at each time point was
determined using similar criteria as RECIST [14], but included
all metastases $5 mm to calculate the sum of diameters (SoD).
Disease progression was defined as the development of new
metastases and/or a $20% and $5 mm increase in the sum of
diameters of all metastases from nadir.
In addition, a response was recorded for each individual
measured metastasis at each time point and classified as complete
response (CR, disappearance or to less than 10 mm for a lymph
node), partial response (PR, $30% reduction), stable disease (SD,
neither CR/PR/PD) or progressive disease (PD, $5 mm and
$20% growth).
At first radiologic assessment, for this study, a uniform response
was predefined as $80% of metastases having a complete or
partial response with no progressing or new metastases. A mixed
response was defined as ,80% of metastases having a complete or
partial response, or the presence of any progressing or new
metastases.
Statistical Analysis
Patient demographic and clinical features were tested for
association with uniform versus mixed response at first scan using
the Fisher’s Exact Test, Pearson’s x
2
, and/or the Mann Whitney
U test as appropriate. Overall survival (OS) and PFS were
calculated from the date of commencement of targeted therapies
to the date of last follow-up or date of progression, respectively.
Univariate time to event analyses were conducted with the
Kaplan-Meier method together with the Log Rank test for
comparison of categorical covariates, and with the Cox propor-
tional hazards method for continuous covariates. Multivariate
overall survival was conducted with the Cox proportional hazards
method. When comparing the two assessment methods (RECIST
and ALL metastasis), best overall response was deemed concor-
dant if there was #10% difference in the percentage degree of best
response and also within the same response category. Time to best
response and progression-free survival were concordant if they
occurred at the same time (on the same scan) by both measures.
All statistical analyses were conducted with IBM SPSS Statistic
v21.
Results
Patient Demographics and Disease Characteristics
Twenty-four patients were included for analysis. The patient
population was typical for patients with BRAF-mutant metastatic
melanoma; the median age of patients was 51 years, 54% of
patients were men, 85% of patients had the V600E genotype, and
58% of patients had stage M1c melanoma (Table 1). All patients
were MAPK inhibitor naı
¨ve. Although several dosing regimens
were administered, 71% of patients were treated with trametinib
at the recommended part two dose of 2 mg daily in combination
with dabrafenib from trial commencement (Table 1). Two patients
received dabrafenib monotherapy until disease progression, after
which 2 mg daily trametinib was added.
Baseline Disease Assessments
135 metastases from the 24 patients were included for
assessment (median 4.5 per patient, range 1–18), substantially
more than included as RECIST targets (N=56, median 2 per
patient, range 1–5) (Table 2). The median diameter of metastases
was the same as RECIST targets (16 mm), but ranged from a
minimum 5 mm rather than 10 mm. Seventy-six percent (N=102)
of metastases were $10 mm, and 46 (45%) of these had not been
Table 2. Baseline disease assessments by examining RECIST
targets versus ALL metastases.
RECIST targets ALL metastases
Total 56 135
Diameter (mm)
Median 16 16
Range 10–108 5–108
Number $10 mm 56 102
Number per patient
Median 2 4.5
Range 1–5 1–18
Sum of Diameters (mm)
Median 48 100
Range 10–174 11–317
Site of metastases (n, %)
SQ 13, 23% 43, 32%
Lymph node 10, 18% 15, 11%
Lung 16, 29% 48, 36%
Liver 12, 21% 24, 18%
Gastrointestinal* 5, 9% 5, 4%
Abbreviations: SQ, subcutaneous and soft tissue.
*Gastrointestinal sites include adrenal (N=3), small bowel (N= 1), pancreas
(N=1).
doi:10.1371/journal.pone.0085004.t002
Figure 1. The proportions of categories of response a) by
patients (
N
= 24), b) by metastases (
N
= 135). Abbreviations: CR,
complete response; PR, partial response; SD, stable disease; PD,
progressive disease.
doi:10.1371/journal.pone.0085004.g001
Heterogeneity of Tumour Response in Melanoma
PLOS ONE | www.plosone.org 3 January 2014 | Volume 9 | Issue 1 | e85004
included as RECIST targets. Most frequent sites of disease
included lung and subcutaneous/soft tissue (SQ) (36% and 32%
respectively) (Table 2).
Overall Patient Response
The majority of patients had a response to treatment. When all
metastases $5 mm were measured, 17% (N= 4) of patients had a
complete response and 75% (N= 18) had a partial response to
treatment (Figure 1a). No patients had progressive disease as best
response. The median time to best response was diverse (median
5.5 months, range 1.7 to 20.1 months), and there was variability in
the degree of response at first assessment (median change 249%,
range +9to295%), the kinetics of response (% change over time),
and the degree of best response (median change 270%, range +9
to 2100) within the patient population (Figure 2). The degree of
best overall response by ALL metastasis and RECIST assessment
measures was concordant in 19/24 (79%) patients (Figure 3), the
category of response was concordant in 20/24 (83%) patients, and
TTBR was concordant in 17/24 (71%) patients.
Individual Metastasis Response
Ninety-three percent (126/135) of metastases had some
reduction in size with treatment and 84.5% (114/135) had either
a complete or partial response. Only 2.2% (3/135) of metastases
demonstrated progressive disease at first assessment, all within the
same patient. Importantly, 52.6% (N= 71) of metastases had a
complete response (Figure 1b, Figure 4). Of 102 metastases
$10 mm diameter, 42% (43/102) had a complete response, and
41% (23/56) RECIST target metastases had complete response.
The median TTBR for all metastases was 12.1 weeks (range
7.3–87.6 weeks) (Table 3). Compared with subcutaneous and soft
tissue metastases (median 8.3 weeks), median TTBR was
significantly longer for lymph nodes (30.3 weeks, P=0.009) and
liver metastases (31.7 weeks, P= 0.038), but not significantly
different for lung metastases (8.0 weeks, P= 0.076). TTBR was
significantly shorter as metastases decreased in size (HR = 0.98,
95% CI 0.96–0.998, P= 0.030), and the degree of response at first
scan correlated with the degree of best response (R
2
= 0.6613,
p,0.001) (Figure 5).
There was no significant difference in the rate of complete
response by disease site (P.0.05) (Table 3). Metastases that had a
complete response were significantly smaller compared with
metastases that had PD/SD/PR (median 11 mm vs 20 mm,
P,0.001). This factor remained significant when stratifying by
disease sites for lung, liver, and SQ metastases (all P,0.05), but
not for lymph nodes (N= 15).
Plots of the response of individual metastases over time within
individual patients (Figure 6) demonstrated the marked variability
in the degree of first response and best response, the kinetics of
Figure 2. Inter-patient heterogeneneity of response and progression with CombiDT. Plot of the percent change in the sum of diameters
of all metastases $5 mm within an individual patient compared to baseline at various time points during treatment with CombiDT until disease
progression. Each line represents an individual patient. Abbreviations: E, disease progressing due to existing lesions; N, new lesions; N+E, new and
existing lesions; OR, ongoing response without progression; T, treatment ceased due to toxicity.
doi:10.1371/journal.pone.0085004.g002
Heterogeneity of Tumour Response in Melanoma
PLOS ONE | www.plosone.org 4 January 2014 | Volume 9 | Issue 1 | e85004
Figure 3. The degree of overall best response for each patient by RECIST and ALL metastasis disease assessments.
doi:10.1371/journal.pone.0085004.g003
Figure 4. The best response of each individual metastasis within each patient. Abbrevations: CR, complete response; PR, partial response;
SD, stable disease; PD, progressive disease.
doi:10.1371/journal.pone.0085004.g004
Heterogeneity of Tumour Response in Melanoma
PLOS ONE | www.plosone.org 5 January 2014 | Volume 9 | Issue 1 | e85004
response, and the time to best response for each individual
metastasis.
Sixty-two percent (15/24) of patients had a uniform response at
first assessment, and 38% (9/24) of patients had a mixed response.
Patient demographics, disease characteristics and CombiDT doses
received were similar in the two groups (Table 1). The two patients
that received dabrafenib monotherapy until disease progression
had a uniform response to treatment.
Patterns of Disease Progression
At the time of analysis 18 (75%) patients had disease progression
(PD) (Figure 2). Median PFS was 8.2 months (range 2.6 to 18.3
months). PFS was highly concordant by ALL metastasis and
RECIST assessment methods (14/18, 77% of patients). Fifty
percent of patients progressed in new metastases only, 44% in
existing metastases only, and 6% in both new and existing
metastases simultaneously. There was no dominant site of disease
progression, but four (22%) patients with no prior history of brain
metastases progressed in new metastases in the brain. At time of
Figure 5. Correlation of the response of individual metastases at first scan versus best response (
N
= 135).
doi:10.1371/journal.pone.0085004.g005
Table 3. Factors influencing individual metastasis response to treament; time to best response by metastasis site, and the effect of
metastasis site and size on response.
Site of
metastasis
Median Time to Best
Response (Range) Weeks CR PR/SD/PD
P
-value*
N
Median Size
(Range) mm
N
Median Size
(Range) mm
All 12.1 (7.3–87.6) 71 11 (5–44) 64 20 (5–108) ,0.001
SQ 8.3 (7.6–56.3) 24 10 (7–30) 19 20 (10–98) ,0.001
LN 30.3 (7.7–87.6) 7 22 (15–31) 8 21 (17–48) 0.38
Lung 8.0 (7.3–63.9) 27 9 (5–44) 21 15 (5–44) 0.036
Liver 31.7 (7.7–56.0) 10 18 (7–27) 14 31 (16–47) 0.006
*P-value for comparison of median size of lesions with CR versus non-CR, Mann Whitney U test.
Abbreviations: CR, complete response; PR, partial response; SD, stable disease; PD, progressive disease; SQ, subcutaneous and soft tissue; LN, lymph node.
doi:10.1371/journal.pone.0085004.t003
Heterogeneity of Tumour Response in Melanoma
PLOS ONE | www.plosone.org 6 January 2014 | Volume 9 | Issue 1 | e85004
Figure 6. Intra-patient heterogeneneity of response and progression with CombiDT. Example plots of the percent change in the diameter
of individual metastases within four patients (a-d) compared to baseline at various time points during treatment until overall disease progression. The
degree and kinetics of response of individual metastases vary within a patient. Similarly, progression often occurs only in a subset of the overall
tumour burden. Patient D had disease progression in new lesions only.
doi:10.1371/journal.pone.0085004.g006
Figure 7. Intra-patient heterogeneity of disease progression. The number and type of metastases progressing at time of disease progression.
doi:10.1371/journal.pone.0085004.g007
Heterogeneity of Tumour Response in Melanoma
PLOS ONE | www.plosone.org 7 January 2014 | Volume 9 | Issue 1 | e85004
PD, the median proportion of metastases progressing in an
individual compared to the total tumor burden ever (including all
metastases at baseline and new metastases) was 49% (range 6 to
100%) (Figure 7). Only one metastasis that underwent complete
response subsequently progressed (1.4%, 1/71).
Survival Analyses
The burden of disease at baseline (SoD of ALL metastasis) and
the degree of overall response at first scan did not correlate with
PFS, 12-month survival or OS (Table 4). The type of initial
response (uniform versus mixed) similarly did not correlate with
PFS. 12-month and OS, however was significantly inferior for
mixed responders (67% and median 14.2 months) compared with
uniform responders (93% and median not reached, P= 0.009), a
result which remained significant when adjusting for baseline
disease burden (HR = 5.1, 95% CI: 1.2–21.1, P= 0.025).
Discussion
This is the first systematic study of patterns of clinical response
and progression to MAPK targeted therapy in all assessable
individual metastases in patients with metastatic melanoma,
demonstrating that melanoma response and progression is
heterogeneous between and within patients. Most individual
metastases undergo a complete response to treatment, yet only a
small proportion of patients achieve an overall complete response.
Disease progression is similarly heterogeneous, both in timing and
nature. Many patients have disease progression in a subset of their
overall tumor burden, and often in new metastases only.
Metastases that initially undergo complete response with treatment
seldom subsequently progress, and a more heterogeneous initial
response to treatment is associated with shorter overall survival.
Results of this study are strengthened by the detailed clinical
assessment of patients on the most highly active targeted therapy
in melanoma [11], the use of a standard modality for disease
assessment (3 mm slice thickness CT) at predetermined specified
time points, inclusion of every metastasis visible and measurable
on CT scan ($5mmor$15 for lymph nodes), and an assessment
of every individual metastasis across every time point from baseline
prior to treatment until disease progression. The inclusion of all
metastases as targets for assessment, as opposed to the maximum 5
target metastases for RECIST (and maximum 2 in any one organ)
provided a more detailed assessment, with increased ability to
assess for intra-patient heterogeneity. This question has been
previously addressed in studies of
18
F-labelled fluorodeoxyglucose
positron emission tomography (PET) metabolic response to single
agent BRAF inhibitors at day 15, with varying results, one study
examining 5 target metastases and observing a homogeneous
response [15], while the other examined every metastasis and
observed heterogeneity [16].
In this study, most metastases achieved a complete response
with treatment. These metastases were located at any body site,
and tended to be smaller than those that did not undergo complete
response, however, some metastases several centimeters in
diameter still had complete response. The reasons why smaller
metastases have a higher complete response rate may be because
they have to shrink less to become clinically occult, however, the
observation that these metastases seldom subsequently progress
perhaps supports alternative hypotheses, for example, they
undergo a more effective secondary immune response [17,18],
or contain less molecular or microenvironmental heterogeneity,
with less resistant tumor subclones. This observation warrants
further research, particularly as larger metastases may be
amenable for resection prior to therapy, and adjuvant trials for
occult metastatic disease are in progress.
Despite heterogeneity observed in the degree and timing of best
overall response amongst patients, most metastases undergo the
majority of tumor shrinkage by 3 months of treatment. Metastases
that have not undergone meaningful initial clinical response (e.g.
persisting local symptoms) by 3 months may therefore warrant
treatment with local therapy (surgery, radiotherapy). Furthermore,
in selected patients where the vast majority of metastases have
undergone complete response, remaining metastases could be
treated locally to render the patient free of overt disease. The
observation that the majority of tumor response occurs early
during treatment also suggests that additional systemic therapies
(e.g. immunotherapy) should be incorporated early in the course of
MAPK inhibitor treatment. Translational data demonstrating
early immune cell infiltration into tumors soon after treatment
commencement (as early as day 3) further supports this, and may
indicate that immunotherapies should be combined from the start
of MAPK inhibitor treatment [17,18].
Disease progression occurred at varying time points among the
patient cohort, and there was a high rate of disease progression
due to the emergence of new metastases. Often, patients
progressed in only a few metastases, with the remainder of disease
under treatment control. In this instance, disease progression may
therefore not equate to overt treatment failure, and local treatment
(e.g. surgery, radiotherapy) may be delivered to progressing
metastases with systemic treatment continued for ongoing clinical
benefit to the remainder of drug-sensitive disease [12,13,19]. This
approach may be more beneficial than a switch to immunotherapy
(e.g. ipilimumab), as little efficacy has been observed in this setting
[20,21], likely at least in part due to the release of MAPK
inhibition, whereas relative ongoing MAPK inhibition still occurs
in resistant tumors with continued MAPK inhibitor treatment
[22].
In this study cohort, a mixed response at first assessment
correlated with shorter overall survival, but not progression-free
survival. This result was likely influenced by small number of
patients, the doses of therapy received, and the fact that many
patients progressed in new metastases alone. Subsequent treat-
ments may have also influenced overall survival. Despite this,
however, this finding warrants validation in future studies, as
biomarkers to predict treatment outcome are scant, and the
method of categorizing response in this study could be reproduced
without additional procedures such as PET.
The clinical heterogeneity of tumor response and progression
demonstrated in this study likely reflects underlying molecular
heterogeneity. The majority of the melanoma burden in patients is
Table 4. Univariate progression-free and overall survival.
Outcome Factor
P
-value
PFS Baseline SoD 0.101
Percent Response at First Scan 0.084
Uniform versus Mixed Response at First Scan* 0.124
OS Baseline SoD 0.349
Percent Response at First Scan 0.105
Uniform versus Mixed Response at First Scan* 0.009
Abbreviations: SoD, sum of diameters.
*Uniform response: $80% of metastases with a complete or partial response
and no progressing or new metastases. Mixed response: ,80% of metastases
with a complete or partial response, or the presence of any progressing or new
metastases.
doi:10.1371/journal.pone.0085004.t004
Heterogeneity of Tumour Response in Melanoma
PLOS ONE | www.plosone.org 8 January 2014 | Volume 9 | Issue 1 | e85004
sensitive to MAPK inhibition, however, a varying proportion of
primarily resistant subclones exist at baseline, and resistance may
also be acquired during treatment. This heterogeneity complicates
clinical management, confounds biopsy driven biomarker re-
search, and remains a barrier to the effective treatment of
melanoma patients, including the deployment of biopsy-driven
adaptive clinical trial design. A broader multi-targeted treatment
approach from the outset (e.g, MAPK and PI3K inhibitors) may
improve response rates and prolong survival, but will likely face
the same problem of clonal drug resistance and treatment failure.
Combinations of MAPK inhibitors and novel immunotherapies
(e.g. PD-1 antibodies) may provide more complete and durable
responses.
Acknowledgments
We acknowledge the assistance of Arthur Clements, Clara Lee, Matthew
Chan, Vicky Wegener, Rebecca Hinshelwood, Amie Cho, Katherine
Carson, Joanna Jackson, Andrea Del Pilar Forero Velandia, Jacob
Cunningham, and Kiran Patel (GSK).
Author Contributions
Conceived and designed the experiments: AMM GVL. Performed the
experiments: AMM. Analyzed the data: AMM LEH GVL. Contributed
reagents/materials/analysis tools: PJAC RFK GVL. Wrote the paper:
AMM LEH MSC MWFA PJAC RFK GVL.
References
1. Meacham CE, Morrison SJ (2013) Tumour heterogeneity and cancer cell
plasticity. Nature 501: 328–337.
2. Vogelstein B, Papadopoulos N, Velculescu VE, Zhou S, Diaz LA Jr, et al. (2013)
Cancer genome landscapes. Science 339: 1546–1558.
3. Yancovitz M, Litterman A, Yoon J, Ng E, Shapiro RL, et al. (2012) Intra- and
inter-tumor heterogeneity of BRAF(V600E))mutations in primary and metastatic
melanoma. PLoS ONE 7: e29336.
4. Takata M, Morita R, Takehara K (2000) Clonal heterogeneity in sporadic
melanomas as revealed by loss-of-heterozygosity analysis. Int J Cancer 85: 492–
497.
5. Lin J, Goto Y, Murata H, Sakaizawa K, Uchiyama A, et al. (2011) Polyclonality
of BRAF mutations in primary melanoma and the selection of mutant alleles
during progression. British Journal of Cancer 104: 464–468.
6. Katona TM, Jones TD, Wang M, Eble JN, Billings SD, et al. (2007) Genetically
heterogeneous and clonally unrelated metastases may arise in patients with
cutaneous melanoma. Am J Surg Pathol 31: 1029–1037.
7. Wilmott JS, Tembe V, Howle JR, Sharma R, Thompson JF, et al. (2012)
Intratumoral Molecular Heterogeneity in a BRAF-Mutant, BRAF Inhibitor-
Resistant Melanoma: A Case Illustrating the Challenges for Personalized
Medicine. Molecular Cancer Therapeutics 11: 2704–2708.
8. Junttila MR, de Sauvage FJ (2013) Influence of tumour micro-environment
heterogeneity on therapeutic response. Nature 501: 346–354.
9. Chapman PB, Hauschild A, Robert C, Haanen JB, Ascierto P, et al. (2011)
Improved survival with vemurafenib in melanoma with BRAF V600E mutation.
New England Journal of Medicine 364: 2507–2516.
10. Hausc hild A, Grob JJ, Demidov LV, Jouary T, Gutzmer R, et al. (2012)
Dabrafenib in BRAF-mutated metastatic melanoma: a multicentre, open-label,
phase 3 randomised controlled trial. Lancet 380: 358–365.
11. Flahert y KT, Infante JR, Daud A, Gonzalez R, Kefford RF, et al. (2012)
Combined BRAF and MEK inhibition in melanoma with BRAF V600
mutations. New England Journal of Medicine 367: 1694–1703.
12. Kim KB, Flaherty KT, Chapman PB, Sosman JA, Ribas A, et al. (2011) Pattern
and outcome of disease progression in phase I study of vemurafenib in patients
with metastatic melanoma (MM). Journal of Clinical Oncology 29: (abstract
8519).
13. Azer MF, Menzies AM, Haydu LE, Kefford RF, Long GV (2013) Patterns of
Response and Progression in Patients with BRAF-mutant Melanoma Metastatic
to the Brain treated with Dabrafenib. Cancer: in press.
14. Eisenhau er EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, et al. (2009)
New response evaluation criteria in solid tumours: revised RECIST guideline
(version 1.1). European Journal of Cancer 45: 228–247.
15. McArthur GA, Puzanov I, Amaravadi R, Ribas A, Chapman P, et al. (2012)
Marked, homogeneous, and early [18F]fluorodeoxyglucose-positron emission
tomography responses to vemurafenib in BRAF-mutant advanced melanoma.
Journal of Clinical Oncology 30: 1628–1634.
16. Carlino MS, Saunders CA, Haydu LE, Menzies AM, Martin Curtis C Jr, et al.
(2013) (18)F-labelled fluorodeoxyglucose-positron emission tomography (FDG-
PET) heterogeneity of response is prognostic in dabrafenib treated BRAF
mutant metastatic melanoma. European Journal of Cancer 49: 395–402.
17. Wilmott JS, Long GV, Howle JR, Haydu LE, Sharma RN, et al. (2012) Selective
BRAF inhibitors induce marked T-cell infiltration into human metastatic
melanoma. Clinical Cancer Research 18: 1386–1394.
18. Frederick DT, Piris A, Cogdill AP, Cooper ZA, Lezcano C, et al. (2013) BRAF
Inhibition Is Associated with Enhanced Melanoma Antigen Expression and a
More Favorable Tumor Microenvironment in Patients with Metastatic
Melanoma. Clinical Cancer Research 19: 1225–1231.
19. Chan M, Haydu L, Menzies AM, Azer MWF, Klein O, et al. (2013) Clinical
characteristics and survival of BRAF-mutant metastatic melanoma patients
treated with BRAF inhibitor dabrafenib or vemurafenib beyond disease
progression. Journal of Clinical Oncology 31: (abstract 9062).
20. Ackerman A, Klein O, McDermott DF, Lawrence DP, Gunturi A, et al. (2013)
Outcomes of patients with metastatic melanoma treated with immunotherapy
prior to or after BRAF inhibitors. Cancer: in press.
21. Ascierto PA, Simeone E, Giannarelli D, Grimaldi AM, Romano A, et al. (2012)
Sequencing of BRAF inhibitors and ipilimumab in patients with metastatic
melanoma: a possible algorithm for clinical use. Journal of Translational
Medicine 10: 107.
22. Carlino MS, Gowrishankar K, Saunders CA, Pupo GM, Snoyman S, et al.
(2013) Antiproliferative effects of continued mitogen-activated protein kinase
pathway inhibition following acquired resistance to BRAF and/or MEK
inhibition in melanoma. Molecular Cancer Therapeutics 12: 1332–1342.
Heterogeneity of Tumour Response in Melanoma
PLOS ONE | www.plosone.org 9 January 2014 | Volume 9 | Issue 1 | e85004
... The efficacy was more pronounced in the lymph node lesions than in the liver and bone lesions (12). In another study, the patterns of response and progression displayed high lesionspecific variation in patients with BRAF-mutant metastatic melanoma treated with a combination of two MAPK-targeted therapies, dabrafenib and trametinib (13). Response variations across anatomic sites could result from divergent clonal genetic, epigenetic, and transcriptional features (14), conferred by different evolutionary pressures and organ-specific tumor microenvironments (5). ...
... Most literature studies investigated the intermetastasis heterogeneity through the distinct genotypic and phenotypic profiles, including genetic mutations, RNA expression, cell proliferation, and metastatic potential (6,12,13,(15)(16)(17)(18). Our study, for the first time, characterized the full response dynamics of each lesion and corroborated their response heterogeneity to inform long-term patient survival. ...
... Patient demographics and clinical characteristics are summarized in Table 1. The median number of metastases was 4 (range [1][2][3][4][5][6][7][8][9][10][11][12][13][14][15][16][17][18][19][20]. The responses of the primary lesions were not included in our dataset because most patients had already received colectomy surgery before enrollment. ...
Article
The sum of target lesions is routinely used to evaluate patient objective responses to treatment in the RECIST criteria, but it fails to address response heterogeneity across metastases. This study argues that spatiotemporal heterogeneity across metastases and organ-specific response is informative for drug efficacy and patient survival. We analyzed the longitudinal data of 11,404 metastatic lesions in 2,802 colorectal cancer patients from five phase III clinical trials. Initially, a metric Gower distance was applied to quantify response heterogeneity across metastases. Next, the spatiotemporal response heterogeneity across anatomic sites, therapies, and KRAS mutation status was assessed and examined for its association with drug efficacy and long-term patient survival. The response of metastatic lesions broadly differed across anatomic sites and therapies. About 60% of patients had at least one lesion respond contrarily from total tumor size. High interlesion heterogeneity was associated with shorter progression-free survival and overall survival. Targeted therapies (bevacizumab or panitumumab) combined with standard chemotherapy reduced interlesion heterogeneity and elicited more favorable effects from liver lesions (P < 0.001) than chemotherapy alone. Moreover, the favorable responses in liver metastases (> 30% shrinkage) were associated with extended patient overall survival (P < 0.001), in contrast to lesions in the lungs and lymph nodes. Altogether, the spatiotemporal response heterogeneity across metastases informed drug efficacy and patient survival, which could improve the current methods for treatment evaluation and patient prognosis. Significance These findings support the modification of RECIST criteria to include individual lesion response to improve assessments of drug efficacy.
... Due to the variable prevalence of etiologies, the global incidence of HCC is different, from 72% in Asia to 5% in North America [4]. Currently, therapeutic strategies such as surgery, chemotherapy, radiotherapy, and ignoring inter-and intra-patient heterogeneity are used to treat HCC patients [5][6][7]. Due to the small size and lack of symptoms of the primary tumors, early diagnosis in most types of cancer remains challenging [8]. Therefore, precision oncology using fluid-phase biopsy is indispensable. ...
Article
Full-text available
Liver cancer is a significant contributor to the cancer burden, and its incidence rates have recently increased in almost all countries. Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer and is the second leading cause of cancer-related deaths worldwide. Because of the late diagnosis and lack of efficient therapeutic modality for advanced stages of HCC, the death rate continues to increase by 2-3 % per year. Circulating tumor cells (CTCs) are promising tools for early diagnosis, precise prognosis, and follow-up of therapeutic responses. They can be considered to be an innovative biomarker for the early detection of tumors and targeted molecular therapy. In this review, we briefly discuss the novel materials and technologies applied for the practical isolation and detection of CTCs in HCC. Also, the clinical value of CTC detection in HCC is highlighted.
... Heterogeneity is frequently attributed to genetics, but it is related to non-genetic influences too [5]. Clearly, this directly impacts therapeutic treatments and outcomes [40,43,59]. Thus, better understanding of heterogeneities will help to improve treatments of metastatic diseases. ...
Article
Full-text available
The growth and treatment of tumors is an important problem to society that involves the manifestation of cellular phenomena at length scales on the order of centimeters. Continuum mechanical approaches are being increasingly used to model tumors at the largest length scales of concern. The issue of how to best connect such descriptions to smaller-scale descriptions remains open. We formulate a framework to derive macroscale models of tumor behavior using the thermodynamically constrained averaging theory (TCAT), which provides a firm connection with the microscale and constraints on permissible forms of closure relations. We build on developments in the porous medium mechanics literature to formulate fundamental entropy inequality expressions for a general class of three-phase, compositional models at the macroscale. We use the general framework derived to formulate two classes of models, a two-phase model and a three-phase model. The general TCAT framework derived forms the basis for a wide range of potential models of varying sophistication, which can be derived, approximated, and applied to understand not only tumor growth but also the effectiveness of various treatment modalities.
... Melanoma is a heterogeneous disease, with differences in the speed of tumor progression and therapeutic responses frequently observed between different metastatic sites within the same patient (11,39). There is evidence that baseline and therapy-induced levels of immune infiltration contribute to this heterogeneity through mechanisms that remain poorly defined (33,34,40). ...
Article
Purpose: Melanoma brain metastases (MBM) and leptomeningeal metastases (LMM) are two different manifestations of melanoma CNS metastasis. Here, we used single cell RNA-Seq (scRNA-Seq) to define the immune landscape of MBM, LMM and melanoma skin metastases. Experimental design: Single cell RNA-sequencing (scRNA-Seq) was undertaken on 43 patient specimens; including 8 skin metastases, 14 MBM and 19 serial LMM specimens. Detailed cell type curation was performed, the immune landscapes mapped and key results validated by IHC and flow cytometry. Association analyses were undertaken to identify immune cell subsets correlated with overall survival. Results: The LMM microenvironment was characterized by an immune-suppressed T-cell landscape distinct from that of brain and skin metastases. An LMM patient with long-term survival demonstrated an immune repertoire distinct from that of poor survivors and more similar to normal CSF. Upon response to PD-1 therapy, this extreme responder showed increased levels of T-cells and dendritic cells in their CSF, whereas poor survivors showed little improvement in their T-cell responses. In MBM patients, therapy led to increased immune infiltrate, with similar T-cell transcriptional diversity noted between skin metastases and MBM. A correlation analysis across the entire immune landscape identified the presence of a rare population of dendritic cells (DC3s) that was associated with increased overall survival and positivelyregulated the immune environment through modulation of activated T-cells and MHC expression. Conclusion: Our study provides the first atlas of two distinct sites of melanoma CNS metastases and defines the immune cell landscape that underlies the biology of this devastating disease.
... Heterogeneous response to treatment in lesions within different metastatic sites or within the same organ has been reported in CRC and other types of cancers. [31][32][33]46 Since the homogeneity in phenotype is crucial in association studies, we used a RECIST-based lesion specific OR to increase the quality and level of detail of phenotypic annotations. It provided an accurate insight into drug response at the level of the lesion and stratified the samples into phenotypic homogeneous subgroups. ...
Article
Full-text available
Background: Therapeutic resistance is the main cause of death in metastatic colorectal cancer. To investigate genomic plasticity, most specifically of metastatic lesions, associated with response to first-line systemic therapy, we collected longitudinal liver metastatic samples and characterized the copy number aberration (CNA) landscape and its effect on the transcriptome. Methods: Liver metastatic biopsies were collected prior to treatment (pre, n = 97) and when clinical imaging demonstrated therapeutic resistance (post, n = 43). CNAs were inferred from whole exome sequencing and were correlated with both the status of the lesion and overall patient progression-free survival (PFS). We used RNA sequencing data from the same sample set to validate aberrations as well as independent datasets to prioritize candidate genes. Results: We identified a significantly increased frequency gain of a unique CN, in liver metastatic lesions after first-line treatment, on chr18p11.32 harboring 10 genes, including TYMS, which has not been reported in primary tumors (GISTIC method and test of equal proportions, FDR-adjusted p = 0.0023). CNA lesion profiles exhibiting different treatment responses were compared and we detected focal genomic divergences in post-treatment resistant lesions but not in responder lesions (two-tailed Fisher's Exact test, unadjusted p ≤ 0.005). The importance of examining metastatic lesions is highlighted by the fact that 15 out of 18 independently validated CNA regions found to be associated with PFS in this study were only identified in the metastatic lesions and not in the primary tumors. Conclusion: This investigation of genomic-phenotype associations in a large colorectal cancer liver metastases cohort identified novel molecular features associated with treatment response, supporting the clinical importance of collecting metastatic samples in a defined clinical setting.
Article
Purpose: Differential tumor response to therapy is partially attributed to tumor heterogeneity. Additional efforts are needed to identify tumor heterogeneity parameters in response to therapy that is easily applicable in clinical practice. We aimed to describe tumor response-speed heterogeneity and evaluate its prognostic value in patients with metastatic colorectal cancer. Patients and methods: Individual patient data from Amgen (NCT00364013) and Sanofi (NCT00305188; NCT00272051) trials were retrieved from Project Data Sphere. Patients in the Amgen 5-fluorouracil, leucovorin, oxaliplatin (FOLFOX) arm were used to establish response-speed heterogeneity. Its prognostic value was subsequently validated in the Sanofi FOLFOX arms and the Amgen panitumumab+FOLFOX arm. Kaplan-Meier method and Cox proportional hazards models were used for survival analyses. Results: Patients with high response-speed heterogeneity in the Amgen FOLFOX cohort had significantly shorter (P<0.001) median progression-free survival (PFS) of 7.27 months (95% CI, 6.12-7.96 mo) and overall survival (OS) of 16.0 months (95% CI, 13.8-18.2 mo) than patients with low response-speed heterogeneity with median PFS of 9.41 months (95% CI, 8.75-10.89 mo) and OS of 22.4 months (95% CI, 20.1-26.7 mo), respectively. Tumor response-speed heterogeneity was a poor prognostic factor of shorter PFS (hazard ratio, 4.17; 95% CI, 2.49-6.99; P<0.001) and shorter OS (hazard ratio, 2.57; 95% CI, 1.64-4.01; P<0.001), after adjustment for other common prognostic factors. Comparable findings were found in the external validation cohorts. Conclusion: Tumor response-speed heterogeneity to first-line chemotherapy was a novel prognostic factor associated with early disease progression and shorter survival in patients with metastatic colorectal cancer.
Article
Full-text available
Objective: We compared therapeutic response of Varlitinib + Capecitabine (VC) versus Lapatinib + Capecitabine (LC) in patients with human epidermal growth factor receptor 2-positive metastatic breast cancer after trastuzumab therapy by assessing changes in target lesion (TL) diameter and volume per location. Methods: We retrospectively analyzed the CT data of the ASLAN001-003 study (NCT02338245). We analyzed TL size and number at each location focusing on therapeutic response from baseline to Week 12. We used TL diameter and volume to conduct an inter-arm comparison of the response according to: RECIST 1.1; stratified per TL location and considering TLs independently. Multiple pairwise intra-arm comparisons of therapeutic responses were performed. Considering TL independently, weighted models were designed by adding weighted mean TL responses grouped by location. Results: We evaluated 42 patients (88 TL) and 35 patients (74 TL), respectively, at baseline and Week 12. We found reductions in breast TL burden in the VC arm compared to the LC arm (p = 0.002 (diameter), p < 0.001 (volume)). Responses and TL sizes at baseline were not correlated. Explained variabilities of volume change per TL location, patient and patient:TL interaction were 36%, 10% and 4% (VC), and 13%, 1% and 23%, (LC). A test of inter-arm difference of responses yielded p = 0.07 (diameter), and p < 0.001 (volume). Conclusions: The therapeutic responses differed across tumors' locations; the magnitude of the differences of responses across the tumors' locations were drug-dependent. Stratified analysis of the response by tumor location improved drug comparisons and is a powerful tool to understand TL heterogeneity.
Article
It has been recently shown that an additional therapeutic gain may be achieved if a radiotherapy plan is altered over the treatment course using a new treatment paradigm referred to in the literature as spatiotemporal fractionation. Because of the nonconvex and large-scale nature of the corresponding treatment plan optimization problem, the extent of the potential therapeutic gain that may be achieved from spatiotemporal fractionation has been investigated using stylized cancer cases to circumvent the arising computational challenges. This research aims at developing scalable optimization methods to obtain high-quality spatiotemporally fractionated plans with optimality bounds for clinical cancer cases. In particular, the treatment-planning problem is formulated as a quadratically constrained quadratic program and is solved to local optimality using a constraint-generation approach, in which each subproblem is solved using sequential linear/quadratic programming methods. To obtain optimality bounds, cutting-plane and column-generation methods are combined to solve the Lagrangian relaxation of the formulation. The performance of the developed methods are tested on deidentified clinical liver and prostate cancer cases. Results show that the proposed method is capable of achieving local-optimal spatiotemporally fractionated plans with an optimality gap of around 10%–12% for cancer cases tested in this study. Summary of Contribution: The design of spatiotemporally fractionated radiotherapy plans for clinical cancer cases gives rise to a class of nonconvex and large-scale quadratically constrained quadratic programming (QCQP) problems, the solution of which requires the development of efficient models and solution methods. To address the computational challenges posed by the large-scale and nonconvex nature of the problem, we employ large-scale optimization techniques to develop scalable solution methods that find local-optimal solutions along with optimality bounds. We test the performance of the proposed methods on deidentified clinical cancer cases. The proposed methods in this study can, in principle, be applied to solve other QCQP formulations, which commonly arise in several application domains, including graph theory, power systems, and signal processing.
Article
Full-text available
Metastatic melanoma is a fatal malignancy with a high mortality and morbidity. Since the early 1970s, available medical therapies were limited in improving survival. Surgery represented the best chance for a cure. However, surgery could only be offered to selected patients. The current landscape of treatment has radically evolved since the introduction of targeted and immunotherapies including BRAF and MEK inhibitors, and checkpoint blockers, like PD-1 and CTLA-4 antibodies. These new therapies have seen survival rates matching, and in some cases surpassing, that of surgery. Anti-PD1 and CTLA-4 combination treatments are associated with severe side effects and BRAF and MEK inhibitor combinations may trigger initial tumour responses but prolonged use have resulted in the development of resistant tumour clones and disease relapse. This review examines the role of pulmonary metastasectomy for lung metastasis from malignant melanoma in the current landscape of effective targeted therapy and immunotherapy.
Article
Full-text available
Inhibitors of the MAPKs, BRAF and MEK, induce tumor regression in the majority of patients with BRAF-mutant metastatic melanoma. The clinical benefit of MAPK inhibitors is restricted by the development of acquired resistance with half of those who benefit having progressed by 6-7 months and long-term responders uncommon. There remains no agreed treatment strategy on disease progression in these patients. Without published evidence, fears of accelerated disease progression on inhibitor withdrawal have led to the continuation of drugs beyond formal disease progression. We now demonstrate that treatment with MAPK inhibitors beyond disease progression can provide significant clinical benefit, and the withdrawal of these inhibitors led to a marked increase in the rate of disease progression in two patients. We also show that MAPK inhibitors retain partial activity in acquired resistant melanoma by examining drug-resistant clones generated to dabrafenib, trametinib or the combination of these drugs. All resistant sublines displayed a markedly slower rate of proliferation when exposed to MAPK inhibitors, and this coincided with a reduction in MAPK signalling, decrease in BrdU incorporation and S-phase inhibition. This cytostatic effect was also associated with diminished levels of cyclin D1 and p-pRb. Two short-term melanoma cultures generated from resistant tumour biopsies also responded to MAPK inhibition with comparable inhibitory changes in proliferation and MAPK signalling. These data provide a rationale for the continuation of BRAF and MEK inhibitors after disease progression and support the development of clinical trials to examine this strategy.
Article
The major obstacle preventing effective treatment of melanoma is the biological heterogeneity of tumor cells. This study was performed to determine clonal genetic heterogeneity within primary melanoma and the evolution of these heterogeneous sub‐clones during disease progression. DNA samples were obtained from 44 morphologically distinct areas identified within 10 primary tumors and from 15 metastases in the same patients. Loss of heterozygosity (LOH) analyses were performed using 17 microsatellite markers that mapped to chromosomes 6q, 9p, 10q and 18q, the most frequently deleted in melanoma. Of 10 primary tumors, 8 were revealed to have intratumoral genetic heterogeneity in terms of LOH of the 4 chromosome arms examined, 7 containing at least 2 different sub‐clones harboring LOH of different chromosome areas, while the remaining one tumor showed prominent intratumoral genetic heterogeneity consisting of at least 6 genetically distinct sub‐clones. LOH of 6q was detected only in a sub‐set of multiple microdissected samples in most of the primary tumors, but was most frequently detected in metastases, suggesting that loss of this chromosome arm occurred late and played an important part in metastatic progression. Comparison of LOH between sub‐clones within primary tumors and within metastases showed the divergence of metastatic clones from dominant populations within the primary tumor in 5 patients, whereas in the remaining three patients parent sub‐clones were not identified, or constituted only a minor sub‐population within the primary tumors. These results, showing considerable genetic heterogeneity in sporadic melanoma, have profound implications for the choice of future therapeutic strategies. Int. J. Cancer 85:492–497, 2000. © 2000 Wiley‐Liss, Inc.
Article
9062 Background: Accelerated tumor growth has been demonstrated on BRAFi cessation in BRAFi-resistant melanoma cell lines. In BRAFi-treated pts initial response rates are high but most have PD at 6+ months. The benefit of ongoing BRAFi after PD is unknown. We sought to describe the characteristics of pts treated beyond progression (TBP) vs those not TBP, and whether TBP prolongs survival. Methods: Clinicopathologic data were collected on 112 pts enrolled in phase I-IV clinical trials at Westmead Hospital and Melanoma Institute Australia from July 2009 to Sept 2012 with unresectable stage IIIC or IV BRAF+ melanoma treated with single agent dabrafenib or vemurafenib. TBP was defined as ongoing BRAFi >28 days beyond RECIST PD. Pt and disease characteristics at baseline and at PD were examined, as well as survival data. Results: 92/112 (82%) pts had RECIST PD. 36/92 (39%) pts were TBP (mean 144 days, median 93 days, range 29–572 days). Pts TBP were significantly more likely to have achieved a RECIST response (CR/PR) prior to PD, have a lower ECOG at PD, presence of brain metastases at PD, have PD treated locally and a smaller RECIST sum of diameters (SoD) at PD, compared with those not TBP (all p<0.05). Median OS from commencement of BRAFi in those TBP was longer than those not TBP (15.0 vs 6.5 months, p<0.001), as was OS from RECIST PD (7.4 vs 1.9 mo, p=0.001). In multivariate analysis of all pts, TBP improved OS from RECIST PD (HR 0.32, p=0.012) even after adjusting for other potential prognostic factors at PD (see table). Within the TBP cohort, RECIST SoD at PD was the only factor that influenced OS from PD (p=0.009), and presence of brain metastases did not. Conclusions: Treatment with BRAFi may be continued after RECIST PD in selected pts, and is associated with a prolonged OS compared with ceasing treatment at PD. [Table: see text]
Conference Paper
Background: Accelerated tumor growth has been demonstrated on BRAFi cessation in BRAFi-resistant melanoma cell lines. In BRAFi-treated pts initial response rates are high but most have PD at 6+ months. The benefit of ongoing BRAFi after PD is unknown. We sought to describe the characteristics of pts treated beyond progression (TBP) vs those not TBP, and whether TBP prolongs survival. Methods: Clinicopathologic data were collected on 112 pts enrolled in phase I-IV clinical trials at Westmead Hospital and Melanoma Institute Australia from July 2009 to Sept 2012 with unresectable stage IIIC or IV BRAF+ melanoma treated with single agent dabrafenib or vemurafenib. TBP was defined as ongoing BRAFi >28 days beyond RECIST PD. Pt and disease characteristics at baseline and at PD were examined, as well as survival data. Results: 92/112 (82%) pts had RECIST PD. 36/92 (39%) pts were TBP (mean 144 days, median 93 days, range 29–572 days). Pts TBP were significantly more likely to have achieved a RECIST response (CR/PR) prior to PD, have a lower ECOG at PD, presence of brain metastases at PD, have PD treated locally and a smaller RECIST sum of diameters (SoD) at PD, compared with those not TBP (all p<0.05). Median OS from commencement of BRAFi in those TBP was longer than those not TBP (15.0 vs 6.5 months, p<0.001), as was OS from RECIST PD (7.4 vs 1.9 mo, p=0.001). In multivariate analysis of all pts, TBP improved OS from RECIST PD (HR 0.32, p=0.012) even after adjusting for other potential prognostic factors at PD (see table). Within the TBP cohort, RECIST SoD at PD was the only factor that influenced OS from PD (p=0.009), and presence of brain metastases did not. Conclusions: Treatment with BRAFi may be continued after RECIST PD in selected pts, and is associated with a prolonged OS compared with ceasing treatment at PD.
Article
The immunotherapy (IT) agents ipilimumab and interleukin-2 as well as BRAF inhibitors (BRAFi) vemurafenib and dabrafenib, with or without trametinib (MEK inhibitors), are all FDA-approved treatments for BRAF metastatic melanoma, but there are few studies to guide optimal sequencing. This retrospective analysis describes the outcomes of patients treated with either BRAFi before IT or IT before BRAFi. A cohort of patients treated with BRAFi alone or with MEK inhibitor was retrospectively identified. Response rate (RR), overall survival (OS), and progression-free survival (PFS) were evaluated for the entire cohort, subdivided by BRAFi prior to or after IT. RR and median PFS and OS calculated from commencement of BRAFi following IT (N = 32) were 57%, 6.7 months (95% confidence interval [CI] = 4.3-9.1 months), and 19.6 months (95% CI = 10.0-undefined months), respectively; whereas for BRAFi initially (N = 242) were 66%, 5.6 months (95% CI = 4.7-6.8 months), and 13.4 months (95% CI = 10.1-17.0 months). Results were similar when controlled for prognostic variables. A total of 193 patients discontinued BRAFi, with OS of 2.9 months (range of 1.8-4.4 months) from day of BRAFi discontinuation. Forty patients subsequently received IT with ipilimumab. Only half could complete 4 doses of ipilimumab; PFS with ipilimumab was 2.7 months (95% CI = 1.8-3.1 months) and OS was 5.0 months (95% CI = 3.0-8.8 months). In this retrospective analysis, prior treatment with IT does not appear to negatively influence response to BRAFi. Outcomes for IT with ipilimumab following BRAFi discontinuation are poor. Randomized controlled trials are needed to define if sequencing IT prior to BRAFi therapy is superior to sequencing BRAFi prior to IT. Cancer 2014. © 2014 American Cancer Society.
Article
Dabrafenib has activity in patients with brain metastases, but little is known of the relative efficacy of treatment within and outside the brain. This study sought to examine the intracranial (IC) and extracranial (EC) patterns of response and progression in patients with active melanoma brain metastases treated with dabrafenib. Clinicopathologic parameters were collected on patients with active brain metastases enrolled in the phase 1 and 2 studies of dabrafenib at a single institution. RECIST (Response Evaluation Criteria In Solid Tumors) response and progression-free survival (PFS) were prospectively assessed by disease site (IC versus EC). Treatments received after disease progression were also assessed. A total of 23 patients were studied. Response rates were similar in IC (78%) and EC (90%) sites (P = .416). IC and EC response was concordant in 71% of patients. Median site-specific PFS was identical in both IC and EC sites (23.6 weeks, P = .465), and exceeded whole-body PFS determined by RECIST (16.3 weeks). Of 20 patients with progressive disease (PD), 6 had IC PD only, 6 had EC PD only, and 8 had PD in both sites. In those with isolated intracranial PD, 5 of 6 underwent local therapy to the brain and continued on dabrafenib longer than 30 days. IC and EC melanoma metastases respond similarly to dabrafenib. There is no dominant site or pattern of disease progression in patients with brain metastases treated with dabrafenib. Salvage local therapy is possible in most patients after IC disease progression, with ongoing dabrafenib treatment possible in a subset of patients. Cancer 2014;120:530-536. © 2013 American Cancer Society.
Article
Tumour formation involves the co-evolution of neoplastic cells together with extracellular matrix, tumour vasculature and immune cells. Successful outgrowth of tumours and eventual metastasis is not determined solely by genetic alterations in tumour cells, but also by the fitness advantage such mutations confer in a given environment. As fitness is context dependent, evaluating tumours as complete organs, and not simply as masses of transformed epithelial cells, becomes paramount. The dynamic tumour topography varies drastically even throughout the same lesion. Heterologous cell types within tumours can actively influence therapeutic response and shape resistance.
Article
Phenotypic and functional heterogeneity arise among cancer cells within the same tumour as a consequence of genetic change, environmental differences and reversible changes in cell properties. Some cancers also contain a hierarchy in which tumorigenic cancer stem cells differentiate into non-tumorigenic progeny. However, it remains unclear what fraction of cancers follow the stem-cell model and what clinical behaviours the model explains. Studies using lineage tracing and deep sequencing could have implications for the cancer stem-cell model and may help to determine the extent to which it accounts for therapy resistance and disease progression.