SlideShare uma empresa Scribd logo
1 de 8
Baixar para ler offline
Genomic signatures to guide the use of
chemotherapeutics
Anil Potti1,2, Holly K Dressman1,3, Andrea Bild1,3, Richard F Riedel1,2, Gina Chan4, Robyn Sayer4,
Janiel Cragun4, Hope Cottrill4, Michael J Kelley2, Rebecca Petersen5, David Harpole5, Jeffrey Marks5,
Andrew Berchuck1,6, Geoffrey S Ginsburg1,2, Phillip Febbo1–3, Johnathan Lancaster4 &
Joseph R Nevins1–3
Using in vitro drug sensitivity data coupled with Affymetrix microarray data, we developed gene expression signatures that predict
sensitivity to individual chemotherapeutic drugs. Each signature was validated with response data from an independent set of cell
line studies. We further show that many of these signatures can accurately predict clinical response in individuals treated with
these drugs. Notably, signatures developed to predict response to individual agents, when combined, could also predict response
to multidrug regimens. Finally, we integrated the chemotherapy response signatures with signatures of oncogenic pathway
deregulation to identify new therapeutic strategies that make use of all available drugs. The development of gene expression
profiles that can predict response to commonly used cytotoxic agents provides opportunities to better use these drugs, including
using them in combination with existing targeted therapies.
Numerous advances have been achieved in the development, selection
and application of chemotherapeutic agents, sometimes with remark-
able clinical successes—as in the case of treatment for lymphomas or
platinum-based therapy for testicular cancers1. In addition, in several
instances, combination chemotherapy in the postoperative (adjuvant)
setting has been curative. However, most people with advanced solid
tumors will relapse and die of their disease. Moreover, administration
of ineffective chemotherapy increases the probability of side effects,
particularly those from cytotoxic agents, and of a consequent decrease
in quality of life1,2.
Recent work has demonstrated the value in using biomarkers to
select individuals for various targeted therapeutics, including tamox-
ifen, trastuzumab and imatinib mesylate. In contrast, equivalent tools
to select those most likely to respond to the commonly used
chemotherapeutic drugs are lacking3.
With the goal of developing genomic predictors of chemotherapy
sensitivity that could direct the use of cytotoxic agents to those most
likely to respond, we combined in vitro drug response data, together
with microarray gene expression data, to develop models that could
potentially predict responses to various cytotoxic chemotherapeutic
drugs4. We now show that these signatures can predict clinical or
pathologic response to the corresponding drugs, including combina-
tions of drugs. We further use the ability to predict deregulated
oncogenic signaling pathways in tumors to develop a strategy that
identifies opportunities for combining chemotherapeutic drugs with
targeted therapeutic drugs in a way that best matches the character-
istics of the individual.
RESULTS
A gene expression–based predictor of sensitivity to docetaxel
To develop predictors of cytotoxic chemotherapeutic drug response,
we used an approach similar to previous work analyzing the NCI-60
panel4 from the US National Cancer Institute (NCI). We first
identified cell lines that were most resistant or sensitive to docetaxel
(Fig. 1a,b) and then genes whose expression correlated most highly
with drug sensitivity, and used Bayesian binary regression analysis to
develop a model that differentiates a pattern of docetaxel sensitivity
from that of resistance. A gene expression signature consisting of 50
genes was identified that classified cell lines on the basis of docetaxel
sensitivity (Fig. 1b, right).
In addition to leave-one-out cross-validation, we used an indepen-
dent dataset derived from docetaxel sensitivity assays in a series
of 30 lung and ovarian cancer cell lines for further validation.
The significant correlation (P o 0.01, log-rank test) between the
predicted probability of sensitivity to docetaxel (in both lung and
ovarian cell lines) (Fig. 1c, left) and the respective 50% inhibitory
concentration (IC50) for docetaxel confirmed the capacity of the
docetaxel predictor to predict sensitivity to the drug in cancer cell
©2008NaturePublishingGrouphttp://www.nature.com/naturemedicine
Received 11 July; accepted 12 September; published online 22 October; corrected online 27 October 2006 (details online) and corrected after print 21 July 2008;
doi:10.1038/nm1491
1Duke Institute for Genome Sciences and Policy, Duke University, Box 3382, Durham, North Carolina 27710, USA. 2Department of Medicine, Duke University Medical
Center, Box 31295, Durham, North Carolina 27710, USA. 3Department of Molecular Genetics and Microbiology, Duke University Medical Center, Box 3054, Durham,
North Carolina 27710, USA. 4Division of Gynecologic Surgical Oncology, H. Lee Moffitt Cancer Center & Research Institute, University of South Florida, 12902
Magnolia Drive, Tampa, Florida 33612, USA. 5Department of Surgery, Duke University Medical Center, Box 3627, Durham, North Carolina 27710, USA. 6Department
of Obstetrics and Gynecology, Duke University Medical Center, Box 3079, Durham, North Carolina 27710, USA. Correspondence should be addressed to
J.R.N. (nevin001@mc.duke.edu).
1294 VOLUME 12 [ NUMBER 11 [ NOVEMBER 2006 NATURE MEDICINE
A R T I C L E S
lines (Supplementary Fig. 1 online). For each set of cell lines, the
accuracy exceeded 80%. Finally, we made use of a second independent
dataset that measured docetaxel sensitivity in a series of 29 lung cancer
cell lines (Gene Expression Omnibus (GEO) database (see URLs)
accession number GSE4127). The docetaxel sensitivity model devel-
oped from the NCI-60 panel again predicted sensitivity in this
independent dataset, also with an accuracy exceeding 80%
(P o 0.001, log-rank test; Fig. 1c, right).
The expression signature predicts clinical docetaxel response
We used published studies with clinical and genomic data that linked
gene expression data with clinical response to docetaxel in a breast
cancer neoadjuvant study5 (Fig. 1d) to test the capacity of the in vitro
docetaxel sensitivity predictor to accurately identify those individuals
that responded to docetaxel. Using a 0.45 predicted probability of
response as the cutoff for predicting positive response, as determined
by receiver operator characteristic curve analysis (Supplementary
Fig. 1), the in vitro–generated profile correctly predicted docetaxel
response in 22 of 24 clinical samples, for an overall accuracy of 91.6%
(Fig. 1d). Applying a Mann-Whitney U-test for statistical significance
demonstrated the capacity of the predictor to distinguish resistant
from sensitive individuals (Fig. 1d, right). We extended this further by
predicting the response to docetaxel as salvage therapy for individuals
with ovarian cancer that was refractory to primary therapy (Fig. 1e),
and the prediction achieved an accuracy exceeding 85% (Fig. 1e,
middle). Further, an analysis of statistical significance demonstrated
the capacity of the predictors to distinguish individuals with resistant
versus sensitive disease (Fig. 1e, right).
We also performed a complementary analysis, using the clinical
response data to generate a signature and then predicting sensitivity
of NCI-60 cell lines to docetaxel (Supplementary Fig. 1). Genes
represented in the initial in vitro–generated docetaxel predictor
and the alternative in vivo predictor showed considerable overlap.
Notably, both predictors were linked to expected targets for docetaxel,
including BCL2, WDR7 (also known as TRAG), ERBB2 and tubulin
genes, all previously described to be involved in taxane chemoresis-
tance6–9 (Supplementary Table 1 online). We also noted that the
predictor of docetaxel sensitivity developed from the NCI-60
data, using the approach described here, was more accurate in
predicting response in the ovarian samples than a predictor developed
from the breast neoadjuvant study data (85.7% versus 64.3%;
Supplementary Fig. 1).
A panel of signatures that predict chemotherapeutic sensitivity
Given the development of a docetaxel response predictor, we exam-
ined the NCI-60 dataset for other opportunities to develop predictors
©2008NaturePublishingGrouphttp://www.nature.com/naturemedicine
NCI-60
cell line panel
Identify resistant and sensitive cells
Build expression predictor of response
Drug response data
Affymetrix
expression data
Neoadjuvant breast
cancer therapy
Response to treatment
Biopsy
Affymetrix expression data
Drug sensitivity prediction
Ovarian salvage therapy
Response to treatment
Biopsy
Affymetrix expression data
Drug sensitivity prediction
100
75
50
25
0
Docetaxel LC50 Docetaxel GI50
Docetaxelconcentration
Resistant Sensitive
5
10
15
20
25
30
35
40
45
2 4 6 8 10 12 14
50
P < 0.01
P < 0.001
6
5
4
3
2
1
0
DocetaxelIC50
(Lungandovariancelllines)
0.1 0.2 0.3
Probability of docetaxel sensitivity
0.4 0.5 0.6 0.7 0.8 0.1 0.2 0.3
Probability of docetaxel sensitivity
0.4 0.5 0.6 0.7 0.8 0.9
8
7
6
5
4
3
2
1
0
DocetaxelIC50
(Lungcancercelllines)
1.0
0.8
0.6
0.4
0.2
0.0
0 5 10 15 20 25
Sample number
Probabilityofdocetaxel
resistance
Accuracy: 22/24 (91.6%)
Docetaxel sensitive
Docetaxel resistant
Accuracy: 12/14 (85.7%)
1.00
0.75
0.50
0.25
0.00
0 3 6 9
Sample number
12 15
Probabilityofdocetaxel
resistance
1.00
0.75
0.50
0.25
0.00
P < 0.001
Probabilityofdocetaxel
resistance
Docetaxel
sensitive
Docetaxel
resistant
Docetaxel
sensitive
Docetaxel
resistant
1.00
0.75
0.50
0.25
0.00
P < 0.01
Probabilityofdocetaxel
resistance
a b c
d
e
Docetaxel-resistant
lines
Docetaxel-sensitive
lines
Docetaxel sensitive
Docetaxel resistant
Figure 1 A gene expression signature that predicts sensitivity to docetaxel. (a) Strategy for generating the chemotherapeutic response predictor. (b) Left,
cell lines from the NCI-60 panel used to develop the in vitro signature of docetaxel sensitivity. There is a statistically significant difference (Mann-Whitney
U-test) in the IC50 and LC50 of the cell lines chosen to represent the sensitive and resistant subsets. Right, expression plots for genes selected for
discriminating the docetaxel-resistant and docetaxel-sensitive NCI-60 cell lines, with blue representing the lowest expression and red the highest. Each
column in the figure represents an individual sample. Each row represents an individual gene, ordered from top to bottom according to regression coefficient.
(c) Left, validation of the docetaxel response prediction model in an independent set of lung and ovarian cancer cell line samples. A collection of lung and
ovarian cell lines were used in a cell proliferation assay to determine the IC50 of docetaxel in the individual cell lines. Right, validation of the docetaxel
response prediction model in another independent set of 29 lung cancer cell line samples. (d) Left, a strategy for assessment of the docetaxel response
predictor as a function of clinical response in the breast neoadjuvant setting. Middle, predicted probability of docetaxel sensitivity in a collection of samples
from a breast cancer single-agent neoadjuvant study. Right, a single-variable scatter plot of a significance test of the predicted probabilities of sensitivity to
docetaxel in the sensitive and resistant tumors (P o 0.001, Mann-Whitney U-test). (e) Left, a strategy for assessment of the docetaxel response predictor as
a function of clinical response in advanced ovarian cancer. Middle, predicted probability of docetaxel sensitivity in a collection of samples from a prospective
single agent salvage therapy study. Right, a single-variable scatter plot showing statistical significance (P o 0.01, Mann-Whitney U-test).
A R T I C L E S
NATURE MEDICINE VOLUME 12 [ NUMBER 11 [ NOVEMBER 2006 1295
of chemotherapeutic response. We developed a series of expression
profiles from the NCI-60 dataset (Fig. 2a) that predict response to
topotecan, adriamycin, etoposide, 5-fluorouracil (5-FU), paclitaxel
and cyclophosphamide (Cytoxan). In each case, the leave-one-out
cross-validation analyses demonstrated the capacity of these profiles to
accurately predict the samples used in the development of the
predictor (Supplementary Fig. 2 online). Each profile was then
further validated using in vitro response data from independent
datasets; in each case, the profile developed from the NCI-60 data
was capable of accurately (485%) predicting response in the separate
dataset of approximately 30 cancer cell lines for which the dose-
response information and relevant Affymetrix U133A gene expression
data were publicly available10 (Supplementary Fig. 2 and Supple-
mentary Table 2 online). Once again, applying a Mann-Whitney
U-test for statistical significance demonstrated the capacity of the
predictor to distinguish resistant from sensitive individuals (Fig. 2b).
We then evaluated the extent to which a signature was also specific
for an individual chemotherapeutic agent. Using the validations of
chemosensitivity seen in the independent European cell line data10, it
was clear that each signature is specific for the drug that was used to
develop the predictor (Supplementary Fig. 3 online).
Given the ability of the in vitro–developed gene expression profiles to
predict response to docetaxel in the clinical samples, we extended this
approach to test the ability of additional signatures to predict responses
to commonly used salvage therapies for ovarian cancer and in another,
independent dataset of samples cultured from adriamycin-treated
individuals (GEO accession nos. GSE2351, GSE649, GSE650 and
GSE651). Each of these predictors was capable of accurately predicting
the response to the drugs in clinical samples, achieving an accuracy in
excess of 74% overall (Fig. 2c). In each case, the positive and negative
predictive values confirmed the validity of the approach (Supplemen-
tary Table 2).
Signatures predict response to multidrug regimens
Many therapeutic regimens make use of combinations of chemother-
apeutic drugs. To evaluate whether individual signatures of response
could predict response to regimens containing combinations of drugs,
we analyzed data from a breast neoadjuvant treatment study that used
a combination of paclitaxel, 5-FU, adriamycin and cyclophosphamide
(TFAC)11,12 (Fig. 3a). The available data from the 51 participants were
used to predict response using each of the single-agent signatures (for
paclitaxel, 5-FU, adriamycin and cyclophosphamide) developed from
the NCI-60 cell line analysis. The predicted response based on each of
the individual chemosensitivity signatures indicated a significant
distinction between the responders (n ¼ 13) and nonresponders
(n ¼ 38), with the exception of the prediction for 5-FU (Fig. 3a,
middle). The sensitivity to the four agents in the TFAC preoperative
(neoadjuvant) regimen was predicted as a combined probability. The
prediction of response based on a combined probability of sensitivity
built from the individual chemosensitivity predictions yielded a
statistically significant (P o 0.0001, Mann-Whitney U-test) distinc-
tion between the responders and nonresponders (Fig. 3a, right).
As a further validation of the capacity to predict response to
combination therapy, we analyzed gene expression data generated
from a collection of breast cancer (n ¼ 45) samples from subjects who
received 5-FU, adriamycin and cyclophosphamide (FAC) as adjuvant
chemotherapy (GEO database accession no. GSE3143). The predicted
response based on the individual signatures indicated a significant
distinction between the responders (n ¼ 34) and nonresponders (n ¼
11; Fig. 3b, left). Furthermore, the combined probability of sensitivity
to the three agents in the FAC regimen yielded a clear and significant
(P o 0.001, Mann-Whitney U-test) distinction between the respon-
ders and nonresponders (accuracy 82.2%, positive predictive value
90.3%, negative predictive value 64.3%; Fig. 3b). Although it is
difficult to interpret the prediction of clinical response in the adjuvant
©2008NaturePublishingGrouphttp://www.nature.com/naturemedicine
403020100
Sample numberSample number
0.00
0.25
Probabilityofpaclitaxel
sensitivity
0.50
0.75
1.00
0.00
0.25
Probabilityoftopotecan
sensitivity
0.50
0.75
1.00
0 10 20 30 40 50
Accuracy: 40/48 (83.3%)
Topotecan resistant
Topotecan sensitive
Accuracy: 28/35 (80%)
Paclitaxel resistant
Paclitaxel sensitive
P < 0.001
P < 0.001
P < 0.001P < 0.0001
P < 0.01
P = 0.001
ResistantSensitiveResistantSensitiveResistantSensitiveResistantSensitiveResistantSensitiveResistantSensitive
1.00
Predictedprobabilityof
sensitivitytotopotecan
Predictedprobabilityof
sensitivitytoadriamycin
Predictedprobabilityof
sensitivitytoetoposide
Predictedprobabilityof
sensitivityto5-FU
Predictedprobabilityof
sensitivitytopaclitaxel
Predictedprobabilityof
sensitivityto
cyclophosphamide
0.75
0.50
0.25
0.00
1.00
0.75
0.50
0.25
0.00
1.00
0.75
0.50
0.25
0.00 0.0
0.1
0.2
0.3
0.4
0.5
0.6
0.7
0.8
0.9
CyclophosphamidePaclitaxel5-FUEtoposideAdriamycinTopotecan
20
40
60
80
100
120
140
10
20
30
40
50
60
70
80
5
10
15
20
25
30
35
40
45
50
5
10
15
20
25
30
35
40
45
5
10
15
20
25
30
35
0.0
0.1
0.2
0.3
0.4
0.5
0.6
0.7
0.8
0.91.00
0.75
0.50
0.25
0.00
a
b
c 1.00
Probabilityofadriamycin
sensitivity
0.75
0.50
0.25
0.00
0 20 40
Sample number
60 80 100
Adriamycin resistant
Adriamycin sensitive
Accuracy: 70/95 (73.7%)
Figure 2 Development of a panel of gene expression signatures that predict sensitivity to chemotherapeutic drugs. (a) Gene expression patterns selected for
predicting response to the indicated drugs. (b) Independent validation of the chemotherapy response predictors in an independent set of cancer cell lines
that have dose-response and Affymetrix expression data10. A single-variable scatter plot of a significance test of the predicted probabilities of sensitivity to
any given drug in the sensitive and resistant cell lines (P values, Mann-Whitney U-test). (c) Prediction of single-agent therapy response in clinical samples
using in vitro cell line–based expression signatures of chemosensitivity. Left, the predicted probability of sensitivity to topotecan when compared to actual
clinical response data (n ¼ 48). Middle, the accuracy of the adriamycin predictor in a cohort of 122 samples. Right, predictive accuracy of the cell line–
based paclitaxel predictor when the drug was used as a salvage chemotherapy in advanced ovarian cancer (n ¼ 35).
A R T I C L E S
1296 VOLUME 12 [ NUMBER 11 [ NOVEMBER 2006 NATURE MEDICINE
setting, as many of these people were probably free of disease after
surgery, the accurate identification of nonresponders is a clear end-
point that does confirm the capacity of the signatures to predict
clinical response. We also examined the prognostic significance of the
prediction of response to FAC: it defined a poor prognosis group
(Fig. 3b, right). Taking these results together, we conclude that the
signatures of chemosensitivity generated from the NCI-60 panel do
indeed have the capacity to predict therapeutic response in individuals
receiving either single agent or combination chemotherapy (Supple-
mentary Tables 2 and 3 online).
An examination of the genes that constituted the paclitaxel pre-
dictor identified microtubule-associated protein tau (MAPT),
described previously as a determinant of paclitaxel sensitivity12. Also
consistent with previous reports5–7, TP53, methylenetetrahydrofolate
reductase (MTHFR) and DNA repair genes constituted the 5-FU
predictor, and excision repair mechanism genes (for example,
ERCC4), retinoblastoma pathway genes and BCL2 constituted the
adriamycin predictor (Supplementary Table 1).
Patterns of predicted chemotherapy response across tumor types
The panel of chemotherapy response predictors described in Figure 3
was used to profile the potential options for use of these drugs, by
predicting the likelihood of sensitivity to the seven agents in a large
collection of breast, lung and ovarian tumor samples. We then
clustered the samples according to patterns of predicted sensitivity
to the various chemotherapeutics and plotted a heatmap. There were
clearly evident patterns of predicted sensitiv-
ity to the various agents (Fig. 4). In many
cases, the predicted sensitivities to the chemo-
therapeutic agents were consistent with the
previously documented efficacy of single-
agent chemotherapies in the individual
tumor types13. For instance, the predicted
response rates for etoposide, adriamycin,
cyclophosphamide and 5-FU approximated
the observed response for these single agents
in individuals with breast cancer (Supple-
mentary Fig. 3). Likewise, the predicted sen-
sitivities to etoposide, docetaxel and paclitaxel
approximated the observed response for these
single agents in individuals with lung cancer
(Supplementary Fig. 3). This analysis also
suggested possibilities for alternate treat-
ments. As an example, it would seem that
individuals with breast cancer who are likely
to respond to 5-FU are resistant to adriamy-
cin and docetaxel (Supplementary Fig. 4
online). Likewise, in lung cancer, docetaxel-
sensitive individuals are likely to be resistant
to etoposide (Supplementary Fig. 4). This is
a potentially useful observation, considering
that both etoposide and docetaxel are viable
front-line options (in conjunction with cis-
platin or carboplatin) for people with lung
cancer1. A similar relationship is seen between
topotecan and adriamycin, both of which are
agents used in salvage chemotherapy for
ovarian cancer (Supplementary Fig. 4).
Thus, by identifying people or cohorts resis-
tant to a certain standard-of-care agent (for
example, topotecan), one could, by choosing
an alternative standard of care agent (for example, adriamycin), avoid
the side effects of the former agent without compromising outcome.
Linking chemotherapy sensitivity to oncogenic pathway status
Most people who are resistant to chemotherapeutic agents are
recruited into a second- or third-line therapy or enrolled in a clinical
trial14,15. Moreover, even those who initially respond to a given agent
are likely to eventually suffer a relapse, and thus, in either case,
additional therapeutic options are needed. As one approach to
identifying such options, we have taken advantage of our recent
work that describes the development of gene expression signatures
that reflect the activation of several oncogenic pathways16 (Supple-
mentary Fig. 5 and Supplementary Table 1 online). To illustrate this
approach, we first stratified the NCI cell lines based on predicted
docetaxel response and then examined the patterns of pathway
deregulation associated with docetaxel sensitivity or resistance (Sup-
plementary Fig. 5). Regression analysis showed a significant relation-
ship between phosphatidylinositol 3-OH (PI3)-kinase pathway
deregulation and docetaxel resistance (Supplementary Fig. 5).
These results indicated an opportunity to use a PI3-kinase inhibitor
in this subgroup, given our recent observations that have demon-
strated a linear positive correlation between the probability of pathway
deregulation and sensitivity to targeted drugs16. To address this
directly, we predicted docetaxel sensitivity and probability of
oncogenic pathway deregulation using DNA microarray data from
17 non–small cell lung cancer cell lines (Fig. 5a, left). Consistent with
©2008NaturePublishingGrouphttp://www.nature.com/naturemedicine
Neoadjuvant breast
cancer therapy
Response to treatment
Biopsy
Affymetrix expression data
Treatment with TFAC
Responders RespondersNonresponders
Responders Nonresponders
Nonresponders
1.00
0.75
0.50
0.25
0.00
1.00
0.75
0.50
0.25
0.00
T + 5-FU + A + C T + 5-FU + A + C
P < 0.0001
T F A AC CT 5-FU
5-FU (F)
Adriamycin (A)
Cyclophosphamide (C)
Probabilityofsensitivity
1.00
0.75
0.50
0.25
0.00
A AC C5-FU5-FU
Probabilityofsensitivity
Combinedprobabilityof
TFACsensitivity
P = 0.002
P = 0.3
P = 0.024
P = 0.003
Paclitaxel (T)
5-FU (F)
Adriamycin (A)
Cyclophosphamide (C)
Accuracy: 42/51 (82.3%)
NPV: 31/33 (94%)
PPV: 11/18 (61.1%)
Responders Nonresponders
1.00
0.75
0.50
0.25
0.00
5-FU + A + C 5-FU + A + C
P < 0.0001
Combinedprobabilityof
FACsensitivity
Accuracy: 37/45 (82.2%)
NPV: 9/14 (64.3%)
PPV: 28/31 (90.3%)
P = 0.003
P = 0.008
P = 0.04
0
0 25 50
Months
P < 0.001
75 100 125 150
25
Diseasefreesurvival(%)
50
75
100
a
b
Figure 3 Prediction of response to combination therapy. (a) Left, strategy for assessment of
chemotherapy response predictors in combination therapy as a function of pathologic response. Middle,
prediction of clinical response to neoadjuvant chemotherapy involving paclitaxel, 5-FU, adriamycin and
cyclophosphamide (TFAC) using the single-agent in vitro chemosensitivity signatures developed for each
of these drugs. Right, prediction of response (38 nonresponders, 13 responders) using a combined
probability predictor assessing the probabilities of all four chemosensitivity signatures in 51 people
treated with TFAC chemotherapy (P o 0.0001, Mann-Whitney U-test). Response was defined as a
complete pathologic response after completion of TFAC neoadjuvant therapy. (b) Left, prediction of
clinical response (n ¼ 45) to adjuvant chemotherapy involving 5-FU, adriamycin and cyclophosphamide
(FAC) using the single-agent in vitro chemosensitivity predictors developed for these drugs. Middle,
prediction of response (34 responders, 11 nonresponders) using a combined probability predictor
assessing the probabilities of all four chemosensitivity signatures in 45 people treated with FAC
chemotherapy. Right, Kaplan-Meier survival analysis for individuals predicted to be sensitive (blue
curve) or resistant (red curve) to FAC adjuvant chemotherapy. PPV, positive predictive value; NPV,
negative predictive value.
A R T I C L E S
NATURE MEDICINE VOLUME 12 [ NUMBER 11 [ NOVEMBER 2006 1297
the analysis of the NCI-60 cell line panel, the cell lines predicted to be
resistant to docetaxel were also predicted to show PI3-kinase pathway
activation (P ¼ 0.03, log-rank test; Supplementary Fig. 5). In parallel,
the lung cancer cell lines were assayed for sensitivity to a PI3-kinase–
specific inhibitor (LY-294002), using a standard measure of cell
proliferation14–16. The cell lines showing an increased probability of
PI3-kinase pathway activation were also more likely to respond to the
PI3-kinase inhibitor (P ¼ 0.001, log-rank test; Fig. 5b, left). The same
relationship held for prediction of resistance to docetaxel: these cells
were more likely to be sensitive to PI3-kinase inhibition (P o 0.001,
log-rank test; Fig. 5b, left).
An analysis of a panel of ovarian cancer cell lines provided a
second example of an opportunity to link predictions of chemo-
therapy sensitivity to oncogenic pathway deregulation. Ovarian cell
lines that were predicted to be topotecan resistant (Fig. 5a, right) had
a higher likelihood of Src pathway deregulation, and there was a
significant linear relationship (P ¼ 0.001, log-rank test) between the
probability of topotecan resistance and sensitivity to a drug (SU6656)
that inhibits the Src pathway (Fig. 5b, right). The results of these
assays clearly demonstrated an opportunity to mitigate drug resistance
(for example, resistance to docetaxel or topotecan) using a specific
pathway–targeted agent (PI3-kinase or Src inhibitor, respectively).
Taken together, these data demonstrate a rational approach to the
identification of therapeutic options for chemotherapy-resistant indivi-
duals, as well as to the identification of new combinations for chemo-
therapy-sensitive individuals, and thus have the potential to change the
current paradigm of cancer care. Prospective validation studies are,
however, needed to confirm the effectiveness of this approach.
DISCUSSION
The practice of oncology continually faces the challenge of matching
the right therapeutic regimen with the right individual, balancing
relative benefit with risk to achieve the most favorable outcome. This
challenge is often daunting, with marginal success rates in many
advanced disease contexts—probably reflecting the enormous com-
plexity of the disease process coupled with an inability to properly
guide the use of available therapeutics1,2,15. The results we present
here, focusing on the development of signatures that predict sensitivity
to common cytotoxic chemotherapeutic drugs, address this limitation
and have the potential to identify drugs—and combinations of
drugs—that best match the individual. The experimental strategy
for analyses used in this study is similar to that used for the
development of oncogenic pathway signatures16: samples representing
extreme cases are used to train the expression data to develop a
signature that can predict drug sensitivity. Of note, we further
demonstrated that these signatures can predict clinical drug response.
The importance of selecting individuals likely to respond to a given
therapeutic agent is perhaps best illustrated by the example of
trastuzumab. In the absence of selection, the overall response rate in
people with breast cancer is approximately 10%. In contrast, for those
selected on the basis of Her2 amplification, the overall response rate
rises to 35–50%17. We suggest that the gene expression signatures
predicting response to various cytotoxic chemotherapeutic agents may
provide an opportunity to optimize the use of these drugs.
Previous work has described the use of gene expression data,
coupled with in vitro drug sensitivity assays, to develop signatures
that could be used to classify response to therapy18,19. This involved
©2008NaturePublishingGrouphttp://www.nature.com/naturemedicine
Breast tumors
(n = 171)
Lung tumors
(n = 91)
Ovarian tumors
(n = 119)
Paclitaxel
Topotecan
5-FU
Docetaxel
Cyclophosphamide
Etoposide
Adriamycin
Etoposide
Paclitaxel
5-FU
Adriamycin
Topotecan
Cyclophosphamide
Docetaxel
Etoposide
Paclitaxel
5-FU
Adriamycin
Topotecan
Cyclophosphamide
Docetaxel
Figure 4 Patterns of predicted sensitivity to common chemotherapeutic drugs in human cancers. Hierarchical clustering of a collection of breast (n ¼ 171),
lung (n ¼ 91) and ovarian cancer (n ¼ 119) samples according to patterns of predicted sensitivity to the various chemotherapeutics. Predictions were
plotted as a heatmap in which high probability of sensitivity, or response, is indicated by red and low probability, or resistance, is indicated by blue.
A R T I C L E S
1298 VOLUME 12 [ NUMBER 11 [ NOVEMBER 2006 NATURE MEDICINE
the development of signatures that could identify chemoresistance to
the combination of drugs used in the treatment of acute lymphoblastic
leukemia. Our findings that individual signatures can accurately
predict response to drugs in both neoadjuvant and adjuvant che-
motherapy settings, where combinations of cytotoxic drugs are used,
clearly demonstrate the utility of the approach. We also describe a
rational approach to identifying new drug regimens, making use of
previously described signatures of oncogenic pathway deregulation16,
to provide a potential strategy for the development of therapeutic
regimens that would be based on knowledge of the status of an
individual’s tumor.
Finally, we note that the availability of predictors of chemotherapy
response, which have been shown to predict clinical response, provides
an opportunity to apply these predictors in present day practice. There
are several instances in which people are treated with one or more
therapeutic regimens that each have equal efficacy. An ability to select
the most effective therapy from among a panel of standard-of-care
agents is a strategy that can be easily applied20. Indeed, we believe this
represents an opportunity for a clinical trial that would evaluate the
performance of ‘random’ selection of agents (current practice) versus a
genomic signature–based selection as an initial step in the process of
achieving a more individualized treatment strategy (Fig. 6). Notably,
such a study may also identify those individuals likely to be resistant to
the available standard-of-care drugs, opening the way to a second-
generation trial that could evaluate the most effective treatment for
these people. Ultimately, we suggest that future treatment strategies
might be based on an analysis of an individual’s tumor, which would
then allow the development of a profile of likely sensitivity to common
chemotherapeutic drugs as well as to targeted therapies. Based on this
information, each individual might then be assigned to a combination
regimen that best matches the profile from the tumor.
METHODS
NCI-60 data. The complete details of the methods involved in the development
of the individual chemosensitivity predictors are available in Supplementary
Methods online. Briefly, the –log10 values of the 50% growth inhibition doses
(IC50; also known as GI50), total growth inhibition doses and 50% cytotoxic
doses (LC50) data were used to populate a matrix in MATLAB software, along
with the relevant expression data for the individual cell lines. Where multiple
entries for a drug screen existed (by NCS number), the entry with the largest
number of replicates was included. Incomplete data were assigned as ‘not a
number’ for statistical purposes. To develop an in vitro gene expression–based
predictor of sensitivity and resistance from the pharmacologic data used in the
NCI-60 drug screen studies, we chose cell lines within the NCI-60 panel that
would represent the extremes of sensitivity to a given chemotherapeutic agent
(mean IC50 ± 1 s.d.). Relevant expression data (revised data gathered using the
Affymetrix U95A2 GeneChip) for the solid tumor cell lines and the respective
pharmacological data for the chemotherapeutics were downloaded from the
NCI website. The individual drug sensitivity and resistance data from the
selected solid tumor NCI-60 cell lines were then used in a supervised analysis
using binary regression methodologies, as described previously21, to develop
models predictive of chemotherapeutic response.
Human ovarian cancer samples. We measured expression of 22,283 genes in
13 ovarian cancer cell lines and 119 advanced (Federation Internationale de
Gynecologie et d’Obstetrique (FIGO) stage III or IV) serous epithelial ovarian
carcinomas using Affymetrix U133A GeneChips. All ovarian cancers were
obtained at initial cytoreductive surgery. All tissues were collected under the
©2008NaturePublishingGrouphttp://www.nature.com/naturemedicine
Clinical presentation of
cancer
Biopsy
Chemotherapy response
prediction
Optimal single
chemotherapy
Combination
chemotherapy
Oncogenic pathway
prediction
Targeted
agent
Figure 6 Scheme for using chemotherapeutic and oncogenic pathway
predictors to identify individualized therapeutic options.
520
1666
125
549
1793
1651
23
2126
358
838
322
1838
441
1734
1650
647
2030
IGROV
OV2008
TOV-21D
A2780-CP
T8
TOV112D
FU-OV-1
OVCAR-5
C13
IMCC3
A2780-S
A2008
OV-90
Docetaxel
Src
Ras
β-Catenin
E2F3
Myc
Pl3K Pl3K
Topotecan
Src
Ras
β-Catenin
E2F3
Myc
Lung cancer cell lines Ovarian cancer cell lines
0.9 200
150
100
50
0.9
0.8
0.8
0.7
0.7
0.6
0.6
0.5
0.5
0.4
0.4
0.3
0.3
0.2
0.2
0.1
0.1
0.0
0.9
0.8
0.7
0.6
0.5
0.4
0.3
0.2
0.1
0.0
20
20
0
40
40
60
60
Src inhibition (µM)
100
100
80
80
0.00
0
25 50 75 100 125 150 175 200
PI3K inhibition (µM) Probability of docetaxel resistance
0.9 1.00.80.70.60.50.40.30.20.10.0
Probability of topotecan resistance
PI3KinhibitionIC50
(µM)SrcinhibitionIC50(µM)
ProbabilityofP13K
pathwayactivation
ProbabilityofSrc
pathwayactivation
Lung cancer cell lines
Ovarian cancer cell lines
a
b
P < 0.001
P < 0.001
P < 0.007 P = 0.001
Figure 5 Relationship between predicted chemotherapeutic sensitivity and
oncogenic pathway deregulation. (a) Left, probability of oncogenic pathway
deregulation as a function of predicted docetaxel sensitivity in a series of
lung cancer cell lines. Right, probability of oncogenic pathway deregulation
as a function of predicted topotecan sensitivity in a series of ovarian cancer
cell lines (red, sensitive; blue, resistant). (b) Top, lung cancer cell lines
showing an increased probability of PI3-kinase (PI3K) activation were also
more likely to respond to a PI3-kinase inhibitor (P ¼ 0.001, log-rank test),
as measured by sensitivity to the drug in assays of cell proliferation.
Furthermore, those cell lines predicted to be resistant to docetaxel were
more likely to be sensitive to PI3-kinase inhibition (P o 0.001, log-rank
test). Bottom, the relationship between Src pathway deregulation and
topotecan resistance in a set of 13 ovarian cancer cell lines. Ovarian cell
lines that are predicted to be topotecan resistant (a, right) have a higher
likelihood of Src pathway deregulation and there is a significant linear
relationship (P ¼ 0.001, log-rank test) between the probability of topotecan
resistance and sensitivity to a drug that inhibits the Src pathway.
A R T I C L E S
NATURE MEDICINE VOLUME 12 [ NUMBER 11 [ NOVEMBER 2006 1299
auspices of respective institutional (Duke University Medical Center and H. Lee
Moffitt Cancer Center) review board–approved protocols involving written
informed consent.
Full details of the methods used for RNA extraction and development of
gene expression signatures representing deregulation of oncogenic pathways in
the tumor samples were recently described16 and are available in Supplemen-
tary Methods. Response to therapy was evaluated using standard criteria for
those with measurable disease, based upon World Health Organization guide-
lines22. Complete details are available in Supplementary Methods.
Validation of in vitro chemotherapy response signatures. Complete details of
the publicly available datasets used for independent validation of the genomic
signatures of chemosensitivity are described in Supplementary Methods.
PI3-kinase signature. A signature representative of PI3-kinase activation was
developed as previously described16. The genes that constitute the PI3-kinase
signature are shown in Supplementary Table 1.
Lung and ovarian cancer cell culture. Total RNA was extracted and oncogenic
pathway predictions were performed similarly to the methods described
previously16. Complete details are available in Supplementary Methods.
Cross-platform Affymetrix Gene Chip comparison. To map the probe sets
across various generations of Affymetrix GeneChip arrays, we used an in-house
program, Chip Comparer (see URLs), as described previously16.
Cell proliferation assays. Methods for drug sensitivity assays are described in
Supplementary Methods.
Statistical analysis methods. Analysis of expression data was performed
as previously described16,21,23,24 and is detailed in Supplementary
Methods. In instances where a combined probability of sensitivity to a
combination chemotherapeutic regimen was required based on the individual
drug sensitivity patterns, we used the probabilities of response to individual
drugs and used the theorem for combined probabilities as described by
William Feller to deduce a probability of response to a combination of the
drugs being studied. The result was then mean-centered to give a probability
between 0 and 1. Hierarchical clustering of tumor predictions was performed
using Gene Cluster 3.0 (ref. 25). Genes and tumors were clustered
using average linkage with the uncentered correlation similarity metric.
Standard linear regression analyses and their significance (log-rank test) were
generated for the drug response data and for correlation between drug
response and probability of chemosensitivity or pathway deregulation using
GraphPad software.
URLs. NCI, http://dtp.nci.nih.gov/docs/cancer/cancer_data.html. GEO data-
base, http://www.ncbi.nlm.nih.gov/geo/. Chip Comparer, http://tenero.duhs.
duke.edu/genearray/perl/chip/chipcomparer.pl.
Note: Supplementary information is available on the Nature Medicine website.
ACKNOWLEDGMENTS
The authors would like to acknowledge and thank L. Pusztai for providing
us with the clinical and expression data on the samples studied from the
M.D. Anderson Cancer Center. Funding for this work was supplied in part by
research grants from the US National Institutes of Health (NCI-U54 CA112952-
02 (J.R.N.) and R01CA106520 (J.R.N.)). A.P. is an American Association for
Cancer Research Translational Research Grant awardee, J.L. receives research
support from Glaxo-Smith-Kline and Sanofi-Aventis Pharmaceuticals and
Orthobiotech, and P.F. is a Damon Runyon Cancer Research Investigator.
Additional sources of funding included the Ovarian Cancer Research Fund and
the Hearing the Ovarian Cancer Whisper Jacquie Liggett Fellowship.
COMPETING INTERESTS STATEMENT
The authors declare competing financial interests (see the Nature Medicine website
for details).
Published online at http://www.nature.com/naturemedicine
Reprints and permissions information is available online at http://npg.nature.com/
reprintsandpermissions/
1. Herbst, R.S. et al. Clinical cancer advances 2005: major research advances in cancer
treatment, prevention, and screening—a report from the American Society of Clinical
Oncology. J. Clin. Oncol. 24, 190–205 (2006).
2. Breathnach, O.S. et al. Twenty-two years of phase III trials for patients with advanced
non-small-cell lung cancer: sobering results. J. Clin. Oncol. 19, 1734–1742
(2001).
3. Blagosklonny, M.V. Why therapeutic response may not prolong the life of a cancer
patient: selection for oncogenic resistance. Cell Cycle 4, 1693–1698 (2005).
4. Staunton, J.E. et al. Chemosensitivity prediction by transcriptional profiling. Proc. Natl.
Acad. Sci. USA 98, 10787–10792 (2001).
5. Chang, J.C. et al. Gene expression profiling for the prediction of therapeutic response to
docetaxel in patients with breast cancer. Lancet 362, 362–369 (2003).
6. Emi, M., Kim, R., Tanabe, K., Uchida, Y. & Toge, T. Targeted therapy against
Bcl-2-related proteins in breast cancer cells. Breast Cancer Res. 7, R940–R952
(2005).
7. Takahashi, T. et al. Cyclin A-associated kinase activity is needed for paclitaxel
sensitivity. Mol. Cancer Ther. 4, 1039–1046 (2005).
8. Modi, S. et al. Phosphorylated/activated HER2 as a marker of clinical resistance to
single agent taxane chemotherapy for metastatic breast cancer. Cancer Invest. 23,
483–487 (2005).
9. Langer, R. et al. Association of pretherapeutic expression of chemotherapy-related
genes with response to neoadjuvant chemotherapy in Barrett carcinoma. Clin. Cancer
Res. 11, 7462–7469 (2005).
10. Gyorffy, B. et al. Gene expression profiling of 30 cancer cell lines predicts resistance
towards 11 anticancer drugs at clinically achieved concentrations. Int. J. Cancer 118,
1699–1712 (2005).
11. Rouzier, R. et al. Breast cancer molecular subtypes respond differently to preoperative
chemotherapy. Clin. Cancer Res. 11, 5678–5685 (2005).
12. Rouzier, R. et al. Microtubule-associated protein tau: a marker of paclitaxel
sensitivity on breast cancer. Proc. Natl. Acad. Sci. USA 102, 8315–8320
(2005).
13. DeVita, V.T., Hellman, S. & Rosenberg, S.A. Cancer: Principles and Practice of
Oncology (Lippincott-Raven, Philadelphia, 2005).
14. Broxterman, H.J. & Georgopapadakou, N.H. Anticancer therapeutics: ‘‘addictive’’
targets, multi-targeted drugs, new drug combinations. Drug Resist. Updat. 8,
183–197 (2005).
15. Minna, J.D., Gazdar, A.F., Sprang, S.R. & Herz, J. Cancer: a bull’s eye for targeted lung
cancer therapy. Science 304, 1458–1461 (2004).
16. Bild, A. et al. Oncogenic pathway signatures in human cancers as a guide to targeted
therapies. Nature 439, 353–357 (2006).
17. Vogel, C.L. et al. Efficacy and safety of trastuzumab as a single agent in first-line
treatment of HER2-overexpressing metastatic breast cancer. J. Clin. Oncol. 20,
719–726 (2002).
18. Lugthart, S. et al. Identification of genes associated with chemotherapy crossresis-
tance and treatment response in childhood acute lymphoblastic leukemia. Cancer Cell
7, 375–386 (2005).
19. Holleman, A. et al. Gene expression patterns in drug resistant acute lympho-
blastic leukemia cells and response to treatment. N. Engl. J. Med. 351, 533–542
(2004).
20. Sargent, D.J., Conley, B.A., Allegra, C. & Collette, L. Clinical trial designs for predictive
marker validation in cancer treatment trials. J. Clin. Oncol. 23, 2020–2027 (2005).
21. Pittman, J., Huang, E., Wang, Q., Nevins, J.R. & West, M. Bayesian analysis of binary
prediction tree models for retrospectively sampled outcomes. Biostatistics 5, 587–601
(2004).
22. Miller, A.B., Hoogstraten, B., Staquet, M. & Winkler, A. Reporting results of cancer
treatment. Cancer 47, 207–214 (1981).
23. West, M. et al. Predicting the clinical status of human breast cancer by using
gene expression profiles. Proc. Natl. Acad. Sci. USA 98, 11462–11467 (2001).
24. Ihaka, R. & Gentleman, R. A language for data analysis and graphics. J. Comput.
Graph. Stat. 5, 299–314 (1996).
25. Eisen, M.B., Spellman, P.T., Brown, P.O. & Botstein, D. Cluster analysis and display of
genome-wide expression patterns. Proc. Natl. Acad. Sci. USA 95, 14863–14868
(1998).
©2008NaturePublishingGrouphttp://www.nature.com/naturemedicine
A R T I C L E S
1300 VOLUME 12 [ NUMBER 11 [ NOVEMBER 2006 NATURE MEDICINE
Corrigendum: Genomic signatures to guide the use of chemotherapeutics
Anil Potti, Holly K Dressman,Andrea Bild, Richard F Riedel, Gina Chan, Robyn Sayer, Janiel Cragun, Hope Cottrill, Michael J Kelley, Rebecca
Petersen, David Harpole, Jeffrey Marks,Andrew Berchuck, Geoffrey S Ginsburg, Phillip Febbo, Johnathan Lancaster & Joseph R Nevins
Nat. Med. 12, 1294–1300 (2006); published online 22 October 2006; corrected after print 10 May and 10 October 2007 and 18 July 2008
In the version of this article initially published,of the 122 samples assayed for sensitivity to daunorubicin for which the authors applied a predictor
of adriamycin sensitivity, 27 samples were replicated owing to the fact that the same samples were included in several separate series files in the
Gene Expression Omnibus generated in 2004 and 2005,which were the source of the data provided for the study.The authors have reanalyzed the
prediction of adriamycin sensitivity using only the 95 unique samples and find a revised accuracy of 74%. Additionally, the authors have added
two more accession numbers (GSE2351 and GSE649) to more clearly identify the sources of the samples. The errors have been corrected in the
HTML and PDF versions of the article.
nature medicine volume 14 | number 8 | august 2008	 889
0 20 40 60 80 100
Sample number
1.00
0.75
0.50
0.25
0.00
Probabilityofadriamycin
sensitivity
Adriamycin resistant
Adriamycin sensitive
Accuracy: 70/95 (73.7%)
co r r i g e n da©2008NaturePublishingGrouphttp://www.nature.com/naturemedicine

Mais conteúdo relacionado

Mais procurados

ATS_NP economic poster S03 (5May15)
ATS_NP economic poster S03 (5May15)ATS_NP economic poster S03 (5May15)
ATS_NP economic poster S03 (5May15)Goran Medic
 
Autologous Bone Marrow Cell Therapy for Autism: An Open Label Uncontrolled C...
Autologous Bone Marrow Cell Therapy for Autism: An  Open Label Uncontrolled C...Autologous Bone Marrow Cell Therapy for Autism: An  Open Label Uncontrolled C...
Autologous Bone Marrow Cell Therapy for Autism: An Open Label Uncontrolled C...remedypublications2
 
Multiscale integrative data analytics in pharmacogenomics
Multiscale integrative data analytics in pharmacogenomicsMultiscale integrative data analytics in pharmacogenomics
Multiscale integrative data analytics in pharmacogenomicsDr. Gerry Higgins
 
Inmunoterapia para carcinoma metastasico renal
Inmunoterapia para carcinoma metastasico renalInmunoterapia para carcinoma metastasico renal
Inmunoterapia para carcinoma metastasico renalDana Pinto Ramos
 
Drug Repurposing Against Infectious Diseases
Drug Repurposing Against Infectious Diseases Drug Repurposing Against Infectious Diseases
Drug Repurposing Against Infectious Diseases Philip Bourne
 
Joseph Levy MedicReS World Congress 2013 - 2
Joseph Levy MedicReS World Congress 2013 - 2Joseph Levy MedicReS World Congress 2013 - 2
Joseph Levy MedicReS World Congress 2013 - 2MedicReS
 
G.Medic ISPOR EU 2015
G.Medic ISPOR EU 2015G.Medic ISPOR EU 2015
G.Medic ISPOR EU 2015Goran Medic
 
Personalized Medicine in Diagnosis and Treatment of Cancer
Personalized Medicine in Diagnosis and Treatment of Cancer Personalized Medicine in Diagnosis and Treatment of Cancer
Personalized Medicine in Diagnosis and Treatment of Cancer Maryam Rafati
 
Moving from Big Data to Better Models of Disease and Drug Response - Joel Dudley
Moving from Big Data to Better Models of Disease and Drug Response - Joel DudleyMoving from Big Data to Better Models of Disease and Drug Response - Joel Dudley
Moving from Big Data to Better Models of Disease and Drug Response - Joel DudleyCityAge
 
Towards Replicable and Genereralizable Genomic Prediction Models
Towards Replicable and Genereralizable Genomic Prediction ModelsTowards Replicable and Genereralizable Genomic Prediction Models
Towards Replicable and Genereralizable Genomic Prediction ModelsLevi Waldron
 
Carcinogenicity of Glyphosate A Systematic Review of the Available Evidence
Carcinogenicity of Glyphosate  A Systematic Review of the Available  EvidenceCarcinogenicity of Glyphosate  A Systematic Review of the Available  Evidence
Carcinogenicity of Glyphosate A Systematic Review of the Available EvidenceAsociación Toxicológica Argentina
 
Methylation Subtypes of EAC/BE
Methylation Subtypes of EAC/BE Methylation Subtypes of EAC/BE
Methylation Subtypes of EAC/BE Sean Maden
 
The case for Genomic Medicine, (Personalized, Individualized Medicine). Medic...
The case for Genomic Medicine, (Personalized, Individualized Medicine). Medic...The case for Genomic Medicine, (Personalized, Individualized Medicine). Medic...
The case for Genomic Medicine, (Personalized, Individualized Medicine). Medic...Mathura Shanmugasundaram PhD
 
Mechanisms and applications of apoptosis based and molecular
Mechanisms and applications of apoptosis based and molecularMechanisms and applications of apoptosis based and molecular
Mechanisms and applications of apoptosis based and molecularDrSatyabrataSahoo
 
SciTech Development Testamonial Brochure
SciTech Development Testamonial BrochureSciTech Development Testamonial Brochure
SciTech Development Testamonial BrochureSciTech Development
 
E1512 Trial Spotlight for May 2013 ECOG-ACRIN Newsletter
E1512 Trial Spotlight for May 2013 ECOG-ACRIN NewsletterE1512 Trial Spotlight for May 2013 ECOG-ACRIN Newsletter
E1512 Trial Spotlight for May 2013 ECOG-ACRIN NewsletterSara Bucknam
 

Mais procurados (20)

ATS_NP economic poster S03 (5May15)
ATS_NP economic poster S03 (5May15)ATS_NP economic poster S03 (5May15)
ATS_NP economic poster S03 (5May15)
 
A Clinical Prediction Rule for Fluoroquinolone Resistance 3 14 11
A Clinical Prediction Rule for Fluoroquinolone Resistance 3 14 11A Clinical Prediction Rule for Fluoroquinolone Resistance 3 14 11
A Clinical Prediction Rule for Fluoroquinolone Resistance 3 14 11
 
Autologous Bone Marrow Cell Therapy for Autism: An Open Label Uncontrolled C...
Autologous Bone Marrow Cell Therapy for Autism: An  Open Label Uncontrolled C...Autologous Bone Marrow Cell Therapy for Autism: An  Open Label Uncontrolled C...
Autologous Bone Marrow Cell Therapy for Autism: An Open Label Uncontrolled C...
 
Oral cancer and Gene therapy
Oral cancer and Gene therapyOral cancer and Gene therapy
Oral cancer and Gene therapy
 
Multiscale integrative data analytics in pharmacogenomics
Multiscale integrative data analytics in pharmacogenomicsMultiscale integrative data analytics in pharmacogenomics
Multiscale integrative data analytics in pharmacogenomics
 
Inmunoterapia para carcinoma metastasico renal
Inmunoterapia para carcinoma metastasico renalInmunoterapia para carcinoma metastasico renal
Inmunoterapia para carcinoma metastasico renal
 
Drug Repurposing Against Infectious Diseases
Drug Repurposing Against Infectious Diseases Drug Repurposing Against Infectious Diseases
Drug Repurposing Against Infectious Diseases
 
Joseph Levy MedicReS World Congress 2013 - 2
Joseph Levy MedicReS World Congress 2013 - 2Joseph Levy MedicReS World Congress 2013 - 2
Joseph Levy MedicReS World Congress 2013 - 2
 
588 Carter
588 Carter588 Carter
588 Carter
 
G.Medic ISPOR EU 2015
G.Medic ISPOR EU 2015G.Medic ISPOR EU 2015
G.Medic ISPOR EU 2015
 
Personalized Medicine in Diagnosis and Treatment of Cancer
Personalized Medicine in Diagnosis and Treatment of Cancer Personalized Medicine in Diagnosis and Treatment of Cancer
Personalized Medicine in Diagnosis and Treatment of Cancer
 
Moving from Big Data to Better Models of Disease and Drug Response - Joel Dudley
Moving from Big Data to Better Models of Disease and Drug Response - Joel DudleyMoving from Big Data to Better Models of Disease and Drug Response - Joel Dudley
Moving from Big Data to Better Models of Disease and Drug Response - Joel Dudley
 
Towards Replicable and Genereralizable Genomic Prediction Models
Towards Replicable and Genereralizable Genomic Prediction ModelsTowards Replicable and Genereralizable Genomic Prediction Models
Towards Replicable and Genereralizable Genomic Prediction Models
 
Carcinogenicity of Glyphosate A Systematic Review of the Available Evidence
Carcinogenicity of Glyphosate  A Systematic Review of the Available  EvidenceCarcinogenicity of Glyphosate  A Systematic Review of the Available  Evidence
Carcinogenicity of Glyphosate A Systematic Review of the Available Evidence
 
Methylation Subtypes of EAC/BE
Methylation Subtypes of EAC/BE Methylation Subtypes of EAC/BE
Methylation Subtypes of EAC/BE
 
srep08308 Bidii
srep08308 Bidiisrep08308 Bidii
srep08308 Bidii
 
The case for Genomic Medicine, (Personalized, Individualized Medicine). Medic...
The case for Genomic Medicine, (Personalized, Individualized Medicine). Medic...The case for Genomic Medicine, (Personalized, Individualized Medicine). Medic...
The case for Genomic Medicine, (Personalized, Individualized Medicine). Medic...
 
Mechanisms and applications of apoptosis based and molecular
Mechanisms and applications of apoptosis based and molecularMechanisms and applications of apoptosis based and molecular
Mechanisms and applications of apoptosis based and molecular
 
SciTech Development Testamonial Brochure
SciTech Development Testamonial BrochureSciTech Development Testamonial Brochure
SciTech Development Testamonial Brochure
 
E1512 Trial Spotlight for May 2013 ECOG-ACRIN Newsletter
E1512 Trial Spotlight for May 2013 ECOG-ACRIN NewsletterE1512 Trial Spotlight for May 2013 ECOG-ACRIN Newsletter
E1512 Trial Spotlight for May 2013 ECOG-ACRIN Newsletter
 

Semelhante a Genomic signatures to guide use of chemotherapeutics

BioIT Drug induced liver injury talk 2011
BioIT Drug induced liver injury talk 2011BioIT Drug induced liver injury talk 2011
BioIT Drug induced liver injury talk 2011Sean Ekins
 
Biomedicine & Pharmacotherapy
Biomedicine & PharmacotherapyBiomedicine & Pharmacotherapy
Biomedicine & PharmacotherapyTrustlife
 
Talk at Yale University April 26th 2011: Applying Computational Models for To...
Talk at Yale University April 26th 2011: Applying Computational Modelsfor To...Talk at Yale University April 26th 2011: Applying Computational Modelsfor To...
Talk at Yale University April 26th 2011: Applying Computational Models for To...Sean Ekins
 
The Principle of Rational Design of Drug Combination and Personalized Therapy...
The Principle of Rational Design of Drug Combination and Personalized Therapy...The Principle of Rational Design of Drug Combination and Personalized Therapy...
The Principle of Rational Design of Drug Combination and Personalized Therapy...Jianghui Xiong
 
Assessing the clinical utility of cancer genomic and proteomic data across tu...
Assessing the clinical utility of cancer genomic and proteomic data across tu...Assessing the clinical utility of cancer genomic and proteomic data across tu...
Assessing the clinical utility of cancer genomic and proteomic data across tu...Gul Muneer
 
Establishment and analysis of a disease risk prediction model for chronic kid...
Establishment and analysis of a disease risk prediction model for chronic kid...Establishment and analysis of a disease risk prediction model for chronic kid...
Establishment and analysis of a disease risk prediction model for chronic kid...KrishMendapara1
 
CXCL1, CCL20, STAT1 was Identified and Validated as a Key Biomarker Related t...
CXCL1, CCL20, STAT1 was Identified and Validated as a Key Biomarker Related t...CXCL1, CCL20, STAT1 was Identified and Validated as a Key Biomarker Related t...
CXCL1, CCL20, STAT1 was Identified and Validated as a Key Biomarker Related t...semualkaira
 
MEDICAL IMAGING MUTIFRACTAL ANALYSIS IN PREDICTION OF EFFICIENCY OF CANCER TH...
MEDICAL IMAGING MUTIFRACTAL ANALYSIS IN PREDICTION OF EFFICIENCY OF CANCER TH...MEDICAL IMAGING MUTIFRACTAL ANALYSIS IN PREDICTION OF EFFICIENCY OF CANCER TH...
MEDICAL IMAGING MUTIFRACTAL ANALYSIS IN PREDICTION OF EFFICIENCY OF CANCER TH...cscpconf
 
MEDICAL IMAGING MUTIFRACTAL ANALYSIS IN PREDICTION OF EFFICIENCY OF CANCER TH...
MEDICAL IMAGING MUTIFRACTAL ANALYSIS IN PREDICTION OF EFFICIENCY OF CANCER TH...MEDICAL IMAGING MUTIFRACTAL ANALYSIS IN PREDICTION OF EFFICIENCY OF CANCER TH...
MEDICAL IMAGING MUTIFRACTAL ANALYSIS IN PREDICTION OF EFFICIENCY OF CANCER TH...csandit
 
fonc-04-00078.pdf
fonc-04-00078.pdffonc-04-00078.pdf
fonc-04-00078.pdfsafinur3
 
41467_2022_Article_32816.pdf
41467_2022_Article_32816.pdf41467_2022_Article_32816.pdf
41467_2022_Article_32816.pdfYifeiGuo6
 
CIIProCluster: Developing Read-Across Predictive Toxicity Models Using Big Data
CIIProCluster: Developing Read-Across Predictive Toxicity Models Using Big DataCIIProCluster: Developing Read-Across Predictive Toxicity Models Using Big Data
CIIProCluster: Developing Read-Across Predictive Toxicity Models Using Big DataDaniel Russo
 
QMB_Poster_Tom_Kelly
QMB_Poster_Tom_KellyQMB_Poster_Tom_Kelly
QMB_Poster_Tom_KellyTom Kelly
 
Population variation to drugs
Population variation to drugsPopulation variation to drugs
Population variation to drugsLars Brandén
 

Semelhante a Genomic signatures to guide use of chemotherapeutics (20)

BioIT Drug induced liver injury talk 2011
BioIT Drug induced liver injury talk 2011BioIT Drug induced liver injury talk 2011
BioIT Drug induced liver injury talk 2011
 
P1-01-17_poster
P1-01-17_posterP1-01-17_poster
P1-01-17_poster
 
Biomedicine & Pharmacotherapy
Biomedicine & PharmacotherapyBiomedicine & Pharmacotherapy
Biomedicine & Pharmacotherapy
 
Talk at Yale University April 26th 2011: Applying Computational Models for To...
Talk at Yale University April 26th 2011: Applying Computational Modelsfor To...Talk at Yale University April 26th 2011: Applying Computational Modelsfor To...
Talk at Yale University April 26th 2011: Applying Computational Models for To...
 
10.1128@jcm.00298 17
10.1128@jcm.00298 1710.1128@jcm.00298 17
10.1128@jcm.00298 17
 
The Principle of Rational Design of Drug Combination and Personalized Therapy...
The Principle of Rational Design of Drug Combination and Personalized Therapy...The Principle of Rational Design of Drug Combination and Personalized Therapy...
The Principle of Rational Design of Drug Combination and Personalized Therapy...
 
Assessing the clinical utility of cancer genomic and proteomic data across tu...
Assessing the clinical utility of cancer genomic and proteomic data across tu...Assessing the clinical utility of cancer genomic and proteomic data across tu...
Assessing the clinical utility of cancer genomic and proteomic data across tu...
 
A predictive ligand based Bayesian model for human drug induced liver injury
A predictive ligand based Bayesian model for human drug induced liver injury A predictive ligand based Bayesian model for human drug induced liver injury
A predictive ligand based Bayesian model for human drug induced liver injury
 
Establishment and analysis of a disease risk prediction model for chronic kid...
Establishment and analysis of a disease risk prediction model for chronic kid...Establishment and analysis of a disease risk prediction model for chronic kid...
Establishment and analysis of a disease risk prediction model for chronic kid...
 
Isaac Kohane, "A Data Perspective on Autonomy, Human Rights, and the End of N...
Isaac Kohane, "A Data Perspective on Autonomy, Human Rights, and the End of N...Isaac Kohane, "A Data Perspective on Autonomy, Human Rights, and the End of N...
Isaac Kohane, "A Data Perspective on Autonomy, Human Rights, and the End of N...
 
CXCL1, CCL20, STAT1 was Identified and Validated as a Key Biomarker Related t...
CXCL1, CCL20, STAT1 was Identified and Validated as a Key Biomarker Related t...CXCL1, CCL20, STAT1 was Identified and Validated as a Key Biomarker Related t...
CXCL1, CCL20, STAT1 was Identified and Validated as a Key Biomarker Related t...
 
MEDICAL IMAGING MUTIFRACTAL ANALYSIS IN PREDICTION OF EFFICIENCY OF CANCER TH...
MEDICAL IMAGING MUTIFRACTAL ANALYSIS IN PREDICTION OF EFFICIENCY OF CANCER TH...MEDICAL IMAGING MUTIFRACTAL ANALYSIS IN PREDICTION OF EFFICIENCY OF CANCER TH...
MEDICAL IMAGING MUTIFRACTAL ANALYSIS IN PREDICTION OF EFFICIENCY OF CANCER TH...
 
MEDICAL IMAGING MUTIFRACTAL ANALYSIS IN PREDICTION OF EFFICIENCY OF CANCER TH...
MEDICAL IMAGING MUTIFRACTAL ANALYSIS IN PREDICTION OF EFFICIENCY OF CANCER TH...MEDICAL IMAGING MUTIFRACTAL ANALYSIS IN PREDICTION OF EFFICIENCY OF CANCER TH...
MEDICAL IMAGING MUTIFRACTAL ANALYSIS IN PREDICTION OF EFFICIENCY OF CANCER TH...
 
fonc-04-00078.pdf
fonc-04-00078.pdffonc-04-00078.pdf
fonc-04-00078.pdf
 
41467_2022_Article_32816.pdf
41467_2022_Article_32816.pdf41467_2022_Article_32816.pdf
41467_2022_Article_32816.pdf
 
CIIProCluster: Developing Read-Across Predictive Toxicity Models Using Big Data
CIIProCluster: Developing Read-Across Predictive Toxicity Models Using Big DataCIIProCluster: Developing Read-Across Predictive Toxicity Models Using Big Data
CIIProCluster: Developing Read-Across Predictive Toxicity Models Using Big Data
 
QMB_Poster_Tom_Kelly
QMB_Poster_Tom_KellyQMB_Poster_Tom_Kelly
QMB_Poster_Tom_Kelly
 
Plos
PlosPlos
Plos
 
Genomics: Personalised Medicine in Brain Cancer?
Genomics: Personalised Medicine in Brain Cancer?Genomics: Personalised Medicine in Brain Cancer?
Genomics: Personalised Medicine in Brain Cancer?
 
Population variation to drugs
Population variation to drugsPopulation variation to drugs
Population variation to drugs
 

Mais de Sandra Gallaga (20)

Alteraciones de la circulacion
Alteraciones de la circulacionAlteraciones de la circulacion
Alteraciones de la circulacion
 
2 parcial work
2 parcial work2 parcial work
2 parcial work
 
Apoptosis
ApoptosisApoptosis
Apoptosis
 
2 parcial work
2 parcial work2 parcial work
2 parcial work
 
Angiogenesis
AngiogenesisAngiogenesis
Angiogenesis
 
alteraciones de la circulacion
alteraciones de la circulacionalteraciones de la circulacion
alteraciones de la circulacion
 
Aborto
AbortoAborto
Aborto
 
Infanticidio
InfanticidioInfanticidio
Infanticidio
 
Aborto
AbortoAborto
Aborto
 
Aborto Ml
Aborto MlAborto Ml
Aborto Ml
 
T. Forense
T. ForenseT. Forense
T. Forense
 
Heridas Ocasionadas Por Arma Blanca
Heridas Ocasionadas Por Arma BlancaHeridas Ocasionadas Por Arma Blanca
Heridas Ocasionadas Por Arma Blanca
 
Tanatologia
TanatologiaTanatologia
Tanatologia
 
Policrestos.Docx Alexis
Policrestos.Docx AlexisPolicrestos.Docx Alexis
Policrestos.Docx Alexis
 
Homeopa1
Homeopa1Homeopa1
Homeopa1
 
Medicamentos Policrestos
Medicamentos PolicrestosMedicamentos Policrestos
Medicamentos Policrestos
 
Infecciones Virales[1]
Infecciones Virales[1]Infecciones Virales[1]
Infecciones Virales[1]
 
Inmuno
InmunoInmuno
Inmuno
 
Concepto De Inmunogenicidad Ag
Concepto De Inmunogenicidad AgConcepto De Inmunogenicidad Ag
Concepto De Inmunogenicidad Ag
 
Citotoxicidad Mediada Por CéLulas
Citotoxicidad Mediada Por CéLulasCitotoxicidad Mediada Por CéLulas
Citotoxicidad Mediada Por CéLulas
 

Genomic signatures to guide use of chemotherapeutics

  • 1. Genomic signatures to guide the use of chemotherapeutics Anil Potti1,2, Holly K Dressman1,3, Andrea Bild1,3, Richard F Riedel1,2, Gina Chan4, Robyn Sayer4, Janiel Cragun4, Hope Cottrill4, Michael J Kelley2, Rebecca Petersen5, David Harpole5, Jeffrey Marks5, Andrew Berchuck1,6, Geoffrey S Ginsburg1,2, Phillip Febbo1–3, Johnathan Lancaster4 & Joseph R Nevins1–3 Using in vitro drug sensitivity data coupled with Affymetrix microarray data, we developed gene expression signatures that predict sensitivity to individual chemotherapeutic drugs. Each signature was validated with response data from an independent set of cell line studies. We further show that many of these signatures can accurately predict clinical response in individuals treated with these drugs. Notably, signatures developed to predict response to individual agents, when combined, could also predict response to multidrug regimens. Finally, we integrated the chemotherapy response signatures with signatures of oncogenic pathway deregulation to identify new therapeutic strategies that make use of all available drugs. The development of gene expression profiles that can predict response to commonly used cytotoxic agents provides opportunities to better use these drugs, including using them in combination with existing targeted therapies. Numerous advances have been achieved in the development, selection and application of chemotherapeutic agents, sometimes with remark- able clinical successes—as in the case of treatment for lymphomas or platinum-based therapy for testicular cancers1. In addition, in several instances, combination chemotherapy in the postoperative (adjuvant) setting has been curative. However, most people with advanced solid tumors will relapse and die of their disease. Moreover, administration of ineffective chemotherapy increases the probability of side effects, particularly those from cytotoxic agents, and of a consequent decrease in quality of life1,2. Recent work has demonstrated the value in using biomarkers to select individuals for various targeted therapeutics, including tamox- ifen, trastuzumab and imatinib mesylate. In contrast, equivalent tools to select those most likely to respond to the commonly used chemotherapeutic drugs are lacking3. With the goal of developing genomic predictors of chemotherapy sensitivity that could direct the use of cytotoxic agents to those most likely to respond, we combined in vitro drug response data, together with microarray gene expression data, to develop models that could potentially predict responses to various cytotoxic chemotherapeutic drugs4. We now show that these signatures can predict clinical or pathologic response to the corresponding drugs, including combina- tions of drugs. We further use the ability to predict deregulated oncogenic signaling pathways in tumors to develop a strategy that identifies opportunities for combining chemotherapeutic drugs with targeted therapeutic drugs in a way that best matches the character- istics of the individual. RESULTS A gene expression–based predictor of sensitivity to docetaxel To develop predictors of cytotoxic chemotherapeutic drug response, we used an approach similar to previous work analyzing the NCI-60 panel4 from the US National Cancer Institute (NCI). We first identified cell lines that were most resistant or sensitive to docetaxel (Fig. 1a,b) and then genes whose expression correlated most highly with drug sensitivity, and used Bayesian binary regression analysis to develop a model that differentiates a pattern of docetaxel sensitivity from that of resistance. A gene expression signature consisting of 50 genes was identified that classified cell lines on the basis of docetaxel sensitivity (Fig. 1b, right). In addition to leave-one-out cross-validation, we used an indepen- dent dataset derived from docetaxel sensitivity assays in a series of 30 lung and ovarian cancer cell lines for further validation. The significant correlation (P o 0.01, log-rank test) between the predicted probability of sensitivity to docetaxel (in both lung and ovarian cell lines) (Fig. 1c, left) and the respective 50% inhibitory concentration (IC50) for docetaxel confirmed the capacity of the docetaxel predictor to predict sensitivity to the drug in cancer cell ©2008NaturePublishingGrouphttp://www.nature.com/naturemedicine Received 11 July; accepted 12 September; published online 22 October; corrected online 27 October 2006 (details online) and corrected after print 21 July 2008; doi:10.1038/nm1491 1Duke Institute for Genome Sciences and Policy, Duke University, Box 3382, Durham, North Carolina 27710, USA. 2Department of Medicine, Duke University Medical Center, Box 31295, Durham, North Carolina 27710, USA. 3Department of Molecular Genetics and Microbiology, Duke University Medical Center, Box 3054, Durham, North Carolina 27710, USA. 4Division of Gynecologic Surgical Oncology, H. Lee Moffitt Cancer Center & Research Institute, University of South Florida, 12902 Magnolia Drive, Tampa, Florida 33612, USA. 5Department of Surgery, Duke University Medical Center, Box 3627, Durham, North Carolina 27710, USA. 6Department of Obstetrics and Gynecology, Duke University Medical Center, Box 3079, Durham, North Carolina 27710, USA. Correspondence should be addressed to J.R.N. (nevin001@mc.duke.edu). 1294 VOLUME 12 [ NUMBER 11 [ NOVEMBER 2006 NATURE MEDICINE A R T I C L E S
  • 2. lines (Supplementary Fig. 1 online). For each set of cell lines, the accuracy exceeded 80%. Finally, we made use of a second independent dataset that measured docetaxel sensitivity in a series of 29 lung cancer cell lines (Gene Expression Omnibus (GEO) database (see URLs) accession number GSE4127). The docetaxel sensitivity model devel- oped from the NCI-60 panel again predicted sensitivity in this independent dataset, also with an accuracy exceeding 80% (P o 0.001, log-rank test; Fig. 1c, right). The expression signature predicts clinical docetaxel response We used published studies with clinical and genomic data that linked gene expression data with clinical response to docetaxel in a breast cancer neoadjuvant study5 (Fig. 1d) to test the capacity of the in vitro docetaxel sensitivity predictor to accurately identify those individuals that responded to docetaxel. Using a 0.45 predicted probability of response as the cutoff for predicting positive response, as determined by receiver operator characteristic curve analysis (Supplementary Fig. 1), the in vitro–generated profile correctly predicted docetaxel response in 22 of 24 clinical samples, for an overall accuracy of 91.6% (Fig. 1d). Applying a Mann-Whitney U-test for statistical significance demonstrated the capacity of the predictor to distinguish resistant from sensitive individuals (Fig. 1d, right). We extended this further by predicting the response to docetaxel as salvage therapy for individuals with ovarian cancer that was refractory to primary therapy (Fig. 1e), and the prediction achieved an accuracy exceeding 85% (Fig. 1e, middle). Further, an analysis of statistical significance demonstrated the capacity of the predictors to distinguish individuals with resistant versus sensitive disease (Fig. 1e, right). We also performed a complementary analysis, using the clinical response data to generate a signature and then predicting sensitivity of NCI-60 cell lines to docetaxel (Supplementary Fig. 1). Genes represented in the initial in vitro–generated docetaxel predictor and the alternative in vivo predictor showed considerable overlap. Notably, both predictors were linked to expected targets for docetaxel, including BCL2, WDR7 (also known as TRAG), ERBB2 and tubulin genes, all previously described to be involved in taxane chemoresis- tance6–9 (Supplementary Table 1 online). We also noted that the predictor of docetaxel sensitivity developed from the NCI-60 data, using the approach described here, was more accurate in predicting response in the ovarian samples than a predictor developed from the breast neoadjuvant study data (85.7% versus 64.3%; Supplementary Fig. 1). A panel of signatures that predict chemotherapeutic sensitivity Given the development of a docetaxel response predictor, we exam- ined the NCI-60 dataset for other opportunities to develop predictors ©2008NaturePublishingGrouphttp://www.nature.com/naturemedicine NCI-60 cell line panel Identify resistant and sensitive cells Build expression predictor of response Drug response data Affymetrix expression data Neoadjuvant breast cancer therapy Response to treatment Biopsy Affymetrix expression data Drug sensitivity prediction Ovarian salvage therapy Response to treatment Biopsy Affymetrix expression data Drug sensitivity prediction 100 75 50 25 0 Docetaxel LC50 Docetaxel GI50 Docetaxelconcentration Resistant Sensitive 5 10 15 20 25 30 35 40 45 2 4 6 8 10 12 14 50 P < 0.01 P < 0.001 6 5 4 3 2 1 0 DocetaxelIC50 (Lungandovariancelllines) 0.1 0.2 0.3 Probability of docetaxel sensitivity 0.4 0.5 0.6 0.7 0.8 0.1 0.2 0.3 Probability of docetaxel sensitivity 0.4 0.5 0.6 0.7 0.8 0.9 8 7 6 5 4 3 2 1 0 DocetaxelIC50 (Lungcancercelllines) 1.0 0.8 0.6 0.4 0.2 0.0 0 5 10 15 20 25 Sample number Probabilityofdocetaxel resistance Accuracy: 22/24 (91.6%) Docetaxel sensitive Docetaxel resistant Accuracy: 12/14 (85.7%) 1.00 0.75 0.50 0.25 0.00 0 3 6 9 Sample number 12 15 Probabilityofdocetaxel resistance 1.00 0.75 0.50 0.25 0.00 P < 0.001 Probabilityofdocetaxel resistance Docetaxel sensitive Docetaxel resistant Docetaxel sensitive Docetaxel resistant 1.00 0.75 0.50 0.25 0.00 P < 0.01 Probabilityofdocetaxel resistance a b c d e Docetaxel-resistant lines Docetaxel-sensitive lines Docetaxel sensitive Docetaxel resistant Figure 1 A gene expression signature that predicts sensitivity to docetaxel. (a) Strategy for generating the chemotherapeutic response predictor. (b) Left, cell lines from the NCI-60 panel used to develop the in vitro signature of docetaxel sensitivity. There is a statistically significant difference (Mann-Whitney U-test) in the IC50 and LC50 of the cell lines chosen to represent the sensitive and resistant subsets. Right, expression plots for genes selected for discriminating the docetaxel-resistant and docetaxel-sensitive NCI-60 cell lines, with blue representing the lowest expression and red the highest. Each column in the figure represents an individual sample. Each row represents an individual gene, ordered from top to bottom according to regression coefficient. (c) Left, validation of the docetaxel response prediction model in an independent set of lung and ovarian cancer cell line samples. A collection of lung and ovarian cell lines were used in a cell proliferation assay to determine the IC50 of docetaxel in the individual cell lines. Right, validation of the docetaxel response prediction model in another independent set of 29 lung cancer cell line samples. (d) Left, a strategy for assessment of the docetaxel response predictor as a function of clinical response in the breast neoadjuvant setting. Middle, predicted probability of docetaxel sensitivity in a collection of samples from a breast cancer single-agent neoadjuvant study. Right, a single-variable scatter plot of a significance test of the predicted probabilities of sensitivity to docetaxel in the sensitive and resistant tumors (P o 0.001, Mann-Whitney U-test). (e) Left, a strategy for assessment of the docetaxel response predictor as a function of clinical response in advanced ovarian cancer. Middle, predicted probability of docetaxel sensitivity in a collection of samples from a prospective single agent salvage therapy study. Right, a single-variable scatter plot showing statistical significance (P o 0.01, Mann-Whitney U-test). A R T I C L E S NATURE MEDICINE VOLUME 12 [ NUMBER 11 [ NOVEMBER 2006 1295
  • 3. of chemotherapeutic response. We developed a series of expression profiles from the NCI-60 dataset (Fig. 2a) that predict response to topotecan, adriamycin, etoposide, 5-fluorouracil (5-FU), paclitaxel and cyclophosphamide (Cytoxan). In each case, the leave-one-out cross-validation analyses demonstrated the capacity of these profiles to accurately predict the samples used in the development of the predictor (Supplementary Fig. 2 online). Each profile was then further validated using in vitro response data from independent datasets; in each case, the profile developed from the NCI-60 data was capable of accurately (485%) predicting response in the separate dataset of approximately 30 cancer cell lines for which the dose- response information and relevant Affymetrix U133A gene expression data were publicly available10 (Supplementary Fig. 2 and Supple- mentary Table 2 online). Once again, applying a Mann-Whitney U-test for statistical significance demonstrated the capacity of the predictor to distinguish resistant from sensitive individuals (Fig. 2b). We then evaluated the extent to which a signature was also specific for an individual chemotherapeutic agent. Using the validations of chemosensitivity seen in the independent European cell line data10, it was clear that each signature is specific for the drug that was used to develop the predictor (Supplementary Fig. 3 online). Given the ability of the in vitro–developed gene expression profiles to predict response to docetaxel in the clinical samples, we extended this approach to test the ability of additional signatures to predict responses to commonly used salvage therapies for ovarian cancer and in another, independent dataset of samples cultured from adriamycin-treated individuals (GEO accession nos. GSE2351, GSE649, GSE650 and GSE651). Each of these predictors was capable of accurately predicting the response to the drugs in clinical samples, achieving an accuracy in excess of 74% overall (Fig. 2c). In each case, the positive and negative predictive values confirmed the validity of the approach (Supplemen- tary Table 2). Signatures predict response to multidrug regimens Many therapeutic regimens make use of combinations of chemother- apeutic drugs. To evaluate whether individual signatures of response could predict response to regimens containing combinations of drugs, we analyzed data from a breast neoadjuvant treatment study that used a combination of paclitaxel, 5-FU, adriamycin and cyclophosphamide (TFAC)11,12 (Fig. 3a). The available data from the 51 participants were used to predict response using each of the single-agent signatures (for paclitaxel, 5-FU, adriamycin and cyclophosphamide) developed from the NCI-60 cell line analysis. The predicted response based on each of the individual chemosensitivity signatures indicated a significant distinction between the responders (n ¼ 13) and nonresponders (n ¼ 38), with the exception of the prediction for 5-FU (Fig. 3a, middle). The sensitivity to the four agents in the TFAC preoperative (neoadjuvant) regimen was predicted as a combined probability. The prediction of response based on a combined probability of sensitivity built from the individual chemosensitivity predictions yielded a statistically significant (P o 0.0001, Mann-Whitney U-test) distinc- tion between the responders and nonresponders (Fig. 3a, right). As a further validation of the capacity to predict response to combination therapy, we analyzed gene expression data generated from a collection of breast cancer (n ¼ 45) samples from subjects who received 5-FU, adriamycin and cyclophosphamide (FAC) as adjuvant chemotherapy (GEO database accession no. GSE3143). The predicted response based on the individual signatures indicated a significant distinction between the responders (n ¼ 34) and nonresponders (n ¼ 11; Fig. 3b, left). Furthermore, the combined probability of sensitivity to the three agents in the FAC regimen yielded a clear and significant (P o 0.001, Mann-Whitney U-test) distinction between the respon- ders and nonresponders (accuracy 82.2%, positive predictive value 90.3%, negative predictive value 64.3%; Fig. 3b). Although it is difficult to interpret the prediction of clinical response in the adjuvant ©2008NaturePublishingGrouphttp://www.nature.com/naturemedicine 403020100 Sample numberSample number 0.00 0.25 Probabilityofpaclitaxel sensitivity 0.50 0.75 1.00 0.00 0.25 Probabilityoftopotecan sensitivity 0.50 0.75 1.00 0 10 20 30 40 50 Accuracy: 40/48 (83.3%) Topotecan resistant Topotecan sensitive Accuracy: 28/35 (80%) Paclitaxel resistant Paclitaxel sensitive P < 0.001 P < 0.001 P < 0.001P < 0.0001 P < 0.01 P = 0.001 ResistantSensitiveResistantSensitiveResistantSensitiveResistantSensitiveResistantSensitiveResistantSensitive 1.00 Predictedprobabilityof sensitivitytotopotecan Predictedprobabilityof sensitivitytoadriamycin Predictedprobabilityof sensitivitytoetoposide Predictedprobabilityof sensitivityto5-FU Predictedprobabilityof sensitivitytopaclitaxel Predictedprobabilityof sensitivityto cyclophosphamide 0.75 0.50 0.25 0.00 1.00 0.75 0.50 0.25 0.00 1.00 0.75 0.50 0.25 0.00 0.0 0.1 0.2 0.3 0.4 0.5 0.6 0.7 0.8 0.9 CyclophosphamidePaclitaxel5-FUEtoposideAdriamycinTopotecan 20 40 60 80 100 120 140 10 20 30 40 50 60 70 80 5 10 15 20 25 30 35 40 45 50 5 10 15 20 25 30 35 40 45 5 10 15 20 25 30 35 0.0 0.1 0.2 0.3 0.4 0.5 0.6 0.7 0.8 0.91.00 0.75 0.50 0.25 0.00 a b c 1.00 Probabilityofadriamycin sensitivity 0.75 0.50 0.25 0.00 0 20 40 Sample number 60 80 100 Adriamycin resistant Adriamycin sensitive Accuracy: 70/95 (73.7%) Figure 2 Development of a panel of gene expression signatures that predict sensitivity to chemotherapeutic drugs. (a) Gene expression patterns selected for predicting response to the indicated drugs. (b) Independent validation of the chemotherapy response predictors in an independent set of cancer cell lines that have dose-response and Affymetrix expression data10. A single-variable scatter plot of a significance test of the predicted probabilities of sensitivity to any given drug in the sensitive and resistant cell lines (P values, Mann-Whitney U-test). (c) Prediction of single-agent therapy response in clinical samples using in vitro cell line–based expression signatures of chemosensitivity. Left, the predicted probability of sensitivity to topotecan when compared to actual clinical response data (n ¼ 48). Middle, the accuracy of the adriamycin predictor in a cohort of 122 samples. Right, predictive accuracy of the cell line– based paclitaxel predictor when the drug was used as a salvage chemotherapy in advanced ovarian cancer (n ¼ 35). A R T I C L E S 1296 VOLUME 12 [ NUMBER 11 [ NOVEMBER 2006 NATURE MEDICINE
  • 4. setting, as many of these people were probably free of disease after surgery, the accurate identification of nonresponders is a clear end- point that does confirm the capacity of the signatures to predict clinical response. We also examined the prognostic significance of the prediction of response to FAC: it defined a poor prognosis group (Fig. 3b, right). Taking these results together, we conclude that the signatures of chemosensitivity generated from the NCI-60 panel do indeed have the capacity to predict therapeutic response in individuals receiving either single agent or combination chemotherapy (Supple- mentary Tables 2 and 3 online). An examination of the genes that constituted the paclitaxel pre- dictor identified microtubule-associated protein tau (MAPT), described previously as a determinant of paclitaxel sensitivity12. Also consistent with previous reports5–7, TP53, methylenetetrahydrofolate reductase (MTHFR) and DNA repair genes constituted the 5-FU predictor, and excision repair mechanism genes (for example, ERCC4), retinoblastoma pathway genes and BCL2 constituted the adriamycin predictor (Supplementary Table 1). Patterns of predicted chemotherapy response across tumor types The panel of chemotherapy response predictors described in Figure 3 was used to profile the potential options for use of these drugs, by predicting the likelihood of sensitivity to the seven agents in a large collection of breast, lung and ovarian tumor samples. We then clustered the samples according to patterns of predicted sensitivity to the various chemotherapeutics and plotted a heatmap. There were clearly evident patterns of predicted sensitiv- ity to the various agents (Fig. 4). In many cases, the predicted sensitivities to the chemo- therapeutic agents were consistent with the previously documented efficacy of single- agent chemotherapies in the individual tumor types13. For instance, the predicted response rates for etoposide, adriamycin, cyclophosphamide and 5-FU approximated the observed response for these single agents in individuals with breast cancer (Supple- mentary Fig. 3). Likewise, the predicted sen- sitivities to etoposide, docetaxel and paclitaxel approximated the observed response for these single agents in individuals with lung cancer (Supplementary Fig. 3). This analysis also suggested possibilities for alternate treat- ments. As an example, it would seem that individuals with breast cancer who are likely to respond to 5-FU are resistant to adriamy- cin and docetaxel (Supplementary Fig. 4 online). Likewise, in lung cancer, docetaxel- sensitive individuals are likely to be resistant to etoposide (Supplementary Fig. 4). This is a potentially useful observation, considering that both etoposide and docetaxel are viable front-line options (in conjunction with cis- platin or carboplatin) for people with lung cancer1. A similar relationship is seen between topotecan and adriamycin, both of which are agents used in salvage chemotherapy for ovarian cancer (Supplementary Fig. 4). Thus, by identifying people or cohorts resis- tant to a certain standard-of-care agent (for example, topotecan), one could, by choosing an alternative standard of care agent (for example, adriamycin), avoid the side effects of the former agent without compromising outcome. Linking chemotherapy sensitivity to oncogenic pathway status Most people who are resistant to chemotherapeutic agents are recruited into a second- or third-line therapy or enrolled in a clinical trial14,15. Moreover, even those who initially respond to a given agent are likely to eventually suffer a relapse, and thus, in either case, additional therapeutic options are needed. As one approach to identifying such options, we have taken advantage of our recent work that describes the development of gene expression signatures that reflect the activation of several oncogenic pathways16 (Supple- mentary Fig. 5 and Supplementary Table 1 online). To illustrate this approach, we first stratified the NCI cell lines based on predicted docetaxel response and then examined the patterns of pathway deregulation associated with docetaxel sensitivity or resistance (Sup- plementary Fig. 5). Regression analysis showed a significant relation- ship between phosphatidylinositol 3-OH (PI3)-kinase pathway deregulation and docetaxel resistance (Supplementary Fig. 5). These results indicated an opportunity to use a PI3-kinase inhibitor in this subgroup, given our recent observations that have demon- strated a linear positive correlation between the probability of pathway deregulation and sensitivity to targeted drugs16. To address this directly, we predicted docetaxel sensitivity and probability of oncogenic pathway deregulation using DNA microarray data from 17 non–small cell lung cancer cell lines (Fig. 5a, left). Consistent with ©2008NaturePublishingGrouphttp://www.nature.com/naturemedicine Neoadjuvant breast cancer therapy Response to treatment Biopsy Affymetrix expression data Treatment with TFAC Responders RespondersNonresponders Responders Nonresponders Nonresponders 1.00 0.75 0.50 0.25 0.00 1.00 0.75 0.50 0.25 0.00 T + 5-FU + A + C T + 5-FU + A + C P < 0.0001 T F A AC CT 5-FU 5-FU (F) Adriamycin (A) Cyclophosphamide (C) Probabilityofsensitivity 1.00 0.75 0.50 0.25 0.00 A AC C5-FU5-FU Probabilityofsensitivity Combinedprobabilityof TFACsensitivity P = 0.002 P = 0.3 P = 0.024 P = 0.003 Paclitaxel (T) 5-FU (F) Adriamycin (A) Cyclophosphamide (C) Accuracy: 42/51 (82.3%) NPV: 31/33 (94%) PPV: 11/18 (61.1%) Responders Nonresponders 1.00 0.75 0.50 0.25 0.00 5-FU + A + C 5-FU + A + C P < 0.0001 Combinedprobabilityof FACsensitivity Accuracy: 37/45 (82.2%) NPV: 9/14 (64.3%) PPV: 28/31 (90.3%) P = 0.003 P = 0.008 P = 0.04 0 0 25 50 Months P < 0.001 75 100 125 150 25 Diseasefreesurvival(%) 50 75 100 a b Figure 3 Prediction of response to combination therapy. (a) Left, strategy for assessment of chemotherapy response predictors in combination therapy as a function of pathologic response. Middle, prediction of clinical response to neoadjuvant chemotherapy involving paclitaxel, 5-FU, adriamycin and cyclophosphamide (TFAC) using the single-agent in vitro chemosensitivity signatures developed for each of these drugs. Right, prediction of response (38 nonresponders, 13 responders) using a combined probability predictor assessing the probabilities of all four chemosensitivity signatures in 51 people treated with TFAC chemotherapy (P o 0.0001, Mann-Whitney U-test). Response was defined as a complete pathologic response after completion of TFAC neoadjuvant therapy. (b) Left, prediction of clinical response (n ¼ 45) to adjuvant chemotherapy involving 5-FU, adriamycin and cyclophosphamide (FAC) using the single-agent in vitro chemosensitivity predictors developed for these drugs. Middle, prediction of response (34 responders, 11 nonresponders) using a combined probability predictor assessing the probabilities of all four chemosensitivity signatures in 45 people treated with FAC chemotherapy. Right, Kaplan-Meier survival analysis for individuals predicted to be sensitive (blue curve) or resistant (red curve) to FAC adjuvant chemotherapy. PPV, positive predictive value; NPV, negative predictive value. A R T I C L E S NATURE MEDICINE VOLUME 12 [ NUMBER 11 [ NOVEMBER 2006 1297
  • 5. the analysis of the NCI-60 cell line panel, the cell lines predicted to be resistant to docetaxel were also predicted to show PI3-kinase pathway activation (P ¼ 0.03, log-rank test; Supplementary Fig. 5). In parallel, the lung cancer cell lines were assayed for sensitivity to a PI3-kinase– specific inhibitor (LY-294002), using a standard measure of cell proliferation14–16. The cell lines showing an increased probability of PI3-kinase pathway activation were also more likely to respond to the PI3-kinase inhibitor (P ¼ 0.001, log-rank test; Fig. 5b, left). The same relationship held for prediction of resistance to docetaxel: these cells were more likely to be sensitive to PI3-kinase inhibition (P o 0.001, log-rank test; Fig. 5b, left). An analysis of a panel of ovarian cancer cell lines provided a second example of an opportunity to link predictions of chemo- therapy sensitivity to oncogenic pathway deregulation. Ovarian cell lines that were predicted to be topotecan resistant (Fig. 5a, right) had a higher likelihood of Src pathway deregulation, and there was a significant linear relationship (P ¼ 0.001, log-rank test) between the probability of topotecan resistance and sensitivity to a drug (SU6656) that inhibits the Src pathway (Fig. 5b, right). The results of these assays clearly demonstrated an opportunity to mitigate drug resistance (for example, resistance to docetaxel or topotecan) using a specific pathway–targeted agent (PI3-kinase or Src inhibitor, respectively). Taken together, these data demonstrate a rational approach to the identification of therapeutic options for chemotherapy-resistant indivi- duals, as well as to the identification of new combinations for chemo- therapy-sensitive individuals, and thus have the potential to change the current paradigm of cancer care. Prospective validation studies are, however, needed to confirm the effectiveness of this approach. DISCUSSION The practice of oncology continually faces the challenge of matching the right therapeutic regimen with the right individual, balancing relative benefit with risk to achieve the most favorable outcome. This challenge is often daunting, with marginal success rates in many advanced disease contexts—probably reflecting the enormous com- plexity of the disease process coupled with an inability to properly guide the use of available therapeutics1,2,15. The results we present here, focusing on the development of signatures that predict sensitivity to common cytotoxic chemotherapeutic drugs, address this limitation and have the potential to identify drugs—and combinations of drugs—that best match the individual. The experimental strategy for analyses used in this study is similar to that used for the development of oncogenic pathway signatures16: samples representing extreme cases are used to train the expression data to develop a signature that can predict drug sensitivity. Of note, we further demonstrated that these signatures can predict clinical drug response. The importance of selecting individuals likely to respond to a given therapeutic agent is perhaps best illustrated by the example of trastuzumab. In the absence of selection, the overall response rate in people with breast cancer is approximately 10%. In contrast, for those selected on the basis of Her2 amplification, the overall response rate rises to 35–50%17. We suggest that the gene expression signatures predicting response to various cytotoxic chemotherapeutic agents may provide an opportunity to optimize the use of these drugs. Previous work has described the use of gene expression data, coupled with in vitro drug sensitivity assays, to develop signatures that could be used to classify response to therapy18,19. This involved ©2008NaturePublishingGrouphttp://www.nature.com/naturemedicine Breast tumors (n = 171) Lung tumors (n = 91) Ovarian tumors (n = 119) Paclitaxel Topotecan 5-FU Docetaxel Cyclophosphamide Etoposide Adriamycin Etoposide Paclitaxel 5-FU Adriamycin Topotecan Cyclophosphamide Docetaxel Etoposide Paclitaxel 5-FU Adriamycin Topotecan Cyclophosphamide Docetaxel Figure 4 Patterns of predicted sensitivity to common chemotherapeutic drugs in human cancers. Hierarchical clustering of a collection of breast (n ¼ 171), lung (n ¼ 91) and ovarian cancer (n ¼ 119) samples according to patterns of predicted sensitivity to the various chemotherapeutics. Predictions were plotted as a heatmap in which high probability of sensitivity, or response, is indicated by red and low probability, or resistance, is indicated by blue. A R T I C L E S 1298 VOLUME 12 [ NUMBER 11 [ NOVEMBER 2006 NATURE MEDICINE
  • 6. the development of signatures that could identify chemoresistance to the combination of drugs used in the treatment of acute lymphoblastic leukemia. Our findings that individual signatures can accurately predict response to drugs in both neoadjuvant and adjuvant che- motherapy settings, where combinations of cytotoxic drugs are used, clearly demonstrate the utility of the approach. We also describe a rational approach to identifying new drug regimens, making use of previously described signatures of oncogenic pathway deregulation16, to provide a potential strategy for the development of therapeutic regimens that would be based on knowledge of the status of an individual’s tumor. Finally, we note that the availability of predictors of chemotherapy response, which have been shown to predict clinical response, provides an opportunity to apply these predictors in present day practice. There are several instances in which people are treated with one or more therapeutic regimens that each have equal efficacy. An ability to select the most effective therapy from among a panel of standard-of-care agents is a strategy that can be easily applied20. Indeed, we believe this represents an opportunity for a clinical trial that would evaluate the performance of ‘random’ selection of agents (current practice) versus a genomic signature–based selection as an initial step in the process of achieving a more individualized treatment strategy (Fig. 6). Notably, such a study may also identify those individuals likely to be resistant to the available standard-of-care drugs, opening the way to a second- generation trial that could evaluate the most effective treatment for these people. Ultimately, we suggest that future treatment strategies might be based on an analysis of an individual’s tumor, which would then allow the development of a profile of likely sensitivity to common chemotherapeutic drugs as well as to targeted therapies. Based on this information, each individual might then be assigned to a combination regimen that best matches the profile from the tumor. METHODS NCI-60 data. The complete details of the methods involved in the development of the individual chemosensitivity predictors are available in Supplementary Methods online. Briefly, the –log10 values of the 50% growth inhibition doses (IC50; also known as GI50), total growth inhibition doses and 50% cytotoxic doses (LC50) data were used to populate a matrix in MATLAB software, along with the relevant expression data for the individual cell lines. Where multiple entries for a drug screen existed (by NCS number), the entry with the largest number of replicates was included. Incomplete data were assigned as ‘not a number’ for statistical purposes. To develop an in vitro gene expression–based predictor of sensitivity and resistance from the pharmacologic data used in the NCI-60 drug screen studies, we chose cell lines within the NCI-60 panel that would represent the extremes of sensitivity to a given chemotherapeutic agent (mean IC50 ± 1 s.d.). Relevant expression data (revised data gathered using the Affymetrix U95A2 GeneChip) for the solid tumor cell lines and the respective pharmacological data for the chemotherapeutics were downloaded from the NCI website. The individual drug sensitivity and resistance data from the selected solid tumor NCI-60 cell lines were then used in a supervised analysis using binary regression methodologies, as described previously21, to develop models predictive of chemotherapeutic response. Human ovarian cancer samples. We measured expression of 22,283 genes in 13 ovarian cancer cell lines and 119 advanced (Federation Internationale de Gynecologie et d’Obstetrique (FIGO) stage III or IV) serous epithelial ovarian carcinomas using Affymetrix U133A GeneChips. All ovarian cancers were obtained at initial cytoreductive surgery. All tissues were collected under the ©2008NaturePublishingGrouphttp://www.nature.com/naturemedicine Clinical presentation of cancer Biopsy Chemotherapy response prediction Optimal single chemotherapy Combination chemotherapy Oncogenic pathway prediction Targeted agent Figure 6 Scheme for using chemotherapeutic and oncogenic pathway predictors to identify individualized therapeutic options. 520 1666 125 549 1793 1651 23 2126 358 838 322 1838 441 1734 1650 647 2030 IGROV OV2008 TOV-21D A2780-CP T8 TOV112D FU-OV-1 OVCAR-5 C13 IMCC3 A2780-S A2008 OV-90 Docetaxel Src Ras β-Catenin E2F3 Myc Pl3K Pl3K Topotecan Src Ras β-Catenin E2F3 Myc Lung cancer cell lines Ovarian cancer cell lines 0.9 200 150 100 50 0.9 0.8 0.8 0.7 0.7 0.6 0.6 0.5 0.5 0.4 0.4 0.3 0.3 0.2 0.2 0.1 0.1 0.0 0.9 0.8 0.7 0.6 0.5 0.4 0.3 0.2 0.1 0.0 20 20 0 40 40 60 60 Src inhibition (µM) 100 100 80 80 0.00 0 25 50 75 100 125 150 175 200 PI3K inhibition (µM) Probability of docetaxel resistance 0.9 1.00.80.70.60.50.40.30.20.10.0 Probability of topotecan resistance PI3KinhibitionIC50 (µM)SrcinhibitionIC50(µM) ProbabilityofP13K pathwayactivation ProbabilityofSrc pathwayactivation Lung cancer cell lines Ovarian cancer cell lines a b P < 0.001 P < 0.001 P < 0.007 P = 0.001 Figure 5 Relationship between predicted chemotherapeutic sensitivity and oncogenic pathway deregulation. (a) Left, probability of oncogenic pathway deregulation as a function of predicted docetaxel sensitivity in a series of lung cancer cell lines. Right, probability of oncogenic pathway deregulation as a function of predicted topotecan sensitivity in a series of ovarian cancer cell lines (red, sensitive; blue, resistant). (b) Top, lung cancer cell lines showing an increased probability of PI3-kinase (PI3K) activation were also more likely to respond to a PI3-kinase inhibitor (P ¼ 0.001, log-rank test), as measured by sensitivity to the drug in assays of cell proliferation. Furthermore, those cell lines predicted to be resistant to docetaxel were more likely to be sensitive to PI3-kinase inhibition (P o 0.001, log-rank test). Bottom, the relationship between Src pathway deregulation and topotecan resistance in a set of 13 ovarian cancer cell lines. Ovarian cell lines that are predicted to be topotecan resistant (a, right) have a higher likelihood of Src pathway deregulation and there is a significant linear relationship (P ¼ 0.001, log-rank test) between the probability of topotecan resistance and sensitivity to a drug that inhibits the Src pathway. A R T I C L E S NATURE MEDICINE VOLUME 12 [ NUMBER 11 [ NOVEMBER 2006 1299
  • 7. auspices of respective institutional (Duke University Medical Center and H. Lee Moffitt Cancer Center) review board–approved protocols involving written informed consent. Full details of the methods used for RNA extraction and development of gene expression signatures representing deregulation of oncogenic pathways in the tumor samples were recently described16 and are available in Supplemen- tary Methods. Response to therapy was evaluated using standard criteria for those with measurable disease, based upon World Health Organization guide- lines22. Complete details are available in Supplementary Methods. Validation of in vitro chemotherapy response signatures. Complete details of the publicly available datasets used for independent validation of the genomic signatures of chemosensitivity are described in Supplementary Methods. PI3-kinase signature. A signature representative of PI3-kinase activation was developed as previously described16. The genes that constitute the PI3-kinase signature are shown in Supplementary Table 1. Lung and ovarian cancer cell culture. Total RNA was extracted and oncogenic pathway predictions were performed similarly to the methods described previously16. Complete details are available in Supplementary Methods. Cross-platform Affymetrix Gene Chip comparison. To map the probe sets across various generations of Affymetrix GeneChip arrays, we used an in-house program, Chip Comparer (see URLs), as described previously16. Cell proliferation assays. Methods for drug sensitivity assays are described in Supplementary Methods. Statistical analysis methods. Analysis of expression data was performed as previously described16,21,23,24 and is detailed in Supplementary Methods. In instances where a combined probability of sensitivity to a combination chemotherapeutic regimen was required based on the individual drug sensitivity patterns, we used the probabilities of response to individual drugs and used the theorem for combined probabilities as described by William Feller to deduce a probability of response to a combination of the drugs being studied. The result was then mean-centered to give a probability between 0 and 1. Hierarchical clustering of tumor predictions was performed using Gene Cluster 3.0 (ref. 25). Genes and tumors were clustered using average linkage with the uncentered correlation similarity metric. Standard linear regression analyses and their significance (log-rank test) were generated for the drug response data and for correlation between drug response and probability of chemosensitivity or pathway deregulation using GraphPad software. URLs. NCI, http://dtp.nci.nih.gov/docs/cancer/cancer_data.html. GEO data- base, http://www.ncbi.nlm.nih.gov/geo/. Chip Comparer, http://tenero.duhs. duke.edu/genearray/perl/chip/chipcomparer.pl. Note: Supplementary information is available on the Nature Medicine website. ACKNOWLEDGMENTS The authors would like to acknowledge and thank L. Pusztai for providing us with the clinical and expression data on the samples studied from the M.D. Anderson Cancer Center. Funding for this work was supplied in part by research grants from the US National Institutes of Health (NCI-U54 CA112952- 02 (J.R.N.) and R01CA106520 (J.R.N.)). A.P. is an American Association for Cancer Research Translational Research Grant awardee, J.L. receives research support from Glaxo-Smith-Kline and Sanofi-Aventis Pharmaceuticals and Orthobiotech, and P.F. is a Damon Runyon Cancer Research Investigator. Additional sources of funding included the Ovarian Cancer Research Fund and the Hearing the Ovarian Cancer Whisper Jacquie Liggett Fellowship. COMPETING INTERESTS STATEMENT The authors declare competing financial interests (see the Nature Medicine website for details). Published online at http://www.nature.com/naturemedicine Reprints and permissions information is available online at http://npg.nature.com/ reprintsandpermissions/ 1. Herbst, R.S. et al. Clinical cancer advances 2005: major research advances in cancer treatment, prevention, and screening—a report from the American Society of Clinical Oncology. J. Clin. Oncol. 24, 190–205 (2006). 2. Breathnach, O.S. et al. Twenty-two years of phase III trials for patients with advanced non-small-cell lung cancer: sobering results. J. Clin. Oncol. 19, 1734–1742 (2001). 3. Blagosklonny, M.V. Why therapeutic response may not prolong the life of a cancer patient: selection for oncogenic resistance. Cell Cycle 4, 1693–1698 (2005). 4. Staunton, J.E. et al. Chemosensitivity prediction by transcriptional profiling. Proc. Natl. Acad. Sci. USA 98, 10787–10792 (2001). 5. Chang, J.C. et al. Gene expression profiling for the prediction of therapeutic response to docetaxel in patients with breast cancer. Lancet 362, 362–369 (2003). 6. Emi, M., Kim, R., Tanabe, K., Uchida, Y. & Toge, T. Targeted therapy against Bcl-2-related proteins in breast cancer cells. Breast Cancer Res. 7, R940–R952 (2005). 7. Takahashi, T. et al. Cyclin A-associated kinase activity is needed for paclitaxel sensitivity. Mol. Cancer Ther. 4, 1039–1046 (2005). 8. Modi, S. et al. Phosphorylated/activated HER2 as a marker of clinical resistance to single agent taxane chemotherapy for metastatic breast cancer. Cancer Invest. 23, 483–487 (2005). 9. Langer, R. et al. Association of pretherapeutic expression of chemotherapy-related genes with response to neoadjuvant chemotherapy in Barrett carcinoma. Clin. Cancer Res. 11, 7462–7469 (2005). 10. Gyorffy, B. et al. Gene expression profiling of 30 cancer cell lines predicts resistance towards 11 anticancer drugs at clinically achieved concentrations. Int. J. Cancer 118, 1699–1712 (2005). 11. Rouzier, R. et al. Breast cancer molecular subtypes respond differently to preoperative chemotherapy. Clin. Cancer Res. 11, 5678–5685 (2005). 12. Rouzier, R. et al. Microtubule-associated protein tau: a marker of paclitaxel sensitivity on breast cancer. Proc. Natl. Acad. Sci. USA 102, 8315–8320 (2005). 13. DeVita, V.T., Hellman, S. & Rosenberg, S.A. Cancer: Principles and Practice of Oncology (Lippincott-Raven, Philadelphia, 2005). 14. Broxterman, H.J. & Georgopapadakou, N.H. Anticancer therapeutics: ‘‘addictive’’ targets, multi-targeted drugs, new drug combinations. Drug Resist. Updat. 8, 183–197 (2005). 15. Minna, J.D., Gazdar, A.F., Sprang, S.R. & Herz, J. Cancer: a bull’s eye for targeted lung cancer therapy. Science 304, 1458–1461 (2004). 16. Bild, A. et al. Oncogenic pathway signatures in human cancers as a guide to targeted therapies. Nature 439, 353–357 (2006). 17. Vogel, C.L. et al. Efficacy and safety of trastuzumab as a single agent in first-line treatment of HER2-overexpressing metastatic breast cancer. J. Clin. Oncol. 20, 719–726 (2002). 18. Lugthart, S. et al. Identification of genes associated with chemotherapy crossresis- tance and treatment response in childhood acute lymphoblastic leukemia. Cancer Cell 7, 375–386 (2005). 19. Holleman, A. et al. Gene expression patterns in drug resistant acute lympho- blastic leukemia cells and response to treatment. N. Engl. J. Med. 351, 533–542 (2004). 20. Sargent, D.J., Conley, B.A., Allegra, C. & Collette, L. Clinical trial designs for predictive marker validation in cancer treatment trials. J. Clin. Oncol. 23, 2020–2027 (2005). 21. Pittman, J., Huang, E., Wang, Q., Nevins, J.R. & West, M. Bayesian analysis of binary prediction tree models for retrospectively sampled outcomes. Biostatistics 5, 587–601 (2004). 22. Miller, A.B., Hoogstraten, B., Staquet, M. & Winkler, A. Reporting results of cancer treatment. Cancer 47, 207–214 (1981). 23. West, M. et al. Predicting the clinical status of human breast cancer by using gene expression profiles. Proc. Natl. Acad. Sci. USA 98, 11462–11467 (2001). 24. Ihaka, R. & Gentleman, R. A language for data analysis and graphics. J. Comput. Graph. Stat. 5, 299–314 (1996). 25. Eisen, M.B., Spellman, P.T., Brown, P.O. & Botstein, D. Cluster analysis and display of genome-wide expression patterns. Proc. Natl. Acad. Sci. USA 95, 14863–14868 (1998). ©2008NaturePublishingGrouphttp://www.nature.com/naturemedicine A R T I C L E S 1300 VOLUME 12 [ NUMBER 11 [ NOVEMBER 2006 NATURE MEDICINE
  • 8. Corrigendum: Genomic signatures to guide the use of chemotherapeutics Anil Potti, Holly K Dressman,Andrea Bild, Richard F Riedel, Gina Chan, Robyn Sayer, Janiel Cragun, Hope Cottrill, Michael J Kelley, Rebecca Petersen, David Harpole, Jeffrey Marks,Andrew Berchuck, Geoffrey S Ginsburg, Phillip Febbo, Johnathan Lancaster & Joseph R Nevins Nat. Med. 12, 1294–1300 (2006); published online 22 October 2006; corrected after print 10 May and 10 October 2007 and 18 July 2008 In the version of this article initially published,of the 122 samples assayed for sensitivity to daunorubicin for which the authors applied a predictor of adriamycin sensitivity, 27 samples were replicated owing to the fact that the same samples were included in several separate series files in the Gene Expression Omnibus generated in 2004 and 2005,which were the source of the data provided for the study.The authors have reanalyzed the prediction of adriamycin sensitivity using only the 95 unique samples and find a revised accuracy of 74%. Additionally, the authors have added two more accession numbers (GSE2351 and GSE649) to more clearly identify the sources of the samples. The errors have been corrected in the HTML and PDF versions of the article. nature medicine volume 14 | number 8 | august 2008 889 0 20 40 60 80 100 Sample number 1.00 0.75 0.50 0.25 0.00 Probabilityofadriamycin sensitivity Adriamycin resistant Adriamycin sensitive Accuracy: 70/95 (73.7%) co r r i g e n da©2008NaturePublishingGrouphttp://www.nature.com/naturemedicine