ASCO_2012_files/Berlin Translational GI Cancers ASCO

Download Report

Transcript ASCO_2012_files/Berlin Translational GI Cancers ASCO

Jointly sponsored by Postgraduate Institute for
Medicine and Clinical Care Options, LLC
In partnership with Translational
Research in Oncology
Translational Research 2012:
Using Leading-Edge Science to Make
the Correct Treatment Choice Today
and Tomorrow
Sunday, June 3, 2012
6:30 PM – 8:30 PM
Hyatt Regency Chicago
Chicago, Illinois
This program is supported by educational grants from
Image:
Image:
Image:
Image:
suravid/Copyright©2012 Shutterstock Images LLC. All Rights Reserved
Gunilla Elam/Copyright©2012 Photo Researchers, Inc. All Rights Reserved
Carol and Mike Werner/Copyright©2012 Photo Researchers, Inc. All Rights Reserved
Science Photo Library/Copyright©2012 Custom Medical Stock Photo, Inc. All Rights Reserved
Current and Future Directions in the
Treatment of GI Cancers: Optimizing
Treatment by Genetic Features of Disease
Jordan D. Berlin, MD
Associate Professor of Medicine
Vanderbilt University
Clinical Director, GI Oncology
The Vanderbilt-Ingram Cancer Center
Nashville, Tennessee
Image:
Image:
Image:
Image:
suravid/Copyright©2012 Shutterstock Images LLC. All Rights Reserved
Gunilla Elam/Copyright©2012 Photo Researchers, Inc. All Rights Reserved
Carol and Mike Werner/Copyright©2012 Photo Researchers, Inc. All Rights Reserved
Science Photo Library/Copyright©2012 Custom Medical Stock Photo, Inc. All Rights Reserved
Translational Research 2012
clinicaloptions.com/oncology
Outline
 EGFR-targeted antibodies in CRC
– Rationale for and data supporting KRAS mutation testing
 Biomarkers for future treatment algorithms
– Impact of BRAF mutations on EGFR-targeted therapy
– Other potential biomarkers
– CTC levels
 Role of HER2 in gastric cancer
– Rationale for and data supporting use of HER2-targeted therapies
 Biomarkers for future treatment algorithms
– Other potential biomarkers
EGFR-Targeted Antibodies
in Colorectal Cancer
Translational Research 2012
clinicaloptions.com/oncology
EGFR-Targeted Antibodies in CRC
 EGFR-targeted antibodies approved in CRC
– Cetuximab
– Panitumumab
 EGFR expression status insufficient for discerning which patients
derive most benefit
 KRAS: signaling molecule downstream of EGFR
– Mutated KRAS becomes constitutively active, negating effect of
EGFR inhibition[1]
– ~40% of CRCs have mutated KRAS genotype[1]
– Mutated KRAS associated with poor response to EGFR-targeted
agents[2]
1. Lièvre A, et al. Oncogene. 2010;29:3033-3043. 2. Dahabreh IJ, et al. Ann Intern Med. 2011;154:37-49.
Translational Research 2012
clinicaloptions.com/oncology
KRAS Testing: What Are the
Recommendations?
 NCCN guidelines[1]
– Strongly recommend KRAS testing in all patients with mCRC at
the time of diagnosis of metastatic disease
– Testing should be performed in a CLIA-certified lab
– Testing can be performed on either primary or metastatic tissue
 ASCO Provisional Clinical Opinion[2]
– All patients with mCRC who are candidates for anti-EGFR
antibody therapy should have their tumor tested for KRAS
mutations in a CLIA-accredited laboratory
 In both cases, anti-EGFR agents (cetuximab and panitumumab)
are recommended for wild-type KRAS patients only[1,2]
1. NCCN. Clinical practice guidelines in oncology: colon cancer. 2012.
2. Allegra CJ, et al. J Clin Oncol. 2009;27:2091-2096.
Translational Research 2012
clinicaloptions.com/oncology
Studies Evaluating Addition of EGFRTargeted Antibody to Chemotherapy
 Benefit in KRAS wild-type CRC patients only
– CRYSTAL: cetuximab plus FOLFIRI[1]
– OPUS: cetuximab plus FOLFOX-4[2]
– PRIME: panitumumab plus FOLFOX-4[3]
 No benefit in either KRAS wild-type or mutated CRC
patients
– COIN: cetuximab plus oxaliplatin/fluoropyrimidine[4]
1. Van Cutsem E, et al. J Clin Oncol. 2011;29:2011-2019. 2. Bokemeyer C, et al. Ann Oncol. 2011;22:15351546. 3. Douillard JY, et al. J Clin Oncol. 2010;28:4697-4705. 4. Maughan TS, et al. Lancet.
2011;377:2103-2114.
Expert Insight Into the First-line Treatment of Metastatic Colorectal Cancer
clinicaloptions.com/oncology
CRYSTAL: KRAS Status in Response to
Cetuximab
 Randomized, multicenter phase III trial[1]
– Significant improvement in PFS with addition of cetuximab to
FOLFIRI vs FOLFIRI alone for first-line mCRC treatment
 Updated survival analysis of
CRYSTAL[2]
– Included only subset
of KRAS-evaluable
patients (n = 1063)
Stratified by geographic
region, ECOG PS
EGFR-positive patients
with previously
untreated mCRC
(N = 1218)
*FOLFIRI: irinotecan 180 mg/m2, folinic acid 400 mg/m2, 5-FU 400 mg/m2 bolus,
5-FU infusion 2.4 g/m2 over 46 hrs.
†Cetuximab: 400 mg/m2 on Day 1, then 250 mg/m2/wk.
1. Van Cutsem E, et al. N Engl J Med. 2009;360:1408-1417.
2. Van Cutsem E, et al. J Clin Oncol. 2011;29:2011-2019.
FOLFIRI* 2-wk cycle
(n = 609)
FOLFIRI* 2-wk cycle +
Cetuximab†
(n = 609)
Expert Insight Into the First-line Treatment of Metastatic Colorectal Cancer
clinicaloptions.com/oncology
CRYSTAL: Updated Survival Analysis
Efficacy Results
 Significant interactions between KRAS status and treatment
outcome observed with PFS, OS, and ORR efficacy outcomes
– Survival and response benefits with addition of cetuximab only
achieved in KRAS wild-type patients
Cetuximab +
FOLFIRI
FOLFIRI
Hazard Ratio
P value
Wild-type KRAS
 Median PFS, mos
 Median OS, mos
 ORR, %
9.9
23.5
57.3
8.4
20.0
39.7
0.696
0.796
2.069*
.0012
.0093
< .001
Mutated KRAS
 Median PFS
 Median OS
 ORR, %
7.4
16.7
31.3
7.7
16.2
36.1
1.171
1.035
0.822*
.26
.75
.35
Outcome
*Odds ratio
Van Cutsem E, et al. J Clin Oncol. 2011;29:2011-2019.
Expert Insight Into the First-line Treatment of Metastatic Colorectal Cancer
clinicaloptions.com/oncology
OPUS: KRAS Status in Response to
Cetuximab
 Randomized, open-label, multicenter phase II trial[1]
– Significant improvement in ORR with addition of cetuximab
to FOLFOX-4 vs FOLFOX-4 alone for first-line mCRC
treatment
 Biomarker analysis of
OPUS[2]
– Included only subset
of KRAS-evaluable
patients (n = 315)
EGFR-positive patients
with previously
untreated mCRC
(N = 344)
*FOLFOX-4: oxaliplatin 85 mg/m2, leucovorin 200 mg/m2, 5-FU 400 mg/m2 bolus,
5-FU infusion 600mg/m2 over 22 hrs.
†Cetuximab: 400 mg/m2 on Day 1, then 250 mg/m2/wk.
1. Bokemeyer C, et al. J Clin Oncol. 2009;27:663-671.
2. Bokemeyer C, et al. Ann Oncol. 2011;22:1535-1546.
FOLFOX-4*
(n = 168)
FOLFOX-4* +
Cetuximab†
(n = 169)
Expert Insight Into the First-line Treatment of Metastatic Colorectal Cancer
clinicaloptions.com/oncology
OPUS: Biomarker Analysis OS Results
Bokemeyer C, et al. Ann Oncol. 2011;22:1535-1546.
Expert Insight Into the First-line Treatment of Metastatic Colorectal Cancer
clinicaloptions.com/oncology
Meta-Analysis: KRAS Status in Response
to Cetuximab
 Pooled efficacy analysis of CRYSTAL and OPUS studies
– CRYSTAL: FOLFIRI + cetuximab vs FOLFIRI alone[2]
– OPUS: FOLFOX + cetuximab vs FOLFOX alone[3]
 90% of samples were subjected to KRAS genotype testing
– HRs for benefit of addition of cetuximab highly statistically
significant in patients with wild-type KRAS
– PFS
– HR: 0.66 (P < .0001)
– OS
– HR: 0.81 (P = .0062)
Bokemeyer C, et al. ASCO 2010. Abstract 3506.
Expert Insight Into the First-line Treatment of Metastatic Colorectal Cancer
clinicaloptions.com/oncology
PRIME: KRAS Status in Response to
Panitumumab
 Randomized, global, open-label, phase III trial
Stratified by ECOG PS (0-1 vs 2)
and geographic region (Western
Europe, Canada, and Australia vs all
other locations)
Patients with
previously
untreated mCRC
Panitumumab 6.0 mg/kg q2w +
FOLFOX4 q2w
(n = 593)
(N = 1183)
Douillard JY, et al. J Clin Oncol. 2010;28:4697-4705.
FOLFOX4 q2w
(n = 590)
Expert Insight Into the First-line Treatment of Metastatic Colorectal Cancer
clinicaloptions.com/oncology
PRIME: Efficacy Results


Wild-type KRAS patients
Mutated KRAS patients
– PFS significantly improved with
FOLFOX4 + panitumumab
– Worse PFS outcome with
panitumumab addition
– HR: 0.80,; 95% CI: 0.67-0.96;
P = .01
– HR: 1.27; 95% CI: 1.04-1.55;
P = .02
Panitumumab +
FOLFOX4
FOLFOX4
P value
Wild-type KRAS
 Median PFS
 Median OS
10.0
23.9
8.6
19.7
.01
.17
Mutated KRAS
 Median PFS
 Median OS
7.4
15.5
9.2
19.2
.02
.15
Outcome, mos
Douillard JY, et al. J Clin Oncol. 2010;28:4697-4705.
Expert Insight Into the First-line Treatment of Metastatic Colorectal Cancer
clinicaloptions.com/oncology
MRC COIN: KRAS Status in Response to
Cetuximab
 Randomized, open-label trial
– Largest to assess addition of cetuximab to chemotherapy in
first-line treatment of mCRC
– Prospective analysis of efficacy in relation to KRAS mutation
status
Patients
with previously
untreated mCRC
irrespective of EGFR
status
(N = 344)
OxFP*
(n = 815)
OxFP* +
Cetuximab†
(n = 815)
3rd treatment arm randomized patients
to interittent OxFP (n = 815)
Maughan TS, et al. Lancet. 2011;377:2103-2114.
*OxFP: oxaliplatin 130 mg/m2 plus
capecitabine 850 mg/m2 twice daily
for 2 weeks, or oxaliplatin 85 mg/m2
plus leucovorin 175 or 350 mg and
5-FU 400 mg/m2 bolus then
5-FU infusion 2400 mg/m2 over 46
hrs.
†Cetuximab: 400 mg/m 2 on Day 1,
then 250 mg/m2/wk.
Expert Insight Into the First-line Treatment of Metastatic Colorectal Cancer
clinicaloptions.com/oncology
MRC COIN: Survival Efficacy Results
 No improvement in either OS or PFS with addition of
cetuximab to chemotherapy in KRAS wild-type patients
KRAS
wild-type
OxFP
OxFP + Cetuximab
Maughan TS, et al. Lancet. 2011;377:2103-2114.
KRAS
mutated
Expert Insight Into the First-line Treatment of Metastatic Colorectal Cancer
clinicaloptions.com/oncology
KRAS: Not all Mutations are Equal
 Mutation pattern[1]
– 79% of CRC tumors have mutations in codon 12
– 17.6% of CRC tumors have mutations in codon 13
 Anecdotal reports suggest < 10% of KRAS-mutated CRC patients can
respond to anti-EGFR therapy[2,3]
– Codon 13 mutations overrepresented in these tumors
 Codon 13 mutations show weaker transforming activity in vitro[4]
 KRAS G13D mutation associated with longer OS and PFS vs other
KRAS mutations[1]
1. De Roock W, et al. JAMA. 2010;304:1812-1820. 2. Benvenuti S, et al. Cancer Res. 2007;67:2643-2648.
3. Moroni M, et al. Lancet Oncol. 2005;6:279-286. 4. Guerrero S, et al. Cancer Res. 2000;60:6750-6756.
A Vision of Tomorrow’s
Treatment Algorithm:
Colorectal Cancer
Translational Research 2012
clinicaloptions.com/oncology
Impact of BRAF Mutations on Anti-EGFR
Therapy
 5% to 9% of CRC tumors have specific BRAF V600E mutation[1,2]
– Results in constitutive activation of BRAF protein
– Typically do not occur in tumors with KRAS exon 2 mutations[2]
 Outcomes with BRAF V600E mutation
– Non-first-line setting: BRAF V600E mutation associated with
resistance to EGFR-targeted agents[3-5]
– First-line setting: BRAF V600E mutation associated with poorer
prognosis, but some patients may still benefit from addition of
cetuximab to chemotherapy[6-7]
1. Van Cutsem E, et al. ASCO 2010. Abstract 3570. 2. Tol J, et al. NEJM. 2009;361:98-99. 3. Di
Nicolantonio F, et al. J Clin Oncol. 2008;26:5705-5712. 4. Laurent-Puig P, et al. J Clin Oncol.
2009;27:5924-5930. 5. Loupakis F, et al. Br J Cancer. 2009;101:715-721. 6. Van Cutsem E, et al. J Clin
Oncol. 2011;29:2011-2019. 7. Bokemeyer C, et al. ASCO 2010. Abstract 3506.
Translational Research 2012
clinicaloptions.com/oncology
BRAF Mutations in First-Line Setting
 CRYSTAL and OPUS meta-analysis
– Patients with wild-type KRAS/mutated BRAF
– Worse outcomes vs wild-type KRAS/wild-type BRAF
– Still experienced non-significant improvements (small N)
Outcome
Wild-type KRAS/
Mutated BRAF
Wild-type KRAS/
Wild-type BRAF
Cetuximab
+ CT
CT Only
P Value
Cetuximab
+ CT
CT Only
P Value
Median PFS,
mos
7.1
3.7
.267
10.9
7.7
< .001
Median OS,
mos
14.1
9.9
.079
24.8
21.1
.041
Median ORR, %
21.9
13.2
.4606
60.7
40.9
< .0001
Bokemeyer C, et al. ASCO 2010. Abstract 3506.
Translational Research 2012
clinicaloptions.com/oncology
Other Potential Biomarkers in CRC

PI3K: 39%[1]
– Associated with significantly
decreased PFS in patients treated
with FOLFIRI plus cetuximab[1]


– Associated with poor prognosis[5]

EGFR ligands
– Surrogate markers for EGFR
inhibition with cetuximab[3]
Defective mismatch repair: 15%[6]
– Improved prognosis and stagedependent survival[7]
NRAS: 2%[2]
– Prognostic/predictive value
unclear[2]

DNA hypermethylation: 30%[4]
– Predictive for lack of efficacy of 5FU-based adjuvant therapy[8]

VEGF receptors and ligand
– VEGF expression may be
associated with poorer prognosis[9]
1. Linot B, et al. ASCO 2010. Abstract 365. 2. Irahara N, et al. Diagn Mol Pathol. 2010;19:157-163. 3. Cremolini C, et al.
ASCO 2010. Abstract 99. 4. Kulendran M, et al. Cancers. 2011;3:1622-1638. 5. Barault L, et al. Cancer Res. 2008;68:85418546. 6. Kerr DJ. J Clin Oncol. 2010;28:3210-3212. 7. Popat S, et al. J Clin Oncol. 2005;23:609-618. 8. Sargent D, et al. J
Clin Oncol. 2010;28:3219-3227. 9. Bendardaf R, et al. Anticancer Res. 2008;28:3865-3870.
Translational Research 2012
clinicaloptions.com/oncology
CTC Levels
 mCRC patients treated with chemotherapy plus targeted agents in
CAIRO2 phase III trial (N = 467)[1]
– Higher CTC count at baseline and during treatment associated
with lower survival vs lower CTC count
– PFS HR: 1.5
– OS HR: 2.2
 mCRC patients (N = 430)[2]
– Higher CTC count at baseline and during treatment associated
with lower survival vs lower CTC count
– PFS: 4.4 vs 7.8 months, P = .004
– OS: 9.4 vs 20.6 months, P < .0001
1. Tol J, et al. Ann Oncol. 2010;21:1006-1012. 2. Cohen SJ, et al. Ann Oncol. 2009;20:1223-1229.
Role of HER2 in Gastric Cancer
Translational Research 2012
clinicaloptions.com/oncology
Rationale for Targeting HER2 in Gastric
Cancer
 10% to 25% tumors overexpress HER21
 HER2 overexpression associated with poor prognosis2
– Poor survival
– Serosal invasion
– Lymph node metastases
– Disease stage
– Distant metastases
1. Yano T, et al. Oncol Rep. 2006;15:65-71.
2. Jorgensen JT, et al. J Cancer. 2012;3:137-144.
Translational Research 2012
clinicaloptions.com/oncology
Chemotherapy Treatment of HER2Positive Gastric Cancer Patients
 Patients with initially metastatic or recurrent gastric
cancer, treated with first-line mFOLFOX-6
Outcome, mos
HER2-positive
HER2-negative
P value
Median TTP
4.3
5.9
0.177
Median OS
7.5
10.8
0.068
 TTP particularly worse in HER2-positive patients without
diffuse-type histology
Kim JW, et al. Anticancer Res. 2012;32:1547-1553.
Translational Research 2012
clinicaloptions.com/oncology
Challenges Determining HER2 Status in
Gastric Cancer
 HER2 testing in gastric cancer differs from breast cancer
– More frequent HER2 heterogeneity (focal staining)
– Incomplete membrane staining
 Testing protocol
– Initial testing with IHC
– Retest IHC 2+ samples with FISH
Rüschoff J, et al. Mod Pathol. 2012;25:637-650.
Translational Research 2012
clinicaloptions.com/oncology
HER2 Positivity: IHC
Score
Surgical Specimen
Staining Pattern
Biopsy Specimen Staining
Pattern
HER2 Overexpr.
Assessment
0
No reactivity or
membranous
reactivity in < 10% of
tumor cells
No reactivity or no
membranous reactivity
in any tumor cell
Negative
1+
Faint or barely perceptible
membranous reactivity in
≥ 10% of tumor cells; cells
are reactive only in part of
their membrane
Tumor cell cluster with faint or
barely perceptible membranous
reactivity irrespective of % of
tumor cells stained
Negative
2+
Weak to moderate
complete, basolateral or
lateral membranous
reactivity in ≥ 10% of
tumor cells
Tumor cell cluster with weak to
moderate complete, basolateral
or lateral membranous reactivity
irrespective of % of tumor cells
stained
Equivocal
3+
Strong complete,
basolateral or lateral
membranous reactivity in
≥ 10% of tumor cells
Tumor cell cluster with strong
complete, basolateral or lateral
membranous reactivity
irrespective of % of tumor cells
stained
Positive
Bang YJ, et al. Lancet. 2010;376:687-697.
Bang YJ, et al. ASCO 2009. Abstract 4556.
Translational Research 2012
clinicaloptions.com/oncology
Trastuzumab + Chemotherapy in
Advanced HER2+ Gastric Cancer: ToGA
 Rationale: a subpopulation of gastric cancers overexpress HER2
Stratified by ECOG PS,
advanced vs metastatic, gastric vs GEJ,
measurable disease, capecitabine vs 5-FU
Patients with
advanced
gastric cancer
screened for
HER2 status
(N = 3803)
Patients with
HER2+
advanced
gastric cancer
(n = 810; 22% of
successful
screenings)
R
(n = 584)
5-FU or Capecitabine* +
Cisplatin 80 mg/m2 q3w x 6 +
Trastuzumab 6 mg/kg q3w until PD
(8 mg/kg loading dose)
(n = 294)
5-FU or Capecitabine* +
Cisplatin 80 mg/m2 q3w x 6
(n = 290)
 Primary endpoint: OS
*Selected at investigator’s discretion: 5-FU 800 mg/m2/day infusional on Days 1-5 q3w x 6;
capecitabine 1000 mg/m2 BID on Days 1-14 q3w x 6.
Bang YJ, et al. Lancet. 2010;376:687-697.
Translational Research 2012
clinicaloptions.com/oncology
ToGA: HER2 Positivity Worldwide
Asian/Pacific
35
Europe
South/Middle
America
Other
Patients, %
30
25
20
15
10
5
0
Bang YJ, et al. ASCO 2009. Abstract 4556.
HER2 positivity in ToGA: 22.1%
Translational Research 2012
clinicaloptions.com/oncology
ToGA: HER2 Positivity Histological Type
Histological type
Localization
Subtype
HER2 Positivity (%)
P Value
Intestinal
Diffuse
Mixed
32.2
6.1
20.4
< .001
GEJ
Gastric
33.2
20.9
< .002
Histological subtype and tumor sublocalization are important factors
for HER2 expression/amplification in gastric cancer
Bang YJ, et al. ASCO 2009. Abstract 4556.
Translational Research 2012
clinicaloptions.com/oncology
ToGA: HER2 Positivity GEJ vs Gastric
Cancer
100
Gastric cancer
GEJ cancer
Samples (%)
80
60
40
20
0
N = 2759; *P < .001
HER2 positive, %
Bang YJ, et al. ASCO 2009. Abstract 4556.
Gastric Cancer
GEJ Cancer
Total
20.9
33.2*
21.3
Translational Research 2012
clinicaloptions.com/oncology
Survival Probability
ToGA: Primary Endpoint OS
1.0
0.9
0.8
0.7
0.6
0.5
0.4
0.3
0.2
0.1
0
Median
Events OS
HR
FC + T
FC
11.1
0
2
4
6
167
182
13.8
11.1
95% CI P Value
0.74 0.60-0.91
13.8
8 10 12 14 16 18 20 22 24 26 28 30 32 34 36
Mos
Pts at Risk, n
294 277 246 209 173 147 113 90
290 266 223 185 143 117 90 64
Bang YJ, et al. Lancet. 2010;376:687-697.
71
47
56
32
43
24
30
16
21
14
13
7
12
6
6
5
4
0
1
0
0
0
.0046
Translational Research 2012
clinicaloptions.com/oncology
ToGA: Efficacy Outcomes
Outcome
Chemotherapy
+ Trastuzumab
(n = 294)
Chemotherapy
Alone
(n = 290)
HR (95% CI)
P Value
Median OS, mos
13.8
11.1
0.74 (0.60-0.91)
.0046
Median PFS, mos
6.7
5.5
0.71 (0.59-0.85)
.0002
ORR, %
47
35
-
.0017

CR
5
2
-
.0599

PR
42
32
-
.0145
 Preplanned subgroup analysis indicated improved OS benefit with
increasing HER2 expression by IHC
 Exploratory analysis of IHC 2+/FISH+ and IHC 3+ cohort
demonstrated a 4-mo increase in OS with trastuzumab
– HR: 0.65 (95% CI: 0.51-0.83)
Bang YJ, et al. Lancet. 2010;376:687-697.
Translational Research 2012
clinicaloptions.com/oncology
ToGA: OS by HER2 Status
HER2 Status Subgroup
All patients (N = 584)
Median OS, Mos
(CT + T vs CT Alone)
HR* (95% CI)
13.8 vs 11.1
0.74 (0.60-0.91)
Preplanned analysis

IHC 0/FISH+ (n = 61)
10.6 vs 7.2
0.92 (0.48-1.76)

IHC 1+/FISH+ (n = 70)
8.7 vs 10.2
1.24 (0.70-2.20)

IHC 2+/FISH+ (n = 159)
12.3 vs 10.8
0.75 (0.51-1.11)

IHC 3+/FISH+ (n = 256)
17.9 vs 12.3
0.58 (0.41-0.81)

IHC3+/FISH- (n = 15)
17.5 vs 17.7
0.83 (0.20-3.38)
Exploratory analysis

IHC 0 or 1+/FISH+ (n = 131)
10.0 vs 8.7
1.07 (0.70-1.62)

IHC 2+/FISH+ or IHC 3+ (n = 446)
16.0 vs 11.8
0.65 (0.51-0.83)
*HR < 1 favors chemotherapy + trastuzumab; HR > 1 favors chemotherapy alone.
Bang YJ, et al. Lancet. 2010;376:687-697.
Translational Research 2012
clinicaloptions.com/oncology
Survival Probability
ToGA: OS in IHC 2+/FISH+ or IHC 3+
(Exploratory Analysis)
1.0
0.9
0.8
0.7
0.6
0.5
0.4
0.3
0.2
0.1
0
Median
Events OS
HR
FC + T
FC
11.8
0
2
4
6
120
136
16.0
11.8
95% CI
0.65 0.51-0.83
16.0
8 10 12 14 16 18 20 22 24 26 28 30 32 34 36
Mos
Pts at Risk, n
228 218 196 170 142 122 100 84
218 198 170 141 112 96 75 53
Bang YJ, et al. Lancet. 2010;376:687-697.
65
39
51
28
39
20
28
13
20
11
12
4
11
3
5
3
4
0
1
0
0
0
Translational Research 2012
clinicaloptions.com/oncology
ToGA: Select Toxicities
Adverse Event, %
Chemotherapy + Trastuzumab
(n = 294)
Chemotherapy Alone
(n = 290)
Grade 3/4 hematologic events

Neutropenia
27
30

Anemia
12
10
Grade 3/4 nonhematologic
events

Diarrhea
9
4

Nausea
7
7
Cardiac events
6
6

1
3
5
1
Grade 3/4
LVEF reduction of ≥ 10% to
absolute value < 50%*
*Chemotherapy plus trastuzumab: n = 237; chemotherapy alone: n = 187.
Bang YJ, et al. Lancet. 2010;376:687-697.
A Vision of Tomorrow’s
Treatment Algorithm:
Gastric Cancer
Translational Research 2012
clinicaloptions.com/oncology
Other Potential Biomarkers in Gastric
Cancer

MicroRNAs
– Unique microRNAs associated with distinct prognostic and predictive values [1,2]
– 7-microRNA signature independent predictor of OS and RFS[3]

DNA methylation
– Prognostic/predictive value[4]

CEA
– Elevated levels correspond with worse prognosis and survival[5]

Cyclin D1
– Elevated levels correspond with worse prognosis[6]
1. Ueda T, et al. Lancet Oncol. 2010;11:136-146. 2. Li X, et al. Gut. 2010;59:579-585. 3. Brenner B, et al.
World J Gastroenterol. 2011;17:3976-3985. 4. Sapari NS, et al. PLoS One. 2012;7:e36275. 5. Mihmanli M,
et al. Hepatogastroenterology. 2004;51:1544-1547. 6. Gao P, et al. World J Gastroenterol. 2004;10:29362939.
Translational Research 2012
clinicaloptions.com/oncology
Summary

KRAS mutation status critical determinant of benefit with EGFR-targeted
antibody therapy
– BRAF mutations potentially predictive of reduced benefit in non-first-line
setting

Future biomarkers in CRC
– PI3K, NRAS, EGFR ligand, DNA hypermethylation, mismatch repair,
VEGF, CTC levels

HER2 expression confers worse prognosis in gastric cancer
– HER2 expression challenging to assess and should be performed only by
experienced pathologists

Future biomarkers in gastric cancer
– MicroRNAs, DNA methylation, CEA, cyclin D1
Go Online for More CCO
Coverage of GI Cancers!
Capsule Summaries of all the key data, plus Expert Analysis panel
discussions exploring the clinical implications
Expert Highlights: download mp3 files and listen to our experts review
the highlights of this conference
Expert Recap (slides and audio) plus
downloadable PowerPoint slides
clinicaloptions.com/oncology