ArticlePDF Available

Any progress in the management of advanced pancreatic cancer? Highlights from the 45th ASCO annual meeting. Orlando, FL, USA. May 29-June 2, 2009

Authors:

Abstract and Figures

Majority of the patients with pancreatic cancer present with advanced disease that is lethal and notoriously difficult to treat. Survival has not improved dramatically despite routine use of chemotherapy and radiotherapy; this situation signifies an urgent need for novel therapeutic approaches. The treatment of advanced disease with gemcitabine has only a modest activity on survival with a favorable impact on quality of life. So far, the current targeted agents that have been used in combination with gemcitabine have failed to improve clinical outcomes. This failure may stem from the heterogeneous molecular pathogenesis of pancreatic cancers, which involves several oncogenic pathways and defined genetic mutations. However, recent data support the evidence that the combination of gemcitabine with erlotinib, capecitabine or platinum compounds could be more active than gemcitabine alone in advanced pancreatic cancer. New therapeutic strategies, particularly using molecular target agents, are under evaluation. A number of molecular mechanisms responsible of transformation and progression of pancreatic cancer have been identified, opening the possibility to identify also possible pharmacological targets. Pancreatic cancer remains the 4th leading cause of cancer death in the U.S.A.. How to treat a non-resectable pancreatic cancer has been a challenging topic for all medical oncologists. Historical 5-fluorouracil has been replaced by single agent gemcitabine since 1997. Numerous combinations using gemcitabine as a backbone have been tested in clinical trials; unfortunately, none of the combinations including the ones with biological agents was proved to be significantly superior to gemcitabine alone. This year, more combinations were investigated and the results were presented on the meeting. In first-line setting, two large phase III trials (Abstracts #4504 and #4601) failed to prove any additional benefit of a second cytotoxic agent or a vaccine. Folinic acid plus 5-FU plus oxaliplatin (FOLFOX) and 5-fluorouracil plus leucovorin plus irinotecan (FOLFIRI) could be considered in the second-line setting after failure of gemcitabine therapy (Abstract #4618). Novel agents (Abstracts #4501, #4625, #4626, #4617) provide some hope; however, in general, all combinations are still significantly relying on the backbone of gemcitabine. Thinking beyond the gemcitabine box and exploring novel agents are very crucial now.
Content may be subject to copyright.
JOP. J Pancreas (Online) 2009 Jul 6; 10(4):361-365.
JOP. Journal of the Pancreas - http://www.joplink.net - Vol. 10, No. 4 - July 2009. [ISSN 1590-8577] 361
HIGHLIGHT ARTICLE
Any Progress in the Management of Advanced Pancreatic Cancer?
Highlights from the “45th ASCO Annual Meeting”. Orlando, FL, USA. May 29 - June 2, 2009
Jia Li, Muhammad Wasif Saif
Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
Summary
Majority of the patients with pancreatic cancer present with advanced disease that is lethal and notoriously difficult to treat. Survival
has not improved dramatically despite routine use of chemotherapy and radiotherapy; this situation signifies an urgent need for novel
therapeutic approaches. The treatment of advanced disease with gemcitabine has only a modest activity on survival with a favorable
impact on quality of life. So far, the current targeted agents that have been used in combination with gemcitabine have failed to
improve clinical outcomes. This failure may stem from the heterogeneous molecular pathogenesis of pancreatic cancers, which
involves several oncogenic pathways and defined genetic mutations. However, recent data support the evidence that the combination
of gemcitabine with erlotinib, capecitabine or platinum compounds could be more active than gemcitabine alone in advanced
pancreatic cancer. New therapeutic strategies, particularly using molecular target agents, are under evaluation. A number of
molecular mechanisms responsible of transformation and progression of pancreatic cancer have been identified, opening the
possibility to identify also possible pharmacological targets. Pancreatic cancer remains the 4th leading cause of cancer death in the
U.S.A.. How to treat a non-resectable pancreatic cancer has been a challenging topic for all medical oncologists. Historical 5-
fluorouracil has been replaced by single agent gemcitabine since 1997. Numerous combinations using gemcitabine as a backbone
have been tested in clinical trials; unfortunately, none of the combinations including the ones with biological agents was proved to be
significantly superior to gemcitabine alone. This year, more combinations were investigated and the results were presented on the
meeting. In first-line setting, two large phase III trials (Abstracts #4504 and #4601) failed to prove any additional benefit of a second
cytotoxic agent or a vaccine. Folinic acid plus 5-FU plus oxaliplatin (FOLFOX) and 5-fluorouracil plus leucovorin plus irinotecan
(FOLFIRI) could be considered in the second-line setting after failure of gemcitabine therapy (Abstract #4618). Novel agents
(Abstracts #4501, #4625, #4626, #4617) provide some hope; however, in general, all combinations are still significantly relying on
the backbone of gemcitabine. Thinking beyond the gemcitabine box and exploring novel agents are very crucial now.
Introduction
American Cancer Society has estimated in 2009, there
will be 21,050 new pancreatic cancer cases in men and
21,420 in women, while 35,240 (about 83%) will die of
pancreatic cancer in 2009 [1]. Pancreatic cancer
remains the 4th cause of death by cancer after lung,
prostate (breast in women), colorectal cancer since
1970s in the USA, although it represents only 2-3% of
all cancers. Endless effort has been put on this
aggressive disease; however, surgical resection remains
the only curative option. Locally advanced or
metastatic diseases are considered non-curable,
palliative chemotherapies are often administered for
alleviating symptoms. Fluorouracil (5-FU) had been
the only active drug in pancreatic cancer for over
decades until the emerging of gemcitabine in 1997 [2].
A significantly higher clinical benefit response
associated with gemcitabine treatment was observed
(23.8% vs. 4% in 5-FU arm) although the overall
objective response rate remained modest [2]. Based on
these results, FDA approved gemcitabine as the first
line therapy for advanced pancreatic cancer in 1997.
Since then, various combinations using gemcitabine as
a backbone were designed and tested in clinical trials.
Unfortunately, none of the combinations is proved to
be superior to gemcitabine monotherapy.
With the advances in molecular biology, newer
biologic agents such as erlotinib, cetuximab and
bevacizumab are adding some benefit to the
conventional cytotoxic agents. Unfortunately, these
agents all failed to show any significant superiority
over gemcitabine except the combination of erlotinib
plus gemcitabine [3]; however, the clinical impact of
this combination remains very controversial until now.
The disappointing results did not discourage
investigators but stimulated them to look for more
Key words Clinical Trials as Topic; Disease-Free Survival;
erlotinib; gemcitabine; Pancreatic Neoplasms; Survival
Abbreviations CONKO: Charité Onkologie; LMWH: low
molecular weight heparin
Correspondence Muhammad Wasif Saif
Section of Medical Oncology, Yale University School of
Medicine, 333 Cedar Street; FMP:116, New Haven, CT 06520,
USA
Phone: +1-203.737.1875; Fax: +1-203.785.3788
E-mail: wasif.saif@yale.edu
Document URL http://www.joplink.net/prev/200907/24.html
JOP. J Pancreas (Online) 2009 Jul 6; 10(4):361-365.
JOP. Journal of the Pancreas - http://www.joplink.net - Vol. 10, No. 4 - July 2009. [ISSN 1590-8577] 362
pharmaceutical agents or combinations. We have
gladly seen over 80 abstracts presented in the 2009
annual meeting of the American Society of Clinical
Oncology (ASCO) in the field of pancreatic cancer. In
this highlight article, we will focus on the management
of advanced (locally advanced and metastatic)
pancreatic cancer.
Since the approval of gemcitabine, true progress in the
management of pancreatic cancer has been very
minimal. There has been persistent effort in the field of
medical oncology regards to explore novel agents
based on better understanding of the diseases.
1. First-Line Therapies
Current standard first-line therapies for advanced
pancreatic cancer are gemcitabine or gemcitabine plus
erlotinib. A number of abstracts are exploring further
first-line options. Interestingly, gemcitabine remains
the core of the combinations.
1.1 Phase III Trials
Three large trials were presented (Table 1) [4, 5, 6];
unfortunately, two large trials (Abstracts #4504 and
#4601) failed to prove any additional benefit of a
second cytotoxic agent or a vaccine. The third trial
(Abstract #4604) comparing erlotinib plus capecitabine
with erlotinib plus gemcitabine only presented interim
toxicity data from 127 patients, efficacy data are
pending. To think beyond the gemcitabine box and
search for novel agents have become crucially urgent
in order to conquer this very aggressive disease.
1.1.1 Gemcitabine vs. Gemcitabine plus Cisplatin
The “Gruppo Oncologico dell’Italia Meridionale
conducted a phase III trial to compare gemcitabine
with or without oxaliplatin, the benefit was only
observed in progression-free survival but not overall
survival, however later pooled- and meta-analysis
proved that the addition of platinum to gemcitabine did
offer survival benefit in selected patients [7, 8, 9]. The
Gruppo Italiano Pancreas” (GIP) conducted another
superiority study to compare gemcitabine monotherapy
with gemcitabine plus cisplatin in advanced pancreatic
cancer patients [4]. The data were presented in this
annual meeting. A total of 400 patients were enrolled
from 46 Italian institutions. One-hundred and ninety-
nine patients received gemcitabine single agent (1,000
mg/m2 weekly x 7, then weekly x 3 every 4 weeks),
whereas the other 201 patients received combination
therapy of gemcitabine plus cisplatin (in addition to
gemcitabine administered as above, cisplatin was given
at 25 mg/m2 weekly). Surprisingly, this large trial did
not demonstrate any survival benefit by adding
cisplatin to gemcitabine. The results not only
confirmed a previously published negative phase III
trial, but also warned all clinicians to carefully interpret
pooled or meta-analyses.
1.1.2 Gemcitabine vs. GV1001 plus Gemcitabine
GV1001 is a telomerase peptide vaccine which showed
a median overall survival of 8.6 months in non-
resectable pancreatic cancer [10]. In order to compare
the efficacy of a combination therapy of GV1001 and
gemcitabine with gemcitabine monotherapy, a phase III
trial was designed [5]. A total of 520 patients were
planned. Patients were randomly assigned to either
gemcitabine monotherapy (1,000 mg/m2 over 30 min
weekly x 7, then weekly x 3 every 4 weeks) or a
sequential combination of GV1001 and gemcitabine
(GV1001 0.56 mg subcutaneous plus granulocyte-
macrophage colony-stimulating factor as immune
adjuvant on days 1, 3, 5, 8, 15, 22, 36, then every 4
weeks, gemcitabine was added when disease
progressed on GV1001). Unfortunately, after 365
patients were enrolled, a preliminary analysis indicated
no survival benefit by giving GV1001. Thus this trial
was prematurely terminated.
1.1.3 Erlotinib plus Capecitabine vs. Erlotinib plus
Gemcitabine
Erlotinib has been proved to have effect in combination
with gemcitabine for advanced pancreatic cancer.
Whether erlotinib can be combined with other
cytotoxic agents such as capecitabine in treating
advanced pancreatic cancer was investigated in a phase
III trial conducted by the “Arbeitsgemeinschaft
Internistische Onkologie” (AIO) group [6]. Two-
hundred and eighty-one patients randomly received
either capecitabine (200 mg/m2/day, days 1-14 every 3
weeks) plus erlotinib (150 mg/day) or gemcitabine
(1,000 mg/m2 over 30 min weekly x 7, then weekly x 3
every 4 weeks) plus erlotinib. The first interim analysis
was reported on the meeting. Sixty patients received
capecitabine plus erlotinib, 67 patients received
gemcitabine plus erlotinib. Toxicity data indicated that
erlotinib can be safely combined with capecitabine;
however, the efficacy data are not completed yet.
Whether this combination could achieve similar
efficacy in terms of progression free survival and/or
overall survival as the combination of erlotinib with
gemcitabine, we will have to wait for the final results.
Table 1. Randomized phase III trials of gemcitabine-based first-line therapies.
Abstract Study design PFS (months) OS (months) Comments
#4504 [4] Arm A: gemcitabine,
Arm B: gemcitabine + cisplatin 3.9 vs. 3.8
(P=0.8) 8.3 vs. 7.2
(P=0.38) Combination therapy did not provide any benefit in PFS, OS or
clinical benefit, but increased toxicities
#4601 [5] Arm A: gemcitabine,
Arm B: GV1001 + gemcitabine 3.7 vs. 1.9 7.3 vs. 5.9 GV1001 has no benefit in treating pancreatic cancer benfit when
administered in sequential combination with gemcitabine
#4604 [6] Arm A: capecitabine plus erlotinib,
Arm B: gemcitabine plus erlotinib Not presented Not presented The first interim analysis only presented toxicity data from the first
127 patients. The combination of erlotinib and capecitabine seems to
be tolerable; however, the efficacy data are not finalized yet
OS: overall survival; PFS: progression-free survival
JOP. J Pancreas (Online) 2009 Jul 6; 10(4):361-365.
JOP. Journal of the Pancreas - http://www.joplink.net - Vol. 10, No. 4 - July 2009. [ISSN 1590-8577] 363
1.2 Phase I/II Trials
Several phase I/II trials studied more combinations,
including four novel agents which will be discussed in
more details in next section (Tables 2 and 3).
2. Second-Line Therapies
Lack of attention to second line treatment strategy in
advanced pancreatic cancer is due to the fact that we
still do not have first line option that renders true
survival benefit; therefore, development of novel
therapeutic agents should be an obvious area of our
focus in the future. However, there is growing evidence
supporting benefit of chemotherapy after gemcitabine
failure in selected patients with good performance
status [14].
Few clinical trials investigating second-line options in
patients with advanced pancreatic cancer after failure
of gemcitabine were presented at the meeting. One
study aimed at exploring folinic acid plus 5-FU plus
oxaliplatin (FOLFOX) and 5-fluorouracil plus
leucovorin plus irinotecan (FOLFIRI.3), two
commonly used regimens in colorectal cancer in this
setting (Aabstract #4618) [15]. Sixty patients were
randomly assigned to either FOLFOX (oxaliplatin 85
mg/m2 over 120 min on day 1, leucovorin 400 mg/m2
on day 1, 5-FU 2,000 mg/m2 over 46 hours every two
weeks) or FOLFIRI.3 (irinotecan 70 mg/m2 over 60
min on day 1, leucovorin 400 mg/m2 over 2 hours on
day 1, 5-FU 2,000 mg/m2 over 46 hours from day 1,
then irinotecan 70 mg/m2 over 60 min at the end of the
5-FU infusion every two weeks). Six-month overall
survival rate in both arms were 25% and 20%,
respectively. Based on patients’ overall performance
status, and prior chemotherapy toxicities, these two
regimens can certainly be considered as second-line
option; however, the clinical benefit needs to be
validated in larger trials.
Table 2. Phase I/II trials of gemcitabine-based first-line therapies.
Abstract Study design Phase
level Efficacy PFS
(months) OS
(months) Severe
toxicities Comments
#4607 [11] Triple combination of
gemcitabine + erlotinib +
capecitabine (n=43)
II PR: 32.6%
SD: 51.2% 6.5 12.0 Cytopenia,
GI toxicity, and
rash
EGFR expression is poor
prognostic factor
#4614 [12] Arm A: PDXG regimen (n=46)
Arm B: PEXG regimen (n=46) II PR:
61% vs. 37% 6-month PFS:
58% vs. 54% 1-year OS:
41% vs. 41% Cytopenia,
fatigue Capecitabine is equivalent to
5-FU, docetaxel seems to be
slightly superior to epirubicin in
terms of response rate
#4623 [13] GTX regimen (n=41) II PR: 21.9%
SD: 41.5% 6.9 14.5 Cytopenia,
infections, and
mucositis
Large trial is warranted to
validate this promising regimen
GTX: gemcitabine, docetaxel and capecitabine; OS: overall survival; PDXG: cisplatin, docetaxel, 5-FU, gemcitabine; PEXG: epirubicin replacing
docetaxel; PFS: progression-free survival; PR: partial response; SD: stable disease
Table 3. Novel agents in the treatment of advanced pancreatic cancer.
Novel agents;
Abstract# Rationale Administration/schedule Clinical
trials Results Future directions
AMG655
#4501 [18] AMG655 is an agonist monoclonal
antibody against human death receptor 5
(DR5), activates caspases, and
subsequently induces apoptosis in
sensitive tumor cells.
Preclinical studies showed synergistic
effect of AMG655 and gemcitabine.
AMG655 at 3 mg/kg or
10 mg/kg on day 1 and 15
plus gemcitabine at
1,000 mg/m2on days 1, 8
and 15 every 28 days
Phase I,
first-line
therapy
13 patients.
PR: 31%. PFS: 5.3 months.
6-month survival rate: 76.2%.
Severe toxicities: 9 (69%);
especially cytopenia.
Same group is
conducting a phase
II trial to compare
gemcitabine with
or without
AMG655.
Nab-paclitaxel
#4525 [19] Pancreatic cancer cells and surrounding
stroma overexpress SPARC.
A new formulated paclitaxel, nab-P, an
albumin-bound nanoparticle form of
paclitaxel increased tumor accumulation
of paclitaxel through binding of albumin
to SPARC
Nab-paclitaxel
at 100-150 mg/m2
plus gemcitabine
at 1,000 mg/m2
were given on days 1, 8,
and 15 every 28 days
Phase I/II,
first-line
therapy
63 patients.
CR: 2%, PR: 12%, SD: 41%.
PFS: 4.8 months for SPARC-.
PFS: 6.2 months for SPARC+.
mOS: 9 months.
Severe toxicities 12 patients;
especially cytopenia.
Nab-paclitaxel is
very promising.
SPARC could be a
predictive factor.
A phase III trial in
larger populations
is warranted.
EndoTAG-1
#4526 [20] EndoTAG-1 is a novel cationic
liposomal formulation of paclitaxel
which targets negatively charged
endothelial cells of tumor blood vessels
Weekly gemcitabine at
1,000 mg/m2,
with or without twice
weekly endoTAG-1 at
3 dose levels:
11, 22 and 44 mg/m2
Phase II,
first-line
therapy
200 patients.
Response rate and PFS were not
presented.
mOS: 11.5 months for gemcitabine
plus high dose endoTAG-1.
More infusion-reaction is associated
with endoTAG-1 treatment groups.
Needs large trial to
confirm the data.
Masitinib
#4617 [21] Masitinib is a tyrosine kinase inhibitor
targeting c-Kit, PDGFR, FGFR3 and
affecting the FAK pathway.
Masitinib was found to enhance the
antiproliferative effects of gemcitabine
in preclinical studies.
Masitinib at 9 mg/kg/day
plus weekly gemcitabine
at 1,000 mg/m2
Phase II,
first-line
therapy
22 patients.
Clinical benefit: 16%.
mPFS: 6.4 months.
mOS: 7.1 months.
18-month survival rate: 23%.
Severe toxicities were:
cytopenia, diarrhea and rash.
The same group is
conducting a phase
III trial to compare
gemcitabine with
or without
masitinib.
CR: complete response; FAK: focal adhesion kinase; FGFR3: fibroblast growth factor receptor 3; mOS: median overall survival; mPFS: median
progression-free survival; PDGFR: platelet-derived growth factor receptor; PFS: progression-free survival; PR: partial response; SD: stable disease;
SPARC: secreted protein acid rich in cysteine
JOP. J Pancreas (Online) 2009 Jul 6; 10(4):361-365.
JOP. Journal of the Pancreas - http://www.joplink.net - Vol. 10, No. 4 - July 2009. [ISSN 1590-8577] 364
Current standard dose of erlotinib is 100 mg/day in
combination with gemcitabine [3]. Skin acne-like rash
has been proposed to be a “surrogate” marker for
response to biologic agents such as erlotinib and
cetuximab. In the 2007 ASCO Gastrointestinal Cancers
Symposium (Orlando, FL, U.S.A.; January 20th, 2007),
Van Cutsem et al. presented a dose-escalation study of
cetuximab in colorectal cancer (EVEREST). The
higher grade of skin rash correlating with increased
response rate was observed [17]. Whether this
“surrogate” marker can be used to maximize the
benefit from erlotinib was studied by Tang et al. in a
phase II trial [16]. Fifty patients with gemcitabine-
refractory pancreatic cancer were orally administered
erlotinib starting at 150 mg/day, dose-escalating by 50
mg every two weeks until rash more than grade 1 or
maximum dose of 300 mg/day (Figure 1). Twenty-five
percent of eligible patients achieved stable disease for
more than 8 weeks which met the primary end-point of
this trial. This trial certainly revolutionized our
understanding of erlotinib. It is worthwhile to perform
a large trial to validate these results and re-compare
gemcitabine with or without erlotinib in which the dose
of erlotinib should be based on skin rash.
3. Novel Agents
Development of novel therapeutic agents is an obvious
area of focus of research in pancreatic cancer. Several
novel agents either new biologic target agents
(AMG655 and masitinib) or newly formulated
conventional cytotoxic agents (endoTAG-1 and nab-
paclitaxel) are tested and results are promising (Table
3).
4. Supportive Therapy
Palliative care represents an important aspect of care in
patient with pancreatic malignancy. Identifying and
treating disease related symptomatology are priorities
[22].
The incidence of venous thromboembolism in
pancreatic cancer patients ranges from 17% to 57%.
Clinical data also suggest that the occurrence of venous
thromboembolism may be associated with poorer
prognosis in such patients. Recent data suggest that
anticoagulant treatments may improve cancer patient
survival by decreasing thromboembolic complications
as well as by anticancer effects [23]. Riess et al.
conducted the “Charité Onkologie” (CONKO-004)
trial to investigate whether the addition of enoxaparin,
a low molecular weight heparin (LMWH) improves
overall survival (Abstract #LBA4506) [24]. Safety and
feasibility of adding enoxaparin to chemotherapy have
been completed in their previously published pilot
study “Prospective, Randomized trial Of Simultaneous
Pancreatic cancer treatment with Enoxaparin”
(PROSPEC-CONKO-004) [25]. The primary endpoint
was to decrease the incidence of symptomatic venous
thromboembolic events. Three-hundred and twelve
patients were enrolled, 160 patients were treated with
chemotherapy plus enoxaparin. The occurrence of
venous thromboembolic events were 8/160 (5.0%)
compared with 14.5% in the non-LMWH arm (Table 4).
Clearly, enoxaparin is effective and safe for prevention
of symptomatic venous thromboembolic events;
however, whether the low incidence of venous
thromboembolic events is associated with some
survival benefit is still unclear. CONKO-004
preliminary data showed no difference in median
overall survival with or without exnoxaparin. We are
looking forward to their final results.
Future Directions
Options for pancreatic cancer in advanced/metastatic
setting are still very limited. Gemcitabine remains the
standard of care despite so many combinations were
examined. The two large phase III trials failed to show
any benefit beyond gemcitabine monotherapy by
adding a second cytotoxic agent such as cisplatin or a
vaccine GV1001. These combinations were promising
in early phase trials or pooled/meta-analysis. Again, we
should be careful when interpreting results from early
phase trials. Many promising results from phase II
trials were unable to be translated into phase III trials.
Over the last 12 years, we have extensively and
intensively explored all possible agents to combine
with gemcitabine, it is the time to think out of the
gemcitabine box and put more effort on novel agents.
Nab-paclitaxel, “an old drug in a new bottle”, seems to
be very promising when combined with gemcitabine.
Table 4. Results of CONKO-004 trial after a median follow-up of 30.4 weeks.
Primary/secondary end-points Chemotherapy arm
(n=152)
Chemotherapy plus
enoxaparin arm
(n=160)
Comments
Venous thromboembolic events 22 (14.5%) 8 (5.0%) P<0.05
Bleeding 15 (9.9%) 10 (6.3%) P=0.6
Median overall survival 29 weeks 31 weeks Preliminary results, not statistically calculated yet
Figure 1. Schema of phase II erlotinib single agent as second-line
therapy.
JOP. J Pancreas (Online) 2009 Jul 6; 10(4):361-365.
JOP. Journal of the Pancreas - http://www.joplink.net - Vol. 10, No. 4 - July 2009. [ISSN 1590-8577] 365
We are looking forward to the phase III results. New
biologic target agent such as AMG655, a monoclonal
antibody against human death receptor-5, also achieved
encouraging results. However, the current designs of
clinical trials in advanced pancreatic cancer still rely on
gemcitabine, even for the aforementioned novel agents.
Nevertheless, gemcitabine is the only cytotoxic agent
providing significant clinical benefit for pancreatic
cancer. We encourage more novel agents should be
tested in second-line setting.
Conflict of interest The authors have no potential
conflicts of interest
References
1. National Cancer Institute, Surveillance Epidemiology and End
Results. Pancreas. SEER Stat Fact Sheets
(http://seer.cancer.gov/statfacts/html/pancreas.html).
2. Burris HA 3rd, Moore MJ, Andersen J, Green MR, Rothenberg
ML, Modiano MR, et al. Improvements in survival and clinical
benefit with gemcitabine as first-line therapy for patients with
advanced pancreas cancer: a randomized trial. J Clin Oncol 1997;
15:2403-13. [PMID 9196156]
3. Moore MJ, Goldstein D, Hamm J, Figer A, Hecht JR, Gallinger
S, et al. Erlotinib plus gemcitabine compared with gemcitabine alone
in patients with advanced pancreatic cancer: a phase III trial of the
National Cancer Institute of Canada Clinical Trials Group. J Clin
Oncol 2007; 25:1960-6. [PMID 17452677]
4. Colucci G, Labianca R, Di Costanzo F, Gebbia V, Cartenì G,
Massidda B, et al. A randomized trial of gemcitabine (G) versus G
plus cisplatin in chemotherapy-naive advanced pancreatic
adenocarcinoma: The GIP-1 (Gruppo Italiano Pancreas-
GOIM/GISCAD/GOIRC) study. J Clin Oncol 2009; 27(15 Suppl.):
Abstract 4504.
5. Buanes T, Maurel J, Liauw W, Hebbar M, Nemunaitis J. A
randomized phase III study of gemcitabine (G) versus GV1001 in
sequential combination with G in patients with unresectable and
metastatic pancreatic cancer (PC). J Clin Oncol 2009; 27(15 Suppl.):
Abstract 4601.
6. Heinemann V, Vehling-Kaiser U, Waldschmidt D, Kettner E,
Marten A, Winkelmann, et al. Gemcitabine plus erlotinib followed
by capecitabine versus capecitabine plus erlotinib followed by
gemcitabine: Interim toxicity analysis of a multicenter, randomized,
cross-over phase III trial of the Arbeitsgemeinschaft Internistische
Onkologie (AIO). J Clin Oncol 2009; 27(15 Suppl.): Abstract 4604.
7. Louvet C, Labianca R, Hammel P, Lledo G, Zampino MG,
André T, et al. Gemcitabine in combination with oxaliplatin
compared with gemcitabine alone in locally advanced or metastatic
pancreatic cancer: results of a GERCOR and GISCAD phase III trial.
J Clin Oncol 2005; 23:3509-16. [PMID 15908661]
8. Heinemann V, Labianca R, Hinke A, Louvet C. Increased
survival using platinum analog combined with gemcitabine as
compared to single-agent gemcitabine in advanced pancreatic cancer:
pooled analysis of two randomized trials, the GERCOR/GISCAD
intergroup study and a German multicenter study. Ann Oncol 2007;
18:1652-9. [PMID 17660491]
9. Heinemann V, Boeck S, Hinke A, Labianca R, Louvet C. Meta-
analysis of randomized trials: evaluation of benefit from
gemcitabine-based combination chemotherapy applied in advanced
pancreatic cancer. BMC Cancer 2008; 8:82. [PMID 18373843]
10. Bernhardt SL, Gjertsen MK, Trachsel S, Møller M, Eriksen JA,
Meo M, et al. Telomerase peptide vaccination of patients with non-
resectable pancreatic cancer: A dose escalating phase I/II study. Br J
Cancer 2006; 95:1474-82. [PMID 17060934]
11. Oh D, Lee K, Lee K, Sohn C, Park Y, Zang D, et al. A phase II
trial of erlotinib in combination with gemcitabine and capecitabine in
previously untreated metastatic/recurrent pancreatic cancer:
Combined analysis with translational research. J Clin Oncol 2009;
27(15 Suppl.): Abstract 4607.
12. Cereda S, Rognone A, Ghidini M, Rezzonico S, Passoni P,
Mazza E, et al. A randomized phase II trial of two different four-drug
combinations in advanced pancreatic adenocarcinoma: Cisplatin,
capecitabine, gemcitabine plus either epirubicin or docetaxel. J Clin
Oncol 2009; 27(15 Suppl.): Abstract 4614.
13. Fine R, Moorer G, Sherman W, Chu, Maurer KM, Chabot J, et
al. Phase II trial of GTX chemotherapy in metastatic pancreatic
cancer. J Clin Oncol 2009; 27(15 Suppl.): Abstract 4623.
14. Kang SP, Saif MW. Optimal second line treatment options for
gemcitabine refractory advanced pancreatic cancer patients. Can we
establish standard of care with available data? JOP. J Pancreas
(Online) 2008; 9:83-90. [PMID 18326918]
15. Hwang JY, Yoo C, Kim T, Lee J, Park D, Seo D, et al. A
randomized phase II study of FOLFOX or FOLFIRI.3 as second-line
therapy in patients with advanced pancreatic cancer previously
treated with gemcitabine-based chemotherapy. J Clin Oncol 2009;
27(15 Suppl.): Abstract 4618.
16. Tang P, Gill S, Au HJ, Chen EX, Hedley D, Leroux M, et al.
Phase II trial of erlotinib in advanced pancreatic cancer (PC). J Clin
Oncol 2009; 27(15 Suppl.): Abstract 4609.
17. Van Cutsem E, Humblet Y, Gelderblom H, Vermorken JB, Vire
Ft, Glimelius B, et al. Cetuximab dose-escalation study in patients
with metastatic colorectal cancer (mCRC) with no or slight skin
reactions on cetuximab standard dose treatment (EVEREST):
Pharmacokinetic and efficacy data of a randomized study. 2007
ASCO Gastrointestinal Cancers Symposium. Abstract #237.
18. Kindler HL, Garbo L, Stephenson J, Wiezorek J, Sabin T, Hsu
M, et al. A phase Ib study to evaluate the safety and efficacy of AMG
655 in combination with gemcitabine (G) in patients (pts) with
metastatic pancreatic cancer (PC). J Clin Oncol 2009; 27(15 Suppl.):
Abstract 4501.
19. Von Hoff DD, Ramanathan R, Borad M, Laheru D, Smith L,
Wood T, et al. SPARC correlation with response to gemcitabine (G)
plus nab-paclitaxel (nab-P) in patients with advanced metastatic
pancreatic cancer: A phase I/II study. J Clin Oncol 2009; 27(15
Suppl.): Abstract 4525.
20. Loehr M, Bodoky G, Fölsch U, Märten A, Karrasch M, Lilla C,
et al. Cationic liposomal paclitaxel in combination with gemcitabine
in patients with advanced pancreatic cancer: A phase II trial. J Clin
Oncol 2009; 27(15 Suppl.): Abstract 4526.
21. Hammel P, Mornex F, Deplanque G, Mitry E, Levy P, Seitz J, et
al. Oral tyrosine kinase inhibitor masitinib in combination with
gemcitabine in patients with advanced pancreatic cancer: A
multicenter phase II study. J Clin Oncol 2009; 27(15 Suppl.):
Abstract 4617.
22. Fazal S, Saif MW. Supportive and palliative care of pancreatic
cancer. JOP. J Pancreas (Online) 2007; 8:240-53. [PMID 17356251]
23. Sohail MA, Saif MW. Role of anticoagulation in the
management of pancreatic cancer. JOP. J Pancreas (Online) 2009;
10:82-7. [PMID 19287098]
24. Riess H, Pelzer U, Deutschinoff G, Opitz B, Stauch M, Reitzig
P, et al. A prospective, randomized trial of chemotherapy with or
without the low molecular weight heparin (LMWH) enoxaparin in
patients (pts) with advanced pancreatic cancer (APC): Results of the
CONKO 004 trial. J Clin Oncol 2009; 27(18 Suppl.): Abstract
LBA4506.
25. Riess H, Pelzer U, Hilbig A, Stieler J, Opitz B, Scholten T, et al.
Rationale and design of PROSPECT-CONKO 004: a prospective,
randomized trial of simultaneous pancreatic cancer treatment with
enoxaparin and chemotherapy). BMC Cancer. 2008 Dec 5;8:361.
[PMID 19055847]
... Pancreatic cancer is the fourth leading cause of cancer death in the United States, and portends a poor prognosis, in part due to delays in diagnosis caused by the subtleties in presentation and the limited efficacy of therapeutic options [1]. This is especially true with advanced-stage cancer. ...
Article
Full-text available
Pancreatic cancer is the fourth leading cause of cancer deaths in the United States, and is considered uniformly fatal when patients present with unresectable, advanced-stage disease at the time of diagnosis. Long-term survival of patients with advanced-stage pancreatic adeno-carcinoma remains rare, despite advances in adjuvant chemoradiation protocols. A 73-year-old male presented to our emergency department with abdominal pain and a history of biopsy-proven, stage III pancre-atic adenocarcinoma. His initial staging CT scan and trans-duodenal ultrasound had demonstrated a stage IIa (T3, N0, Mx) lesion. On surgical exploration, he was up-staged to stage III (T4, N0, Mx), noting encasement of the superior mesenteric vessels and involvement of the portal vein. He underwent palliative choledochojejunostomy and was treated with 4 months of oxaliplatin and capecitabine, with concurrent radiation therapy (50.4 Gy), followed by 4 months of gemcitabine. After 7 months, the patient withdrew from therapy due to treatment intolerance. He then turned to self-medication with non-traditional herbal therapies. After 3 years of surveillance, he was lost to follow-up until presenting to our facility with abdominal pain 8 years after his initial diagnosis. On diagnostic CT scan during his current presentation for abdominal pain, he was found to have no evidence of pancreatic cancer. Based on our review of the literature, we present the longest known survival of a patient with surgically unresectable pancreatic adenocarcinoma. Further study of this patient's phenotypic or genotypic characteristics may provide insight into better therapeutic agents, or a predictive subset of patients who will benefit from specific chemotherapeutic options.
... In contrast, RTK+/MMP7+ or RTK-/MMP7+ tumors would be intrinsically resistant to combinations of EGFR and IGF- 1R inhibitors due to alternative IGF-1R-independent pathways regulated by MMP-7 [34] such as TRAIL [35, 36] or absence of target expression (RTK), respectively. It is remarkable that, considering our hypothesis-based of IGF- 1R resistant pattern, only 36% of our models are expected to respond to combinations of gemcitabine plus erlotinib and IGF-1R inhibitors combined therapies, in accordance with recent data in early phase II clinical trials in PDAC [13, 37]. An inherent limitation of our study is that this platform is derived from resected patients and then, only represents 20% of all patients with PDAC. ...
Article
Full-text available
Efforts to identify novel therapeutic options for human pancreatic ductal adenocarcinoma (PDAC) have failed to result in a clear improvement in patient survival to date. Pancreatic cancer requires efficient therapies that must be designed and assayed in preclinical models with improved predictor ability. Among the available preclinical models, the orthotopic approach fits with this expectation, but its use is still occasional. An in vivo platform of 11 orthotopic tumor xenografts has been generated by direct implantation of fresh surgical material. In addition, a frozen tumorgraft bank has been created, ensuring future model recovery and tumor tissue availability. Tissue microarray studies allow showing a high degree of original histology preservation and maintenance of protein expression patterns through passages. The models display stable growth kinetics and characteristic metastatic behavior. Moreover, the molecular diversity may facilitate the identification of tumor subtypes and comparison of drug responses that complement or confirm information obtained with other preclinical models. This panel represents a useful preclinical tool for testing new agents and treatment protocols and for further exploration of the biological basis of drug responses.
Chapter
Pancreas adenocarcinoma is now the 10th-leading cause of cancer in the United States but the fourth leading cause of cancer-related death. By 2030, it is expected to be the second-leading cause of cancer-related death in the United States after lung cancer. Understanding the genetics, molecular biology, and immunology of pancreas cancer has been key to making inroads in developing new therapies. This chapter highlights recent progress.
Article
Full-text available
Chemotherapy (CT) options in pancreatic cancer (PC) are limited to gemcitabine and 5-fluorouracil (5-FU). Several identified molecular targets in PC represent client proteins of HSP90. HSP90 is a promising target since it interferes with many oncogenic signaling pathways simultaneously. The aim of this study was to evaluate the efficacy of different HSP90 inhibitors in gemcitabine and 5-FU resistant PC. PC cell lines 5061, 5072 and 5156 were isolated and brought in to culture from patients being operated at our institution. L3.6pl cell line served as a control. Anti-proliferative efficacy of three different HSP90 inhibitors (17-AAG, 17-DMAG and 17-AEPGA) was evaluated by the MTT assay. Alterations in signaling pathway effectors and apoptosis upon HSP90 inhibition were determined by western blot analysis and annexin V/PI staining. The cell lines 5061, 5072 and 5156 were resistant to gemcitabine and 5-FU. In contrast 17-AAG and the water-soluble derivates 17-DMAG and 17-AEPGA displayed high anti-proliferative activity in all tested cell lines. The calculated IC50 was below 1 µM. Highly significant down regulation of epidermal-growth-factor-receptor, insulin-like-growth-factor-receptor-1, AKT and MAPK reflected the intracellular molecular signaling-network disruption. Furthermore, besides HSP70 also HSP27 was upregulated in all cell lines. Apoptosis occurred early under HSP90 inhibition and was determined by annexin V/PI staining and CASPASE-3 and PARP assay. In contrast, gemcitabine treated cells did not show any apoptosis. HSP90 inhibition disrupts multiple signaling cascades in gemcitabine and 5-FU resistant PC simultaneously and promotes cancer cell apoptosis. Watersoluble 17-DMAG is equally effective as 17-AAG. HSP27, besides HSP70, may represent an effective response marker of successful HSP90 inhibition.
Article
Background: There are indications that pancreatic cancer survival may differ according to sociodemographic factors, such as residential location. This may be due to differential access to curative resection. Understanding factors associated with the decision to offer a resection might enable strategies to increase the proportion of patients undergoing potentially curative surgery. Methods: Data were extracted from medical records and cancer registries for patients diagnosed with pancreatic cancer between July 2009 and June 2011, living in one of two Australian states. Among patients clinically staged with non-metastatic disease we examined factors associated with survival using Cox proportional hazards models. To investigate survival differences we examined determinants of: 1) attempted surgical resection overall; 2) whether patients with locally advanced disease were classified as having resectable disease; and 3) attempted resection among those considered resectable. Results: Data were collected for 786 eligible patients. Disease was considered locally advanced for 561 (71%) patients, 510 (65%) were classified as having potentially resectable disease and 365 (72%) of these had an attempted resection. Along with age, comorbidities and tumour stage, increasing remoteness of residence was associated with poorer survival. Remoteness of residence and review by a hepatobiliary surgeon were factors influencing the decision to offer surgery. Conclusions: This study indicated disparity in survival dependent on patients' residential location and access to a specialist hepatobiliary surgeon. Accurate clinical staging is a critical element in assessing surgical resectability and it is therefore crucial that all patients have access to specialised clinical services.
Article
Full-text available
Pancreatic cancer continues to be the fourth leading cause of death despite advancements in surgical and adjuvant therapeutic approaches. In the present review, the current cytotoxic therapeutic approaches and advanced targeted therapies are objectively discussed with consideration to the current literature.
Article
Purpose: The taurine derivative taurolidine (TRD) exerts anti-neoplastic effects in a variety of tumor models. On the other hand, TRD at low doses was shown to reduce cell-cell adhesion, a prerequisite for metastasis. The aim of this study was to elucidate the effects of low-dose TRD on pancreatic cancer. Methods: Human pancreatic cancer cell lines representing diverse states of differentiation were exposed to TRD for 24 h. Cell viability was assessed by MTT assay and trypan blue staining, apoptosis by caspase-3/7 activity, and flow-cytometric cell cycle analysis. Expression of Snail and E-cadherin was analyzed by polymerase chain reaction and Western blotting. Results: MTT-tested viability of all pancreatic cancer cell lines decreased dose-dependently up to 50 % of the untreated control. In contrast to staurosporine TRD (100 and 250 μM) did not induce apoptosis but increased the percentage of cells in G1/G0 arrest. Correlation of MTT test and trypan blue staining revealed a decreased adherence of vital tumor cells at 250 μM TRD. This was associated with reduced expression of the adhesion molecule E-cadherin and an increased expression of the transcription factor Snail, a regulator of epithelial-mesenchymal transition (EMT). Conclusion: Low-dose TRD reduces not only viability but also cell-cell adherence and E-cadherin expression of pancreatic cancer cells, whereas the expression of the EMT inducer Snail was increased. By induction of these EMT hallmarks, low-dose TRD may promote metastasis in pancreatic cancer.
Article
Pancreatic cancer is a lethal disease mostly affecting elderly people. Clinical trials on treatment contain disproportionately fewer elderly patients compared with everyday practice. This retrospective study evaluates differences in the rates of chemotherapy delivered and associated survival in different age groups. Data were collected from the Cancer Information Resource Files on patients diagnosed with pancreatic cancer from 1993 to 2008. Patients were divided into 3 age groups, namely, A with younger than 50 years, B with 50 to 70 years old, and C with older than 70 years. Complete data were available on 16,694 patients. Forty-four percent were in group C.Chemotherapy was given to 38% of patients in group C versus 69% of patients in group A. A multivariate analysis revealed a similar chemotherapy benefit in all groups, as follows: group C (hazard ratio [HR], 0.51), group A (HR, 0.74), and group B (HR, 0.55). We found that elderly patients with pancreatic cancer receive treatment less frequently compared with younger patients. However, elderly patients receiving chemotherapy derive similar benefits. Randomized clinical trials are needed to evaluate pancreatic cancer treatment in the elderly patients, particularly given the increasing occurrence of pancreatic cancer in later life.
Article
4501 Background: AMG 655 is an investigational, fully human agonist monoclonal antibody (IgG1) that binds human death receptor 5 (DR5), activates caspases, and induces apoptosis in sensitive tumor cells. In preclinical PC models, cooperative activity is observed when G is added to AMG 655. We performed a multi-center phase I trial to evaluate AMG 655 + G in metastatic PC pts. The primary endpoint was dose-limiting toxicity (DLT). Secondary endpoints included toxicity, pharmacokinetics, antibody formation, objective response rate, progression-free survival (PFS), 6-month and overall survival. Methods: Eligible pts had previously untreated metastatic PC and ECOG PS 0 or 1. Pts enrolled into sequential cohorts and received AMG 655 3 or 10 mg/kg IV days (D) 1 and 15 and G 1000 mg/m ² IV D 1, 8, and 15 every 28 D. CT scans were obtained Q8 weeks. Results: 13 pts (3 mg/kg cohort = 6; 10 mg/kg cohort = 7) enrolled from 7/07–11/07. Pt characteristics: females 61%; ECOG PS 1 69%; median age 65 (range 35–81); liver metastases 77%. Median number of cycles: 6 (range 2–12). There were no DLT. Nine (69%) pts had grade 3–4 toxicity, the most common were: thrombocytopenia (4 pts), neutropenia (2 pts), and abdominal pain (2 pts). No anti-AMG 655 antibodies were detected. After one 3 or 10 mg/kg dose of AMG 655 after G, the C max and AUC of AMG 655 were similar to those in the first- in-human single-agent study (LoRusso JCO 2007; 25: abstract 3534). Preliminary data indicate no effect of AMG 655 on PK of G. Partial response 31% (4 pts, 2 unconfirmed); stable disease 38%. Median PFS: 5.3 months (95% CI, 3.5, 6.2); 6-month survival rate: 76.2% (95% CI: 42.7%-91.7%). Conclusions: AMG 655 + G is well-tolerated and may have activity in metastatic pancreatic cancer. A randomized phase II trial of G ± AMG 655 at 10-mg/kg is currently enrolling. [Table: see text]
Article
4504 Background: Single-agent gemcitabine (G) remains standard treatment for advanced pancreatic adenocarcinoma (APC). The GIP-1 randomized phase III trial (clinicaltrials.gov ID NCT00813696 ) was performed to compare the combination of cisplatin (P) and G vs. G alone as 1st-line treatment. Methods: Patients (pts) with locally advanced and/or metastatic pancreatic adenocarcinoma, age 18–75, Karnofsky Performance Status (KPS) ≥50, were randomized to receive G (arm A) or G+P (arm B). In arm A, G was administered at 1000 mg/m2 weekly for 7 consecutive wks, and, after a 2-week rest, on day 1, 8, 15 every 4 wks. In Arm B, P 25 mg/m2 weekly (with the exception of day 22) was added to G, same dose used in Arm A (Colucci et al, Cancer 2002; 94:902–10). No maximum number of cycles was planned. Primary endpoint was overall survival (OS). Clinical benefit (CB), objective response rate (ORR), progression-free survival (PFS), toxicity and quality of life were secondary endpoints. To have 80% power of detecting a 0.74 Hazard Ratio (HR) of death (corresponding to increase in median OS from 4.8 to 6.5 months, with bilateral alpha=0.05, 400 pts were planned and 355 deaths were required for final analysis. Results: From April 2002 to April 2007, 400 pts were enrolled (A:199, B; 201) in 46 Italian Institutions. Median age was 63 yrs (range 35–75), 59% were males, 84% stage IV, 83% KPS≥80. After a median follow-up of 38.2 months and 357 deaths, median OS was 8.3 vs 7.2 months in arm A and B, respectively (HR 1.10, 95% CI 0.89–1.35, p=0.38). Median PFS was 3.9 vs 3.8 months in arm A and B, respectively (HR 0.97, 95% CI 0.80–1.19, p=0.80). ORR was 10.1% in arm A and 12.9% in B (p=0.37). CB response was experienced by 23.0% and 15.1% (Arm A vs B, p=0.057). Patients assigned to combination arm experienced more anaemia (all grades: 50% vs 39%, G3: 5% vs 1%), more neutropenia (all grades: 44% vs 36%, G3&4: 25% vs 14%) and more thrombocytopenia (all grades: 57% vs 29%, G3&4: 16% vs 5%). No relevant differences were seen in non-haematological toxicity. Conclusions: Weekly combination of P and G, compared to single-agent G as 1st-line treatment of APC, failed to demonstrate any improvement in OS, PFS, ORR and clinical benefit. No significant financial relationships to disclose.
Article
4618 Background: Only few clinical trials have been conducted in patients with advanced pancreatic cancer after failure of first-line gemcitabine-based chemotherapy. We conducted a randomized phase II trial of modified FOLFOX vs. modified FOLFIRI.3 as second-line regimen for the patients with gemcitabine refractory pancreatic cancer ( NCT00786006 ). Methods: Patients with advanced pancreatic adenocarcinoma previously treated with gemcitabine were randomly assigned to FOLFOX or FOLFIRI.3 stratifying by age (≤ 65 vs. >65), performance status (0–1 vs. 2) and prior response to gemcitabine (PR/SD vs. PD). FOlFIRI.3 regimen consisted of Irinotecan 70 mg/m ² (over 60 min) D1, leucovorin 400 mg/m ² (over 2h) D1, 5-FU 2000 mg/m ² (over 46 hours) from D1, then irinotecan 70 mg/m ² (over 60 min) at the end of the 5-FU infusion every two week. FOLFOX regimen is composed of oxaliplatin 85 mg/m ² (over 120 min) D1, LV 400 mg/m ² D1, 5-FU 2,000 mg/m ² (over 46 hours) every two week. The primary end-point was 6-month overall survival (P 0 =20%) and Simon-Wittes-Ellenberg design was used to calculate the sample size (29 evaluable patients for each treatment arm). Results: From January 2007 to December 2008, sixty patients were enrolled and randomized to FOLFOX (N=30) or FOLFIRI.3 (N=30). Baseline characteristics were well balanced between each arm; median age 56 (35–60) vs. 56 yo (37–73); ECOG PS 0/1/2, 5/24/1 vs. 5/25/0; prior response to gemcitabine-based chemotherapy PR/SD/PD 10/13/7 vs. 10/11/9. With a median follow-up period of 6.0 months (95% CI, 4.7–7.3) the median overall survival was 4.0 months in both group (HR=0.95, 95% CI 0.52–1.75) with 6-month survival rates of 25% and 20%, respectively. The median PFS was 1.4 months for FOLFOX and 1.9 months for FOLFIRI.3 (HR=1.11, 95% CI, 0.64–1.92). Disease control (PR+SD) was achieved in 20% (5/25 in FOLFOX) and 28% (7/25 in FOLFIRI.3) of patients with measurable disease. The incidences of grade 3/4 toxicities were similar in both groups. Conclusions: Both FOLFOX and FOLFIRI.3 were tolerated with manageable toxicity, offering modest activity as second-line treatment of patients with advanced or metastatic pancreatic cancer, previously treated with gemcitabine. No significant financial relationships to disclose.
Article
4609 Background: The epidermal growth factor receptor (EGFR) is a potentially important target in PC. Benefit from erlotinib (Tarceva), an oral EGFR tyrosine kinase inhibitor has been associated with the presence of a skin rash. The purpose of this study was to determine the efficacy of erlotinib, dosed to achieve a rash, in patients (pts) with PC. Methods: Erlotinib was given at an initial dose of 150 mg/day to eligible pts with locally advanced (LA) or metastatic PC who had progressed or were unable to tolerate gemcitabine-based chemotherapy. The dose of erlotinib was increased by 50mg every 2 weeks (maximum 300 mg/day) until > grade 1 rash (CTCAE v 3.0) or other dose-limiting toxicities occurred. Erlotinib pharmacokinetic (PK) studies were performed. Baseline tumor tissue was collected for analysis of Kras mutations, EGFR by IHC and FISH. The primary endpoint of this two- stage phase II trial was prolonged disease control (PR + SD > 8 wks) with a rate of >20% assumed to be significant . Results: Fifty pts were accrued (median age 61, M:F = 25:25, ECOG 0:1:2 = 5:41:4, LA:Metastatic = 5:45, prior gemcitabine none:adjuvant:palliative = 2:16:35). 47 and 40 pts were evaluable for toxicity and response, respectively. Dose-escalation to 200–300 mg of erlotinib was possible in 9 pts. Most common treatment adverse events (TAEs) of any grade were: rash (35 pts, 74.5%), diarrhea (18 pts, 38.3 %), and fatigue (8 pts, 17%). Grade 3+ TAEs were rash in 2 pts and diarrhea in 2 pts. Best response was SD in 14 pts, 0.35 (95% CI: 0.2–0.5). Prolonged disease control (SD > 8 wks) was observed in 10/40 evaluable pts, 0.25 (95% CI: 0.12–0.38), which met the primary study endpoint. Median TTP was 1.6 mo (95% CI:1.6–2.1), mOS 4.1 mo (95% CI:3.2–7.3), and 6 mo OS rate was 39% (95%CI: 24–61%). PK and correlative data are being analyzed and will be presented. Conclusions: Erlotinib is associated with prolonged stable disease in a subset of pts with advanced refractory PC. Dose escalation in the absence of toxicity is feasible and safe. [Table: see text]
Article
4525 Background: Pancreatic cancer cells and surrounding stroma are known to overexpress SPARC (secreted protein acid rich in cysteine), which is associated with poor clinical outcomes. nab-P, an albumin-bound nanoparticle form of paclitaxel increased tumor accumulation of paclitaxel through binding of albumin to SPARC. This disease specific phase 1 study was designed to evaluate the safety and efficacy of G + nab-P and the correlation of response with tumor SPARC and serum CA19–9 levels. Methods: nab-P doses (100–150 mg/m ² ) + (G) (1000 mg/m ² ) were given on days 1, 8, and 15 of a 28-day cycle to pts with metastatic pancreatic adenocarcinoma and with no prior chemotherapy for their metastatic disease. Level 3 SPARC staining by immunohistochemistry was considered positive. Results: 63 pts received treatment. The most common grade 3 and 4 adverse event that occurred in >20% of pts was neutropenia. Nine (18%) pts and 4 (8%) pts had a grade 3/4 event, respectively. Neuropathy was also observed. One combination-associated death due to sepsis occurred at the 150 mg/m ² nab-P level. Serial PET scans of 53 pts with outside adjudication to date showed 12 (23%) complete responses, 29 (55%) partial responses (PRs) and 4 (8%) stable disease (SD). By RECIST criteria, of the 49 pts evaluable to date, 1(2%) had CR, 12 (24%) had PR, and 20 (41%) had SD. The median survival was 9 months to date. SPARC data were available for 35 pts, of which 10 (29%) were SPARC+ and 25 (71%) were SPARC-. Of these, 27 pts had evaluable response data. Pts that were SPARC+ (8/27) were more likely to be responders (6/8, 75%) than pts who were SPARC- (5/19, 26%), P = 0.03, Fisher's exact test. Median progression-free survival (PFS) increased from 4.8 months for SPARC- pts (22 pts) to 6.2 months for SPARC+ pts (9 pts); however, these data are still immature. Of 45 pts with elevated CA19–9 at baseline, 42 (93%) had maximum decrease of >40% with a median maximum decrease of 92%. Conclusions: The combination of nab-P and G was generally well tolerated and had substantial enough antitumor activity in patients with pancreatic cancer to warrant a phase III clinical trial. SPARC+ status in these patients was associated with higher response rate and longer PFS. [Table: see text]
Article
4623 Background: Preclinical studies suggest synergy between gemcitabine (G), docetaxel (T) and capecitabine (X). This GTX regimen was designed to inhibit MEK-ERK phosphorylation and increase BAX and BAK and also decrease BCL-2 in pancreatic cancer cell lines. Based on these findings, we pursued a prospective clinical trial evaluating the activity of GTX in previously untreated patients with metastatic pancreatic cancer. Methods: Patients with histologically confirmed metastatic adenocarcinoma of the pancreas, median age 60, 63% male, ECOG PS 0–2, received capecitabine 1500mg/m2/day total orally in divided doses, days 1 thru 14, gemcitabine 750mg/m2 IV over 75 minutes followed by docetaxel 30mg/m2 IV on days 4 and 11 on a 21 day cycle. Scans were completed every 9 to 10 weeks to assess for tumor response by RECIST criteria. Treatment was continued until evidence of disease progression, intolerable toxicity, surgery or a delay in treatment greater than 6 weeks. The primary endpoint was response rate. Secondary endpoints were overall survival (OS) measured as time from start of GTX to death, time to treatment failure (TTF) measured as time from start of GTX to disease progression or other reason for a halt in therapy. Results: Forty-three patients were enrolled at two centers between May 2004 and January 2007. Forty-one patients were eligible for assessment by intent to treat analysis. 35 patients (85%) had liver metastases. 9 patients (21.9%) had partial responses and 17 patients (41.5%) had stable disease. The one year survival rate was 56%. Two year survival rate was 14.6%. The median OS was 14.5 months. The median TTF was 6.9 months. Grade 3 and 4 toxicities included leukopenia (31.6%), neutropenia (29.2%), thrombocytopenia (12.2%), infection (12.5%), and mucositis (7.5%). Conclusions: The combination of gemcitabine, docetaxel, and capecitabine has activity in metastatic pancreatic cancer with a median survival over 1 year. A randomized phase III trial is in planning. [Table: see text]
Article
4526 Background: EndoTAG-1 is a novel cationic liposomal formulation of paclitaxel being developed for the treatment of solid malignancies. It acts by targeting activated negatively charged endothelial cells of tumor blood vessels. We present safety and efficacy data of a randomized, controlled phase II trial in pancreatic cancer (PC). Methods: 200 patients with advanced PC were randomized to 1 st line treatment with weekly gemcitabine (GEM: 1000 mg/m ² ) and twice weekly infusions of EndoTAG-1 (E) at 3 different dose levels (E low : 11 mg/m ² , E med : 22 mg/m ² , E high : 44 mg/m ² ) or GEM monotherapy. Patients were treated for 7 weeks and followed up for overall survival (OS) for at least 1 year. After finishing study treatment, any anti-tumor therapy was allowed. A subgroup of patients had the option to receive repeated cycles of combination therapy in case of at least stable disease according to RECIST until disease progression. Results: Median OS was substantially higher in the GEM+E med and GEM+ E high groups than the GEM monotherapy group. Adjusted hazard ratios for OS were 0.72 (95% CI 0.46–1.13) and 0.67 (0.43–1.07). In patients receiving >1 treatment cycle, median OS was 11.5 months (GEM+E high ); in the GEM+E med group 75% of patients were alive at 1 year. Treatment with EndoTAG-1 and gemcitabine was generally well tolerated. A trend for increasing adverse event frequency with EndoTAG-1 dose was observed for infusion-related reactions associated with chills and pyrexia, and thrombocytopenia. The overall frequency of serious adverse events in the GEM+E groups was low, the most frequent SAE being pyrexia in 4 (8%) patients in the GEM+E high group. There was no indication for significant organ toxicity associated with EndoTAG-1, even in patients receiving multiple treatment cycles. Conclusions: This phase II trial indicates a considerable survival benefit for patients with advanced PC receiving EndoTAG-1 in combination with gemcitabine and a favourable safety profile warranting further development of EndoTAG-1 in this indication. [Table: see text]
Article
4617 Background: Masitinib, a tyrosine kinase inhibitor targeting c-Kit, PDGFR, FGFR3 and affecting the FAK pathway, can enhance the antiproliferative effects of gemcitabine (GEM) in human pancreatic cancer cells. This multicenter phase 2 study aimed to determine the efficacy and safety of masitinib in combination with GEM in the first-line treatment of patients with locally advanced (LAPC) or metastatic (MPC) pancreatic cancer. Methods: Patients received oral masitinib (9 mg/kg/d) and standard weekly infusion of GEM (1,000 mg/m2). Primary endpoint was time-to-progression (TTP). Our hypothesis for efficacy was a TTP over 2.1 months. Secondary endpoints included survival rates, tumor response (RECIST) and clinical benefit. Results: 22 patients, with LAPC (n=9) or MPC (n=13), KPS[80–100]/[70] (18/4) were enrolled and treated with masitinib plus GEM. Median TTP was 6.4 months, well beyond our threshold for efficacy (LAPC: 8.3 months, MPC: 2.7 months, KPS[80–100]: 6.4 months, KPS[70]: 0.8 months). At 12 months, 17% of LAPC and 14% of KPS[80–100] were progression-free; all MPC and KPS[70] patients had progressed. The disease control rate was 73% (LAPC: 89%, MPC: 62%, KPS[80–100]: 89%; KPS[70] patients progressed immediately). Median OS was 7.1 months (LAPC: 8.4 months, MPC: 6.8 months, KPS[80–100]: 8.0 months, KPS[70]: 4.4 months). At 18 months, the survival rate was 23%. However, when considering KPS[80–100] alone, it reached 28%. The 18-months survival rates were similar for LAPC (22%) and MPC (23%). 16% of the 19 patients evaluated experienced clinical benefit (LAPC: 38%, KPS[80–100]: 18%). One patient (5%) presented suspected grade 4 neutropenia. Main suspected grade 3 toxicity were anemia, lymphopenia (23%), leucopenia, neutropenia (18%), asthenia (14%), diarrhea, cytolytic hepatitis, and skin rash (9%). Altogether, the combination masitinib plus GEM did not seem to increase the toxicity commonly reported with GEM alone. Conclusions: The antitumor activity of the combination masitinib plus GEM is very promising and does not present limiting toxicities. Based on those encouraging data, a randomized phase III trial comparing masitinib plus GEM with GEM alone is now actively recruiting patients in the US and in Europe. [Table: see text]
Article
4037 Background: Response to epidermal growth factor receptor (EGFR) inhibitor, cetuximab appears to correlate with the intensity of the associated skin reaction. This randomized study investigated cetuximab dose-escalation in patients (pts) with EGFR-expressing mCRC failing irinotecan-including therapy (I). Methods: Pts were randomized 22 days after starting cetuximab (400 mg/m ² initial dose then 250 mg/m ² /week [w]) with I (180 mg/m ² q 2 w) if they had not experienced >grade (G) 1 skin reaction, any other >G 2 cetuximab-related adverse event and were tolerant to I. Randomization was to standard cetuximab dose (Arm A; 250 mg/m ² /w) or dose-escalation (Arm B; cetuximab dose increased by 50 mg/m ² q 2 w, until >G 2 toxicity, tumor response or dose = 500 mg/m ² ). Pts not randomized (Arm C) continued on standard cetuximab dose. Primary endpoint was to compare in skin and tumor biopsies, taken before and during treatment, the effects of dose-escalation on EGFR and downstream signalling markers with those of the standard cetuximab regimen. Secondary endpoints were PK, efficacy, safety, tolerability, biomarker analyses on tumor biopsies and plasma samples. Results: 284 pts screened, 221 pts EGFR-expressing, 166 pts enrolled and randomized: 45 to Arm A; 44 to Arm B. 77 non-randomized pts were included in Arm C. M/F 106/60, median age 60 years [25–79], median KPS 90 [70–100]. 24 pts in Arm B reached 500 mg/m ² /w. By the cut-off date of July 31 st 2006 the preliminary response rate (RR) in Arm B was 30% vs. 13% in Arm A. RR in arm C was 22%. Preliminary progression-free survival in Arm A was 3,9 mo vs. 4,8 mo in Arm B and 3,9 mo in Arm C. In Arm B, 9% pts had G 3/4 skin reactions, 0% in Arm A (14% in Arm C). Dose related increases in C max and AUC were observed. T 1/2 values were dose independent. Conclusions: Cetuximab dose- escalation up to 500 mg/m ² /w improves RR in pts with no or slight skin reactions on standard dose treatment. Overall cetuximab PK behavior is in good agreement with previous experience. Treatment was generally well tolerated. PD and biomarker results will be presented at the meeting. No significant financial relationships to disclose.