ArticlePDF Available

Efficacy and safety of gemcitabine-capecitabine combination therapy for pancreatic cancer

Authors:

Abstract and Figures

Background Recent randomized controlled trials revealed the combination of gemcitabine and capecitabine (GemCap) regime shows promising efficacy in pancreatic cancer patients. Here, we conducted a meta-analysis to compare the efficacy and safety of gemcitabine (Gem) with GemCap for pancreatic cancer. Methods The database of MEDLINE (PubMed), EMBASE, Cochrane Central Controster of Controlled Trials, Web of Science was searched for relevant randomized controlled trials before 8 April, 2020. The outcomes were overall survival (OS), 12-month survival rate, progress free survival (PFS), partial response rate (PRR), objective response rate (ORR), and Grade 3/4 toxicities. Results Five randomized controlled trials involving 1879 patients were included in this study. The results showed that GemCap significantly improves the OS (hazard ratio = 1.15, 95% CI: 1.037-1.276, P = .008), PFS (hazard ratio = 1.211, 95% CI 1.09-1.344, P = 0), PRR (relative risk (RR) = 0.649, 95% CI 0.488-0.862, P = .003), ORR (RR = 0.605, 95% CI 0.458-0.799, P = 0), and the overall toxicity (RR = 0.708, 95% CI 0.620-0.808, P = .000) compared to Gem alone. However, no significant difference was found in 12-month survival. Conclusions Despite a higher incidence of Grade 3/4 toxicity, GemCap was associated with better outcomes of OS, PFS, PRR, ORR, as compared with Gem, which is likely to become a promising therapy for pancreatic cancer.
Content may be subject to copyright.
Efcacy and safety of gemcitabine-capecitabine
combination therapy for pancreatic cancer
A systematic review and meta-analysis of randomized controlled
trials
Guoqing Ouyang, MD, Yongrong Wu, MD, Zhen Liu, MD, Wuchang Lu, MD, Shuai Li, MD,
Shuqing Hao, MD, Guangdong Pan, MD
Abstract
Background: Recent randomized controlled trials revealed the combination of gemcitabine and capecitabine (GemCap) regime
shows promising efcacy in pancreatic cancer patients. Here, we conducted a meta-analysis to compare the efcacy and safety of
gemcitabine (Gem) with GemCap for pancreatic cancer.
Methods: The database of MEDLINE (PubMed), EMBASE, Cochrane Central Controster of Controlled Trials, Web of Science was
searched for relevant randomized controlled trials before 8 April, 2020. The outcomes were overall survival (OS), 12-month survival
rate, progress free survival (PFS), partial response rate (PRR), objective response rate (ORR), and Grade 3/4 toxicities.
Results: Five randomized controlled trials involving 1879 patients were included in this study. The results showed that GemCap
signicantly improves the OS (hazard ratio =1.15, 95% CI: 1.037-1.276, P=.008), PFS (hazard ratio =1.211, 95% CI 1.09-1.344,
P=0), PRR (relative risk (RR) =0.649, 95% CI 0.488-0.862, P=.003), ORR (RR =0.605, 95% CI 0.458-0.799, P=0), and the overall
toxicity (RR =0.708, 95% CI 0.620-0.808, P=.000) compared to Gem alone. However, no signicant difference was found in 12-
month survival.
Conclusions: Despite a higher incidence of Grade 3/4 toxicity, GemCap was associated with better outcomes of OS, PFS, PRR,
ORR, as compared with Gem, which is likely to become a promising therapy for pancreatic cancer.
Abbreviations: 5-FU =5-uorouracil, Gem =gemcitabine, GemCap =gemcitabine and capecitabine, HR =hazard ratio, ORR =
objective response rate, OS =overall survival, PFS =progress free survival, PRR =partial response rate, RCTs =recent randomized
controlled trials, RR =relative risk.
Keywords: capecitabine, gemcitabine, meta-analysis, pancreatic cancer
1. Introduction
Pancreatic cancer is the fourth leading cause of cancer-related
death in the world,
[1]
and the incidence of pancreatic cancer has
been increasing rapidly in recent years. Almost 96% of pancreatic
cancer was constituted with pancreatic ducal adenocarcinoma.
[2]
It is expected that pancreatic cancer will become the 2
nd
leading
cause of cancer-related death by 2030.
[3]
The prognosis of
pancreatic cancer was extremely poor with a 1-year survival rate
was around 18% and 5-year survival rate was less than 8%.
[46]
Surgical resection remains the only treatment that can achieve
long-term survival. Because the majority of patients are
diagnosed with local advanced or metastatic, only 15% to
20% of patients are amenable to surgical resection.
[7,8]
Therefore, chemotherapy becomes the rst-line treatment for
advanced and metastatic pancreatic cancer.
Gemcitabine (Gem) was recommended as the gold
standard treatment for pancreatic cancer due to a Phase III
clinical trial that found Gem achieved a better survival rate and
more clinical benets than 5-uorouracil (5-FU). Gem has
acquired about a 20% increase in 1-year survival rate and
a median overall survival of 5 to 7 months in metastatic
pancreatic cancers.
[9]
However, the therapeutic effect of Gem
monopoly is still not satisfactory,
[9,10]
it calls for the need of
better treatment strategies for pancreatic cancer. To improve
clinical benet of pancreatic cancer, various anti-tumor agents
combined with Gem were recently attempted in numerous
clinical setti ng, such as Nab-paclitaxel and FOL FIRINOX (5-FU,
Oxaliplatin, Irinotecan, folinic acid), cisplatin, oxaliplatin,
docetaxel et al, but most studies cannot show a signicant
improvement in overall survival (OS) or a better tolerance of
Editor: Maya Saranathan.
This study was supported by the Hubei Chen Xiaoping Technology Development
Fund (CXPJJH1190000-2019321).
The authors have no conicts of interest to disclose.
The datasets generated during and/or analyzed during the current study are
publicly available.
Department of Hepatobiliary Surgery, Liuzhou Peoples Hospital, Liuzhou,
Guangxi, China.
Correspondence: Guangdong Pan, Department of Hepatobiliary Surgery,
Liuzhou Peoples Hospital, Liuzhou, Guangxi 545006, China
(e-mail: pgdhx@126.com).
Copyright ©2021 the Author(s). Published by Wolters Kluwer Health, Inc.
This is an open access article distributed under the terms of the Creative
Commons Attribution-Non Commercial License 4.0 (CCBY-NC), where it is
permissible to download, share, remix, transform, and buildup the work provided
it is properly cited. The work cannot be used commercially without permission
from the journal.
How to cite this article: Ouyang G, Wu Y, Liu Z, Lu W, Li S, Hao S, Pan G.
Efcacy and safety of gemcitabine-capecitabine combination therapy for
pancreatic cancer: a systematic review and meta-analysis of randomized
controlled trials. Medicine 2021;100:48(e27870).
Received: 28 March 2021 / Received in nal form: 23 September 2021 /
Accepted: 3 November 2021
http://dx.doi.org/10.1097/MD.0000000000027870
Systematic Review and Meta-Analysis Medicine®
OPEN
1
toxicities than Gem monopoly, except for combing capecitabine
or erlotinib.
[1114]
Capecitabine is a designed oral uoropyrimidine carbamate
that is associated with a lower incidence of toxicities and achieved
similar efcacy compared with intravenous 5-FU.
[15]
The
convenience of oral administration makes capecitabine widely
used in various tumors. Capecitabine is currently approved by the
FDA for the treatment of breast cancer and colorectal cancer.
[16]
Capecitabine monopoly has been found to be safe and effective in
advanced pancreatic cancer in a phase II study and demonstrated
similar benets in tumor-related symptoms with Gem.
[9,17]
Though capecitabine led to a modest clinical benet, it failed
to improve the terrible prognosis. To obtain better clinical
benets, various capecitabine-based schemes have been
attempted in multiple clinical research.
Gemcitabine and capecitabine (GemCap) are both nucleoside
analogs and the combination show synergism to antitumor.
Furthermore, both drugs have nonoverlapped toxicity and are
well tolerated.
[16,18,19]
Previous studies have shown Gem
combined capecitabine improved overall survival rate, progres-
sion-free survival (PFS) and achieved a better tumor response
than Gem alone.
[11,16,1921]
However, some clinical benet in the
5 studies are quite contrary. In this study, we conducted this
systematic review and meta-analysis of the currently available
randomized controlled studies to evaluate the efcacy and safety
of the gemcitabine-capecitabine (Gem-Cap) and Gem alone for
the patients of pancreatic cancer.
2. Methods
2.1. Search strategy
From inception to 8 April, 2020, comprehensive electronic
searches were performed with the database of MEDLINE
(PubMed), EMBASE, Cochrane Central Register of Controlled
Trials, Web of Science. The search terms and strategy are based
on the combination of the following keyword: (pancreatic
cancer) AND (gemcitabineor Gemzar) AND (capecita-
bineor Xeloda) AND (randomized controlled trial). The
searching language was restricted to English. This meta-analysis
was conducted following the guidelines provided by the PRISMA
statement.
2.2. Inclusion and exclusion criteria
Trials, were included in this meta-analysis should fulll the
following criteria: The study was randomized controlled trials
(RCTs; Phase II or III) with Gem and GemCap treatment.
Cytologically or histologically ascertained pancreatic cancer; Age
was between 18 and 85years; Karnofsky performance status
score 50% (or WHO Health Organization performance status
2), adequate bone marrow, hepatic, and renal functions; No
previous chemotherapy or radiotherapy.
The exclusions are as follows: Studies were not RCTs such as
reviews and case reports, full text unavailable and non-published
conference was also excluded. For Duplicate publications, the
most comprehensive article was selected. Single-arm studies.
2.3. Data extraction
The data extracted and quality assessment was performed
independently by 2 authors (i.e., GQOY and WC L). Any
disagreement between 2 authors was resolved after discussing
with a third reviewer (YRW) and a consensus was achieved. The
following data were extracted: The rst authors name, year of
publication, gender distribution, study design, treatment group,
number of patients, patient characteristics. The outcome of
treatment, such as CRR, objective response rate (ORR), partial
response rate (PRR), OS, PFS, toxicities. If the same trail reported
in different publications, the most recent publication or
comprehensive one was chosen. If log hazard ratio (HR) and
its variance of overall survival or forgeprogression-free survival
was not provided directly in the text but only in gures, Engauge
Digitizer version 4.1 (http://digitizer.sourceforge.net/) was used
to read the Kaplan-Meier curves.
2.4. Quality assessment
The Jadad score was used to assess the quality of the included
RCTs. The 3 items of Jadad are as follows: randomization,
double blinding, withdrew wals and dropouts. Jadad score
ranged from 0 to 5, and 3 was considered high-quality
literature.
[22]
2.5. Statistical analysis
All analyses were performed with Stata version 12.0 software
(Stata Corporation, College Station, TX) according to the
Cochrane Handbook for Systematic Review. The primary
endpoint of this meta-analysis was included OS and PFS. OS
was dened as the time between date of random assignment and
the date of death from any cause. PFS was dened as the time by
random assignment to disease progression. The second endpoints
included 12months survival rate, ORR, PRR, CRR, Grade 3-4
toxicities. The HRs with 95% CIs were used to express the results
of OS and PFS. The pooled odds ratio with 95% CI was
calculated for the second point. The x
2
-based Q-test and I
2
statistics were used to assess the heterogeneity. If there were
statistical differences in terms of heterogeneity (I
2
>50%,
P<.10), a random effects model was used
[23]
; otherwise, a xed
effects model was selected. A Pvalue less than .05 was considered
statistically signicant. The possibility of publication bias was
ascertained by visually funnels plots.
2.6. Ethical statement
The data analyzed in this study was extracted from previously
published studies, and therefore ethical approval was not
necessary.
3. Results
3.1. Literature search
After searching literature within several databases, a total of
1382 potentially relevant studies was eventually identied for
screening (Fig. 1). After duplicating and screening the titles and
abstracts, 15 studies were enrolled for full-text screening. Of the
15 remaining studies, 8 were excluded because they were non-
randomize trials and 2 were excluded because they were single-
arm control study. Finally, 5 RCTs compared Gem alone with
GemCap in pancreatic cancer were included in this meta-
analysis.
[11,16,1921]
Ouyang et al. Medicine (2021) 100:48 Medicine
2
3.2. Study characteristics
The baseline characteristic of the enrolled studies is summarized
in Table 1. Five studies including a total of 1879 patients with
pancreatic cancer were included in the nal analysis, 939 patients
were assigned to the Gem group and 940 to the GemCap group.
All included trials were considered high qualities, with a score of
3 in Jadad score. Of the 5 trials, 4 were randomized phase II trials
and 1 were phase II trial. The patient of these trials came from the
UK, Germany, France, Sweden, Austria, Italy, Switzerland, and
South Korea. Four studies were done in Europe and 1 in Asia. The
Table 1
Characteristics of 5 included trials in the meta-analysis.
Male/female Median age (range)
Studies Year Publication type Inclusion period Total number Gem GemCap Gem GemCap Jadad score
Scheithauer W 2003 Phase 2 1996.6-2001.5 83 23/19 27/14 66 (39-75) 66 (40-75) 3
Herrmann R 2007 Phase 3 2001.6-2004.6 319 85/74 86/74 NA NA 3
Cunningham D 2009 Phase 3 2002.2-2005.1 533 153/113 160/167 62 (26-83) 62 (37-82) 3
Lee HS 2017 Phase 3 NA 214 57/49 63/45 64 (43-85) 64 (37-80) 3
Neoptolemos JP 2017 Phase 3 2008.11-2014.9 730 212/154 202/162 65 (37-80) 65 (39-81) 3
Gem =gemcitabine, GemCap=gemcitabine plus capecitabine, NA=not available.
Figure 1. Flow diagram for identifying and including studies in the meta-analysis.
Ouyang et al. Medicine (2021) 100:48 www.md-journal.com
3
period of publication ranged from 2003 to 2017. The ratio of
male/female in the Gem group was 530/409 and in GemCap
group was 538/402.
3.3. Twelve-month survival rate and overall survival
The 12-month survival data were reported for 4 trials. A total of
1665 pancreatic cancer patients from these 4 trials, 833 from the
Figure 2. A: Twelve-month survival rate results are not associated with the Gem group and the GemCap group. B: Meta-analysis of OS results for the Gem group
and the GemCap group. CI=condence interval, Gem =gemcitabine, GemCap =gemcitabine and capecitabine, HR =hazard ratio, RR =relative risk.
Ouyang et al. Medicine (2021) 100:48 Medicine
4
Gem group and 832 from the GemCap group, was enrolled in our
meta-analysis. The consequences of this meta-analysis shows that
no statistically signicant difference was found in the 12-month
survival rate between 2 groups (relative risk (RR) =0.958, 95%
CI 0.886-1.035, P=.278) (Fig. 2A). And no inter-group
signicant heterogeneity was detected in the 12-months survival
rate (I
2
=0, P=.88).
HR and its 95% CI for OS was available in all included
studies. After performing a meta-analysis, the pooled HR
between the Gem group and the GemCap group was 1.15
(95% CI: 1.037-1.276, P=.008) (Fig. 2B). The result revealed
that Gem group was associated with a statistically signicant
19% increase in HR for OS than the GemCap group. There
was no signicant heterogeneity has observed in the OS (I
2
=
0, P=.998,), so a xed-effects model was employed to pool
the data.
3.4. Progression-free survival
The relevant PFS data were reported for 4 trials. A total of 1073
patients from these 4 studies, 537 from the Gem alone group and
576 from the GemCap group, was enrolled in this meta-analysis.
After pooling the data, no heterogeneity among the studies was
detected (I
2
=0, P=.86); therefore, a xed-effects model was
used. There was a signicant difference exist between Gem-alone
groups and GemCap groups. The overall meta-analysis revealed
that Gem increase 21.1% of HR for PFS (HR =1.211, 95% CI
1.09-1.344, P=.860) (Fig. 3).
3.5. Partial response rate and objective response rate
In this meta-analysis, both PRR and ORR were analyzed,
whereas CRR was not analyzed because of data decient. Four
studies involving a total of 1133 patients compared the PRR and
ORR between the Gem alone group and the GemCap group.
After pooling the data, the results revealed that GemCap group
signicantly improved the PRR than in the Gem-alone group (RR
=0.649, 95% CI 0.488-0.862, P=.003) (Fig. 4A), and there was
no heterogeneity was detected regarding the outcome of PRR (I
2
=16.3%, P=.31), a xed-effects model was employed. The
combined group of GemCap was associated with higher ORR
than Gem monopoly (RR =0.605, 95% CI 0.458-0.799, P=0),
and xed-effects model was used because no heterogeneity was
found (I
2
=0, P=.44) (Fig. 4B).
3.6. Toxicity
Five trials reported the incidence of Grade 3/4 neutropenia,
anemia, and diarrhea
[11,16,1921]
; 4 trials reported the incidence of
Grade 3/4 thrombocytopenia and stomatitis.
[11,16,20,21]
Three
trials reported the incidence of Grade 3/4 Nausea
[11,16,21]
and
febrile neutropenia.
[16,20,21]
The Grade 3/4 toxic effects were calculated with dichotomous
data (RR, 95% CI) and the results of 2 arms were summarized
in Table 2. As shown in Table 2, the most common toxicity of
2 arms was neutropenia (25.8%), and the incidence of the
remaining toxicity was all below 7%. After pooling the data,
there was no heterogeneity was found except for thrombocyto-
Figure 3. Meta-analysis of PFS results for the Gem group and the GemCap group. CI =condence interval, Gem =gemcitabine, GemCap =gemcitabine and
capecitabine, HR =hazard ratio, PFS =progress free survival.
Ouyang et al. Medicine (2021) 100:48 www.md-journal.com
5
penia while the I
2
was 62.6% (Table 2). Therefore, thrombocy-
topenia used a random effect model and the other 6 toxic events
used the xed-effects model. The pooled results show that as
compared with Gem alone, GemCap group signicantly
increased the incidence of neutropenia, diarrhea; whereas no
signicant difference was found in the incidence of anemia,
neubrimbocytopenia, febrile neutropenia, nausea, diarrhea
between 2 groups (as shown in Table 2). The results of
this meta-analysis based on the merging of the 7 toxic events
revealed that Gem alone associate with lower toxicity than the
GemCap group (RR =0.708, 95% CI 0.620-0.808, P=.000)
(Fig. 5).
Figure 4. A: Meta-analysis of PRR results for the Gem group and the GemCap group. B: Meta-analysis of ORR results for the Gem group and the GemCap group.
CI =condence interval, Gem =gemcitabine, GemCap =gemcitabine and capecitabine, ORR =objective response rate, PRR =partial response rate, RR =
relative risk.
Ouyang et al. Medicine (2021) 100:48 Medicine
6
3.7. Publication bias
Begg funnel plot and Egger test were used to evaluating the
potential publication bias of the included studies. The result
indicated that there was no obvious publication bias for OS,
analysis (Bgger test: P=.806 for OS) (Fig. 6A). The Egger test did
not demonstrate any publication bias (P=.373 for OS) (Fig. 6B).
4. Discussion
Pancreatic cancer is one of the most fatal malignant neoplasms
with an overall 5-year survival rate for all stages is no more than
8% and the ratio of mortality/incidence is 98%.
[6,24]
Although
surgical resection is the only way to provide curative treatment,
more than 80% of patients with pancreatic cancer are ineligible
to be resected at diagnosed, because they usually metastasize to
the distant organ or invading the major vessel when diag-
nosed.
[25]
Therefore, chemotherapy has become an alternative
choice for pancreatic cancer patients. Since 1997, Gem has
become the mainstay for the treatment of pancreatic cancer.
[9]
After that, Gem has become cornerstone treatment for patients
with pancreatic cancer. In recent years, many randomized
controlled trials have aimed at assessing the potential benets of
Gem-based combination over Gem monopoly. According to
meta-analysis, Gem-based combination chemotherapy signi-
cantly improved the OS, ORR, and PFS in advancer pancreatic
cancer patients. However, the combined group was associated
with increased toxicity.
[26]
Capecitabine is widely used in many
solid malignancies, particularly in gastrointestinal and breast
Figure 5. Meta-analysis of toxicity results in the Gem group and the GemCap group. CI =condence interval, Gem =gemcitabine, GemCap =gemcitabine and
capecitabine, RR =relative risk.
Table 2
Toxicities of Gem and GemCap.
Toxicity Gem
n/N
GemCap
n/N
RR 95% CI I
2
P
Neutropenia 188/909 282/912 0.667 0.569-0.781 0 .818
Anemia 42/909 36/912 1.174 0.762-1.809 0 .481
Thrombocytopenia 32/543 38/553 1.083 0.396-2.961 62.6% .046
Febrile neutropenia 4/296 7/302 0.614 0.197-1.913 0 .693
Diarrheas 20/909 43/912 0.479 0.288-0.799 6.5% .370
Nausea 22/442 25/450 0.895 0.513-1.561 0 .993
Stomatitis 3/543 11/553 0.367 0.126-1.071 0 .506
CI =condence interval, Gem =gemcitabine, GemCap=gemcitabine plus capecitabine, RR=relative ratios.
Ouyang et al. Medicine (2021) 100:48 www.md-journal.com
7
tumors, but also in pancreatic cancer.
[27]
The question of whether
GemCap can achieve a better benet than Gem alone is still
unclear.
We performed the present meta-analysis to compare the
efcacy and tolerability of GemCap vs Gem alone for pancreatic
cancer patients. Five randomized trials include a total of 1879
patients enrolled into this study. Our pooled analysis revealed
that Gem alone may lower the RR for 12-month survival rate
than GemCap; however, no signicant difference was found and
heterogeneity was detected (I
2
=0, P=.88). It was indicated that
the combination group could not prolong the 12-months survival
rate. The pooled HR for OS in our analysis was 1.19, indicating a
19% decline in the risk of death in pancreatic cancer patients
treated with GemCap regimen. The median OS reported by the
included 5 studies varied from 6.2 to 25.5 months in Gem group
and from 7.1 to 28.0 months in GemCap regimen. However,
Neoptolemos JP
[19]
reported the median OS in Gem and GemCap
was 25.5 months and 28.0 months which was higher than the rest
of the 4 studies. The difference between the included articles was
due to the fact that the patients included in Neoptolemos JP
[19]
research were undergo surgical resection, while the enrolled
patients of other studies were metastatic or advanced pancreatic
cancer. As a result, we performed a subgroup analysis based on
whether conducted resection and nd that resection did not affect
the result of OS between Gem and GemCap group (HR 1.15,
P=.008). The median PFS in Gem alone group ranged from 3.8
to 5.3 months and 4.3 to 6.2 months in the GemCap group. The
result shows that GemCap signicantly reduces the hazard of
death by 21.1% compared to Gem alone. Our results were also
consistent with those of Cunningham Ds study.
[11]
A meta-
analysis
[28]
that reported Gem-based combination therapy could
improve the OS and PFS, but not the 12-months survival rate was
similar to our study. Based on the above results, though GemCap
might not achieve a better outcome in 12-months survival rate,
the OS and PFS were superior in GemCap compared to Gem
alone. Thus, Gem plus capecitabine is more recommended over
Gem monopoly.
The present meta-analysis showed that GemCap signicantly
increases PRR and ORR by 35.1% and 39.5% as compared with
Gem monopoly. Our previous meta-analysis compared Gem plus
cisplatin vs Gem alone revealed that the combination regime
improves the outcome of ORR.
[29]
It was indicated that Gem
combines with capecitabine or cisplatin might achieve synergistic
effects. Another meta-analysis also suggested that Gem-based
combination therapy increased 58% of ORR than Gem
monotherapy, especially when combined with Gem or oxalipla-
tin; moreover, the combinations of Gem plus platinum
uoropyrimidine improved ORR by 47% as compared with
Gem alone.
[30]
Capecitabine is an oral uoropyrimidine seems to
be a substitute intravenous 5-Fu, with the advantage of no need
injection.
[27]
Lee et al
[20]
reported that GemCap signicantly
improved ORR (43.7% vs 17.6%; P=.001), but fail to improve
OS (HR, 0.82; 95% CI, 0.67-1.01; P=.06) and PFS (HR, 0.87;
95% CI, 0.73-1.03; P=.08) compared with Gem monopoly.
Some of the above results are different from ours because we
found that GemCap not only improves not ORR but also
improves OS and PFS.
In the present study, we found that the GemCap was associated
with higher odds for Grade 3/4 toxic effects compared with Gem
alone. Pooled adverse data signicantly increased by 29.2% in
GemCap over Gem alone (RR =0.708, 95% CI 0.620-0.808,
P=.000).
Previous studies proved that several new combination chemo-
therapies offer superior survival outcome and PFS than Gem
alone regimens; however, it could not apply to all pancreatic
cancer patients due to its severe toxicity.
[3133]
The incidence rate
of toxicity was under 7% except for neutropenia (25.8%);
nevertheless, the incidence of neutropenia is still very low when
compared to Gem plus S-1(63.3%).
[34]
It is well known that if
patients experience serious adverse events during chemotherapy,
the physician may reduce the dosage or even stop the
chemotherapy, so lower toxicity effects were important for
pancreatic cancer patients to improve the tolerance of chemo-
therapy.
Our meta-analysis reveals a statistically signicant greater
incidence of grades III/IV toxic effects in the GemCap regime, the
12-months survival, OS, PFS, PRR, and ORR, however,
signicantly increased may make the adverse events generally
tolerable and reversible. However, due to the specic data that
could not be obtained, more studies are required to further verify
the greatest beneciary from this treatment.
There are some limitations in the present meta-analysis and
there were: our study only enrolled 5 RCTs and the sample size of
each regimen relatively too small to provide sufcient evidence
for the safety and efciency of pancreatic cancer between
GemCap and Gem alone. Therefore, more RCTs with larger
sample sizes are required. Our study was based on literature and
was associated with publication bias; the present meta-analysis
was an inference from published abstracted data rather than
individual patient proles. Different dose and modication
Figure 6. A: Begg funnel plot of public ation bias test. B: Egger publication bias
plot. HR =hazard ratio.
Ouyang et al. Medicine (2021) 100:48 Medicine
8
schemes of Gem or capecitabine may generate divergent
outcomes. The status of pancreatic cancer in 4 included studies
was advanced or metastasis and only treated with chemotherapy,
however, the status in Neoptolemos JP
[19]
was resected
pancreatic cancer and patients were conducted surgical resection
before chemotherapy. We hope further RCTs may resolve the
above problems and provide much more high-quality clinical
evidence.
5. Conclusions
This meta-analysis of randomized control studies revealed that
GemCap signicantly improves OS, PFS, PRR, ORR; however,
no signicant difference was found in the 12-months survival
rate. Though the incidence of Grade 3/4 toxicity was higher in
GemCap compared with Gem alone, the adverse events of
GemCap were tolerable. In conclusion, Gem plus capecitabine
may be considered as promising chemotherapy for pancreatic
cancer. However, owing to the above limitations, more
convincing large-scale RCTs are needed.
Author contributions
Conceptualization: Guoqing Ouyang, Guangdong Pan.
Data curation: Zheng Zhou, Yongrong Wu.
Formal analysis: Zhen Liu, Wuhang Lu, Shuqing Hao.
Resources: Guangdong Pan.
Validation: Shuai Li.
Writing original draft: Guoqing Ouyang, Guangdong Pan,
Yongrong Wu.
Writing review & editing: Guangdong Pan
References
[1] Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J Clin
2019;69:734.
[2] Siegel RL, Miller KD, Jemal A. Cancer statistics, 2015. CA Cancer J Clin
2015;65:529.
[3] Rahib L, Smith BD, Aizenberg R, et al. Projecting cancer incidence and
deaths to 2030: the unexpected burden of thyroid, liver, and pancreas
cancers in the United States. Cancer Res 2014;74:291321.
[4] Hidalgo M, Cascinu S, Kleeff J, et al. Addressing the challenges of
pancreatic cancer: future directions for improving outcomes. Pancrea-
tology 2015;15:818.
[5] Taherian N, Saam J, Larson K, et al. Hereditary cancer genetic testing
among patients with pancreatic cancer. J Clin Oncol 2019;37
(15_suppl):4134.
[6] Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin
2018;68:730.
[7] Ryan DP, Hong TS, Bardeesy N. Pancreatic adenocarcinoma. N Engl J
Med 2014;371:103949.
[8] Chao Y, Wu CY, Wang JP, et al. A randomized controlled trial of
gemcitabine plus cisplatin versus gemcitabine alone in the treatment of
metastatic pancreatic cancer. Cancer Chemother Pharmacol 2013;
72:63742.
[9] Burris HA, Moore MJ, Andersen J, et al. Improvements in survival and
clinical benet with gemcitabine as rst-line therapy for patients with
advanced pancreas cancer: a randomized trial. J Clin Oncol 1997;
15:240313.
[10] Wang D, Bai T, Chen G, et al. Upregulation of long non-coding RNA
FOXP4-AS1 and its regulatory network in hepatocellular carcinoma.
Onco Targets Ther 2019;12:702538.
[11] Cunningham D, Chau I, Stocken DD, et al. Phase III randomized
comparison of gemcitabine versus gemcitabine plus capecitabine in
patients with advanced pancreatic cancer. J Clin Oncol 2009;27:55138.
[12] Moore MJ, Goldstein D, Hamm J, 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;25:19606.
[13] Poplin E, Feng Y, Berlin J, et al. Phase III, randomized study of
gemcitabine and oxaliplatin versus gemcitabine (xed-dose rate infusion)
compared with gemcitabine (30-minute infusion) in patients with
pancreatic carcinoma E6201: a trial of the Eastern Cooperative
Oncology Group. J Clin Oncol 2009;27:377885.
[14] Peixoto RD, Ho M, Renouf DJ, et al. Eligibility of metastatic pancreatic
cancer patients for rst-line palliative intent nab-paclitaxel plus
gemcitabine versus FOLFIRINOX. Am J Clin Oncol 2017;40:50711.
[15] Miwa M, Ura M, Nishida M, et al. Design of a novel oral
uoropyrimidine carbamate, capecitabine, which generates 5-uoroura-
cil selectively in tumours by enzymes concentrated in human liver and
cancer tissue. Eur J Cancer (Oxford, England: 1990) 1998;34:127481.
[16] Herrmann R, Bodoky G, Ruhstaller T, et al. Gemcitabine plus
capecitabine compared with gemcitabine alone in advanced pancreatic
cancer: a randomized, multicenter, phase III trial of the Swiss Group for
Clinical Cancer Research and the Central European Cooperative
Oncology Group. J Clin Oncol 2007;25:22127.
[17] Cartwright TH, Cohn A, Varkey JA, et al. Phase II study of oral
capecitabine in patients with advanced or metastatic pancreatic cancer. J
Clin Oncol 2002;20:1604.
[18] Schilsky RL, Bertucci D, Vogelzang NJ, et al. Dose-escalating study of
capecitabine plus gemcitabine combination therapy in patients with
advanced cancer. J Clin Oncol 2002;20:5827.
[19] Neoptolemos JP, Palmer DH, Ghaneh P, et al. Comparison of adjuvant
gemcitabine and capecitabine with gemcitabine monotherapy in patients
with resected pancreatic cancer (ESPAC-4): a multicentre, open-label,
randomised, phase 3trial. Lancet (London, England) 2017;389:101124.
[20] Lee HS, Chung MJ, Park JY, et al. A randomized, multicenter, phase III
study of gemcitabine combined with capecitabine versus gemcitabine
alone as rst-line chemotherapy for advanced pancreatic cancer in South
Korea. Medicine 2017;96:e5702.
[21] Scheithauer W, Schull B, Ulrich-Pur H, et al. Biweekly high-dose
gemcitabine alone or in combination with capecitabine in patients with
metastatic pancreatic adenocarcinoma: a randomized phase II trial. Ann
Oncol 2003;14:97104.
[22] Jadad AR, Moore RA, Carroll D, et al. Assessing the quality of reports of
randomized clinical trials: is blinding necessary? Control Clin Trials
1996;17:112.
[23] Higgins JP, Thompson SG, Deeks JJ, et al. Measuring inconsistency in
meta-analyses. BMJ (Clin Res ed) 2003;327:55760.
[24] Ilic M, Ilic I. Epidemiology of pancreatic cancer. World J Gastroenterol
2016;22:9694705.
[25] Lee YS, Lee JC, Yang SY, et al. Neoadjuvant therapy versus upfront
surgery in resectable pancreatic cancer according to intention-to-treat
and per-protocol analysis: a systematic review and meta-analysis. Sci Rep
2019;9:18.
[26] Zhang XW, Ma YX, Sun Y, et al. Gemcitabine in combination with a
second cytotoxic agent in the rst-line treatment of locally advanced or
metastatic pancreatic cancer: a systematic review and meta-analysis.
Target Oncol 2017;12:30921.
[27] Siddiqui NS, Godara A, Byrne MM, et al. Capecitabine for the treatment
of pancreatic cancer. Expert Opin Pharmacother 2019;20:399409.
[28] Jin SF, Fan ZK, Pan L, et al. Gemcitabine-based combination therapy
compared with gemcitabine alone for advanced pancreatic cancer: a
meta-analysis of nine randomized controlled trials. Hepatobiliary
Pancreat Dis Int 2017;16:23644.
[29] Ouyang G, Liu Z, Huang S, et al. Gemcitabine plus cisplatin versus
gemcitabine alone in the treatment of pancreatic cancer: a meta-analysis.
World J Surg Oncol 2016;14:19.
[30] Hu J, Zhao G, Wang HX, et al. A meta-analysis of gemcitabine
containing chemotherapy for locally advanced and metastatic pancreatic
adenocarcinoma. J Hematol Oncol 2011;4:115.
[31] Okusaka T, Ikeda M, Fukutomi A, et al. Phase II study of FOLFIRINOX
for chemotherapy-naive Japanese patients with metastatic pancreatic
cancer. Cancer Sci 2014;105:13216.
[32] Van Loon K, Espinoza AM, Fogelman DR, et al. Should combination
chemotherapy serve as the backbone in clinical trials of advanced
pancreatic cancer? A pooled analysis of phase II trials of gemcitabine-
containing doublets plus bevacizumab. Pancreas 2014;43:3439.
[33] Liu GF, Li GJ, Zhao H. Efcacy and toxicity of different chemotherapy
regimens in the treatment of advanced or metastatic pancreatic cancer: a
network meta-analysis. J Cell Biochem 2018;119:51123.
[34] Sudo K, Hara R, Nakamura K, et al. Phase II study of induction
gemcitabine and S-1 followed by chemoradiotherapy and systemic
chemotherapy using S-1 for locally advanced pancreatic cancer. Cancer
Chemother Pharmacol 2017;80:195202.
Ouyang et al. Medicine (2021) 100:48 www.md-journal.com
9
... Current studies have shown some clinical benefit against traditional tumor angiogenesis-targeting drugs, but resistance has been shown to be a problem (Beijnum et al. 2015;Gotink et al. 2011). Clinical data show that gemcitabine has not significantly improved survival in patients with advanced PC (Ouyang et al. 2021;Thummuri et al. 2022). Poor perfusion is one of the main features of PC tissue, and the molecules behind this state of low angiogenesis are not known (Tao et al. 2021). ...
Article
Full-text available
Background: Vascular mimicry (VM) epitomizes an innovative tumor angiogenesis pathway, potentially serving as an alternate conduit under the assumption of traditional tumor angiogenesis pathway inhibition. The role of VM in pancreatic cancer (PC), however, remains unexplored. Methods: Using differential analysis and Spearman correlation, we identified key long non-coding RNAs (lncRNAs) signatures in PC from the collected set of VM-associated genes in the literature. We identified optimal clusters using the non-negative matrix decomposition (NMF) algorithm, and then compared clinicopathological features and prognostic differences between clusters. We also assessed tumor microenvironmental (TME) differences between clusters using multiple algorithms. Using univariate Cox regression analyses as well as lasso regression, we constructed and validated new lncRNA prognostic risk models for PC. We used Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) to analyze model-enriched functions and pathways. Nomograms were then developed to predict patient survival in association with clinicopathological factors. In addition, single-cell RNA-sequencing (scRNA-seq) analysis was used to analyze the expression patterns of VM-related genes and lncRNAs in the PC of TME. Finally, we used the Connectivity Map (cMap) database to predict local anaesthetics that could modify the VM of PC. Results: In this study, we developed a novel three-cluster molecular subtype using the identified VM-associated lncRNA signatures of PC. The different subtypes have significantly different clinical characteristics and prognostic value, and also show differential treatment response and TME. Following an in-depth analysis, we constructed and validated a novel prognostic risk model for PC based on the VM-associated lncRNA signatures. Enrichment analysis suggested that high riskscores were significantly associated with functions and pathways, including extracellular matrix remodeling, et al. In addition, we predicted eight local anaesthetics that could modulate VM in PC. Finally, we discovered differential expression of VM-related genes and lncRNAs across various cell types within pancreatic cancer. Conclusion: VM has a critical role in PC. This study pioneers the development of a VM-based molecular subtype that demonstrates substantial differentiation in PC populations. Furthermore, we highlighted the significance of VM within the immune microenvironment of PC. Moreover, VM might contribute to PC tumorigenesis through its mediation of mesenchymal remodeling and endothelial transdifferentiation-related pathways, which offers a new perspective on its role in PC.
... The exosomes have clear role in communications between tumor and immune cells and supposed to have a dynamic role in tumor immunity regulation. Gemcitabine chemotherapy is considered a standard treatment for PC either in combination or monotherapy, based on evidence from many studies which shown a better survival rate and more clinical benefits with median overall survival (OS) of 5 mo to 7 mo [45]. Most of patients with PC ultimately present with rapid disease progression even following chemotherapy with gemcitabine. ...
Article
Full-text available
Despite the development of newer oncological treatment, the survival of patients with pancreatic cancer (PC) remains poor. Recent studies have identified exosomes as essential mediators of intercellular communications and play a vital role in tumor initiation, metastasis and chemoresistance. Thus, the utility of liquid biopsies using exosomes in PC management can be used for early detection, diagnosis, monitoring as well as drug delivery vehicles for cancer therapy. This review summarizes the function, and clinical applications of exosomes in cancers as minimally invasive liquid biomarker in diagnostic, prognostic and therapeutic roles.
... At the same time, in order to improve the therapeutic effect of pancreatic cancer treatment, since GEM entered the market, researchers have been trying to treat advanced pancreatic cancer based on GEM combined with many drugs, which has significantly improved the overall survival (OS) and progression-free survival (PFS) of patients (11)(12)(13). In particular, there have been many studies conducted on the combination of GEM with platinum-or fluorouracilbased drugs. ...
Article
Full-text available
Background: Gemcitabine (GEM) is used as a standard first-line drug to effectively alleviate symptoms and prolong survival time for advanced pancreatic cancer. Most randomized controlled trials (RCTs) show that GEM-based combination therapy is better than GEM alone, while some RCTs have the opposite conclusion. This study aimed to investigate whether GEM-based combination therapy would be superior to GEM alone by a systematic review and meta-analysis. Methods: According to the PICOS principles, RCTs (S) focused on comparing GEM-based combination therapy (I) vs. GEM alone (C) for advanced pancreatic cancer (P) were collected from eight electronic databases, outcome variables mainly include survival status and adverse events (AEs) (O). Review Manager 5.4 was used to evaluate the pooled effects of the results among selected articles. Pooled estimate of hazard ratio (HR) and odds ratio (OR) with 95% confidence interval (CI) were used as measures of effect sizes. Quality assessment for individual study was performed using the Cochrane tool for risk of bias. Results: A total of 17 studies including 5,197 patients were selected in this analysis. The pooled results revealed that GEM-based combination therapy significantly improved the overall survival (OS; HR =0.84; 95% CI: 0.79 to 0.90; P<0.00001), progression-free survival (PFS; HR =0.78; 95% CI: 0.72 to 0.84; P<0.00001), overall response rate (ORR; OR =1.92; 95% CI: 1.61 to 2.30; P<0.00001), 1-year survival rate (OR =1.44; 95% CI: 1.02 to 2.03; P=0.04), respectively. Subgroup analysis showed that the efficacy of GEM plus capecitabine (CAP) and GEM plus S-1 was better than that of GEM alone, while GEM plus cisplatin (CIS) did not achieve an improved effect. GEM-based combination therapy can significantly increase the incidence of AEs, such as leukopenia (P<0.001), neutropenia (P<0.001), anemia (P<0.05), nausea (P<0.001), diarrhea (P<0.05), and stomatitis (P<0.001). No publication bias existed in our meta-analysis (P>0.10). Discussion: Our study supported that GEM-based combination therapy was more beneficial to improve patient's survival than GEM alone, while there was no additional benefits in GEM plus CIS. We also found that GEM-based combination therapy increased the incidence of AEs. Clinicians need to choose the appropriate combination therapy according to the specific situation.
Article
Full-text available
The effectiveness of neoadjuvant therapy (NAT) remains unclear in resectable pancreatic cancer (PC) as compared with upfront surgery (US). The aim of this study was to investigate the survival gain of NAT over US in resectable PC. PubMed and EMBASE were searched for studies comparing survival outcomes between NAT and US for resectable PC until June 2018. Overall survival (OS) was analyzed according to treatment strategy (NAT versus US) and analytic methods (intention-to-treat analysis (ITT) and per-protocol analysis (PP)). In 14 studies, 2,699 and 6,992 patients were treated with NAT and US, respectively. Although PP analysis showed the survival gain of NAT (HR 0.72, 95% CI 0.68–0.76), ITT analysis did not show the statistical significance (HR 0.96, 95% CI 0.82–1.12). However, NAT completed with subsequent surgery showed better survival over US completed with adjuvant therapy (HR 0.82, 95% CI 0.71–0.93). In conclusion, the supporting evidence for NAT in resectable PC was insufficient because the benefit was not demonstrated in ITT analysis. However, among the patients who completed both surgery and chemotherapy, NAT showed survival benefit over adjuvant therapy. Therefore, NAT could have a role of triaging the patients for surgery even in resectable PC.
Article
Full-text available
Objective FOXP4-AS1 (FOXP4 antisense RNA 1) is putatively a functional oncogene in colorectal cancer. This study constructed a regulatory network involving FOXP4-AS1 for better understanding of its function in hepatocellular carcinoma (HCC). Methods FOXP4-AS1 was assessed in HCC and adjacent normal (control) liver samples via quantitative real-time PCR. Differentially expressed micro RNAs (DEmiRNAs) were predicted. Their target genes were verified via the gene expression profiling interaction analysis (GEPIA) database, and subjected to gene ontology (GO) annotation and KEGG (Kyoto Encyclopedia of Genes and Genome) pathway enrichment analysis. Protein-protein interaction (PPI) networks were established and hub genes identified with Cytoscape software. The GEPIA database was used to assess the prognostic roles of 20 hub genes in liver cancer. The cBioPortal database was used to exhibit alterations of the genes. Results The HCC samples had significantly higher levels of FOXP4-AS1 compared with the control (P=0.001). Six upregulated and 4 downregulated DEmiRNAs were identified. Over- and under-expressed predicted target genes (183 and 147, respectively) were selected for GO annotation and KEGG pathway enrichment analysis. The downregulated genes were significantly prominent in the PI3K-Akt signaling pathway; the upregulated genes in the cell cycle. The PPI networks indicated IGFBP3 and PRC1 as hub genes with the highest node degrees. Higher expressions of 9 (6) genes were associated with worse (better) prognosis in HCC. Conclusion An HCC-associated FOXP4-AS1-miRNA-mRNA regulatory network was constructed, and molecular mechanisms involved in HCC development were elucidated. This work provides direction for finding new HCC therapeutic targets.
Article
Full-text available
Each year, the American Cancer Society estimates the numbers of new cancer cases and deaths that will occur in the United States and compiles the most recent data on cancer incidence, mortality, and survival. Incidence data, available through 2015, were collected by the Surveillance, Epidemiology, and End Results Program; the National Program of Cancer Registries; and the North American Association of Central Cancer Registries. Mortality data, available through 2016, were collected by the National Center for Health Statistics. In 2019, 1,762,450 new cancer cases and 606,880 cancer deaths are projected to occur in the United States. Over the past decade of data, the cancer incidence rate (2006‐2015) was stable in women and declined by approximately 2% per year in men, whereas the cancer death rate (2007‐2016) declined annually by 1.4% and 1.8%, respectively. The overall cancer death rate dropped continuously from 1991 to 2016 by a total of 27%, translating into approximately 2,629,200 fewer cancer deaths than would have been expected if death rates had remained at their peak. Although the racial gap in cancer mortality is slowly narrowing, socioeconomic inequalities are widening, with the most notable gaps for the most preventable cancers. For example, compared with the most affluent counties, mortality rates in the poorest counties were 2‐fold higher for cervical cancer and 40% higher for male lung and liver cancers during 2012‐2016. Some states are home to both the wealthiest and the poorest counties, suggesting the opportunity for more equitable dissemination of effective cancer prevention, early detection, and treatment strategies. A broader application of existing cancer control knowledge with an emphasis on disadvantaged groups would undoubtedly accelerate progress against cancer.
Article
Full-text available
Purpose: S-1 has systemic activity for locally advanced pancreatic cancer (LAPC). Here, the efficacy and safety of induction gemcitabine (GEM) and S-1 (GS) followed by chemoradiotherapy (CRT) and systemic chemotherapy using S-1 for LAPC were assessed. Methods: The treatment consisted of four cycles of induction GS (S-1 60, 80, or 100 mg/day based on body surface area for 14 days every 3 weeks plus GEM 1000 mg/m(2) on days 8 and 15), followed by S-1 (80, 100, or 120 mg/day based on body surface area on days 1-14 and 22-35) and concurrent radiotherapy (50.4 Gy in 28 fractions). Maintenance chemotherapy with S-1 was started 1-4 weeks after CRT until disease progression or unacceptable toxicity was observed. The primary endpoint was 1-year survival. Results: A total of 30 patients with LAPC were enrolled. The median survival and progression-free survival were 21.3 and 12.7 months, respectively. Overall survival rates at 1, 2, 3, and 4 years were 73.3, 36.7, 23.3, and 16.7%, respectively. The median survival of 23 patients who received CRT was 22.9 months, with a 3-year survival rate of 30.4%. The two most common grade 3 or 4 adverse events during induction GS were neutropenia (63.3%) and biliary tract infection (20%). Toxicities during CRT or maintenance chemotherapy were generally mild. Conclusions: This regimen was feasible and highly active resulting in encouraging survival in patients with LAPC. Further investigations are warranted to elucidate the effectiveness of this treatment strategy in future studies. Clinical trials information: UMIN000006332.
Article
4134 Background: Pancreatic cancer (PC) is typically diagnosed at a late, untreatable stage, with a 5-year survival rate of only ~8%. Genetic testing for individuals with PC may aid in therapy decisions, as those with a germline or somatic pathogenic variant (PV) in a DNA-repair gene may benefit from PARP inhibitors. In addition, germline genetic testing for unaffected family members can identify high risk individuals who may be appropriate for surveillance studies. We assessed the results of hereditary cancer genetic testing among individuals with a personal history of PC and evaluated several possible risk factors. Methods: Individuals with PC who had germline testing for 25-29 cancer-susceptibility genes between September 2013 and November 2018 were included in this analysis (N = 1,676). Clinical characteristics were obtained from provider-completed test request forms and included personal cancer history (PHx), family cancer history (FHx), and age at diagnosis. Results: Overall, 12.6% (212/1676) of patients with PC carried a PV, most commonly in BRCA2 (3.8%), ATM (2.7%), and PALB2 (1.2%). PVs were more common in men and for individuals who had a PHx of additional cancer(s) (see Table). Age at PC diagnosis did not impact the positive PV rate. The PV positive rate was elevated among individuals with PC and at least two relatives with PC (15.1%) and for individuals with a FHx of cancer at an early age (14.2%). The PV positive rate remained > 10% regardless of nearly all other FHx characteristics evaluated, including the absence of any FHx (see Table). Conclusions: In this cohort, a substantial proportion of individuals with a PHx of PC carried PVs, regardless of age at diagnosis and personal and family cancer history. [Table: see text]
Article
Introduction: Capecitabine is an oral prodrug of 5-fluorouracil (5-FU) which is converted to 5FU by a series of reactions catalyzed by different enzymes, the last of the enzymes being thymidine phosphorylase (TP). TP is found to be elevated in tumor cells in comparison to normal cells, which consequently tumor-localizes the production of 5-FU, thereby limiting its systemic toxicity. Today, capecitabine is extensively used for the treatment of many solid malignancies, with a particular focus in breast and gastrointestinal tumors, but also in pancreatic cancer. Areas covered: This review summarizes the pharmacology and the clinical evidence relevant to the use of capecitabine in the treatment of pancreas cancer. The authors provide, furthermore, provide their expert perspectives on its use. Expert opinion: Capecitabine has the advantage over other therapeutics in so much that it has both convenient oral administration and a favorable toxicity profile. Current data has promised the use of capecitabine in all stages of pancreatic cancer. However, predictive markers for outcome and toxicity remain to be validated.
Article
Each year, the American Cancer Society estimates the numbers of new cancer cases and deaths that will occur in the United States and compiles the most recent data on cancer incidence, mortality, and survival. Incidence data, available through 2014, were collected by the Surveillance, Epidemiology, and End Results Program; the National Program of Cancer Registries; and the North American Association of Central Cancer Registries. Mortality data, available through 2015, were collected by the National Center for Health Statistics. In 2018, 1,735,350 new cancer cases and 609,640 cancer deaths are projected to occur in the United States. Over the past decade of data, the cancer incidence rate (2005-2014) was stable in women and declined by approximately 2% annually in men, while the cancer death rate (2006-2015) declined by about 1.5% annually in both men and women. The combined cancer death rate dropped continuously from 1991 to 2015 by a total of 26%, translating to approximately 2,378,600 fewer cancer deaths than would have been expected if death rates had remained at their peak. Of the 10 leading causes of death, only cancer declined from 2014 to 2015. In 2015, the cancer death rate was 14% higher in non-Hispanic blacks (NHBs) than non-Hispanic whites (NHWs) overall (death rate ratio [DRR], 1.14; 95% confidence interval [95% CI], 1.13-1.15), but the racial disparity was much larger for individuals aged <65 years (DRR, 1.31; 95% CI, 1.29-1.32) compared with those aged ≥65 years (DRR, 1.07; 95% CI, 1.06-1.09) and varied substantially by state. For example, the cancer death rate was lower in NHBs than NHWs in Massachusetts for all ages and in New York for individuals aged ≥65 years, whereas for those aged <65 years, it was 3 times higher in NHBs in the District of Columbia (DRR, 2.89; 95% CI, 2.16-3.91) and about 50% higher in Wisconsin (DRR, 1.78; 95% CI, 1.56-2.02), Kansas (DRR, 1.51; 95% CI, 1.25-1.81), Louisiana (DRR, 1.49; 95% CI, 1.38-1.60), Illinois (DRR, 1.48; 95% CI, 1.39-1.57), and California (DRR, 1.45; 95% CI, 1.38-1.54). Larger racial inequalities in young and middle-aged adults probably partly reflect less access to high-quality health care. CA Cancer J Clin 2018.
Article
Background Pancreatic cancer is one of the most aggressive malignancies and chemotherapy is an effective strategy for advanced pancreatic cancer. Gemcitabine (GEM) is one of first-line agents. However, GEM-based combination therapy has shown promising efficacy in patients with advanced pancreatic cancer. This meta-analysis aimed to compare the efficacy and safety of GEM-based combination therapy versus GEM alone in the treatment of advanced pancreatic cancer. Data sources A comprehensive search of literature was performed using PubMed, EMBASE, Web of Science and Cochrane Central Register of Controlled Trials. A quantitative meta-analysis was performed based on the inclusion criteria from all eligible randomized controlled trials. The outcome indicators included overall survival (OS), 6-month survival, 1-year survival, progression-free survival/time-to-progression (PFS/TTP), and toxicities. Results A total of nine randomized controlled trials involving 1661 patients were included in this meta-analysis. There was significant improvement in the GEM-based combination therapy with regard to the OS (HR=0.85, 95% CI: 0.76–0.95, P=0.003), PFS (HR=0.76, 95% CI: 0.65–0.90, P=0.002), 6-month survival (RR=1.09, 95% CI: 1.01–1.17, P=0.03), and the overall toxicity (RR=1.68, 95% CI: 1.52–1.86, P<0.01). However, there was no significant difference in the 1-year survival. Conclusions GEM-based combination chemotherapy might improve the OS, 6-month survival, and PFS in advanced pancreatic cancer. However, combined therapy also added toxicity.
Article
Objective: A network meta-analysis was conducted to compare the efficacy and toxicity of different chemotherapy regimens in treating advanced or metastatic pancreatic cancer (PC). Methods: PubMed, Cochrane Library and EMBASE databases from inception to June 2016 were searched. A combination of direct and indirect evidences was referred to for calculating the weighted mean difference (WMD) or the odds ratio (OR) and to establish surface under the cumulative ranking (SUCRA) curves, so as to evaluate the efficacy and toxicity of different chemotherapy regimens in treating advanced or metastatic PC. Twenty randomized controlled trials were enrolled. Twelve chemotherapy regimens included Gemcitabine, S-1 (Tegafur), Gemcitabine + Cisplatin, Gemcitabine + Capecitabine, Gemcitabine + S-1, Gemcitabine + 5-FU (5-fluorouracil), Gemcitabine + Exatecan, Gemcitabine + Irinotecan, Gemcitabine + Nab-paclitaxel, FOLFIRINOX (Oxaliplatin + Irinotecan + Fluorouracil + Leucovorin), Gemcitabine + Oxaliplatin and Gemcitabine + Pemetrexed. Higher overall response rate (ORR) was observed in patients treated with the gemcitabine + S-1 and FOLFIRINO regimens. Thrombocytopenia reduced in patients treated with the S-1 regimen. Results: The Gemcitabine + S-1 and FOLFIRINO regimens had better short- and long-term efficacies than the other regimens; S-1 regimen had the lowest hematologic toxicity, while Gemcitabine + Nab-paclitaxel, FOLFIRINOX and Gemcitabine + Pemetrexed regimens had higher incidence of non-hematologic toxicity among twelve chemotherapy regimens. Conclusion: The efficacy of Gemcitabine + S-1 and FOLFIRINOX regimens may be better in treating patients with advanced or metastatic pancreatic cancer, while FOLFIRINOX and Gemcitabine + Pemetrexed regimens may have relatively higher incidence of toxicity than other regimens. This article is protected by copyright. All rights reserved.
Article
Background It remains controversial whether the addition of a second cytotoxic agent can further improve the therapeutic effect of gemcitabine monotherapy in advanced or metastatic pancreatic cancer (LA/MPC). Objective The objective of the present systematic review and meta-analysis was to investigate the efficacy and safety of gemcitabine-based doublet chemotherapy regimens compared to single-agent gemcitabine in the first-line treatment of unresectable LA/MPC. Methods We searched for randomized controlled trials (RCTs) of gemcitabine monotherapy versus gemcitabine in combination with a second cytotoxic agent in patients with LA/MPC. The last search date was December 31, 2016. ResultsTwenty-seven RCTs were identified and included in the present systematic review and meta-analysis, involving a total of 7343 patients. The meta-analysis showed that gemcitabine-based combination therapy significantly improved overall survival (OS) (HR: 0.89; 95% confidence interval (CI): 0.85-0.94; P < 0.0001), progression-free survival (PFS) (HR: 0.80; 95% CI: 0.73-0.88; P < 0.0001), and overall response rate (ORR) (RR: 1.83; 95% CI: 1.62-2.07; P < 0.0001) in comparison to single-agent gemcitabine. Subgroup analysis suggested that the antitumor activity differed between gemcitabine-based combination regimens: doublet regimens of gemcitabine plus a taxoid, and gemcitabine plus a fluoropyrimidine, in particular an oral fluoropyrimidine, resulted in a significant OS benefit for the patients. However, the combination of gemcitabine with other cytotoxic agents, such as platinum compounds or topoisomerase inhibitors failed to reduce the mortality risk. Combination therapy caused more grade 3/4 toxicities, including neutropenia, thrombocytopenia, vomiting, diarrhea, and fatigue. Conclusions Gemcitabine-based doublet regimens demonstrated superiority over gemcitabine monotherapy in overall efficacy, but were associated with increased toxicity. Different gemcitabine-based combinations showed different antitumor activity, and doublet regimens of gemcitabine in combination with a taxoid or a fluoropyrimidine, in particular an oral fluoropyrimidine provided significant survival benefits in the first-line treatment of unresectable LA/MPC.