Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Seminars in Oncology 46 (2019) 19–27

Contents lists available at ScienceDirect

Seminars in Oncology
journal homepage: www.elsevier.com/locate/seminoncol

Neoadjuvant Treatment for Pancreatic Cancer

Alexander G. Raufi, MD a,∗, Gulam A. Manji, MD, PhD a, John A. Chabot, MD b, Susan E. Bates, MD a
a
The Division of Medical Oncology, Columbia University Medical Center, New York, NY
b
The Department of Surgery, Columbia University Medical Center, New York, NY

a r t i c l e i n f o a b s t r a c t

Article history: Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy with limited effective therapeu-
Received 11 December 2018 tic options and exceedingly high mortality rates. Currently, cure can only be achieved through resection,
Accepted 11 December 2018
however the vast majority of patients present with advanced disease for which upfront surgery is not
an option. In an effort to improve surgical candidacy, neoadjuvant chemotherapy, with or without ra-
Keywords: diation therapy, is often used in an effort to downstage borderline resectable and locally advanced tu-
Pancreatic Cancer mors, and some argue for its use even in patients with resectable tumors. Underlying this thinking is the
Neoadjuvant therapy recognition that pancreatic cancer is simultaneously both a locally invasive and systemic disease, even
Resectable in patients without evidence of metastasis on imaging. Current evidence to date is largely retrospec-
Borderline resectable
tive, but suggests that neoadjuvant therapy can increase R0 (pathologically negative margin) resection
Locally advanced
rates and improve overall survival. The standard approach to neoadjuvant treatment involves choosing
between the two most active combination regimens for metastatic disease, namely modified FOLFIRNOX
and gemcitabine/nab-paclitaxel. Nonrandomized data indicate that these regimens can yield resection
rates up to 68% and 36%, in borderline resectable and locally advanced PDAC, respectively. Furthermore,
randomized data in patients with resectable PDAC treated with gemcitabine-based neoadjuvant therapy
suggests that despite an approximate 10% drop in resection rates, there is a significant improvement in
median overall survival. Herein, we will discuss the rationale for neoadjuvant therapy, current and for-
mer treatment regimens, common issues faced by clinicians when using these combinations, and several
ongoing clinical trials.
© 2018 Elsevier Inc. All rights reserved.

Contents

Background. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
Rationale for neoadjuvant therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
Data supporting neoadjuvant therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
Defining resectable, borderline resectable, and locally advanced pancreatic cancer and treatment effect . . . . . . . . . . . . . . . . . . . . . 20
Trials with neoadjuvant therapy in patients with resectable disease. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21
Trials with neoadjuvant therapy in patients with borderline resectable disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22
Trials with neoadjuvant therapy in patients with locally advanced disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 23
Currently used regimens for neoadjuvant chemotherapy in patients with BR or LA disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 23
FOLFIRINOX. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 23
Gemcitabine/nab-paclitaxel . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24
Gemcitabine/docetaxel/capecitabine (GTX). . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24
Complexities of neoadjuvant therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 25
Ongoing trials with neoadjuvant therapy in patients with resectable, BR, or LA pancreatic cancer . . . . . . . . . . . . . . . . . . . . . . . . . . 25
Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 25
Conflict of interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26
Supplementary materials. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26

Corresponding author. Columbia University, 161 Fort Washington Avenue at W.
165th Street, New York, NY 10032. E-mail address: agr2139@cumc.columbia.edu (A.G. Raufi).

https://doi.org/10.1053/j.seminoncol.2018.12.002
0093-7754/© 2018 Elsevier Inc. All rights reserved.

Descargado para Anonymous User (n/a) en Universidad El Bosque de ClinicalKey.es por Elsevier en abril 08, 2021. Para uso personal
exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
20 A.G. Raufi, G.A. Manji and J.A. Chabot et al. / Seminars in Oncology 46 (2019) 19–27

Background with a diagnosis of PDAC were included and were grouped into
two cohorts: resectable and nonresectable disease. Of those pa-
The aggressive nature of pancreatic ductal adenocarcinoma tients initially diagnosed with nonresectable disease, 46.9% under-
(PDAC) makes it a particularly challenging disease to treat. At the went surgical exploration after neoadjuvant therapy and 69.9% of
time of diagnosis, approximately 9% of cases will have localized these were able to undergo successful resection, with an R0 re-
disease and 29% regional disease; but in total only 15%–20% will section rate of 79.2%. The authors estimated a median overall sur-
be deemed resectable [1]. Pancreaticoduodenectomy, developed by vival (mOS) for these patients of 20.5 months, which mirrors that
Allen Whipple at Columbia University, is the primary procedure of patients with resectable disease at diagnosis. A major drawback
performed for patients with resectable tumors of the pancreatic of this study was that the analysis was not done by intention-to-
head, where over 65% of pancreatic cancer is discovered [2]. It is a treat which, if performed, would reduce potential bias in treat-
technically challenging operation, given the proximity of the pan- ment effect (survival bias), since clearly not all patients proceed
creas to major vascular structures, and it carries substantial opera- to surgery. In 2017, D’Angelo et al performed one of the first meta-
tive and postoperative risks. Unfortunately, even with a successful analyses to report survival using ITT analysis [14]. Twelve prospec-
R0 resection (pathologically negative margin), neither pancreatico- tive neoadjuvant studies with both resectable and either border-
duodenectomy nor distal pancreatectomy, can guarantee a cure and line resectable and locally advanced PDAC (BR/LA disease), pub-
rates of recurrence approach 80% [3]. lished between 2008 and 2015, were included and the authors
reported a resection rate of 65%, and notably, a similar mOS of
Rationale for neoadjuvant therapy 22.78 months. More recently, Versteijne et al performed a meta-
analysis on pooled data from 38 trials, again using ITT analysis,
1. To increase R0 resection rates by reducing tumor bulk and and included a total of 3,484 patients diagnosed with resectable or
involvement of nearby structures. BR pancreatic cancer [15]. They reported no meaningful difference
2. To downstage disease. in mOS for patients with resectable disease treated with upfront
3. To decrease surgical complexity. surgery versus neoadjuvant therapy (17.14 v 18.2 months) however,
4. To give patients suffering from weight loss or biliary ob- there was a difference in mOS, favoring neoadjuvant therapy over
struction time to improve their performance status for upfront surgery, in patients with BR disease (19.2 v 12.8 months).
surgery. R0 resection rates favored those receiving neoadjuvant therapy, in
5. To ensure that all patients receive chemotherapy. both resectable (85% v 71.4%) and BR disease (88.6% v 63.9%). Sim-
6. To allow emergence of resistant, already present metastatic ilar results were also reported in a large meta-analysis by Suker et
disease, if such exists. al (see below), who examined outcomes with neoadjuvant FOLFIRI-
7. To treat locally invasive disease before resection, as surgery NOX in 315 patients from 11 studies, and reported that 25% of pa-
may alter perfusion in the area. tients were effectively downstaged and able to undergo resection
8. To treat micrometastatic disease earlier, at the time of diag- [16].
nosis. In 2017, Mokdad et al published a unique retrospective study
There are several compelling reasons to consider neoadjuvant utilizing propensity score matched analysis to investigate the role
therapy (Text Box). Since surgical resection represents the only of neoadjuvant therapy in patients with early stage pancreas can-
treatment modality that can lead to cure, increasing resectability cer. The authors queried the National Cancer Database for patients
of borderline resectable (BR) and locally advanced (LA) tumors has with stage I or II PDAC who underwent surgery between 2006 and
been a major focus of recent trials. In addition, by decreasing tu- 2012. A total of 2,005 patients treated with neoadjuvant therapy
mor bulk and vessel involvement, neoadjuvant therapy may fulfill followed by surgery were matched with 6,015 patients who under-
another goal, it may also increase R0 resection rates, which unsur- went upfront resection and the authors reported a median overall
prisingly is associated with improved survival [4]. It is widely rec- survival difference of 5 months (26 v 21 months) favoring those
ognized that pancreatic cancer is both a locally invasive and sys- who received neoadjuvant therapy. Although this study was lim-
temic disease in the majority of patients at presentation, which is ited by inherent selection bias, as the neoadjuvant therapy group
in part supported by the fact that, despite complete resection and was populated with only those patients who tolerated neoadjuvant
adjuvant therapy, disease recurs in the majority of patients and therapy, it demonstrates the utility of neoadjuvant therapy as a se-
5-year survival rates approach 30% [5–9]. Therefore, neoadjuvant lection strategy for management of patients with early stage dis-
therapy offers an opportunity to treat micrometastatic disease ear- ease [17]. A major challenge to the interpretation of these studies
lier, which can be particularly relevant in cases of prolonged sur- is the varying definitions of resectable, BR, and LA disease; a prob-
gical recovery. Significant delays in initiating adjuvant therapy are lem that has only recently been addressed in the NCCN guidelines.
not uncommon and approximately 20% of patients are unable to
recover sufficiently following surgery to receive any adjuvant ther- Defining resectable, borderline resectable, and locally advanced
apy [10]. pancreatic cancer and treatment effect
The two most frequently used neoadjuvant regimens are
mFOLFIRINOX (5-FU, leucovorin, irinotecan and oxaliplatin) and Prior to 2010, no universally adopted criteria defining BR or LA
gemcitabine/nab-paclitaxel. While the choice of one versus the pancreatic cancer existed and definitions varied considerably from
other depends on several factors, there is good evidence that ei- study to study. In an effort to rectify this problem the NCCN for-
ther of these “modern” regimens outperforms older combinations, mally adopted a set of guidelines based on criteria established by
or single agent therapy [11,12]. an expert consensus group (Table 2) [18,19]. Although this has im-
proved clinical trial reporting, it has not eliminated all interin-
Data supporting neoadjuvant therapy stitutional differences impacting decisions regarding resectability.
For example, variability remains in imaging methods and inter-
The increasing focus on neoadjuvant options is supported pri- pretation. In 2012, Katz et al reviewed 122 cases of BR pancre-
marily by data from small, single institution trials and several large atic cancer imaged after neoadjuvant therapy; 84 (69%) had sta-
meta-analyses (Table 1). In one of the largest meta-analyses per- ble disease, 15 (12%) had a partial response, and 23 patients (19%)
formed to-date, Gillen et al pooled data from 111 neoadjuvant tri- had progressive disease [20]. Although only one patient (0.8%)
als conducted between 1966 and 2009 [13]. In total, 4,394 patients had his/her disease downstaged to resectable status by imaging,

Descargado para Anonymous User (n/a) en Universidad El Bosque de ClinicalKey.es por Elsevier en abril 08, 2021. Para uso personal
exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
A.G. Raufi, G.A. Manji and J.A. Chabot et al. / Seminars in Oncology 46 (2019) 19–27 21

Table 1
Meta-analyses supporting a neoadjuvant therapy strategy.

Study Patient population Data Comment


Gillen et al. (2010) • Pooled data from 111 neoadjuvant trials • Amongst patients initially diagnosed with • Estimated mOS of 20.5 months, mirrors
[13] conducted between 1966 and 2009 nonresectable disease who underwent that of patients with resectable disease
neoadjuvant therapy: at diagnosis
• 4,394 patients with PDAC grouped into → 46.9% underwent surgical exploration • Major drawback that analysis not done
two cohorts: resectable and → 69.9% had successful resection by ITT which would reduce potential bias
nonresectable disease → 79.2% had R0 resection in treatment effect (survival bias), since
• Estimated mOS = 20.5 months not all patients proceed to surgery
D’Angelo et al. • Pooled data from 12 prospective • ITT analysis • One of the first meta-analyses to report
(2017) [14] neoadjuvant studies published between survival using ITT analysis
2008 and 2015 • Resection rate = 65%
• 624 patients with resectable, BR, and LA • Similar mOS of 22.78 months • Focused solely on prospective trials
PDAC
Versteijne et al. • Pooled data from 38 trials • ITT analysis • One of the largest ITT meta-analysis
(2018) [15] • 3,484 patients with resectable or BR • Resectable disease performed to date
PDAC → Similar mOS for upfront surgery (17.4
months) v neoadjuvant therapy (18.2
months)
→ R0 rates favored neoadjuvant therapy
(85% v 71.4%)
• BR disease
• mOS favored neoadjuvant therapy (19.2
months) over upfront surgery, (12.8
months)
• R0 rates favored neoadjuvant therapy
(88.6 v 63.9%)
Suker et al. (2016) • Pooled data from 11 studies of • 25% of patients effectively downstaged • Focused on solely on LA PDAC
[16] neoadjuvant FOLFIRINOX and able to undergo resection
• 315 patients with LA PDAC • Estimated mOS = 24.2 months
Mokdad et al., • Retrospective study of National Cancer • Median overall survival of 26 months • Limited by inherent selection bias, as
(2017) [17] Database between 2006 and 2012 with neoadjuvant therapy v 21 months neoadjuvant therapy group populated
• Utilized propensity score matched with upfront resection with those patients who tolerated
analysis to investigate role of neoadjuvant therapy
neoadjuvant therapy in stage I or II PDAC • Demonstrates the utility of neoadjuvant
who underwent surgery therapy as a selection strategy for
• 2,005 patients treated with neoadjuvant management of patients with early stage
therapy followed by surgery matched disease
with 6,015 patients with upfront
resection
Abbreviations: mOS, median overall survival; ITT, intention-to-treat; BR, borderline resectable; LA, locally advanced; PDAC, pancreatic ductal adenocarcinoma

Table 2
Classification for of pancreatic cancer involving the head or uncinated process (NCCN/AHPBA/SSO/SSAT).

Resectable Borderline resectable Locally advanced/Unresectable


SMV/PV No abutment/encasement or ≤180° contact without vein contour Abutment/encasement (≥1800 ), reconstructible Not reconstructible
irregularity
SMA No abutment/encasement Abutment (≤180°) Encasement (≥180°)
CHA No abutment/encasement Abutment or short-segment encasement Long-segment encasement
Celiac trunk No abutment/encasement No abutment/encasement Abutment

SMV, superior mesenteric vein; PV, portal vein; SMA, superior mesenteric artery; CHA, common hepatic artery.

85 patients (66%) underwent pancreatectomy with 81/85 (95%) [25]. To date, no large randomized phase III control trial compar-
achieving an R0 resection, illustrating how inaccurate current ing neoadjuvant therapy to upfront surgery in resectable pancre-
imaging techniques are at predicting disease status postneoadju- atic cancer has been published; however, several small trials sug-
vant therapy. To mitigate these discrepancies and facilitate patient gest the merit of this strategy (Table 3). A nonrandomized study of
selection for surgery, structured reporting strategies have been de- neoadjuvant gemcitabine plus cisplatin given to 28 patients with
veloped, but have yet to be widely adopted [21]. Ultimately, the resectable disease found that 93% remained resectable and 25/28
question of what a surgeon feels comfortable with will continue to (89%) completed pancreaticoduodenectomy with an R0 resection
vary, and will likely continue to impact treatment decisions [22– rate in 20/25 (80%) [26]. At 26.5 months, the mOS in this trial
24]. was significantly higher than seen a decade earlier with the same
agents [27]. A similar 27.2 month mOS was reported in 2014 by
Trials with neoadjuvant therapy in patients with resectable disease O’Reilly et al combining gemcitabine and oxaliplatin [28].
A currently unresolved question in resectable pancreatic can-
Whether patients with resectable disease should undergo cer treatment is whether or not the addition of neoadjuvant ra-
neoadjuvant treatment remains highly controversial. While there diation to chemotherapy improves outcomes. Several early phase
are compelling reasons to treat (Text Box), there is a risk that some clinical trials have studied the tolerability and efficacy of com-
patients will not only fail to benefit but progress to unresectable bining radiation and chemotherapy in this setting (Table 3). Two
disease. This “failure rate” has been reported to be as high as 16% studies randomized patients to upfront surgery or neoadjuvant

Descargado para Anonymous User (n/a) en Universidad El Bosque de ClinicalKey.es por Elsevier en abril 08, 2021. Para uso personal
exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
22 A.G. Raufi, G.A. Manji and J.A. Chabot et al. / Seminars in Oncology 46 (2019) 19–27

Table 3
Neoadjuvant clinical trials for patients with resectable pancreatic cancer.

Trial N Therapy Resection rate (%) R(0) Resection rate (%) Median survival
Palmer et al. (2007) [27] 50 A: Gemcitabine 1 g/m2 every 7 days for 43 days A: 9/24 (38) A: 6/9 (75) A: 9.9 months
B: Gemcitabine 1 g/m2 + cisplatin 25 mg/m2 every 7 days B: 18/26 (70) B: 12/18 (75) B: 16.6 months
(omitting day 22 or days 15 and 36 [revised schedule])
Heinrich et al. (2008) [26] 28 Gemcitabine 1 g/m2 + cisplatin 50 mg/m2 biweekly for 4 25/28 (89) (20/25) 80 26.5 months
cycles
O’Reilly et al. (2014) [28] 38 Gemcitabine 1 g/m2 + oxaliplatin 80 mg/m2 biweekly for 4 27/38 (71) 20/27 (74) 27.2 months
cycles
Golcher et al. (2015) [29] 66 A: Surgery A: 23/33 (70) A: 16/23 (70) A: 14.4 months
B: Gemcitabine 300 mg/m2 + cisplatin 30 mg/m2 days 1, 8, B: 19/33 (58) B: 17/19 (89) B: 17.4 months
22, 29 with RT
Casadei et al. (2015) [30] 38 A: Surgery A: 15/20 (75) A: 5/15 (33) A: 19.5 months
B: Gemcitabine 1 g/m2 days 1, 8, every 21 days x 2 cycles B: 11/18 (61) B: 7/11 (64) B: 27.5 months
followed by gemcitabine 50 mg/m2 twice weekly for a
total of 6 weeks + RT

Table 4
Neoadjuvant clinical trials for patients with borderline resectable pancreatic cancer.

Trial N Therapy Resection rate (%) R(0) Resection rate (%) Median survival
2 2
Motoi et al. (2013) [31] 35 Gemcitabine 1,0 0 0 mg/m days 1 and 8 with S-1 40 mg/m BID 30/35 (86) 26/30 (87) 19.7 months
for 14 days every 21 days x 2 cycles
Landry et al. (2010) 21 A: Gemcitabine 500 mg/m2 weekly for 6 weeks with RT A: 3/10 (30) Not reported A: 19.4 months
[32]
2
B: Gemcitabine 175 mg/m on days 1, 5, 29, and 33, cisplatin 20 B: 2/11 (18) B: 13.4 months
mg/m2 on days 1–5 and 29–32, 5-FU 600 mg/m2 on days 1–5
and 29–32 followed by radiation with continuous infusion 5-FU
225 mg/m2 for 6 weeks
Kim et al. (2013) [33] 68 Gemcitabine 1 g/m2 on days 1, 8, 15 + oxaliplatin 85 mg/m2 on 43/68 (63) 36/43 (84) 18.2 months
days 1, 15 every 21 days for 2 cycles with RT, 30 Gy

Katz et al. (2016) [34] 22 modified FOLFIRINOX (85 mg/m2 of oxaliplatin, 180 mg/m2 of 15/22 (68) 14/15 (93) 21.7 months
irinotecan hydrochloride, 400 mg/m2 of leucovorin calcium, and
2,400 mg/m2 of 5-FU) followed by 5.5 weeks of external-beam
RT (50.4 Gy) concurrently with capecitabine 825 mg/m2 BID
Murphy et al. (2018) 48 FOLFIRINOX (85 mg/m2 oxaliplatin, 400 mg/m2 bolus 5-FU on day 32/48 (66) 31/32 (97) 37.7 months
[35] 1, then 2,400 mg/m2 continuous infusion for 46 hours.
Leucovorin calcium, 400 mg/m2 , and irinotecan hydrochloride,
180 mg/m2 ) x8 cycles. Upon restaging, patients with resolution
of vascular involvement received RT (5 Gy × 5) with
capecitabine. Patients with persistent vascular involvement
received RT with 5-FU or capecitabine.
van Tienhoven et al. 246 A: Surgery A: 91/127 (72) A: 28/91 (31) A: 13.5 months
(2018) [36]
B: 15 times of 2.4 Gray (Gy) combined with gemcitabine, 1,0 0 0 B: 72/119 (60) B: 45/72 (63) B: 17.1 months
mg/m2 on days 1, 8, and 15, preceded and followed by a cycle of
gemcitabine

Trial included patients with resectable disease.

therapy with chemo-RT suggested better overall survival with which patients were treated with neoadjuvant modified FOLFIRI-
neoadjuvant therapy, notwithstanding lower resection rates were NOX with RT [34]. Using current NCCN classifications, 22 patients
reported in this group. Results from the two studies were simi- with BR disease were selected and treated with four cycles of mod-
lar: 70%–75% of those assigned to upfront surgery compared with ified FOLFIRINOX followed by 5.5 weeks of external-beam radiation
57.5%–61% of those assigned to the neoadjuvant arm underwent therapy concurrently with capecitabine prior to surgery. The au-
resection [29,30]. Thus, among patients with resectable disease, thors reported that despite significant ≥ grade 3 (64%), 15 (68%)
neoadjuvant therapy does not significantly reduce resection rates, patients were able to undergo pancreatectomy with an R0 resec-
but with a highest reported mOS of 27.5 months, the addition of tion in 14/15 (93%). The mOS for these patients was 21.7 months
RT to chemotherapy does not appear to significantly outperform from time of registration, supporting further study of this regimen
chemotherapy alone. As noted above, patient populations in these in a phase III trial. Another single arm phase II trial with 8 cycles
studies are often not comparable and the question of neoadjuvant of neoadjuvant FOLFIRINOX followed by RT given to 48 patients
therapy for resectable disease remains far from settled. with BR pancreatic cancer showed similar findings. [35]. Among
the 32 patients who underwent resection, 31/32 (97%) achieved an
Trials with neoadjuvant therapy in patients with borderline R0 resection and the overall mOS was 37.7 months. The random-
resectable disease ized phase III PREOPANC trial, which compared upfront surgery to
neoadjuvant combination gemcitabine and RT in patients with re-
The role of neoadjuvant chemotherapy for the treatment of bor- sectable and BR disease, was presented at the 2018 ASCO Meeting
derline resectable pancreatic cancer has also been investigated in [36]. The study demonstrated superiority of neoadjuvant chemoRT
several phase II clinical trials, many of which have focused on with a mOS of 17.1 months compared to 13.5 months in those re-
the addition of RT to chemotherapy (Table 4) [31–33]. Katz et al ceiving surgery alone (P = 0.074). Furthermore, although the re-
reported the results of the Alliance for Clinical Trials in Oncol- section rate in the preoperative arm was lower than that of the
ogy Trial A021101, a prospective, multicenter, single-arm trial in surgery arm (60% v 72%), the R0 resection rate was substantially

Descargado para Anonymous User (n/a) en Universidad El Bosque de ClinicalKey.es por Elsevier en abril 08, 2021. Para uso personal
exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
A.G. Raufi, G.A. Manji and J.A. Chabot et al. / Seminars in Oncology 46 (2019) 19–27 23

Table 5
Neoadjuvant clinical trials for patients with locally advanced pancreatic cancer.

Trial N Therapy Resection R(0) Resection Median


rate (%) rate (%) survival
Herman et al. (2015) [37] 49 Gemcitabine (1,0 0 0 mg/m2 ) x3 doses followed by RT (33.0 Gy) 4/49 (8) 4/4 (100) 13.9 months
Ikeda et al. (2013) [39] 60 S-1 80 mg/m2 BID with RT (50.4 Gy) in 28 fractions over 5.5 weeks 2/60 (3) Not reported 16.2 months
Marthey et al. (2015) [43] 77 Folfirinox oxaliplatin (85 mg/m2 ), leucovorin (400 mg/m2 ), irinotecan 28/77 (36) 25/28 (89) 22 months
(180 mg/m2 ), 5-FU (400 mg/m2 bolus with a 46-hour continuous
infusion of 2.4 g/m2 ) every 2 weeks
Sherman et al. (2015) [38] 45 GTX capecitabine (1500 mg/m2 , days 1–14), gemcitabine (750 mg/m2 , 40/45 (89) 28/40 (70) 29 months
days 4 and 11) and docetaxel (30 mg/m2 , days 4 and 11) followed by
GX/RT if extensive arterial involvement

higher (63% v 31%). A follow-up Alliance study, A021501, is cur- ment was delivered every 2 weeks until disease progression, pa-
rently ongoing for BR disease, randomizing 134 patients to re- tient refusal, unacceptable toxicity, or consolidation therapy with
ceive FOLFIRINOX with or without stereotactic body frame radia- external radiotherapy/surgery. While no complete responses were
tion therapy (SBRT). The added value of RT in the BR setting has reported, partial responses, based on RECIST (Response Evaluation
been difficult to discern; notably the Alliance study was recently Criteria in Solid Tumors) criteria, were noted in 22/77 (28%) pa-
suspended following interim analysis. tients, and 43/77 (56%) patients had stable disease. In total, 28/77
(36%) patients underwent a surgical resection with a reported R0
resection rate of 89% (25/28). Notably, 24/28 (86%) subjects were
Trials with neoadjuvant therapy in patients with locally advanced
treated with external radiotherapy prior to surgery and the mOS
disease
for all patients enrolled on this trial was 22 months.

A large proportion of patients diagnosed with PDAC are found


to have unresectable LA disease. Although by definition these pa- Currently used regimens for neoadjuvant chemotherapy in
tients have no evidence of metastatic spread, treatment strategies patients with BR or LA disease
have focused on the use of systemic chemotherapy alone. Conver-
sion to BR or resectable disease has traditionally been exceedingly FOLFIRINOX
rare, however, several studies have reported more promising re-
sults (Table 5) [37,38]. A Japanese trial investigating S-1 (a combi- The two preferred regimens for treating patients with BR or
nation of tegafur, gimeracil, and oteracil potassium) combined with LA PDAC are modified FOLFIRINOX and gemcitabine/nab-paclitaxel.
RT in 60 patients with LA PDAC demonstrated favorable responses The rationale for using these combinations is based on data gath-
and tolerability [39]. In this trial, 16 (27%) patients achieved a ered in the metastatic setting. The activity of FOLFIRINOX was ini-
partial response but only two (3%) patients had disease that was tially validated by Conroy et al who, in 2011, published their land-
downstaged and eventually resected. mark findings of a median survival of 11.1 months with this com-
In 2012, Hosein et al published the results of a retrospective bination compared to 6.8 months with gemcitabine alone [44]. Pa-
series evaluating the role of full-dose neoadjuvant FOLFIRINOX in tients in the FOLFIRINOX group had higher rates of toxicity com-
18 patients with locally advanced PDAC [40]. Although this trial pared to gemcitabine and in the United States most clinicians
did include several patients with borderline resectable disease, it have found it more toxic than originally reported. This has re-
represents the first published data including North American pa- sulted in preferential use of modified regimens, with dose-reduced
tients with LA PDAC treated with FOLFIRINOX. At maximum re- irinotecan or omission of the bolus portion of the 5-FU infusion.
sponse or tolerability, 7/18 (39 %) were considered resectable and These “modified FOLFIRINOX” (mFOLFIRINOX) regimens with vary-
went to surgery. More recently, Blazer et al published a retro- ing drug doses and intensity (Table 6) have been studied in both
spective study on 43 patients with BR and LA PDAC treated with the metastatic and the neoadjuvant settings, but have never been
modified FOLFIRINOX [41]. Overall the regimen was well toler- directly compared to the original full dose regimen [40,41,45].
ated, and of those who were initially diagnosed with LA disease Given that the goals of chemotherapy in the metastatic and neoad-
based on NCCN guidelines, resection was accomplished in 11 of juvant setting are different, with the former to slow and control
25 (44%) cases. Of these patients, 10/11 (91%) had an R0 resec- disease and the latter to shrink and eradicate micrometastatic dis-
tion. The median OS had not been reached at the time of publi- ease, different therapeutic strategies should be acknowledged. Be-
cation. In 2015, Nanda et al performed a single institution retro- cause of the steep dose response curve for cytotoxic chemother-
spective study that included 29 patients with LA pancreatic cancer apy, we cannot assume that modifying FOLFIRINOX in all patients
who were also treated with modified FOLFIRINOX with or without will provide comparable benefit. Without data to the contrary, in
additional chemoRT prior to surgery [42]. Among these patients, considering FOLFIRINOX for neoadjuvant therapy, physicians should
41.3% were able to undergo surgery and 83% achieved an R0 re- strive for full and on-schedule dosing.
section. One-year PFS was 49.2% and OS was 65.5% while the mOS However, there is evidence that mFOLFIRINOX may be suffi-
for this cohort was 18.6 months. These results have been further cient for neoadjuvant therapy. Suker et al, reviewed 11 studies with
validated in several prospective studies described below. The high 315 treatment-naive patients who received FOLFIRINOX as first-line
R0 rates suggest that while chemotherapy may not convert locally therapy for LA PDAC, finding that approximately one-quarter of pa-
advanced disease to a resectable state, when it does, the response tients were able to undergo surgical resection after treatment with
can be profound. an R0 resection rate of 75% and mOS of 24.2 months [16]. Impor-
Radiation therapy has also been utilized to aid chemotherapy in tantly, two of these studies used dose-modified FOLFIRINOX yet
down staging of LA disease. Marthey et al reported a prospective reported an mOS of 21.2 months (Mahaseth et al, which did not
phase II study in 77 patients with LA PDAC treated with full dose give bolus 5-FU [46]) and 26 months (Sadot et al, which adminis-
FOLFIRINOX with or without neoadjuvant radiation therapy [43]. tered chemotherapy at 80% dose intensity [47]). Furthermore, pre-
Patients were enrolled at 11 French medical centers where treat- liminary data from the PRODIGE 24/CCTG PA.6 trial, presented at

Descargado para Anonymous User (n/a) en Universidad El Bosque de ClinicalKey.es por Elsevier en abril 08, 2021. Para uso personal
exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
24 A.G. Raufi, G.A. Manji and J.A. Chabot et al. / Seminars in Oncology 46 (2019) 19–27

Table 6
Modified FOLFIRINOX dosing regimens.

Trial N Therapy Dose (MG/M2) Resection R(0) Resection Median survival


rate (%) rate (%)
Mahaseth et al. (2013) [46]∗ 24 Oxaliplatin 85 42 83.3 17.8 months
Irinotecan 180
5-FU CIV 2,400
Marthey et al. (2015) [43] 77 Oxaliplatin 85 36 89 22 months
Irinotecan 180
5-FU bolus 400
5-FU CIV 2,400
Nanda et al. (2015) [42] 29 Oxaliplatin 85 41.3 83 18.6 months
Irinotecan 180
5-FU CIV 2,400
Sadot et al. (2015) [47] 101 Oxaliplatin 68 31 55 25 months
Irinotecan 144
5-FU bolus 320
5-FU CIV 1,920
Blazer et al. (2015) [41] 39 Oxaliplatin 85 51.1 86.4 18 months
Irinotecan 165
5-FU CIV 2,400
Katz et al. (2016) [34] 22 Oxaliplatin 85 68 93 21.7 months
Irinotecan 180
5-FU CIV 2,400
Stein et al. (2016) [49]∗ 31 Oxaliplatin 85 41.9 100 26.6 months
Irinotecan 135
5-FU bolus 300
5-FU CIV 2,400
Conroy et al. (2018) [48]∗∗ 247 Oxaliplatin 85 N/A N/A 54 months
Irinotecan 180 (reduced to 150 after patient #162)
5-FU CIV 2,400

Abbreviations: 5-FU, 5-fluorouracil; CIV, continuous intravenous infusion.



Trial included patients with borderline resectable and locally advanced (BR/LA) as well as metastatic disease—reported here are the results of the BR/LA cases.
∗∗
mFOLFRINOX given as adjuvant therapy.

the 2018 ASCO conference, revealed the striking activity of modi- these studies are being conducted in the metastatic setting,
fied FOLFIRINOX in the adjuvant setting [48]. Several phase II stud- and several have reported negative findings, MM-141 [51]; necu-
ies also lend support to the notion that modified FOLFIRINOX may paranib [52]; and demcizumab [53], leaving the combination of
have significant efficacy in both the metastatic and neoadjuvant gemcitabine/nab-paclitaxel as the only proven combination to
settings (Table 6) [49]. Nonetheless, studies are needed to deter- date.
mine best dose and schedule for modified FOLFIRINOX. In the neoadjuvant setting, only single institution experiences
have been reported. In 2016, Ielpo et al reported on 25 patients
Gemcitabine/nab-paclitaxel with resectable and BR PDAC defined according to the NCCN guide-
lines who received neoadjuvant gemcitabine/nab-paclitaxel [54].
Gemcitabine plus nab-paclitaxel (Abraxane) is the regimen con- The resection rate among all patients was 68% (17/25 patients),
sidered to most closely approximate the activity of FOLIRINOX and 73% (8/11 patients) among those with BR disease; the R0
in metastatic PDAC. Its role in pancreatic cancer was established resection rate was 100% in all resectable cases. Among patients
by a large phase III trial that randomized 861 patients with who underwent surgery, mOS was 21 months and mDFS was 19
metastatic PDAC to either the combination of gemcitabine and nab- months.
paclitaxel or gemcitabine alone and demonstrated an improvement
in median overall survival of 8.5 months from the gemcitabine Gemcitabine/docetaxel/capecitabine (GTX)
plus nab-paclitaxel combination compared to 6.7 months for sin-
gle agent gemcitabine [50]. Because gemcitabine/nab-paclitaxel Investigators at Columbia University first described the 3-drug
is well tolerated, and has been studied in patients over 70 regimen GTX for use in the PDAC in 2004 and this combination has
years of age with slightly worse performance status, this com- subsequently been tested in the neoadjuvant setting [55]. Sherman
bination has rapidly been adopted in the community. Further- et al described 45 patients with LA pancreatic cancer (American
more, unlike FOLFIRINOX, this regimen can be easily combined Joint Committee on Cancer stage III) who were treated with 6 cy-
with other agents, and many investigators are currently com- cles of neoadjuvant GTX [38]. Among the 34 patients with arte-
bining the gemcitabine/nab-paclitaxel platform with other ther- rial involvement, 29 (85%) patients underwent subsequent resec-
apies in an effort to further improve outcomes. Examples in- tion, with 20/29 (69%) patients achieving an R0 resection. All 11
clude addition of: (1) the immune checkpoint inhibitor nivolumab patients with venous-only involvement underwent resection. The
(NCT02309177); (2) the matrix degrading hyaluronidase PEGPH20, median OS among those with arterial involvement was 29 months
(NCT02715804); (3) the anti-IGF-1R/anti-ErbB3 bispecific mon- and in those with venous involvement, the median OS had not
oclonal antibody MM-141 (NCT02399137); (4) the multitarget- been reached at the time of publication (>42 months). A follow-up
ing heparan sulfate mimetic, necuparanib (NCT01621243); (5) the analysis failed to reveal any presenting parameters capable of pre-
selective inhibitor of nuclear export selinexor (NCT02178436); dicting response to neoadjuvant treatment with this regimen [56].
(6) the inhibitor of notch-mediated signaling and transcrip- From a health economic perspective, GTX is significantly less costly
tion demcizumab (NCT02289898); and (7) the hormone in- when compared to gemcitabine/nab-paclitaxel or FOLFIRINOX but
hibitor enzalutamide (NCT02138383). Of note, the majority of has yet to become widely adopted.

Descargado para Anonymous User (n/a) en Universidad El Bosque de ClinicalKey.es por Elsevier en abril 08, 2021. Para uso personal
exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
A.G. Raufi, G.A. Manji and J.A. Chabot et al. / Seminars in Oncology 46 (2019) 19–27 25

Table 7
Selected ongoing neoadjuvant clinical trials.

Trial Number Phase Target Therapy Primary endpoints


planned population
NCT02839343 134 II BR A: mFOLFIRINOX Overall survival
B: mFOLFIRINOX + RT
NCT02676349 90 II BR A: mFOLFIRINOX R0 resection rate
B: mFOLFIRINOX + RT
NCT03199144 70 II BR Nab-paclitaxel + gemcitabine + RT Overall survival
NCT03099265 28 II BR mFOLFIRINOX + SBRT R0 resection rate
NCT02723331 50 II R/BR Nab-paclitaxel and gemcitabine + SBRT R0 resection rate
NCT02125136 168 II BR/LA A: Nab-paclitaxel + gemcitabine Conversion rate
B: Nab-paclitaxel + gemcitabine followed by FOLFIRINOX
NCT02562716 112 II R A: mFOLFIRINOX Overall survival
B: Nab-paclitaxel + gemcitabine
NCT02305186 56 Ib/II R/BR Neoadjuvant CRT + pembrolizumab Safety and number of TILs
per HPF
NCT02749136 44 II BR mFOLFIRINOX PFS at 1 year

Abbreviations: R, resectable; BR, borderline resectable; LA, locally advanced; TILS, tumor infiltrating lymphocytes; HPF, high-power field; PFS, progression-free survival;
NAB-PACLITAXEL, nanoparticle albumin-bound paclitaxel (Abraxane).

Complexities of neoadjuvant therapy domized or real-world comparative data are desperately needed.
Until such data are available, treatment practices will vary.
The above studies demonstrate the many complexities sur-
rounding neoadjuvant trials in pancreatic cancer. Trials performed
Ongoing trials with neoadjuvant therapy in patients with resectable,
to date have often been underpowered and performed at sin-
BR, or LA pancreatic cancer
gle institutions and comparing results between trials is difficult
given the variability in treatment regimens, dosing, and lack of
Ongoing phase II clinical trials incorporating patients with bor-
uniform metrics defining resectable, BR, and LA disease. Tolerabil-
derline resectable pancreatic cancer will hopefully shed light on
ity of FOLFIRINOX is a prime example of this issue. Studies per-
optimal neoadjuvant regimens in this setting (Table 7). Several
formed in Europe typically have used one standard dosing regimen
investigate the role of newer neoadjuvant combinations such as
of FOLFIRINOX whereas those performed in the United States have
FOLFIRINOX or gemcitabine/nab-paclitaxel with or without RT.
often modified and dose reduced this combination. It remains un-
For example, the phase II Alliance A021501 (NCT02839343) trial,
clear what effect, if any, significant dose reductions have on out-
which is currently underway in the United States, randomizes pa-
comes. As discussed above, while reports of modified versions of
tients with BR pancreatic cancer to either neoadjuvant treatment
FOLFIRINOX have suggested little impact of dose reduction, large
with mFOLFIRINOX × 8 cycles or mFOLFIRINOX × 7 cycles + 5 days
randomized studies may contest these findings in the neoadjuvant
of RT followed by FOLFOX x 4 cycles. The PANDAS-PRODIGE44
setting, where maximum efficacy is needed. In addition, many tri-
study (NCT02676349), which opened in France in 2016, also com-
als under-report performance status of patients, which plays an
pares neoadjuvant mFOLFIRINOX to mFOLFIRINOX with RT. In this
important role in patient selection, especially with FOLFIRINOX or
study, patients with BR disease are randomly selected for treat-
radiation therapy. Another factor impacting interpretation and gen-
ment with neoadjuvant mFOLFIRINOX with or without preopera-
eralizability of these studies is the fact that many have been per-
tive chemoRT with capecitabine. There are also multiple single-
formed at single institutions with significant experience in treating
arm trials combining gemcitabine and nab-paclitaxel with SBRT.
patients with pancreatic cancer. This hampers the ability to apply
These include the CKPancreas (NCT03199144) trial, open in Luxem-
these results to real world practice.
bourg; and a University of Colorado trial (NCT02723331) which is
Determining the role of radiation therapy has been a complex
enrolling both resectable and BR PDAC. The phase III NEOLAP trial
issue and due to changes in technology, data from early trials can-
(NCT02125136) will intensify therapy by following nab-paclitaxel
not be easily extrapolated. Unfortunately, few trials directly com-
and gemcitabine with FOLFIRINOX in patients with BR or LA tu-
pare chemotherapy to chemoRT in the neoadjuvant setting and
mors. In addition, the SWOG 1505 (NCT02562716) trial, which
those that do are often underpowered to detect statistically sig-
has completed accrual, directly compares neoadjuvant mFOLFIRI-
nificant differences. Conventional external beam radiation therapy
NOX to gemcitabine plus nab-paclitaxel in resectable disease.
(EBRT) is rapidly being replaced by SBRT, which provides pre-
Lastly, an interesting randomized multicenter phase Ib/II study
cise delivery of radiation while sparing surrounding normal tis-
(NCT02305186) is underway and will assess the safety and the im-
sues. Two recent retrospective case series evaluated outcomes in
munological effects of chemoRT in combination with immunother-
patients with either BR or LA disease who received SBRT in the
apy (pembrolizumab) in patients with resectable or BR pancreatic
neoadjuvant setting. The first, reported data from Moffitt Cancer
cancer.
Center where a total of 159 patients were treated with chemother-
apy for 2–3 months followed by SBRT (25–30 Gy) [57]. The esti-
mated mOS was 19.2 months for BRPC patients and 15.0 months Conclusions
for LA PDAC patients (P = 0.402). In the second study, performed at
Johns Hopkins University, 88 patients (14 patients with BR and 74 Despite recent ground-breaking advances in the field of on-
patients with LA PDAC) were treated with neoadjuvant chemother- cology as a whole, considerable gains are needed for patients
apy followed by SBRT (25–33 Gy) [58]. Of the 19 (22%) patients diagnosed with pancreatic cancer. There is mounting evidence
who underwent surgery following SBRT, 84% had an R0 resection. that neoadjuvant therapy is able to not only downstage tumors
The mOS of resected patients was 20.2 months. While many in- and increase resectability, but also treats micrometastatic cancer
vestigators consider SBRT valuable in the neoadjuvant setting, ran- earlier in the disease course. Currently, there are considerable

Descargado para Anonymous User (n/a) en Universidad El Bosque de ClinicalKey.es por Elsevier en abril 08, 2021. Para uso personal
exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
26 A.G. Raufi, G.A. Manji and J.A. Chabot et al. / Seminars in Oncology 46 (2019) 19–27

challenges for clinicians presented with cases of BR or LA pancre- [18] Tempero MA, Malafa MP, Al-Hawary M, et al. Pancreatic adenocarcinoma, ver-
atic cancer and much of this stems from inexact data. This disease sion 2.2017, NCCN clinical practice guidelines in oncology. J Natl Compr Cancer
Netw: JNCCN 2017;15(8):1028–61.
requires a multidisciplinary approach to optimize outcomes. The [19] Callery MP, Chang KJ, Fishman EK, Talamonti MS, William Traverso L,
groundwork to studying neoadjuvant therapy for this patient Linehan DC. Pretreatment assessment of resectable and borderline re-
population depends on clear staging and imaging guidelines and sectable pancreatic cancer: expert consensus statement. Ann Surg Oncol
2009;16(7):1727–33.
well designed, properly powered, phase III control trials. Until [20] Katz MH, Fleming JB, Bhosale P, et al. Response of borderline resectable pan-
more convincing evidence arises, regimens with proven activ- creatic cancer to neoadjuvant therapy is not reflected by radiographic indica-
ity in the metastatic setting, such as modified FOLFIRINOX and tors. Cancer 2012;118(23):5749–56.
[21] Al-Hawary MM, Francis IR, Chari ST, et al. Pancreatic ductal adenocarci-
gemcitabine/nab-paclitaxel, will remain the treatment of choice
noma radiology reporting template: consensus statement of the Society of
for this patient population. Abdominal Radiology and the American Pancreatic Association. Radiology
2014;270(1):248–60.
[22] Simunovic M, To T, Theriault M, Langer B. Relation between hospital surgi-
Conflict of interest cal volume and outcome for pancreatic resection for neoplasm in a publicly
funded health care system. CMAJ: Can Med Assoc J 1999;160(5):643–8.
No known conflicts of interest for Raufi, Chabot and Bates. [23] Finlayson EV, Goodney PP, Birkmeyer JD. Hospital volume and operative mor-
tality in cancer surgery: a national study. Arch Surg (Chicago, Ill: 1960)
Dr. Manji has received research funding for a clinical trial from 2003;138(7):721–5 discussion 726.
Plexxikon, MERCK, Roche/Genentech. Research funding has also [24] Gooiker GA, Lemmens VE, Besselink MG, et al. Impact of centralization
been received from MERCK and Roche/Genentech. of pancreatic cancer surgery on resection rates and survival. Br J Surg
2014;101(8):10 0 0–5.
[25] Zhan HX, Xu JW, Wu D, et al. Neoadjuvant therapy in pancreatic cancer:
Supplementary materials a systematic review and meta-analysis of prospective studies. Cancer Med.
2017;6(6):1201–19.
[26] Heinrich S, Pestalozzi BC, Schafer M, et al. Prospective phase II trial of neoad-
Supplementary material associated with this article can be juvant chemotherapy with gemcitabine and cisplatin for resectable adenocar-
found, in the online version, at doi:10.1053/j.seminoncol.2018.12. cinoma of the pancreatic head. J Clin Oncol 2008;26(15):2526–31.
002. [27] Palmer DH, Stocken DD, Hewitt H, et al. A randomized phase 2 trial of neoad-
juvant chemotherapy in resectable pancreatic cancer: gemcitabine alone versus
gemcitabine combined with cisplatin. Ann Surg Oncol 2007;14(7):2088–96.
References [28] O’Reilly EM, Perelshteyn A, Jarnagin WR, et al. A single-arm, nonrandomized
phase II trial of neoadjuvant gemcitabine and oxaliplatin in patients with re-
[1] Howlader N NA, Krapcho M, Miller D, Bishop K, Altekruse SF, Kosary CL, Yu M, sectable pancreas adenocarcinoma. Ann Surg 2014;260(1):142–8.
Ruhl J, Tatalovich Z, Mariotto A, Lewis DR, Chen HS, Feuer EJ, Cronin KA, ed- [29] Golcher H, Brunner TB, Witzigmann H, et al. Neoadjuvant chemoradiation ther-
itors. SEER Cancer Statistics Review, MD: National Cancer Institute Bethesda; apy with gemcitabine/cisplatin and surgery versus immediate surgery in re-
1975. 2013. sectable pancreatic cancer: results of the first prospective randomized phase
[2] Greenlee RT, Murray T, Bolden S, Wingo PA. Cancer statistics, 20 0 0. CA II trial. Strahlentherapie und Onkologie: Organ der Deutschen Rontgengesellschaft
20 0 0;50(1):7–33. [et al] 2015;191(1):7–16.
[3] Kleeff J, Korc M, Apte M, et al. Pancreatic cancer. Nat Rev Dis Primers [30] Casadei R, Di Marco M, Ricci C, et al. Neoadjuvant chemoradiotherapy and
2016;2:16022. surgery versus surgery alone in resectable pancreatic cancer: a single-center
[4] Fatima J, Schnelldorfer T, Barton J, et al. Pancreatoduodenectomy for duc- prospective, randomized, controlled trial which failed to achieve accrual tar-
tal adenocarcinoma: implications of positive margin on survival. Arch Surg gets. J Gastrointest Surg 2015;19(10):1802–12.
2010;145(2):167–72. [31] Motoi F, Ishida K, Fujishima F, et al. Neoadjuvant chemotherapy with gem-
[5] Carpelan-Holmstrom M, Nordling S, Pukkala E, et al. Does anyone survive pan- citabine and S-1 for resectable and borderline pancreatic ductal adenocarci-
creatic ductal adenocarcinoma? A nationwide study re-evaluating the data of noma: results from a prospective multi-institutional phase 2 trial. Ann Surg
the Finnish Cancer Registry. Gut 2005;54(3):385–7. Oncol 2013;20(12):3794–801.
[6] Oettle H, Neuhaus P, Hochhaus A, et al. Adjuvant chemotherapy with gemc- [32] Landry J, Catalano PJ, Staley C, et al. Randomized phase II study of gemcitabine
itabine and long-term outcomes among patients with resected pancreatic can- plus radiotherapy versus gemcitabine, 5-fluorouracil, and cisplatin followed by
cer: the CONKO-001 randomized trial. JAMA 2013;310(14):1473–81. radiotherapy and 5-fluorouracil for patients with locally advanced, potentially
[7] Neoptolemos JP, Stocken DD, Bassi C, et al. Adjuvant chemotherapy with fluo- resectable pancreatic adenocarcinoma. J Surg Oncol 2010;101(7):587–92.
rouracil plus folinic acid vs gemcitabine following pancreatic cancer resection: [33] Kim EJ, Ben-Josef E, Herman JM, et al. A multi-institutional phase 2 study
a randomized controlled trial. JAMA 2010;304(10):1073–81. of neoadjuvant gemcitabine and oxaliplatin with radiation therapy in patients
[8] Bilimoria KY, Bentrem DJ, Ko CY, Stewart AK, Winchester DP, Talamonti MS. with pancreatic cancer. Cancer 2013;119(15):2692–700.
National failure to operate on early stage pancreatic cancer. Ann Surg [34] Katz MHG, Shi Q, Ahmad SA, et al. Preoperative modified FOLFIRINOX treat-
2007;246(2):173–80. ment followed by capecitabine-based chemoradiation for borderline resectable
[9] Neoptolemos JP, Palmer DH, Ghaneh P, et al. Comparison of adjuvant gemc- pancreatic cancer: alliance for clinical trials in oncology trial A021101. JAMA
itabine and capecitabine with gemcitabine monotherapy in patients with re- Surg 2016;151(8) e161137–e161137.
sected pancreatic cancer (ESPAC-4): a multicentre, open-label, randomised, [35] Murphy JE, Wo JY, Ryan DP, et al. Total neoadjuvant therapy with FOLFIRINOX
phase 3 trial. Lancet 2017;389(10073):1011–24. followed by individualized chemoradiotherapy for borderline resectable pan-
[10] Klinkenbijl JH, Jeekel J, Sahmoud T, et al. Adjuvant radiotherapy and 5-fluo- creatic adenocarcinoma: a phase 2 clinical trial. JAMA Oncol 2018;4(7):963–9.
rouracil after curative resection of cancer of the pancreas and periampullary [36] van Tienhoven G. Journal of clinical oncology: official journal of the American
region: phase III trial of the EORTC gastrointestinal tract cancer cooperative Society of Clinical Oncology. 2018;36(18). doi:10.1200/JCO.2018.36.18_suppl.
group. Ann Surg 1999;230(6):776–82 discussion 782-774. LBA4002.
[11] Bednar F, Zenati MS, Steve J, et al. Analysis of predictors of resection and [37] Herman JM, Chang DT, Goodman KA, et al. Phase 2 multi-institutional
survival in locally advanced Stage III pancreatic cancer: does the nature of trial evaluating gemcitabine and stereotactic body radiotherapy for pa-
chemotherapy regimen influence outcomes? Ann Surg Oncol 2017;24:1406–13. tients with locally advanced unresectable pancreatic adenocarcinoma. Cancer
[12] Wang Y, Camateros P, Cheung WY. A real-world comparison of FOLFIRINOX, 2015;121(7):1128–37.
gemcitabine plus nab-Paclitaxel, and gemcitabine in advanced pancreatic can- [38] Sherman WH, Chu K, Chabot J, et al. Neoadjuvant gemcitabine, docetaxel, and
cers. J Gastrointest Cancer 2017. doi:10.1007/s12029- 017- 0028- 5. capecitabine followed by gemcitabine and capecitabine/radiation therapy and
[13] Gillen S, Schuster T, Meyer Zum Buschenfelde C, Friess H, Kleeff J. Preopera- surgery in locally advanced, unresectable pancreatic adenocarcinoma. Cancer
tive/neoadjuvant therapy in pancreatic cancer: a systematic review and meta– 2015;121(5):673–80.
analysis of response and resection percentages. PLoS Med 2010;7(4):e10 0 0267. [39] Ikeda M, Ioka T, Ito Y, et al. A multicenter phase II trial of S-1 with concurrent
[14] D’Angelo F, Antolino L, Farcomeni A, et al. Neoadjuvant treatment in pancre- radiation therapy for locally advanced pancreatic cancer. Int J Radiat Oncol Biol
atic cancer: evidence-based medicine? A systematic review and meta-analysis. Phys 2013;85(1):163–9.
Med Oncol 2017;34(5):85. [40] Hosein PJ, Macintyre J, Kawamura C, et al. A retrospective study of neoadjuvant
[15] Versteijne E, Vogel JA, Besselink MG. Meta-analysis comparing upfront surgery FOLFIRINOX in unresectable or borderline-resectable locally advanced pancre-
with neoadjuvant treatment in patients with resectable or borderline re- atic adenocarcinoma. BMC Cancer 2012;12:199.
sectable pancreatic cancer. Br J Surg 2018;105:946–58. [41] Blazer M, Wu C, Goldberg RM, et al. Neoadjuvant modified (m) FOLFIRINOX for
[16] Suker M, Beumer BR, Sadot E, et al. FOLFIRINOX for locally advanced pancre- locally advanced unresectable (LAPC) and borderline resectable (BRPC) adeno-
atic cancer: a systematic review and patient-level meta-analysis. Lancet Oncol carcinoma of the pancreas. Ann Surg Oncol 2015;22(4):1153–9.
2016;17(6):801–10. [42] Nanda RH, El-Rayes B, Maithel SK, Landry J. Neoadjuvant modified FOLFIRINOX
[17] Mokdad AA, Minter RM, Zhu H, et al. Neoadjuvant therapy followed by re- and chemoradiation therapy for locally advanced pancreatic cancer improves
section versus upfront resection for resectable pancreatic cancer: a propensity resectability. J Surg Oncol 2015;111(8):1028–34.
score matched analysis. J Clin Oncol 2017;35(5):515–22.

Descargado para Anonymous User (n/a) en Universidad El Bosque de ClinicalKey.es por Elsevier en abril 08, 2021. Para uso personal
exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
A.G. Raufi, G.A. Manji and J.A. Chabot et al. / Seminars in Oncology 46 (2019) 19–27 27

[43] Marthey L, Sa-Cunha A, Blanc JF, et al. FOLFIRINOX for locally advanced pan- [52] O’Reilly EM, Roach J, Miller P, et al. Safety, pharmacokinetics, pharmacodynam-
creatic adenocarcinoma: results of an AGEO multicenter prospective observa- ics, and antitumor activity of necuparanib combined with nab-paclitaxel and
tional cohort. Ann Surg Oncol 2015;22(1):295–301. gemcitabine in patients with metastatic pancreatic cancer: phase I results. On-
[44] Conroy T, Desseigne F, Ychou M, et al. FOLFIRINOX versus gemcitabine for col 2017;22(12):1429–139.
metastatic pancreatic cancer. N Engl J Med 2011;364(19):1817–25. [53] https://globenewswire.com/news-release/2017/04/10/958259/0/en/OncoMed-
[45] Stein SM, James ES, Deng Y, et al. Final analysis of a phase II study of modified s- Phase- 2- Demcizumab- Pancreatic- Cancer- Trial- Misses- Primary- Endpoint.
FOLFIRINOX in locally advanced and metastatic pancreatic cancer. Br J Cancer html.
2016;114(7):737–43. [54] Ielpo B, Duran H, Diaz E, et al. Preoperative treatment with gemcitabine plus
[46] Mahaseth H, Brutcher E, Kauh J, et al. Modified FOLFIRINOX regimen with im- nab-paclitaxel is a safe and effective chemotherapy for pancreatic adenocarci-
proved safety and maintained efficacy in pancreatic adenocarcinoma. Pancreas noma. Eur J Surg Oncol 2016;42(9):1394–400.
2013;42(8):1311–15. [55] Frucht H, Stevens PD, Fogelman DR, et al. Advances in the genetic screening,
[47] Sadot E, Doussot A, O’Reilly EM, et al. FOLFIRINOX induction therapy for stage work-up, and treatment of pancreatic cancer. Curr Treatment Opt Gastroen-
3 pancreatic adenocarcinoma. Ann Surg Oncol 2015;22(11):3512–21. terol 2004;7(5):343–54.
[48] J Clin Oncol 2018;36(18) (suppl; abstr LBA4001). doi:10.1200/JCO.2018.36.18_ [56] Sherman WH, Hecht E, Leung D, Chu K. Predictors of response and survival in
suppl.LBA4001. locally advanced adenocarcinoma of the pancreas following neoadjuvant GTX
[49] Stein SM, James ES, Deng Y, et al. Final analysis of a phase II study of modified with or without radiation therapy. Oncol 2018;23(1):4–e10.
FOLFIRINOX in locally advanced and metastatic pancreatic cancer. Br J Cancer [57] Mellon EA, Hoffe SE, Springett GM, et al. Long-term outcomes of induction
2016;114(7):737–43. chemotherapy and neoadjuvant stereotactic body radiotherapy for borderline
[50] Von Hoff DD, Ervin T, Arena FP, et al. Increased survival in pancreatic cancer resectable and locally advanced pancreatic adenocarcinoma. Acta Oncol (Stock-
with nab-paclitaxel plus gemcitabine. N Engl J Med 2013;369(18):1691–703. holm, Sweden) 2015;54(7):979–85.
[51] https://www.prnewswire.com/news-releases/merrimack- announces- top- line- [58] Moningi S, Dholakia AS, Raman SP, et al. The role of stereotactic body radiation
results-from-randomized-phase-2-trial-of-mm-141-in-front-line-metastatic- therapy for pancreatic cancer: a single-institution experience. Ann Surg Oncol
pancreatic- cancer- 300671289.html. 2015;22(7):2352–8.

Descargado para Anonymous User (n/a) en Universidad El Bosque de ClinicalKey.es por Elsevier en abril 08, 2021. Para uso personal
exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.

You might also like