Skip to main content
Therapeutic Advances in Medical Oncology logoLink to Therapeutic Advances in Medical Oncology
. 2020 Jul 7;12:1758835920936093. doi: 10.1177/1758835920936093

Novel strategies using modern radiotherapy to improve pancreatic cancer outcomes: toward a new standard?

Christelle Bouchart 1,, Julie Navez 2, Jean Closset 3, Alain Hendlisz 4, Dirk Van Gestel 5, Luigi Moretti 6, Jean-Luc Van Laethem 7
PMCID: PMC7343368  PMID: 32684987

Abstract

Pancreatic ductal adenocarcinoma (PDAC) remains one of the most aggressive solid tumours with an estimated 5-year overall survival rate of 7% for all stages combined. In this highly resistant disease that is located in the vicinity of many radiosensitive organs, the role of radiotherapy (RT) and indications for its use in this setting have been debated for a long time and are still under investigation. Although a survival benefit has yet to be clearly demonstrated for RT, it is the only technique, other than surgery, that has been demonstrated to lead to local control improvement. The adjuvant approach is now strongly challenged by neoadjuvant treatments that could spare patients with rapidly progressive systemic disease from unnecessary surgery and may increase free margin (R0) resection rates for those eligible for surgery. Recently developed dose-escalated RT treatments, designed either to maintain full-dose chemotherapy or to deliver a high biologically effective dose, particularly to areas of contact between the tumour and blood vessels, such as hypofractionated ablative RT (HFA-RT) or stereotactic body RT (SBRT), are progressively changing the treatment landscape. These modern strategies are currently being tested in prospective clinical trials with encouraging preliminary results, paving the way for more effective treatment combinations using novel targeted therapies. This review summarizes the current literature regarding the use of RT for the treatment of primary PDAC, describes the limitations of conventional RT, and discusses the emerging role of dose-escalated RT and heavy-particle RT.

Keywords: heavy-particle radiotherapy, neoadjuvant therapy, pancreatic cancer, radiotherapy, stereotactic radiotherapy

Introduction

Pancreatic ductal adenocarcinoma (PDAC) remains one of the most aggressive solid tumours with an estimated 5-year overall survival (OS) rate of 7% for all stages combined.1 Most patients have asymptomatic early disease, a factor that contributes to the discovery of PDAC at a locally advanced or metastatic stage in more than 70% of cases. The remaining patients are diagnosed with potentially resectable disease, classified as either resectable (around 20% of cases) or borderline resectable (BR; up to 10% of cases); resectability status being determined by the tumour’s relationship with the surrounding vascular structures.1,2 The only potentially curative option is to obtain a complete surgical resection, but even in this case, the 5-year OS is only about 20%.3 Currently, across the United States and Europe, with nearly 57,000 and 44,000 deaths per year, respectively, PDAC ranks fourth in number of cancer deaths and is expected to reach second place in this ranking by 2030.46 This situation is exacerbated by delays in the emergence of effective systemic treatments compared with many other cancers, highlighting the fact that new strategies are urgently needed.6 In this treatment-resistant disease, located in the vicinity of many radiosensitive organs, the role of radiotherapy (RT) and indications for its use in this setting have been debated for some time and are still under investigation. The results of several important RT trials did not meet expectations and, thus, many have predicted that the use of RT in pancreatic cancer has ended.710 Although a survival benefit has yet to be clearly demonstrated for RT, it is the only technique, other than surgery, that has been demonstrated to lead to local control improvement.10 The lack of survival benefit is likely hidden by poorly effective prior systemic therapies that do not allow sufficient survival for RT to play its role, with the exception of a minority of poorly identified long-term survivors.1113 However, the integration of dose-escalated RT treatments, such as hypofractionated ablative RT (HFA-RT) and stereotactic body RT (SBRT), into innovative multidisciplinary neoadjuvant approaches has renewed interest in the use of RT in PDAC. These modern strategies, currently being tested in several single- and multi-centre trials with encouraging preliminary results but without level I evidence, are already changing the way radiation oncologists treat PDAC. These promising combinations, with more effective systemic therapies and progressive improvements in patient selection, are paving the way for RT to become relevant to survival outcomes. This review will summarize the current literature regarding the use of RT for the treatment of primary PDAC, describe the limitations of conventional RT in this setting, and highlight the emerging roles of dose-escalated RT and heavy-particle RT.

RT for primary pancreatic cancer

Non-stereotactic RT

For several decades, conventional RT has been used for the treatment of PDAC but the lack of high-level evidence of its added survival value has made the place of RT in the management of this disease uncertain, particularly for potentially resectable PDAC.14 The introduction of intensity-modulated RT (IMRT) into the clinical routine a decade ago has limited the treatment-related toxicity of these procedures.15 However, many issues have been, and are still being, encountered in RT trials for PDAC owing to the lack of consensus with regard to delineation, dose and fractionation to be used, leading to significant treatment variations between radiation oncologists with possible effects on survival.16 Recently, efforts have been made to conduct PDAC studies using contemporary RT techniques and modern quality assurance.

Resected pancreatic cancer: adjuvant approach

Given the residual high risk of loco-regional failure (LF) in the resection bed and lymph nodes after surgery and standard adjuvant chemotherapy (CT), which was estimated to be as high as 53% in the recent ESPAC-4 trial, adjuvant RT strategies have been explored over the last 30 years.17 Compared with surgery followed by observation, a phase III trial published in 1985 by the Gastrointestinal Study Group (GITSG) demonstrated that adjuvant chemoradiotherapy (CRT) offered an advantage in survival.18 However, the three phase III studies that followed were not able to replicate the results of the GITSG trial and did not support a survival benefit of adjuvant CRT compared with observation.79,19 In comparison with adjuvant CT alone, prospective phase II/III studies and meta-analyses reported no advantage of adding RT8,9,2022 except in subgroup analyses of patients with positive resection margins for which CRT might still have a role.23 However, while a survival advantage has not been demonstrated, adjuvant CRT appears to reduce local recurrence rates at first progression, as suggested in a randomized phase II trial (11% versus 24% for CT alone).20

It is important to note that the above-mentioned phase III trials have been strongly criticized for the use of an inadequate split-course scheme, the delivery of a total dose of 40 Gy that was likely to be insufficient for providing disease control, and inadequate RT quality control. In addition, two of these phase III studies included a large number of patients with other types of peri-ampullary cancers, known to be associated with a better prognosis than PDAC.24 The ESPAC-1 phase III trial, which concluded that RT was detrimental to survival,8,9 was particularly criticized for an unexpectedly high local recurrence rate (62%), poor adherence to treatment (30% of patients did not receive the planned treatment) and no uniformity of treatment. Moreover, only 53% of the patients were included in the final analysis and modifications of the primary design resulted in three underpowered parallel studies rather than a real 2 × 2 randomization.2527

In an attempt to close this debate, the ongoing RTOG 0848 phase III trial aims to demonstrate that modern adjuvant CRT [50.4 Gy in 28 fractions with concomitant 5-fluorouracil (5-FU)] with high-quality control can increase the survival of resected patients who remain free of disease after five cycles of adjuvant gemcitabine ± erlotinib.28,29 The results of the first randomization of 336 resected patients evaluating the addition of erlotinib to adjuvant gemcitabine were presented in 2017 and did not demonstrate any increase in OS.29 We are now awaiting the results of the second randomization comparing adjuvant CT with or without concurrent RT.

However, this adjuvant approach is now being strongly challenged by more aggressive neoadjuvant treatments that could spare patients with rapidly progressive systemic disease from unnecessary invasive surgery and might increase free margin (R0) resection rates.30

Potentially resectable pancreatic cancer: neoadjuvant approach

Resectable pancreatic adenocarcinoma

Less than 20% of PDACs are diagnosed as initially resectable due to the close vicinity of major arterial and venous trunks. However, even for these patients with relatively favourable disease, the risk of positive margin at surgery (R1) remains high (around 20–50%), especially at the retroperitoneal margin and due to underestimated contact between the tumour and blood vessels.3133 Pathological margin status is a crucial prognostic factor and the survival rate of patients with direct involvement of a margin is similar to that of patients with locally advanced disease.3437 When tumour within 1 mm of the resection margin is included in the definition of R1 margins, the rate of R1 resections increases dramatically up to 80% and this also correlates with poor survival.3841 Consequently, we are now progressively moving toward developing clinical trials in resectable PDAC that investigate the role of neo-adjuvant therapies, including CRT with or without induction CT. These approaches offer several hypothetical advantages including tumour down-staging, maximizing CRT efficacy on well-oxygenated tissues, increasing R0 resection rates, eradicating micrometastases and selecting patients without rapidly progressive disease.4244 The results of the randomized phase II/III Prep-02/JSAP05 trial have been recently presented at the American Society of Clinical Oncology (ASCO) meeting and are gradually changing the paradigm. The authors reported a statistically significant survival benefit for the CT arm (gemcitabine/S1) compared with upfront surgery for resectable PDAC (median OS 36.7 versus 26.6 months, p = 0.015).45 Regarding RT, several single-arm studies and meta-analyses exploring neo-adjuvant CRT in this setting have demonstrated promising results with regard to R0 resection rates (84–100%) and OS,4651 but results are still conflicting (Table 1).44,5255 Nevertheless, upfront surgery followed by adjuvant CT remains the standard of care for resectable PDAC.17,56,57 Currently, upfront surgery followed by adjuvant CT [gemcitabine ± capecitabine and, more recently, FOLFIRINOX (oxaliplatin, irinotecan, leucovorin and fluorouracil) in fit patients] is still the standard option for resectable PDAC. However, National Comprehensive Cancer Network (NCCN) and ASCO guidelines do recommend that neoadjuvant treatment be considered in patients with high-risk features including: large primary tumours, very highly elevated CA19-9 levels, large regional lymph nodes, radiographic interface between tumour and mesenteric vasculature, excessive weight loss and extreme pain.58,59

Table 1.

Selected modern trials evaluating the role of non-stereotactic CRT in potentially resectable (R – BR) pancreatic adenocarcinoma.

Study Study design N Resectability status (classification used) RT regimen: dose (Gy)/# RT technique CT Resection rate (%)
R0 resection rate (%)
Median survival (months)
Surgery alone CRT Surgery alone CRT All Res. Not Res.
Versteijne et al.60 Phase III Arm A: 127
Arm B: 119
R/BR
(DPCG, 2012)
Arm A: –
Arm B: 36/15
VMAT, IMRT or 3D Arm A: –
Arm B: gem (S + 1 cycle after CRT)
72 61 40 71 Arm A: 14.3
Arm B: 16
Arm A: 19.2
Arm B: 35.2
NR
Jang et al.61 Phase II/III Arm A: 23
Arm B: 27
BR
(NCCN)
Arm A: –
Arm B: 54/30
3D Arm A: –
Arm B: gem (S)
78 63 33.3 82.4 Arm A: 12
Arm B: 21
NR NR
Pietrasz et al.62 Retrospective Group A: 57
Group B: 49
BR
(NCCN)
Group A: –
Group B: 54/30
IMRT (80%) – 3D (20%) Group A: I = FOLFIRINOX (median 6 cycles)
Group B: I + S = 5-FU or capecitabine
NA NA NA Group A & B: 80.2 NA 44.7
Group A: 35.5
Group B: 62.9
NA
Nagakawa et al.63 Phase II 27 BR-A
(own definition, derived from NCCN)
50.4/28 IMRT I: gem (2 cycles)
S: gem/S-1
NA 70 NA 95 22.4 22.9 9.3
Fujii et al.47 Observational Group A: 416
Group B: 88
R/BR-A/BR-PV
(own definition, derived from NCCN)
Group A: –
Group B: 50.4/28
NR Group A: –
Group B: S = S-1
R: 88
BR-PV: 82
BR-A: 68
R: 92
BR-PV: 93
BR-A: 70
R: 70
BR-PV: 61
BR-A: 31
R: 86
BR-PV: 96
BR-A: 71
R:
CRT: 28.6
Surgery alone: 33.7
BR-PV:
CRT: 28.4
Surgery alone: 20.1
BR-A:
CRT: 18.1
Surgery alone: 10
NR NR
Katz et al.64 (Alliance A021101) Phase II 22 BR
(Intergroup criteria)
50.4/28 IMRT or 3D I: mFOLFIRINOX (4 cycles)
S: capecitabine
NA 68 NA 93 21.7 NR NR
Casadei et al.*55 Phase III Arm A: 20
Arm B: 18
R
(Ishikawa)
Arm A: –
Arm B: 54/30
3D Arm A: –
I: gem (2 cycles)
S: gem
75 61 25 39 Arm A: 19.5
Arm B: 22.4
NR NR
Golcher et al.*65 Randomised phase II Arm A: 33
Arm B: 33
R/BR
(own definition)
Arm A: –
Arm B: 55.8/31
3D S: gem/cisplatin 67 58 48 52 Arm A: 14.4
Arm B: 17.4
Arm A: 18.9
Arm B: 25
NR
Dholakia et al.66 Retrospective 50 BR
(AHPBA/SSO/SSAT)
NR VMAT or IMRT or 3D I: 5-FU/oxaliplatin or FOLFIRINOX
S: gem +/–oxaliplatin or capecitabine
NA 58 NA 93 17.2 22.9 13
Kim et al.48 Phase II 68 R/BR/LAPC
(NCCN 2008)
30/15 3D S: gem/oxaliplatin (+ 1 cycle after) NA R: 57
BR: 72
NA 84 18.2
R: 26.5
BR: 18.4
27.1 10.9
Turrini et al.49 Phase II 34 R
(own definition)
45/25 3D S: docetaxel NA 50 NA 100 15.5 32 11

#, number of fractions; 5-FU, 5-fluorouracil; AHPBA/SSO/SSAT, American Hepatopancreaticobiliary Association/Society of Surgical Oncology/Society for surgery of the Alimentary Tract; BR(-A/PV), borderline resectable (due to arterial abutment/due to exclusive involvement of the portal vein system); CRT, chemoradiotherapy; CT, chemotherapy; DPCG, Dutch Pancreatic Cancer Group; FOLFIRINOX, oxaliplatin, irinotecan, leucovorin and fluorouracil, Gy, Gray; I, induction; S, sensitizer; Gem, gemcitabine; IMRT, intensity-modulated radiation therapy; N, number of patients; NA, not applicable; NCCN, National Comprehensive Cancer Network; NR, not reported; R, resectable; RT, radiation therapy; S-1, combination Tegafur/gimeracil/oteracil; VMAT, volumetric modulated arc therapy.

Early termination due to efficacy.

Statistically positive results in favour of CRT arm.

*

Closed early due to poor accrual.

Results for BR population only.

Borderline resectable pancreatic adenocarcinoma

One of the main issues so far has been that the definition of resectability varies considerably from one study to another and that most trials include resectable, borderline resectable (BR) and even locally advanced pancreatic cancer (LAPC) patients. Theoretically, BR tumours can potentially be resected but contacts between the primary tumour and the surrounding vasculature are more extended, impeding a curative surgery.67 The lack of standardization for this sub-population is of critical importance as various BR definitions have been used, sometimes with significant differences. For example, the MD Anderson group includes patients with poor performance status or severe comorbidities (BR type C) and patients with a suspicion of extra-pancreatic metastatic disease (BR type B).68,69 Therefore, cautious interpretation should be made when analysing and comparing trials that include patients with BR tumours. The neoadjuvant approach appears to be particularly beneficial for BR tumours since several non-randomized trials and meta-analyses have demonstrated promising results regarding R0 resection rates and survival (Table 1).4856,6266,68,69 However, the four published randomized phase II/III trials comparing primary surgery with neoadjuvant CRT in potentially resectable PDAC closed early and/or were largely underpowered due to poor accrual.57,61,65,70 Another example is a Korean phase II/III trial that aimed to compare initial surgery versus neoadjuvant CRT (54 Gy in 30 fractions with gemcitabine) for BR tumours only, defined according to 2012 NCCN guidelines. Results of the interim analysis were recently published: 50 patients were enrolled out of the 110 required and, in the intention-to-treat analysis, the experimental arm showed a significant improvement in R0 resection rates (51.8% versus 26.1%, p = 0.004) and median survival (21 versus 12 months, p = 0.028). Consequently, owing to these positive results, the study was prematurely discontinued based on efficacy.61 In addition, the phase III PREOPANC-1 study completed inclusion of patients in July 2017 with 248 resectable and BR patients randomized between immediate surgery and preoperative hypofractionated CRT with modern quality assurance (36 Gy in 15 fractions; 3 cycles of gemcitabine 1 g/m2, concurrent during the second cycle).71,72 This particular hypofractionated RT scheme was chosen to maintain a full dose of gemcitabine during CRT and was determined according to preliminary phase I/II studies.73,74 The results demonstrated a higher R0 resection rate (40% versus 71%, p < 0.001), benefits in terms of median disease-free survival (DFS; 7.7 versus 8.1 months, p = 0.032) and median LF-free interval (LFFI; 13.4 months versus not reached, p = 0.003) and significantly lower rates of pathologic lymph nodes (78% versus 33%, p < 0.001) and perineural and venous invasion (73% versus 39%, p< 0.001 and 36 versus 19%, p = 0.024, respectively) in favour of the neoadjuvant CRT arm. However, median OS by intention-to-treat, the primary end point, including survival results of patients with metastatic lesions at diagnostic laparoscopy and unexpected LAPC, was not significantly improved for this arm (16 versus 14.3 months, p = 0.096).60 A sub-group analysis of patients undergoing R0/R1 surgery after CRT and having started adjuvant CT was performed to allow for comparison with adjuvant trials and showed a large benefit in median OS (35.2 versus 19.2 months, if not resected, p = 0.029). When looking at the BR subgroup only, the median OS is also in favour of the preoperative CRT arm (17.6 versus 13.2 months, p = 0.029).60 Once again, these results only suggest but do not definitively prove the superiority of the neoadjuvant CRT approach. Furthermore, beyond gemcitabine, new combined CT regimens, such as gemcitabine/nab-paclitaxel and, particularly, FOLFIRINOX, have shown promising improved outcomes in the metastatic and adjuvant settings but now also in the neoadjuvant approach.7577 Recent studies and meta-analyses have shown neoadjuvant FOLFIRINOX to be the most effective, providing significantly better resection rates and OS compared with other CT regimens.43,78,79 In 2016, Katz et al. published a unique prospective series of 22 BR patients treated with induction CT with 4 cycles of modified FOLFIRINOX followed by CRT (50.4 Gy in 28 fractions with capecitabine). The authors reported a resection rate of 68% and an R0 resection rate of 93% with 13% complete pathological response. The median OS was 21.7 months, demonstrating that this neoadjuvant sequence was feasible despite significant toxicity associated with preoperative treatments (grade ⩾3: 64%).64

Main ongoing trials

While upfront surgery seems to be less commonly recommended today, favouring the neoadjuvant approach for potentially resectable disease, high-level evidence is still needed and it remains unclear which modality or strategy is the most effective, CT only or CT followed by CRT. Results of randomized phase II/III trials are urgently required. The PREOPANC-2 phase III study from a Dutch group began in June 2018 with a goal of randomising 368 resectable or BR patients in order to compare 8 cycles of neoadjuvant FOLFIRINOX versus preoperative hypofractionated CRT (as previously described) followed by surgery and 4 cycles of adjuvant gemcitabine.80 In addition, the randomized phase II PANDAS-PRODIGE 44 trial from a French group is recruiting BR patients who will be allocated to 6 cycles of neoadjuvant FOLFIRINOX ± classical CRT (50.4 Gy in 28 fractions with concurrent capecitabine) and will also contribute to answering that question.81

Locally advanced (unresectable) pancreatic cancer

As we have described for potentially resectable tumours, the role of RT in the management of LAPC is not yet well defined and is an important topic of debate. While the two initial GITSG trials in the 1980s demonstrated a survival advantage for CRT over CT or RT alone,82,83 further randomized trials and meta-analyses have not confirmed these first results8489 with the exception of the Eastern Cooperative Oncology Group (ECOG) E4201 study.89 This randomized trial reported a significant improvement in median OS for treatment with CRT compared to gemcitabine alone (11.1 versus 9.2 months, p = 0.017). However, the results of the ECOG E4201 trial should be considered with great caution due to incomplete accrual, including 74 patients instead of the 316 planned.89 Recent meta-analyses of available randomized trials were not able to demonstrate a statistically significant difference in OS between patients treated with neoadjuvant CT alone versus CRT, except on a subgroup analysis of consolidation CRT after an induction CT of at least 3 months.90,91 The largest phase III study to date to explore this question was the LAP07 trial from the Groupe Coopérateur Multidisciplinaire en Oncologie (GERCOR) that aimed to investigate the benefit of CRT after 4 months of induction CT with gemcitabine ± erlotinib. The trial did not meet its primary endpoint (median OS: 15.2 for CRT versus 16.5 months for CT alone, p = 0.830) despite the fact that CRT was associated with a statistically significant decrease in local progression (32% versus 46%, p = 0.030) and a trend toward improved median progression-free survival (PFS: 9.9 versus 8.4 months for CT alone, p = 0.060).10 This study had several limitations due to the choice of CT regimen used and the quality of RT. For RT, 88% of patients treated with CRT were assessable for RT quality analysis and, of these, 50% and 18% presented with minor and major RT protocol deviations, respectively. For CT, the LAP07 trial was designed in 2005 before the advent of FOLFIRINOX and gemcitabine/nab-paclitaxel and, therefore, used a non-optimal gemcitabine regimen. In this trial, although loco-regional progression was decreased, the rate of metastatic progression was higher in the CRT arm (60% versus 44% for CT alone, p = 0.040). This implies that with the use of chemotherapies allowing for better control of distant disease, CRT could add more survival benefit. It should also be noted that in the LAP07 trial, the resection rate obtained either after CT alone or after CRT was very low (7% and 3%, respectively) because these treatments were delivered mainly in a conclusive manner with no intention of further surgical exploration except for the few cases where an important response was shown.10

Interestingly, a meta-analysis by Gillen et al. in 2010 reported that one-third of patients initially classified as unresectable at diagnosis can be successfully resected after neoadjuvant treatment with an estimated median survival of 20.5 months, approximately equivalent to patients who underwent immediate resection.46 However, given the evolution and high variability of the definitions of resectability of PDAC, some of these patients considered to be LAPC at the time of this trial could currently be classified as BR. More recent non-randomized studies exploring induction with modern combinations of CT over 4–8 cycles followed by CRT in LAPC have reported even more impressive results regarding resection rates (up to 89%), R0 resection rates (70–100%), and median survival (18.1–58 months) (Table 2).10,68,88,89,92,93,94,95,96 In 2018, a large French retrospective trial of highly selected patients with resected PDAC (106 BR and 97 LAPC) treated with induction FOLFIRINOX ± CRT (54 Gy in 30 fractions with concurrent 5-FU or capecitabine) was published. Significant differences were demonstrated in favour of neoadjuvant treatment with FOLFIRINOX+CRT, both in BR and LAPC populations with greatly improved median OS (57.8 versus 35.5 months; p = 0.007), R0 resection rates (89.2% versus 76.3%, p = 0.017), and ypN0 rates (76.2% versus 48.5%, p < 0.001).62 This neoadjuvant approach, providing true R0 resection, tumour downstaging and downsizing and major pathological response in a selected number of “good” patients with better outcomes suggests that this represents a multi-step selective process for patient selection that could offer a way to improve the prognosis of pancreatic cancer.

Table 2.

Selected modern trials evaluating the role of CRT for locally advanced pancreatic adenocarcinoma (LAPC).

Study Study design N Res.
status
RT regimen: dose (Gy)/# RT technique CT Resection rate (%)
R0 resection rate (%)
Median survival (months)
CT alone CRT CT alone CRT All Res. Not res.
Reyngold et al.(abstract)97 Prospective 136 LAPC 75/25 or 67.5/15 HFA-IMRT I: NR NA NA NA NA Not reached (2y. OS:55%) NA NA
Sherman et al.92 Phase II 45 LAPC 50.4/28 3D or IMRT I: GTX (6 cycles)
S: gem/capecitabine
NA 89 NA 70 24.8 NR NR
Wo et al.93 Retrospective 74
25
LAPC
BR
58.8/28 3D or IMRT (dose painting) +/– IORT I: FOLFIRINOX or FOLFOX or gem-based (4–8 cycles)
S: capecitabine or 5-FU or gem
NA 37
BR: 32
LAPC: 39
NA 87 18.1 30.9 NR
Pietrasz et al.62 Retrospective Group A: 44
Group B: 53
LAPC Group A: –
Group B: 54/30
3D (20%) – IMRT (80%) I: FOLFIRINOX (median: 6 cycles)
S: 5-FU or capecitabine
NA NA 86.6 NA 49
Group A: 37.8
Group B: 57.8
NA
Sudo et al.94 Phase II 30
(CRT if not M+ after induction: 23)
LAPC 50.4/28 3D I: S-1/gem (4 cycles)
S: S-1
NA 10 NA 100 21.3
CRT: 22.9
NR NR
Huguet et al.95 Retrospective 134 LAPC 56/28 IMRT I: gem-based or FOLFIRINOX (median: 3.2 months)
S: gem or 5-FU
NA 19 NA 85 23 NR NR
Hammel et al.10
(LAP07)
Phase III 442
1st random:
Arm A: 223 – gem
Arm B: 219 – gem + erlotinib
2nd random. if no progression at 4mo.: (269/442):
Arm C: 136 – CT
Arm D: 133 – CRT
LAPC Arm D: 54/30 3D I: gem +/– erlotinib
S: capecitabine
7 3 61 12.8
Arm C (CT): 16.5
Arm D (CRT): 15.2
30.9 NR
Habermehl et al.96 Retrospective 215 LAPC 52.2/29 3D S: gem
Consolidation: gem
NA 26 NA 39 12.3 22.1 11.9
Barhoumi et al.89
2000-01 FFCD/SFRO
Phase III Arm A: 59
Arm B: 59
LAPC Arm A: –
Arm B: 60/30
NR Arm A: gem
Arm B: S = 5-FU/cisplatin
5 3 NR NR Arm A: 13
Arm B: 11.1
NR NR
Loehrer et al.*89
ECOG E4201
Phase III Arm A: 37
Arm B: 34
LAPC Arm A: –
Arm B: 50.4/28
3D Arm A: gem (7 cycles)
Arm B: S/C: gem
0 0 NA NA Arm A: 9.2
Arm B: 11.1
NA NA

#, number of fractions; 5-FU, 5-fluorouracil; BR, borderline resectable; C, consolidation; CRT, chemoradiotherapy; CT, chemotherapy; FOLFIRINOX, oxaliplatin, irinotecan, leucovorin and fluorouracil; FOLFOX, oxaliplatin, leucovorin and fluorouracil; Gem, gemcitabine; GTX, gemcitabine/docetaxel/capecitabine; Gy, Gray; HFA, hypofractionated ablative; I, induction; IMRT, intensity-modulated radiation therapy; IORT, intra-operative radiotherapy; LAPC, locally advanced pancreatic cancer; M+, metastatic; mo., months; N, number of patients; NA, not applicable; NR, not reported; OS, overall survival; Res., Resection; RT, radiation therapy; S, sensitizer; S-1, combination Tegafur/gimeracil/oteracil.

*

Closed early due to poor accrual.

Statistically positive results in favour of CRT arm.

Results for LAPC population only.

The use of hypofractionated ablative (HFA)-RT for selected patients after induction CT is also an interesting approach. In a study by Krishnan et al., 200 LAPC patients were treated with induction CT followed by either conventional CRT (50.4 Gy in 28 fractions with concomitant CT) or HFA-IMRT [delivery of biologically effective doses (BED) > 70 Gy, mainly by using a simultaneous integrated boost technique (SIB) in 15 or 28 fractions]. Only 47 patients with tumours more than 1 cm from the closest gastrointestinal (GI) mucosa were treated with HFA-IMRT. The authors reported promising OS (median: 17.8 versus 15 months, p = 0.030; 3-year OS: 31% versus 9%) and local control (median local-regional recurrence-free survival: 10.2 versus 6.2 months, p = 0.050) for HFA-IMRT. Interestingly, no additional toxicity in the HFA-IMRT group was observed and delivery of high BED was the only predictor of improved OS on multivariate analysis. The authors also suggested that concomitant capecitabine could be better tolerated with dose escalation than gemcitabine, cisplatin/5-FU or 5-FU/mitomycin C.98 Recently, the results of a prospective study from the Sloan Kettering group including 136 LAPC patients treated with definitive HFA-IMRT (BED ⩾ 100 Gy; 75 Gy in 25 fractions or 67.5 Gy in 15 fractions) were presented. With a median follow-up of 12 months, the median OS and freedom from local progression (FFLP) were not reached and the impressive 2-year OS and FFLP were 55% and 78%, respectively, with a safe toxicity profile.97

Regarding these data, while (C)RT was initially only used as a definitive treatment to prevent or delay local progression, there is now a gradual shift for patients with LAPC toward a neoadjuvant approach followed by surgical exploration in cases of no progression. New RCTs are underway to evaluate the role of modern (HFA)-CRT after induction therapy with more active CT versus CT alone, such as the CONKO-007 trial using FOLFIRINOX, the SCALOP-2 trial using gemcitabine/nab-paclitaxel for induction, and the phase II MAIBE trial that is further exploring the role of HFA-RT (67.5 Gy in 15 fractions or 75 Gy in 25 fractions with concomitant capecitabine).99101

SBRT

Background

SBRT allows precise delivery of high doses to the tumour in a few sessions (1–5), reducing the dose and toxicity to neighbouring organs at risk (OARs).102 The SBRT technique is already successfully used, particularly for intracranial tumours and for the treatment of early-stage non-small cell lung cancer in patients who are inoperable or refuse surgery.103 However, performing an SBRT treatment that targets a pancreatic lesion that may move during respiration in the middle of the upper abdomen is more challenging. Several useful tools and techniques have recently been developed, making SBRT possible for PDAC, including: modern planning methods [IMRT/volumetric modulated arc therapy (VMAT), four-dimensional computed tomography (4D-CT) to assess the amplitude and direction of tumour movement during the respiratory cycle, abdominal compression and breath-hold techniques to restrain tumour movement, endoscopic implantation of fiducial markers into the tumour, and the use of on-board cone-beam computed tomography (CBCT) for daily tumour position verification or tracking.104,105 All these innovations provide reductions in margin expansion and dose escalation to the target volume while safely limiting and controlling dose and toxicity to OARs. In contrast to conventional RT, the SBRT approach seeks to avoid irradiation of large volumes and, therefore, does not include prophylactic irradiation of neighbouring lymph node areas. Owing to the shorter duration of treatment compared to conventional RT (1 week versus 4–6 weeks), patients receiving SBRT can resume systemic therapy more quickly, reducing long interruptions of full-dose chemotherapy.103,104 Another goal of SBRT is to improve local control with the delivery of higher BED to the tumour, since up to 30% of PDAC patients die due to local progression only.106 Therefore, SBRT has been tested in the treatment of PDAC since the early 2000s with interesting results in terms of feasibility, safety and efficacy, providing high local control, improved (R0-) resection rates and survival.

Clinical data in pancreatic cancer and challenges

History

The development of SBRT for PDAC took place in two main phases. During the first decade of the 2000s, studies explored pancreatic SBRT in different sequences (alone, with induction CT, as a boost after CRT) with very good local control rates (LCR, generally > 85% at 1 year) but with little or no impact on median survival. These early studies, usually using a single fraction with very high dose per fraction (15–25 Gy), reported unacceptably high rates of grade 3–4 GI toxicity up to 22.6%, leading to a new disappointment for radiation oncologists.107113 Subsequently, during the second decade of the 2000s, further retrospective or phase II trials reported results on the use of fractionated SBRT (3–5 fractions) with softer hypofractionation (mainly 5–6.6 Gy per fraction), that attempted to respect strict GI constraints, particularly for the duodenum. These studies reported acceptable rates of acute and late grade ⩾3 GI toxicity, with risks generally between 0% and 10% and consisting mainly of GI bleeding, ulcers, stenosis, perforations or gastroparesis.114128 Overall, fractionated SBRT treatments are well tolerated, showing less acute toxicity than IMRT-CRT and similar perioperative complication rates.129132 The main studies that have reported the use of this approach in this setting are summarized in Table 3.

Table 3.

Selected historical and modern trials evaluating the role of SBRT for pancreatic adenocarcinoma.

Study Study design N Res. status Dose (Gy)/# RT machine CT Resection rate (%) R0 resection rate (%) LC (at 1 year, %) Grade ⩾3 GI toxicity (%)
Median survival from diagnosis (months)
Acute Late All Res
Main historical studies with single fraction
Koong et al.107 Phase I 15 LAPC 15–25/1 Cyberknife I (20%): gem-based / / 77
100 (for the six patients at 25 Gy)
33 / 11 /
Koong et al.108 Phase II 16 LAPC 25/1 Cyberknife / / / 94 12.5 / 7.7 /
Schellenberg et al.109 Case series 16 LAPC 25/1 Cyberknife I/C: gem / / 100 18.6 / 11.4
Chang et al.110 Retrospective 77 R (3%)/LAPC (78%)/M+ (19%) 25/1 Cyberknife / 1 NR 84 5 9 11.9
LAPC: 11.5
NR
Schellenberg et al.111 Phase II 20 LAPC 25/1 Cyberknife I/C: gem / / 94 15 5 11.8 /
Goyal et al.112 Case series 19 LAPC/Recurrence (10%) 20–30/1–3 Cyberknife I (68%): 5-FU or gem-based / / 65 0 16 14.4 /
Rajagopalan et al.113 Retrospective 12 BR (58%)/LAPC (42%) 24–36/1–3 Cyberknife/Linac I (92%): gem-based 11.4 91.7 NR 0 16.7 47.2 NR
Main modern hypofractionated SBRT studies including mainly LAPC ¥
Moningi et al.114 Retrospective 74
14
LAPC
BR
25–33/5 Linac I (87.5%): gem-based or FOLFIRINOX LAPC: 20
BR: 29
All: 84 All: 61 3.4 5.7 LAPC: 18.4
BR: 14.4
All: 20.2
Herman et al.115 Phase II 49 LAPC 33/5 Linac I: gem (up to 3weeks)
C: gem
8 100 78 12.2 10 13.9 22.2
Comito et al.116 Phase II 45 LAPC 45/6 Linac I: gem-based (71%) / / 87 0 0 19 /
Zhong et al.117 Retrospective 631 LAPC 40/5 NR I (87%): NR 10.8 92 NR NR NR 13.9 NR
Gurka et al.118 Retrospective 28
6
4
LAPC
BR
Medically inoperable
25–30/5 Cyberknife I/S: 5-FU, capecitabine, gem or FOLFOX 0 / 79 5 8 All: 14.3 /
Jumeau et al.119 Retrospective 17
4
LAPC
Local recurrence
30–35/5–6 Cyberknife I (38%): gem or FOLFIRINOX / / 67 5 5 22 /
Jung et al.120 Retrospective 95 LAPC 24–36 (median 28)/4 Linac I (14%)/C (81%): gem-based or FOLFIRINOX 7.4 57 80 3.2 3.2 16.7 NR
Suker et al.128 Phase II 50 LAPC 40/5 NR I: FOLFIRINOX 12 100 Median: 20 months 10 NR 15 23
Main modern hypofractionated SBRT trials including mainly BR PDAC
Chuong et al.121 Retrospective 57
16
BR
LAPC
25–30/5 (SIB TVI 35–50 Gy) Linac I: gem-based (3 cycles) 56
0
97
/
81 (Not resected only) 0 5.3 16.4
15
19.3
/
Mellon et al.122 Retrospective 110
49
BR
LAPC
28–30/5 (SIB TVI 50 Gy) Linac I: gem-based or FOLFIRINOX 51
10
96
100
78 (Not resected only) 2 7 19.2
15
All: 34.2
Chuong et al.123 Retrospective 36 BR 25–30/5 (SIB TVI up to 40 Gy) Linac I: gem-based, mainly GTX 100 97.2 100 0 NR 22.5 /
Mellon et al.124 Retrospective 150
72
BR
LAPC
25–30/5 (SIB TVI 50 Gy) Linac I: gem-based mainly GTX or FOLFIRINOX (3 cycles) 51
11
All: 97.5 NR NR NR NR All: 37.5
Quan et al.125 Phase II 19
16
BR
LAPC
36/3 NR I: gem/capecitabine (4 cycles) 53
12.5
All: 91.7 63
78
0 0 28.3
14.3
31
24.6
Palta et al.121 Prospective (Abstract) 16
9
BR
R
25/5 NR I: gem/nab-paclitaxel (2 cycles) All: 68 All: 93 77 0 / All: 24 NR
Kharofa et al.127 Phase II 15
3
BR
R
25/5 (SIB TVI 33 Gy) NR I: gem/nab-paclitaxel or FOLFIRINOX (3 months) All: 67 All: 92 50 0 0 All: 21 All: 31

#, number of fractions; BR, borderline resectable; C, consolidation; CRT, chemoradiotherapy; CT, chemotherapy; FOLFIRINOX, oxaliplatin, irinotecan, leucovorin and fluorouracil; FOLFOX, oxaliplatin, leucovorin, and fluorouracil; Gem, gemcitabine; GI, gastrointestinal; GTX, gemcitabine/docetaxel/capecitabine; Gy, Gray; I, induction; LAPC, locally advanced pancreatic cancer; LC, local control; Linac, linear accelerator; N, number of patients; NA, not applicable; NR, not reported; R, resectable; RT, radiation therapy; S, sensitizer; SBRT, stereotactic body radiation therapy; SIB, simultaneous-integrated boost; TVI, tumour–vessel interface.

¥

Results concerns LAPC only even if other PDAC status were included in the studies, excepted if clearly mentioned in the table.

SBRT in the neoadjuvant PDAC setting

While the main indication for SBRT was initially the definitive treatment of LAPC, some trials have investigated the role of SBRT for potentially resectable PDAC and LAPC in a neoadjuvant setting and these are summarized in Table 3.113,114,123127 The SBRT technique can be easily integrated into a neoadjuvant approach to target the tumour with a particular concern for the areas of contact between the tumour and blood vessels that are called the tumour–vessel interfaces (TVIs) where an integrated boost should be applied whenever possible. These TVIs are very important for limiting the possibility of curative resection outside the metastatic context and attempts to sterilize these areas could increase (R0) resection rates.132 This hypothesis seems to be confirmed by the results of available phase I/II and retrospective studies showing very high rates of R0 resection for BR and LAPC patients treated with SBRT (84–97.5%) (Table 3).113,114,120127 However, it is now well known that a major issue with the use of modern multi-agent CT and RT is the difficulty of predicting resectability after neoadjuvant treatment by imaging assessment.133,134 Indeed, few patients show an improvement in the number and degree of TVIs after neoadjuvant treatment due to insufficient differentiation of residual tumour versus desmoplasia, particularly at TVIs.66,133,134 For illustration, in the study by Kluger et al., 61 LAPC patients who failed to regress from >180° encasement of coeliac, superior mesenteric or hepatic arteries after receiving neoadjuvant therapy (induction CT principally gemcitabine-based or FOLFIRINOX followed by RT with IMRT or SBRT technique) were systematically surgically explored. While 8% were metastatic at laparoscopy, the remaining 56 patients were resected with an R0 resection rate of 80.4% (defined as no malignant cells within 1 mm of any margin) and an impressive median OS from the beginning of neoadjuvant therapy of 28.9 months.135 New imaging modalities are currently being explored to improve appropriate selection of patients for surgery such as positron emission tomography (PET) or advanced diffusion-weighted imaging magnetic resonance imaging (DWI-MRI).136,137 It is now uniformly recommended to perform systematic surgical exploration in non-metastatic and non-locally progressive operable patients with localized PDAC, even if there is no evidence of radiographic down-staging after modern-era neoadjuvant strategies.

Clinical development and limitations of SBRT in PDAC

The only analysis that pooled 19 SBRT trials for LAPC patients demonstrated a median OS of 17 months (range 5.7–47 months), a 1-year OS rate of 51.6% and a 1-year LCR of 72.3%. Heterogeneity was high between studies and most of them were small retrospective series.138 The dose and the number of fractions delivered were highly variable and an optimal scheme for pancreatic fractionated SBRT was not clearly established. However, reducing the dose too much to ensure safety, as is commonly done in current practice, is not a solution, as the fractionation schemes of 25–33 Gy in 5 fractions correspond to a maximal BED of 55 Gy, well below the ablative doses sought with SBRT. It is, therefore, not surprising that the survival benefit usually obtained with this low-BED SBRT is modest and that the local control is weaker than that observed with the first historical SBRT analyses of non-randomized studies available (Table 3).107128 This modest survival benefit of SBRT over CRT seems to be confirmed by the results of three large retrospective studies using the National Cancer Center Database and one recent meta-analysis.117,139,140 To illustrate for LPAC, in the meta-analysis by Tchelebi et al., SBRT, defined as RT delivered at ⩾5 Gy per fraction, was associated with a modest benefit for 2-year OS (26.9 versus 13.7 months for CRT, p = 0.004) but OS differences were not statistically significant at 1 year (53.7% versus 49.3%, p = 0.630).140 For resectable and BR PDAC, Jiang et al. retrospectively studied 5828 patients treated with different neoadjuvant treatments before pancreatectomy including 332 with CT followed by SBRT (defined as ⩾6 Gy per fraction). Although the SBRT group contained more stage cT3–T4 patients, a survival improvement was shown compared with CT alone or CT+conventional CRT and persisted after propensity score-matching with median OS of 32.1, 27.5 and 27.1 months (p = 0.013), respectively. The R0 resection rate was higher in the RT groups than with CT alone (p < 0.001) and the SBRT group was also associated with better T/N-stage downstaging (p < 0.001).141 Substantial expectations were placed upon the randomized phase II Alliance A021501 trial designed to compare the outcomes of BR patients treated with induction with FOLFIRINOX alone or followed by SBRT (33 Gy in 5 fractions with SIB up to 40 Gy at TVI or 25 Gy in 5 fractions).142 Initially this trial included three arms, but due to the results of the LAP07 trial, the FOLFIRINOX followed by conventional CRT arm was cancelled.143 Unfortunately, the study was recently suspended following an interim analysis of 30 patients revealing crossing of the futility boundary for R0 resection rates for the SBRT arm.127,144 Taking into account all these data, and despite the lack of level I evidence, the NCCN, ASCO, ASTRO and the American College of Radiobiology (ACR) guidelines have already listed SBRT as an optional treatment for localized PDAC in experienced, high-volume centres.2,14,58,59,145

It is essential to continue to improve the pancreatic SBRT technique, in particular by trying to deliver very high BED to the target while maintaining a safe toxicity profile. Indeed, delivery of a BED10 ⩾60 Gy (α/β = 10) seems correlated with improved OS and PFS on multivariate analysis.146 The progressive availability of magnetic resonance linear accelerator (MR-linac) systems in high-volume RT centres could be an elegant option for providing high-dose delivery. Using the stereotactic MR-guided adaptive RT (SMART) technique, Rudra et al. treated 44 BR or LAPC patients with different RT schemes including high-dose SBRT (BED ⩽70 Gy group: 40–55 Gy in 25–28 fractions or 30–35 Gy in 5 fractions; BED > 70 Gy group: 50–67.5 Gy in 10–15 fractions or 40–52 Gy in 5 fractions). The authors reported a 2-year OS of 49% versus 30% (p = 0.030) in favour of the high-dose group but only six patients underwent surgery after completion of RT and high-dose RT did not predict OS in multivariate analysis.147 A prospective phase II multi-institutional trial opened in 2019 with the goal of investigating the SMART technique (50 Gy in 5 fractions) for BR and LAPC patients.148

Another limitation of SBRT in this setting could be the failure of durable local control following low-BED SBRT owing to the limited irradiated volume. Dholakia et al. generated a map of local recurrences from 202 heterogeneously treated PDAC with or without RT. Forty-five per cent of these patients presented with an LF and 90% of these LFs could be covered using a method of asymmetric Boolean extension from the superior mesenteric artery (SMA) and coeliac trunk of 1–3 cm.149 These perivascular tissues are not systematically covered by pancreatic SBRT and could explain the important rates of LF reported in some studies.127,150 A recent example is provided by the phase II study from Kharofa et al. in which 18 BR patients were treated with 3 cycles of multi-agent CT followed by SBRT (33 Gy in 5 fractions with an optional elective 25 Gy volume to the at-risk vasculature). Surgery was performed in 12 patients with 92% of R0 resections. The median OS and PFS were 21 and 11 months, respectively, with LF predominantly observed outside the planning target volume (PTV) 33 Gy.127

Numerous studies are underway to further explore and evaluate the role of SBRT in PDAC, especially in the neoadjuvant setting. A randomized phase II study was recently opened for recruitment by the Medical College of Wisconsin, the SOFT Preop study, for resectable and BR patients who will be randomized between neoadjuvant SBRT versus CRT + CT.151 For LAPC patients, a phase III trial from Stanford University is recruiting 172 patients randomized between induction with up to 4 cycles of FOLFIRINOX alone versus FOLFIRINOX + SBRT.152

Future challenges

As distant and regional recurrence remains a problem for a majority of localized PDAC patients, even when they are treated with the best current standard of care, the emergence of new systemic innovative targeted therapies is awaited. Until now, results of trials using single- or dual-checkpoint immunotherapy in PDAC have not made the expected breakthrough as PDACs, except for a few patients with MSI, appear to be unresponsive.153156 This failure is largely explained by the high capacity of immune escape owing to the complex tumour microenvironment of PDAC that contains abundant desmoplastic stroma and immunosuppressive cells.157 Various combinations with checkpoint immunotherapy are currently under study, including: a/plus CT with some encouraging results, such as the recent phase Ib studying triple combination of gemcitabine/nab-paclitaxel with anti-CD40 and anti-PD-1 in a first-line metastatic setting158 or the phase Ib study combining FOLFIRINOX with anti-CCR2 and additional neoadjuvant CRT at the discretion of the tumour board for BR and LAPC patients;159 b/plus modulators of the stroma, such as pegvorhyaluronidase alfa (PEGPH20);160 c/plus immunocytokines in an effort to boost anti-tumoural immunity;157,161 d/plus RT, particularly SBRT as an immune priming treatment. Modern, highly localized SBRT techniques can partly spare local lymphatics and have the potential to trigger immune responses through multiple pathways such as: induction of immune cell death, delivery of new antigens, activation of the cGAS-STING pathway, local recruitment of T cells and transient overexpression of specific receptors at the surface of tumour cells rendering them more vulnerable to cytotoxic T-cell killing.157,162,163 Although optimal dose, fractionation and timing between RT and immunotherapy are not well established and appear to vary widely depending on the context, a multitude of phase I/II trials are underway to explore these innovative immuno-SBRT combinations, including some for PDAC.162,164

Heavy-particle therapy: proton and carbon-ion therapy

Heavy-particle beam therapy takes advantage of a particular physical characteristic with an energy release inversely proportional to the square of its velocity. The result is a very conformal dose deposit, as the particle delivers a low dose at the entry and most of its energy to a peak called the ‘Bragg peak’ just before it stops. Therefore, particle therapy is expected to offer interesting perspectives in PDAC by limiting toxicity to the many surrounding OARs and allowing dose escalation at the tumour level.165 Most of the heavy-particle centres in the world are equipped with proton therapy facilities but there are many other particles that can be exploited in RT, including carbon ions (12C-ions). Compared with photon- or proton-based RT, 12C-ion therapy may also offer the advantage of better local control of radioresistant tumours through higher linear energy transfer, resulting in more effective DNA damage to cancer cells, reduced oxygen effect and less cell cycle-related radiosensitivity.166 As 12C-ion facilities are increasing but still limited worldwide, studies exploring this technique for PDAC are rare. Results of the few published phase I/II and retrospective studies have demonstrated promising results with median OS up to 25.1 months for LAPC patients included in the higher-dose groups (Table 4).167174 However, toxicity outcomes should be carefully considered as some trials have reported high GI toxicity following CRT with particle therapy. A dosimetric analysis between IMRT and proton plans in LAPC reported a decrease in low and intermediate doses at OAR volumes, but an increase in the high-dose region to the duodenum and stomach.174 Another in silico analysis using fraction dose calculations on CBCT for PDAC with photon, proton and 12C-ion plans reported that photon plans were highly robust regarding interfractional anatomical changes in contrast to heavy-particle plans where severe adaptive reductions in target dose coverage were observed.175 Similarly, a prospective study in which endoscopy was performed after proton-based CRT with concurrent gemcitabine demonstrated appearance of RT-induced ulcers in the stomach and duodenum in 49.4% of the 91 patients investigated, although no bleeding or perforations were described.176 Therefore, particle therapy should be used with caution for moving GI targets, particularly for investigating dose escalation as this technique could deliver very high BED to OAR in cases of slight inter/intrafractional and set-up modifications of their position. Use of strict management of tumour and OAR motion with dedicated devices such as tracking or 4D treatment planning is, therefore, highly recommended and the development of new delivery techniques such as pencil-beam scanning intensity-modulated proton therapy (IMPT) could improve the dosimetric advantage of particle therapy over photon.177179 Regarding outcomes, although a retrospective data-based model of CRT in LAPC predicted an advantage of hypofractionated particle therapy over standard photon RT for 1-year OS, there are still no available results from randomized trials comparing the efficacy and toxicity outcomes of photon versus proton or 12C-ion therapy.180 A phase III trial comparing 12C-ion therapy with photon IMRT in LAPC followed by four cycles of gemcitabine/nab-paclitaxel has recently opened for recruitment.181

Table 4.

Selected trials evaluating the role of heavy-particle therapy for pancreatic cancer.

Study Study design N Resectability status Dose (GyE/RBE)/# Type of particles CT Resection rate (%) R0 resection rate (%) LC (at 1 year, %) Grade ⩾3 GI toxicity (%)
Median survival from diagnosis (months)
Acute Late All Res
LAPC
Terashima et al.167 Phase I/II 50 LAPC 50–70.2/25–26 Protons S/C: gem / / 81.7 18 10 NR /
Sachsman et al.168 Phase I/II 11 LAPC 59.4/33 Protons I: Gem or FOLFIRINOX
CRT: capecitabine
27 33 86 0 0 18.4 NR
Shinoto et al.169 Phase I/II 72 LAPC 43.2–55.2/12 (dose-escalation) Carbon-ion S: Gem (dose-escalation) / / 92 7 1 19.6 /
Jethwa et al.170 Retrospective 13 LAPC 50/25 Protons (IPMT) I: FOLFIRINOX or gem/nab-paclitaxel
S: 5-FU or capecitabine
/ / 66 0 0 NR
At 1 year: 62%
/
Kawashiro et al.171 Retrospective 72 LAPC 52.8–55.2/12 Carbon-ion I (74%): gem-based or FOLFIRINOX
S (78%): gem and/or S-1
/ / 84 3 1 21.5 /
Shinoto et al.172 Retrospective 64 LAPC 55.2/12 Carbon-ion I (76%): gem-based or FOLFIRINOX
S: gem and/or S-1
/ / 75 6 0 25.1 /
Potentially resectable
Shinoto et al.173 Phase I 26 R 30–36.8/8 Carbon-ion / 81 90.5 100 (resected only) 0 0 18.6 Not reached
Hong et al.174 Phase I/II 48 R 25/5 Protons S: capecitabine 77 84 NR 4 NR 17.3 27

#, number of fractions; C, consolidation; CRT, chemoradiotherapy; FOLFIRINOX, oxaliplatin, irinotecan, leucovorin and fluorouracil; Gem, gemcitabine; GI, gastrointestinal; GTX, gemcitabine/docetaxel/capecitabine; GyE, Gray equivalent; I, induction; IPMT, intensity proton modulated therapy; LAPC, locally advanced pancreatic cancer; LC, local control; N, number of patients; NR, not reported; R, resectable; RBE, radiobiological equivalent; S, sensitizer; S-1, combination Tegafur/gimeracil/oteracil.

Conclusions and perspectives

After decades of discussion and investigation, the role of RT in the treatment of primary PDAC has not yet been established. Currently, except for its well-proven benefit in local control, conventional RT treatment cannot be formally recommended in either adjuvant or neoadjuvant approaches as a result of the recently published randomized studies showing no survival benefit of adding RT to CT. However, the final results of ongoing phase III trials using modern high-dose RT techniques, which have already reported encouraging preliminary data, are eagerly awaited. These trials are summarized in Table 5 and a comparison between modern conventional CRT, SBRT and HFA-RT for the treatment of localized PDAC in the neoadjuvant setting is summarized in Table 6. In particular, the integration of hypofractionated SBRT or HFA-RT into modern neoadjuvant treatment regimens (FOLFIRINOX or gemcitabine/nab-paclitaxel) seems to be the most promising for resectable, BR and LAPC pancreatic cancer. These options for neoadjuvant sequence allow optimization of PDAC patient selection and are expected to demonstrate their superiority by improving resectability rates and clinical outcomes of PDAC patients with curative intent. Moreover, while LAPC patients were definitively treated with (R)CT several years ago, it has become clear that these patients can now also benefit from these modern neoadjuvant treatments with resection rates of up to about 40%. These resected LAPC patients now show increased survival times that were previously not considered for this category of patients. Well-designed trials should also focus on and integrate a strict definition of tumour (non-)resectability. In light of the above, the place of RT in the field of localized PDAC is an open question and the development of new techniques (SBRT, HFA-IMRT, heavy-particle therapy and combinations with immunotherapy) must be investigated further in good quality randomized studies.

Table 5.

Radiotherapeutic perspectives for potentially resectable (R & BR) and locally advanced pancreatic adenocarcinoma (LAPC).

Potentially resectable
LAPC
Resectable BR
Incidence 15–20% 7–10% 15–20%
Standard sequence of treatments Upfront surgery → adjuvant therapy
Consider neoadjuvant therapy in case of presence of high-risk features
Neoadjuvant therapy → surgery → adjuvant therapy
Less recommended: upfront surgery → adjuvant therapy
Neoadjuvant therapy → surgery if possible
Adjuvant approach
Perspectives – Main ongoing trials
Conventional CRT: no advantage in survival demonstrated in comparison to adjuvant CT alone (level of evidence: I/A) – Potential remaining benefit for R1/R2 surgery or N+ (only demonstrated on sub-group analysis)
Use of IMRT and modern QA –
Phase III RTOG 0848 trial: Gem ± erlotinib → CRT (50.4 Gy in 28#; 5-FU or capecitabine) versus 1 mo. Gem ± erlotinib [ClinicalTrials.gov identifier: NCT01013649]
NA
Neoadjuvant approach
Perspectives – Main ongoing trials
Conventional CRT: no advantage in survival demonstrated in comparison with neoadjuvant CT alone (level of evidence: II) Conventional CRT: potential advantage in R0 resection rate and survival in comparison with upfront surgery (level of evidence: II) Conventional CRT: no advantage in survival demonstrated in comparison with neoadjuvant CT alone (level of evidence: II) – Potential remaining benefit after induction CT of at least 3 mo. (sub-group analysis)
Induction with modern multi-agents CT + CRT – Randomized phase II PANDAS-PRODIGE 44 trial: 6 cycles FOLFIRINOX ± CRT (50.4 Gy in 28#; capecitabine) [ClinicalTrials.gov identifier: NCT02676349] Induction with modern multi-agents CT + CRT –
Phase III CONKO-007 trial: induction CT (Gem or FOLFIRINOX) + CRT (50.4 Gy in 28#; Gem) versus CT alone [ClinicalTrials.gov identifier: NCT01827553]
Randomized phase II SCALOP-2 trial: 3 mo. induction CT (Gem/Nab-paclitaxel) then Arm A= Nelfinavir+CRT (50.4 Gy in 28#; capecitabine) or Arm B= CRT alone (50.4 Gy in 28#; capecitabine) or Arm C= Nelfinavir+CRT (60 Gy in 30#; capecitabine) or Arm D= CRT alone (60 Gy in 30#; capecitabine) or Arm E= CT alone (Gem/Nab-P) [ClinicalTrials.gov identifier: NCT02024009]
Hypofractionated CRT –
Phase III PREOPANC-2 trial: neoadjuvant FOLFIRINOX versus CRT (36 Gy in 15#; Gem 1000 mg/m2) + adjuvant Gem [EudraCT 2017-002036-17]
Hypofractionated CRT –
Phase III PREOPANC-2 trial
Hypofractionated CRT –
Phase II MAIBE trial: HFA-IMRT (67.5 Gy in 15# or 75 Gy in 25# with concomitant capecitabine or 5FU)
[ClinicalTrials.gov identifier: NCT03523312]
Hypofractionated SBRT –
Randomized phase II SOFT-Preop trial: 2 mo. induction CT + SBRT versus conventional CRT [ClinicalTrials.gov identifier: NCT03704662]
Hypofractionated SBRT –
Randomized phase II SOFT-Preop trial
Randomized phase II Alliance A021501 trial: neoadjuvant FOLFIRINOX versus FOLFIRINOX + SBRT (33–40 Gy in 5#) [ClinicalTrials.gov identifier: NCT02839343]
Randomized phase II BRPCNCC-1 trial: neoadjuvant gem/nab-P versus gem/nab-P + SBRT versus S-1/nab-P + SBRT [ClinicalTrials.gov identifier: NCT03777462]
Phase II SMART trial
Hypofractionated SBRT –
Phase III Stanford trial: induction with up to 4 cycles of FOLFIRINOX ± SBRT [ClinicalTrials.gov identifier: NCT01926197]
Phase II SMART trial: 50 Gy in 5# delivered by MRI-Linac with on-table adaptative re-planning. [ClinicalTrials.gov identifier: NCT03621644]
Heavy-particle therapy Heavy-particle therapy Heavy-particle therapy –
Phase III CIPHER trial: 12C-ion-based CRT (59.4 GyE in 12#; Gem) versus photon-based CRT (50.4 Gy in 28#; Gem) followed by 4 cycles of Gem/Nab-P [ClinicalTrials.gov identifier: NCT03536182]

#, number of fractions; 5-FU, 5-fluorouracil; BR, borderline resectable; CRT, chemoradiotherapy; CT, chemotherapy; FOLFIRINOX, oxaliplatin, irinotecan, leucovorin and fluorouracil; Gem, gemcitabine; GyE, Gray equivalent; HFA, hypofractionated ablative; IMRT, intensity-modulated radiation therapy; LAPC, locally advanced pancreatic cancer; Mo., months; MRI-Linac, magnetic resonance imaging-linear accelerator; Nab-P, nab-paclitaxel; QA, quality assurance; RBE, radiobiological equivalent; SBRT, stereotactic body radiation therapy.

Table 6.

Global comparison between conventional chemoradiotherapy and emerging dose-escalated treatments for the neoadjuvant treatment of localized pancreatic adenocarcinoma.

Conventional CRT Fractionated SBRT HFA-RT
RT equipment Standard installations
Fiducials insertion: not mandatory
Advanced installations: 4D-CT, abdominal compression, DIBH, tracking, CBCT, Linac dedicated to SBRT, MR-Linac. . .
Fiducials insertion: mandatory
Advanced installations
Fiducials insertion: mandatory
RT contouring Large volume
(inclusion of ENI controversial)
Limited volume Intermediate volume
RT planning 3D-CRT or IMRT or VMAT VMAT or IMRT
SIB notably at TVI
VMAT or IMRT
with SIB
Dose prescription Usually 45–54 Gy in 1.8–2 Gy/# Highly variable
Mainly 33–50 Gy in 3–5# with up to 30% dose heterogeneity
Highly recommended: BED10 ⩾70 Gy
Highly variable
Mainly 45–100 Gy in 25# or 37.5–90 Gy in 15#
Highly recommended: BED10⩾70 Gy
Concomitant CT Yes No Yes
Main proven advantages Robust and well-known
Benefit in local control (level A)
Delivery of high to very high BED
Well tolerated
Resume quickly systemic therapy or undergo surgical resection
Greater patient convenience
Increase machine capacity
Delivery of very high BED
No additional toxicity compared to conventional CRT
Main disadvantages Decreased machine capacity
Less convenient for the patients (side-effects, displacements. . .)
No level A evidence of survival benefit
More time-consuming for simulation/contouring/RT planning/treatment
Not recommended for some patients (very large tumour, active GI ulcerations, GI tumour invasion. . .)
Only results of non-randomized trials
Target volume too small?
More time-consuming for simulation/contouring/RT planning/treatment
Not recommended if large GI invasion
Only results of non-randomized trials
Neoadjuvant treatment setting for localized PDAC Resectable/BR (limited data)
LAPC (definitive or neoadjuvant)
Can be used interchangeably
Recommended: in clinical trials or in experienced high-volume RT centres
Only studied in LAPC
Not recommended out of clinical trials

#, fraction; 3D-CRT, 3-dimensional conformal radiotherapy; 4D-CT, four-dimensional computed tomography; BED, biological equivalent dose; BR, borderline resectable; CBCT, cone beam computed tomography; CRT, chemoradiotherapy; CT, chemotherapy; DIBH, deep inspiration breath hold; ENI, elective nodal irradiation; GI, gastrointestinal; Gy, Gray; IMRT, intensity-modulated radiation therapy; LAPC, locally advanced pancreatic cancer; Linac, linear accelerator; MRI, magnetic resonance; PDAC, pancreatic adenocarcinoma; RT, radiotherapy; SBRT, stereotactic body radiation therapy; SIB, simultaneous integrated boost; TVI, tumour–vessel interface; VMAT, volumetric modulated arc therapy.

Acknowledgments

The authors want to thank Tatjana Arsenijevic for her proofreading of the manuscript. The authors would like to acknowledge the contribution of a medical writer, Sandy Field, PhD, for language editing of this manuscript.

Footnotes

Author contributions: CB drafted and designed the manuscript; JN/JC/AH/DVG contributed to major review of the manuscript; LM/JLVL contributed to review and concept supervision of the manuscript; all authors reviewed and edited the manuscript, gave critical input and gave final approval for publication.

Conflict of interest statement: The authors declare that there is no conflict of interest.

Funding: The authors disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This work was supported by a doctoral grant from the ‘Amis de l’Institut Bordet’ [grant number: 2019-31] and by the ‘Fonds de la Recherche Scientifique – FNRS’ [grant number: FC 33593] (CB).

ORCID iD: Christelle Bouchart Inline graphic https://orcid.org/0000-0003-1714-234X

Contributor Information

Christelle Bouchart, Department of Radiation-Oncology, Institut Jules Bordet, Boulevard de Waterloo, 121, Brussels, 1000, Belgium.

Julie Navez, Department of Hepato-Biliary-Pancreatic Surgery, Erasme University Hospital, Université Libre de Bruxelles, Brussels, Belgium.

Jean Closset, Department of Hepato-Biliary-Pancreatic Surgery, Erasme University Hospital, Université Libre de Bruxelles, Brussels, Belgium.

Alain Hendlisz, Department of Gastroenterology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium.

Dirk Van Gestel, Department of Radiation-Oncology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium.

Luigi Moretti, Department of Radiation-Oncology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium.

Jean-Luc Van Laethem, Department of Gastroenterology, Hepatology and Digestive Oncology, Erasme University Hospital, Université Libre de Bruxelles, Brussels, Belgium.

References

  • 1. Kleeff J, Korc M, Apte M, et al. Pancreatic cancer. Nat Rev Dis Primers 2016; 2: 16022. [DOI] [PubMed] [Google Scholar]
  • 2. Balaban EP, Mangu PB, Khorana AA, et al. Locally advanced, unresectable pancreatic cancer: American society of clinical oncology clinical practice guideline. J Clin Oncol 2016; 34: 2654–2668. [DOI] [PubMed] [Google Scholar]
  • 3. He J, Ahuja N, Makary MA, et al. 2564 resected periampullary adenocarcinomas at a single institution: trends over three decades. HBP (Oxford) 2014; 16: 83–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Malvezzi M, Carioli G, Bertuccio P, et al. European cancer mortality predictions for the year 2017, with focus on lung cancer. Ann Oncol 2017; 28: 1117–1123. [DOI] [PubMed] [Google Scholar]
  • 5. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J Clin 2019; 69: 7–34. [DOI] [PubMed] [Google Scholar]
  • 6. 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: 2913–2921. [DOI] [PubMed] [Google Scholar]
  • 7. Klinkenbijl JH, Jeekel J, Sahmoud T, et al. Adjuvant radiotherapy and 5-fluorouracil after curative resection of cancer of the pancreas and periampullary region: phase III trial of the EORTC gastrointestinal tract cancer cooperative group. Ann Surg 1999; 230: 776–782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Neoptolemos JP, Dunn JA, Stocken DD, et al. ; European Study Group for Pancreatic Cancer. Adjuvant chemoradiotherapy and chemotherapy in resectable pancreatic cancer: a randomised controlled trial. Lancet 2001; 358: 1576–1585. [DOI] [PubMed] [Google Scholar]
  • 9. Neoptolemos JP, Stocken DD, Friess H, et al. ; European Study Group for Pancreatic Cancer. A randomized trial of chemoradiotherapy and chemotherapy after resection of pancreatic cancer. N Engl J Med 2004; 350: 1200–1210. [DOI] [PubMed] [Google Scholar]
  • 10. Hammel P, Huguet F, van Laethem JL, et al. ; LAP07 Trial Group. Effect of chemoradiotherapy vs chemotherapy on survival in patients with locally advanced pancreatic cancer controlled after 4 months of gemcitabine with or without erlotinib: the LAP07 randomized clinical trial. JAMA 2016; 315: 1844–1853. [DOI] [PubMed] [Google Scholar]
  • 11. Balachandran VP, Łuksza M, Zhao JN, et al. Identification of unique neoantigen qualities in long-term survivors of pancreatic cancer. Nature 2017; 551: 512–516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Yachida S, White CM, Naito Y, et al. Clinical significance of the genetic landscape of pancreatic cancer and implications for identification of potential long-term survivors. Clin Cancer Res 2012; 18: 6339–6347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Kardosh A, Lichtensztajn DY, Gubens MA, et al. Long-term survivors of pancreatic cancer: a California population-based study. Pancreas 2018; 47: 958–966. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Palta M, Godfrey D, Goodman KA, et al. Radiation therapy for pancreatic cancer: executive summary of an ASTRO clinical practice guideline. Pract Radiat Oncol 2019; 9: 322–332. [DOI] [PubMed] [Google Scholar]
  • 15. Bittner MI, Grosu AL, Brunner TB. Comparison of toxicity after IMRT and 3D-conformal radiotherapy for patients with pancreatic cancer - a systematic review. Radiother Oncol 2015; 114: 117–121. [DOI] [PubMed] [Google Scholar]
  • 16. Abrams RA, Winter KA, Regine WF, et al. Failure to adhere to protocol specified radiation therapy guidelines was associated with decreased survival in RTOG 9704–a phase III trial of adjuvant chemotherapy and chemoradiotherapy for patients with resected adenocarcinoma of the pancreas. Int J Radiat Oncol Biol Phys 2012; 82: 809–816. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Neoptolemos JP, Palmer DH, Ghaneh P, et al. ; European Study Group for Pancreatic Cancer. Comparison of adjuvant gemcitabine and capecitabine with gemcitabine monotherapy in patients with resected pancreatic cancer (ESPAC-4): a multicentre, open-label, randomised, phase 3 trial. Lancet 2017; 389: 1011–1024. [DOI] [PubMed] [Google Scholar]
  • 18. Kalser MH, Ellenberg SS. Pancreatic cancer: adjuvant combined radiation and chemotherapy following curative resection. Arch Surg 1985; 120: 899–903. [DOI] [PubMed] [Google Scholar]
  • 19. Smeenk HG, van Eijck CH, Hop WC, et al. Long-term survival and metastatic pattern of pancreatic and periampullary cancer after adjuvant chemoradiation or observation: long-term results of EORTC trial 40891. Ann Surg 2007; 246: 734–740. [DOI] [PubMed] [Google Scholar]
  • 20. Van Laethem JL, Hammel P, Mornex F, et al. Adjuvant gemcitabine alone versus gemcitabine-based chemoradiotherapy after curative resection for pancreatic cancer: a randomized EORTC-40013-22012/FFCD-9203/GERCOR phase II study. J Clin Oncol 2010; 28: 4450–4456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Liao WC, Chien KL, Lin YL, et al. Adjuvant treatments for resected pancreatic adenocarcinoma: a systematic review and network meta-analysis. Lancet Oncol 2013; 14: 1095–1103. [DOI] [PubMed] [Google Scholar]
  • 22. Xu JB, Jiang B, Chen Y, et al. Optimal adjuvant chemotherapy for resected pancreatic adenocarcinoma: a systematic review and network meta-analysis. Oncotarget 2017; 8: 81419–81429. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Stocken DD, Büchler MW, Dervenis C, et al. ; Pancreatic Cancer Meta-Analysis Group. Meta-analysis of randomised adjuvant therapy trials for pancreatic cancer. Br J Cancer 2005; 92: 1372–1381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Lemke J, Schäfer D, Sander S, et al. Survival and prognostic factors in pancreatic and ampullary cancer. Anticancer Res 2014; 34: 3011–3020. [PubMed] [Google Scholar]
  • 25. Abrams RA, Lillemoe KD, Piantadosi S. Continuing controversy over adjuvant therapy of pancreatic cancer. Lancet 2001; 358: 1565–1566. [DOI] [PubMed] [Google Scholar]
  • 26. O’Reilly EM. Adjuvant therapy for pancreas adenocarcinoma: where are we going? Expert Rev Anticancer Ther 2011; 11: 173–177. [DOI] [PubMed] [Google Scholar]
  • 27. Hoffe S, Rao N, Shridhar R. Neoadjuvant vs adjuvant therapy for resectable pancreatic cancer: the evolving role of radiation. Semin Radiat Oncol 2014; 24: 113–125. [DOI] [PubMed] [Google Scholar]
  • 28. ClinicalTrials.gov. Gemcitabine hydrochloride with or without erlotinib hydrochloride followed by the same chemotherapy regimen with or without radiation therapy and capecitabine or fluorouracil in treating patients with pancreatic cancer that has been removed by surgery, https://clinicaltrials.gov/ct2/show/NCT01013649 (accessed 16 October 2019).
  • 29. Safran H, Winter K, Abrams RA, et al. Results of the randomized phase II portion of NRG Oncology/RTOG 0848 evaluating the addition of erlotinib to adjuvant gemcitabine for patients with resected pancreatic head adenocarcinoma. J Clin Oncol 2017; 35: 4007. [Google Scholar]
  • 30. Russo S, Ammori J, Eads J, et al. The role of neoadjuvant therapy in pancreatic cancer: a review. Future Oncol 2016; 12: 669–685. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Robin TP, Goodman KA. Radiation therapy in the management of pancreatic adenocarcinoma: review of current evidence and future opportunities. Chin Clin Oncol 2017; 6: 28. [DOI] [PubMed] [Google Scholar]
  • 32. Wagner M, Redaelli C, Lietz M, et al. Curative resection is the single most important factor determining outcome in patients with pancreatic adenocarcinoma. Br J Surg 2004; 91: 586–594. [DOI] [PubMed] [Google Scholar]
  • 33. Willett CG, Lewandrowski K, Warshaw AL, et al. Resection margins in carcinoma of the head of the pancreas. Implications for radiation therapy. Ann Surg 1993; 217: 144–148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Sohn TA, Yeo CJ, Cameron JL, et al. Resected adenocarcinoma of the pancreas-616 patients: results, outcomes, and prognostic indicators. J Gastrointest Surg 2000; 4: 567–579. [DOI] [PubMed] [Google Scholar]
  • 35. Howard TJ, Krug JE, Yu J, et al. A margin-negative R0 resection accomplished with minimal postoperative complications is the surgeon’s contribution to long-term survival in pancreatic cancer. J Gastrointest Surg 2006; 10: 1338–1345. [DOI] [PubMed] [Google Scholar]
  • 36. Bilimoria KY, Talamonti MS, Sener SF, et al. Effect of hospital volume on margin status after pancreaticoduodenectomy for cancer. J Am Coll Surg 2008; 207: 510–519. [DOI] [PubMed] [Google Scholar]
  • 37. Wong J, Solomon NL, Hsueh CT. Neoadjuvant treatment for resectable pancreatic adenocarcinoma. World J Clin Oncol 2016; 7: 1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Campbell F, Cairns A, Duthie F, et al. ; The Royal College of Pathologists. Datasets for the histopathological reporting of carcinomas of the pancreas, ampulla of Vater and common bile duct. G091, https://www.rcpath.org/uploads/assets/34910231-c106-4629-a2de9e9ae6f87ac1/g091-pancreasdataset-mar17.pdf (2017, accessed 16 October 2019).
  • 39. Esposito I, Kleeff J, Bergmann F, et al. Most pancreatic cancer resections are R1 resections. Ann Surg Oncol 2008; 15: 1651–1660. [DOI] [PubMed] [Google Scholar]
  • 40. Campbell F, Smith RA, Whelan P, et al. Classification of R1 resections for pancreatic cancer: the prognostic relevance of tumour involvement within 1mm of a resection margin. Histopathology 2009; 55: 277–283. [DOI] [PubMed] [Google Scholar]
  • 41. Strobel O, Hank T, Hinz U, et al. Pancreatic cancer surgery: the new R-status counts. Ann Surg 2017; 265: 565–573. [DOI] [PubMed] [Google Scholar]
  • 42. Fischer R, Breidert M, Keck T, et al. Early recurrence of pancreatic cancer after resection and during adjuvant chemotherapy. Saudi J Gastroenterol 2012; 18: 118–121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Dhir M, Zenati MS, Hamad A, et al. FOLFIRINOX versus gemcitabine/nab-paclitaxel for neoadjuvant treatment of resectable and borderline resectable pancreatic head adenocarcinoma. Ann Surg Oncol 2018; 25: 1896–1903. [DOI] [PubMed] [Google Scholar]
  • 44. Gillen S, Schuster T, Büschenfelde CMZ, et al. Preoperative/neoadjuvant therapy in pancreatic cancer: a systematic review and meta-analysis of response and resection percentages. PLoS Med 2010; 7: e1000267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Unno M, Motoi F, Matsuyama Y, et al. Randomized phase II/III trial of neoadjuvant chemotherapy with gemcitabine and S-1 versus upfront surgery for resectable pancreatic cancer (Prep-02/JSAP-05). J Clin Oncol 2019; 37(Suppl. 4): 189. [DOI] [PubMed] [Google Scholar]
  • 46. Versteijne E, Vogel JA, Besselink MG, et al. ; Dutch Pancreatic Cancer Group. Meta-analysis comparing upfront surgery with neoadjuvant treatment in patients with resectable or borderline resectable pancreatic cancer. Br J Surg 2018; 105: 946–958. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Fujii T, Satoi S, Yamada S, et al. Clinical benefits of neoadjuvant chemoradiotherapy for adenocarcinoma of the pancreatic head: an observational study using inverse probability of treatment weighting. J Gastroenterol 2017; 52: 81–93. [DOI] [PubMed] [Google Scholar]
  • 48. Kim EJ, Ben-Josef E, Herman JM, et al. A multi-institutional phase 2 study of neoadjuvant gemcitabine and oxaliplatin with radiation therapy in patients with pancreatic cancer. Cancer 2013; 119: 2692–2700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Turrini O, Ychou M, Moureau-Zabotto L, et al. Neoadjuvant docetaxel-based chemoradiation for resectable adenocarcinoma of the pancreas: new neoadjuvant regimen was safe and provided an interesting pathologic response. Eur J Surg Oncol 2010; 36: 987–992. [DOI] [PubMed] [Google Scholar]
  • 50. Evans DB, Varadhachary GR, Crane CH, et al. Preoperative gemcitabine-based chemoradiation for patients with resectable adenocarcinoma of the pancreatic head. J Clin Oncol 2008; 26: 3496–3502. [DOI] [PubMed] [Google Scholar]
  • 51. Varadhachary GR, Wolff RA, Crane CH, et al. Preoperative gemcitabine and cisplatin followed by gemcitabine-based chemoradiation for resectable adenocarcinoma of the pancreatic head. J Clin Oncol 2008; 26: 3487–3495. [DOI] [PubMed] [Google Scholar]
  • 52. Zhan HX, Xu JW, Wu D, et al. Neoadjuvant therapy in pancreatic cancer: a systematic review and meta-analysis of prospective studies. Cancer Med 2017; 6: 1201–1219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Andriulli A, Festa V, Botteri E, et al. Neoadjuvant/preoperative gemcitabine for patients with localized pancreatic cancer: a meta-analysis of prospective studies. Ann Surg Oncol 2012; 19: 1644–1662. [DOI] [PubMed] [Google Scholar]
  • 54. Assifi MM, Lu X, Eibl G, et al. Neoadjuvant therapy in pancreatic adenocarcinoma: a meta-analysis of phase II trials. Surgery 2011; 150: 466–473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Casadei R, Di Marco M, Ricci C, et al. Neoadjuvant chemoradiotherapy and surgery versus surgery alone in resectable pancreatic cancer: a single-center prospective, randomized, controlled trial which failed to achieve accrual targets. J Gastrointest Surg 2015; 19: 1802–1812. [DOI] [PubMed] [Google Scholar]
  • 56. Lutz MP, Zalcberg JR, Ducreux M, et al. 3rd St. Gallen EORTC gastrointestinal cancer conference: consensus recommendations on controversial issues in the primary treatment of pancreatic cancer. Eur J Cancer 2017; 79: 41–49. [DOI] [PubMed] [Google Scholar]
  • 57. Conroy T, Hammel P, Hebbar M, et al. ; Canadian Cancer Trials Group and the Unicancer-GI–PRODIGE Group. FOLFIRINOX or gemcitabine as adjuvant therapy for pancreatic cancer. N Engl J Med 2018; 379: 2395–2406. [DOI] [PubMed] [Google Scholar]
  • 58. National Comprehensive Cancer Network. Pancreatic Adenocarcinoma (Version 3.2019), https://www.nccn.org/professionals/physician_gls/pdf/pancreatic.pdf (accessed 16 October 2019).
  • 59. Khorana AA, McKemin SE, Berlin J, et al. Potentially curable pancreatic adenocarcinoma: ASCO clinical practice guideline update. J Clin Oncol 2019; 37: 2082–2088. [DOI] [PubMed] [Google Scholar]
  • 60. Versteijne E, Suker M, Groothuis K, et al. ; Dutch Pancreatic Cancer Group. Preoperative chemoradiotherapy versus immediate surgery for resectable and borderline resectable pancreatic cancer: results of the Dutch randomized phase III PREOPANC trial. J Clin Oncol 2020; 38: 1763–1773. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Jang JY, Han Y, Lee H, et al. Oncological benefits of neoadjuvant chemoradiation with gemcitabine versus upfront surgery in patients with borderline resectable pancreatic cancer: a prospective, randomized, open-label, multicenter phase 2/3 trial. Ann Surg 2018; 268: 215–222. [DOI] [PubMed] [Google Scholar]
  • 62. Pietrasz D, Turrini O, Vendrely V, et al. How does chemoradiotherapy following induction FOLFIRINOX improve the results in resected borderline or locally advanced pancreatic adenocarcinoma? An AGEO-FRENCH multicentric cohort. Ann Surg Oncol 2019; 26: 109–117. [DOI] [PubMed] [Google Scholar]
  • 63. Nagakawa Y, Hosokawa Y, Nakayama H, et al. A phase II trial of neoadjuvant chemoradiotherapy with intensity-modulated radiotherapy combined with gemcitabine and S-1 for borderline-resectable pancreatic cancer with arterial involvement. Cancer Chemother Pharmacol 2017; 79: 951–957. [DOI] [PubMed] [Google Scholar]
  • 64. Katz MHG, Shi Q, Ahmad SA, et al. Preoperative modified FOLFIRINOX treatment followed by capecitabine-based chemoradiation for borderline resectable pancreatic cancer: alliance for clinical trials in oncology trial A021101. JAMA Surg 2016; 151: e161137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Golcher H, Brunner TB, Witzigmann H, et al. Neoadjuvant chemoradiation therapy with gemcitabine/cisplatin and surgery versus immediate surgery in resectable pancreatic cancer: results of the first prospective randomized phase II trial. Strahlenther Onkol 2015; 191: 7–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Dholakia AS, Hacker-Prietz A, Wild AT, et al. Resection of borderline resectable pancreatic cancer after neoadjuvant chemoradiation does not depend on improved radiographic appearance of tumor-vessel relationships. J Radiat Oncol 2013; 2: 413–425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Lopez NE, Prendergast C, Lowy AM. Borderline resectable pancreatic cancer: definitions and management. World J Gastroenterol 2014; 20: 10740–10751. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Katz MHG, Pisters PWT, Evans DB, et al. Borderline resectable pancreatic cancer: the importance of this emerging stage of disease. J Am Coll Surg 2008; 206: 833–846; discussion 846–848. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Stokes JB, Nolan NJ, Stelow EB, et al. Preoperative capecitabine and concurrent radiation for borderline resectable pancreatic cancer. Ann Surg Oncol 2011; 18: 619–627. [DOI] [PubMed] [Google Scholar]
  • 70. Tachezy M, Gebauer F, Petersen C, et al. Sequential neoadjuvant chemoradiotherapy (CRT) followed by curative surgery vs. primary surgery alone for resectable, non-metastasized pancreatic adenocarcinoma: NEOPA- a randomized multicenter phase III study (NCT01900327, DRKS00003893, ISRCTN82191749). BMC Cancer 2014; 14: 411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Versteijne E, van Eijck CH, Punt CJA, et al. ; Dutch Pancreatic Cancer Group. Preoperative radiochemotherapy versus immediate surgery for resectable and borderline resectable pancreatic cancer (PREOPANC trial): study protocol for a multicenter randomized controlled trial. Trials 2016; 17: 127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Versteijne E, Lens E, van der Horst A, et al. Quality assurance of the PREOPANC trial (2012-003181-40) for preoperative radiochemotherapy in pancreatic cancer: the dummy run. Strahlenther Onkol 2017; 193: 630–638. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. McGinn CJ, Zalupski MM, Shureigi I, et al. Phase I trial of radiation dose escalation with concurrent weekly full-dose gemcitabine in patients with advanced pancreatic cancer. J Clin Oncol 2001; 19: 4202–4208. [DOI] [PubMed] [Google Scholar]
  • 74. Small W, Jr, Berlin J, Freedman GM, et al. Full-dose gemcitabine with concurrent radiation therapy in patients with nonmetastatic pancreatic cancer: a multicenter phase II trial. J Clin Oncol 2008; 26: 942–947. [DOI] [PubMed] [Google Scholar]
  • 75. Conroy T, Desseigne F, Ychou M, et al. ; Groupe Tumeurs Digestives of Unicancer, PRODIGE Intergroup. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med 2011; 364: 1817–1825. [DOI] [PubMed] [Google Scholar]
  • 76. Goldstein D, El-Maraghi RH, Hammel P, et al. Nab-paclitaxel plus gemcitabine for metastatic pancreatic cancer: long-term survival from a phase III trial. J Natl Cancer Inst 2015; 107: dju413. [DOI] [PubMed] [Google Scholar]
  • 77. Von Hoff DD, Ervin T, Arena FP, et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med 2013; 369: 1691–1703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Suker M, Beumer BR, Sadot E, et al. FOLFIRINOX for locally advanced pancreatic cancer: a systematic review and patient-level meta-analysis. Lancet Oncol 2016; 17: 801–810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Hackert T, Sachsenmaier M, Hinz U, et al. Locally advanced pancreatic cancer: neoadjuvant therapy with folfirinox results in resectability in 60% of the patients. Ann Surg 2016; 264: 457–463. [DOI] [PubMed] [Google Scholar]
  • 80. Neoadjuvant FOLFIRINOX versus neoadjuvant chemoradiotherapy and adjuvant chemotherapy for (borderline) resectable pancreatic carcinoma: the PREOPANC-2 study, https://www.trialregister.nl/trial/7094 (accessed 10 October 2019).
  • 81. ClinicalTrials.gov. Neoadjuvant mFOLFIRINOX with or without preoperative concomitant chemoradiotherapy in patients with borderline resectable pancreatic carcinoma (PANDAS-PRODIGE 44), https://clinicaltrials.gov/ct2/show/NCT02676349 (accessed 10 October 2019).
  • 82. Moertel CG, Frytak S, Hahn RG, et al. Therapy of locally unresectable pancreatic carcinoma: a randomized comparison of high dose (6000 rads) radiation alone, moderate dose radiation (4000 rads + 5-fluorouracil), and high dose radiation + 5-fluorouracil: the gastrointestinal tumor study group. Cancer 1981; 48: 1705–1710. [DOI] [PubMed] [Google Scholar]
  • 83. Gastrointestinal Tumor Study Group. Treatment of locally unresectable carcinoma of the pancreas: comparison of combined-modality therapy (chemotherapy plus radiotherapy) to chemotherapy alone. J Natl Cancer Inst 1988; 80: 751–755. [PubMed] [Google Scholar]
  • 84. Klaassen DJ, MacIntyre JM, Catton GE, et al. Treatment of locally unresectable cancer of the stomach and pancreas: a randomized comparison of 5-fluorouracil alone with radiation plus concurrent and maintenance 5-fluorouracil–an Eastern cooperative oncology group study. J Clin Oncol 1985; 3: 373–378. [DOI] [PubMed] [Google Scholar]
  • 85. Sultana A, Smith CT, Cunningham D, et al. Systematic review, including meta-analyses, on the management of locally advanced pancreatic cancer using radiation/combined modality therapy. Br J Cancer 2007; 96: 1183–1190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Chauffert B, Mornex F, Bonnetain F, et al. Phase III trial comparing intensive induction chemoradiotherapy (60 Gy, infusional 5-FU and intermittent cisplatin) followed by maintenance gemcitabine with gemcitabine alone for locally advanced unresectable pancreatic cancer. Definitive results of the 2000-01 FFCD/SFRO study. Ann Oncol 2008; 19: 1592–1599. [DOI] [PubMed] [Google Scholar]
  • 87. Huguet F, Girard N, Guerche CSE, et al. Chemoradiotherapy in the management of locally advanced pancreatic carcinoma: a qualitative systematic review. J Clin Oncol 2009; 27: 2269–2277. [DOI] [PubMed] [Google Scholar]
  • 88. Barhoumi M, Mornex F, Bonnetain F, et al. Locally advanced unresectable pancreatic cancer: induction chemoradiotherapy followed by maintenance gemcitabine versus gemcitabine alone: definitive results of the 2000–2001 FFCD/SFRO phase III trial. Cancer Radiother 2011; 15: 182–191. [DOI] [PubMed] [Google Scholar]
  • 89. Loehrer PJ, Sr, Feng Y, Cardenes H, et al. Gemcitabine alone versus gemcitabine plus radiotherapy in patients with locally advanced pancreatic cancer: an Eastern cooperative oncology group trial. J Clin Oncol 2011; 29: 4105–4112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Wang C, Liu X, Wang X, et al. Effects of chemoradiotherapy and chemotherapy on survival of patients with locally advanced pancreatic cancer: a meta-analysis of randomized controlled trials. Medicine (Baltimore) 2018; 97: e12260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Chang JS, Chiu YF, Yu JC, et al. The role of consolidation chemoradiotherapy in locally advanced pancreatic cancer receiving chemotherapy: an updated systematic review and meta-analysis. Cancer Res Treat 2018; 50: 562–574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Sherman WH, Hecht E, Leung D, et al. Predictors of response and survival in locally advanced adenocarcinoma of the pancreas following neoadjuvant GTX with or without radiation therapy. Oncologist 2018; 23: 4–e10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Wo JY, Niemierko A, Ryan DP, et al. Tolerability and long-term outcomes of dose-painted neoadjuvant chemoradiation to regions of vessel involvement in borderline or locally advanced pancreatic cancer. Am J Clin Oncol 2018; 41: 656–661. [DOI] [PubMed] [Google Scholar]
  • 94. 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: 195–202. [DOI] [PubMed] [Google Scholar]
  • 95. Huguet F, Hajj C, Winston CB, et al. Chemotherapy and intensity-modulated radiation therapy for locally advanced pancreatic cancer achieves a high rate of R0 resection. Acta Oncol 2017; 56: 384–390. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Habermehl D, Kessel K, Welzel T, et al. Neoadjuvant chemoradiation with gemcitabine for locally advanced pancreatic cancer. Radiat Oncol 2012; 7: 28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Reyngold M, O’Reilly E, Zinovoy M, et al. Ablative RT results in excellent local control and survival in localized pancreatic cancer. In ASTRO 2019: Int J Radiat Oncol Biol Phys. 2019; 105(Suppl. 1): S206. [Google Scholar]
  • 98. Krishnan S, Chadha AS, Suh Y, et al. Focal radiation therapy dose escalation improves overall survival in locally advanced pancreatic cancer patients receiving induction chemotherapy and consolidative chemoradiation. Int J Radiat Oncol Biol Phys 2016; 94: 755–765. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. ClinicalTrials.gov. Pancreatic carcinoma: chemoradiation compared with chemotherapy alone after induction chemotherapy (CONKO-007), https://clinicaltrials.gov/ct2/show/NCT01827553 (accessed 10 October 2019).
  • 100. ClinicalTrials.gov. Systemic therapy and chemoradiation in advanced localised pancreatic cancer – 2 (SCALOP-2), https://clinicaltrials.gov/ct2/show/NCT02024009 (accessed 10 October 2019).
  • 101. ClinicalTrials.gov. Use of high-dose radiation therapy plus chemotherapy to improve the likelihood of surgical treatment in patients with locally advanced pancreatic cancer (MAIBE), https://clinicaltrials.gov/ct2/show/NCT03523312 (accessed 10 October 2019).
  • 102. Potters L, Kavanagh B, Galvin JM, et al. ; American Society for Therapeutic Radiology and Oncology, American College of Radiology. American society for therapeutic radiology and oncology (ASTRO) and American college of radiology (ACR) practice guideline for the performance of stereotactic body radiation therapy. Int J Radiat Oncol Biol Phys 2010; 76: 326–332. [DOI] [PubMed] [Google Scholar]
  • 103. Timmerman RD, Herman J, Cho LC. Emergence of stereotactic body radiation therapy and its impact on current and future clinical practice. J Clin Oncol 2014; 32: 2847–2854. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Trakul N, Koong AC, Chang DT. Stereotactic body radiotherapy in the treatment of pancreatic cancer. Semin Radiat Oncol 2014; 24: 140–147. [DOI] [PubMed] [Google Scholar]
  • 105. De Bari B, Porta L, Mazzola R, et al. Hypofractionated radiotherapy in pancreatic cancer: lessons from the past in the era of stereotactic body radiation therapy. Crit Rev Oncol Hematol 2016; 103: 49–61. [DOI] [PubMed] [Google Scholar]
  • 106. Iacobuzio-Donahue CA, Fu B, Yachida S, et al. DPC4 gene status of the primary carcinoma correlates with patterns of failure in patients with pancreatic cancer. J Clin Oncol 2009; 27: 1806–1813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Koong AC, Le QT, Ho A, et al. Phase I study of stereotactic radiosurgery in patients with locally advanced pancreatic cancer. Int J Radiat Oncol Biol Phys 2004; 58: 1017–1021. [DOI] [PubMed] [Google Scholar]
  • 108. Koong AC, Christofferson E, Le QT, et al. Phase II study to assess the efficacy of conventionally fractionated radiotherapy followed by a stereotactic radiosurgery boost in patients with locally advanced pancreatic cancer. Int J Radiat Oncol Biol Phys 2005; 63: 320–323. [DOI] [PubMed] [Google Scholar]
  • 109. Schellenberg D, Goodman KA, Lee F, et al. Gemcitabine chemotherapy and single-fraction stereotactic body radiotherapy for locally advanced pancreatic cancer. Int J Radiat Oncol Biol Phys 2008; 72: 678–686. [DOI] [PubMed] [Google Scholar]
  • 110. Chang DT, Schellenberg D, Shen J, et al. Stereotactic radiotherapy for unresectable adenocarcinoma of the pancreas. Cancer 2009; 115: 665–672. [DOI] [PubMed] [Google Scholar]
  • 111. Schellenberg D, Kim J, Christman-Skieller C, et al. Single-fraction stereotactic body radiation therapy and sequential gemcitabine for the treatment of locally advanced pancreatic cancer. Int J Radiat Oncol Biol Phys 2011; 81: 181–188. [DOI] [PubMed] [Google Scholar]
  • 112. Goyal K, Einstein D, Ibarra RA, et al. Stereotactic body radiation therapy for nonresectable tumors of the pancreas. J Surg Res 2012; 174: 319–325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Rajagopalan MS, Heron DE, Wegner RE, et al. Pathologic response with neoadjuvant chemotherapy and stereotactic body radiotherapy for borderline resectable and locally-advanced pancreatic cancer. Radiat Oncol 2013; 8: 254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Moningi S, Dholakia AS, Raman SP, et al. The role of stereotactic body radiation therapy for pancreatic cancer: a single-institution experience. Ann Surg Oncol 2015; 22: 2352–2358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Herman JM, Chang DT, Goodman KA, et al. Phase 2 multi-institutional trial evaluating gemcitabine and stereotactic body radiotherapy for patients with locally advanced unresectable pancreatic adenocarcinoma. Cancer 2015; 121: 1128–1137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Comito T, Cozzi L, Clerici E, et al. Can stereotactic body radiation therapy be a viable and efficient therapeutic option for unresectable locally advanced pancreatic adenocarcinoma? Results of a phase 2 study. Technol Cancer Res Treat 2017; 16: 295–301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Zhong J, Patel K, Switchenko J, et al. Outcomes for patients with locally advanced pancreatic adenocarcinoma treated with stereotactic body radiation therapy versus conventionally fractionated radiation. Cancer 2017; 123: 3486–3493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Gurka MK, Kim C, He AR, et al. Stereotactic body radiation therapy (SBRT) combined with chemotherapy for unresected pancreatic adenocarcinoma. Am J Clin Oncol 2017; 40: 152–157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Jumeau R, Delouya G, Roberge R, et al. Stereotactic body radiotherapy (SBRT) for patients with locally advanced pancreatic cancer: a single center experience. Dig Liver Dis 2018; 50: 396–400. [DOI] [PubMed] [Google Scholar]
  • 120. Jung J, Yoon SM, Park JH, et al. Stereotactic body radiation for locally advanced pancreatic cancer. PLoS One 2019; 14: e0214970. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Chuong MD, Springett GM, Freilich JM, et al. Stereotactic body radiation therapy for locally advanced and borderline resectable pancreatic cancer is effective and well tolerated. Int J Radiat Oncol Biol Phys 2013; 86: 516–522. [DOI] [PubMed] [Google Scholar]
  • 122. Mellon EA, Hoffe SE, Springett GM, et al. Long-term outcomes of induction chemotherapy and neoadjuvant stereotactic body radiotherapy for borderline resectable and locally advanced pancreatic adenocarcinoma. Acta Oncol 2015; 54: 979–985. [DOI] [PubMed] [Google Scholar]
  • 123. Chuong MD, Frakes JM, Figura N, et al. Histopathologic tumor response after induction chemotherapy and stereotactic body radiation therapy for borderline resectable pancreatic cancer. J Gastrointest Oncol 2016; 7: 221–227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Mellon EA, Jin WH, Frakes JM, et al. Predictors and survival for pathologic tumor response grade in borderline resectable and locally advanced pancreatic cancer treated with induction chemotherapy and neoadjuvant stereotactic body radiotherapy. Acta Oncol 2017; 56: 391–397. [DOI] [PubMed] [Google Scholar]
  • 125. Quan K, Sutera P, Xu K, et al. Results of a prospective phase 2 clinical trial of induction gemcitabine/capecitabine followed by stereotactic ablative radiation therapy in borderline resectable or locally advanced pancreatic adenocarcinoma. Pract Radiat Oncol 2018; 8: 95–106. [DOI] [PubMed] [Google Scholar]
  • 126. Palta M, Czito BG, Duffy E, et al. A phase II trial of neoadjuvant gemcitabine/nab-paclitaxel and SBRT for potentially resectable pancreas cancer: an evaluation of acute toxicity. J Clin Oncol 2018; 36: 4121. [Google Scholar]
  • 127. Kharofa J, Mierzwa M, Olowokure O, et al. Pattern of marginal local failure in a phase II trial of neoadjuvant chemotherapy and stereotactic body radiation therapy for resectable and borderline resectable pancreas cancer. Am J Clin Oncol 2018; 42: 247–252. [DOI] [PubMed] [Google Scholar]
  • 128. Suker M, Nuyttens JJ, Eskens FALM, et al. Efficacy and feasibility of stereotactic radiotherapy after folfirinox in patients with locally advanced pancreatic cancer (LAPC-1 trial). EClinicalMedicine 2019; 17: 100200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Park JJ, Hajj C, Reyngold M, et al. Stereotactic body radiation vs. intensity-modulated radiation for unresectable pancreatic cancer. Acta Oncol 2017; 56: 1746–1753. [DOI] [PubMed] [Google Scholar]
  • 130. Blair AB, Rosati LM, Rezaee N, et al. Postoperative complications after resection of borderline resectable and locally advanced pancreatic cancer: the impact of neoadjuvant chemotherapy with conventional radiation or stereotactic body radiation therapy. Surgery 2018; 163: 1090–1096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Chapman BC, Gleisner A, Rigg D, et al. Perioperative outcomes and survival following neoadjuvant stereotactic body radiation therapy (SBRT) versus intensity-modulated radiation therapy (IMRT) in pancreatic adenocarcinoma. J Surg Oncol 2018; 117: 1073–1083. [DOI] [PubMed] [Google Scholar]
  • 132. Rwigema JCM, Parikh SD, Heron DE, et al. Stereotactic body radiotherapy in the treatment of advanced adenocarcinoma of the pancreas. Am J Clin Oncol 2011; 34: 63–69. [DOI] [PubMed] [Google Scholar]
  • 133. Katz MHG, Fleming JB, Bhosale P, et al. Response of borderline resectable pancreatic cancer to neoadjuvant therapy is not reflected by radiographic indicators. Cancer 2012; 118: 5749–5756. [DOI] [PubMed] [Google Scholar]
  • 134. Ferrone CR, Marchegiani G, Hong TS, et al. Radiological and surgical implications of neoadjuvant treatment with FOLFIRINOX for locally advanced and borderline resectable pancreatic cancer. Ann Surg 2015; 261: 12–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Kluger MD, Rashid MF, Rosario VL, et al. Resection of locally advanced pancreatic cancer without regression of arterial encasement after modern-era neoadjuvant therapy. J Gastrointest Surg 2018; 22: 235–241. [DOI] [PubMed] [Google Scholar]
  • 136. Klaassen R, Gurney-Champion OJ, Engelbrecht MRW, et al. Evaluation of six diffusion-weighted MRI models for assessing effects of neoadjuvant chemoradiation in pancreatic cancer patients. Int J Radiat Oncol Biol Phys 2018; 102: 1052–1062. [DOI] [PubMed] [Google Scholar]
  • 137. Okano K, Suto H, Oshima M, et al. 18F-fluorodeoxyglucose positron emission tomography to indicate conversion surgery in patients with initially unresectable locally advanced pancreatic cancer. Jpn J Clin Oncol 2018; 48: 434–441. [DOI] [PubMed] [Google Scholar]
  • 138. Petrelli F, Comito T, Ghidini A, et al. Stereotactic body radiation therapy for locally advanced pancreatic cancer: a systemic review and pooled analysis of 19 trials. Int J Radiat Oncol Biol Phys 2017; 97: 313–322. [DOI] [PubMed] [Google Scholar]
  • 139. de Geus SWL, Eskander MF, Kasumova GG, et al. Stereotactic body radiotherapy for unresected pancreatic cancer: a nationwide review. Cancer 2017; 123: 4158–4167. [DOI] [PubMed] [Google Scholar]
  • 140. Tchelebi LT, Lehrer EJ, Trifiletti DM, et al. Conventionally fractionated radiation therapy versus stereotactic body radiation therapy for locally advanced pancreatic cancer (CRiSP): an international systematic review and meta-analysis. Cancer 2020; 126: 2120–2131. [DOI] [PubMed] [Google Scholar]
  • 141. Jiang W, Hague W, Verma V, et al. Neoadjuvant stereotactic body radiation therapy for nonmetastatic pancreatic adenocarcinoma. Acta Oncol 2019; 58: 1259–1266. [DOI] [PubMed] [Google Scholar]
  • 142. ClinicalTrials.gov. Combination chemotherapy with or without hypofractionated radiation therapy before surgery in treating patients with pancreatic cancer. (Alliance A021501), https://clinicaltrials.gov/ct2/show/NCT02839343 (accessed 6 April 2020).
  • 143. Rosati LM, Kumar R, Herman JM. Integration of stereotactic body radiation therapy into the multidisciplinary management of pancreatic cancer. Semin Radiat Oncol 2017; 27: 256–267. [DOI] [PubMed] [Google Scholar]
  • 144. Janssen QP, O’Reilly EM, van Eijck CHJ, et al. Neoadjuvant treatment in patients with resectable and borderline resectable pancreatic cancer. Front Oncol 2020; 10: 41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145. Expert Panel on Radiation Oncology-Gastrointestinal, Small W, Jr, Hayes JP, et al. ACR appropriateness criteria® borderline and unresectable pancreas cancer. Oncology (Williston Park) 2016; 30: 619–624. [PubMed] [Google Scholar]
  • 146. Zhu X, Shi D, Li F, et al. Prospective analysis of different combined regimens of stereotactic body radiation therapy and chemotherapy for locally advanced pancreatic cancer. Cancer Med 2018; 7: 2913–2924. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147. Rudra S, Jiang N, Rosenberg SA, et al. Using adaptive magnetic resonance image-guided radiation therapy for treatment of inoperable pancreatic cancer. Cancer Med 2019; 8: 2123–2132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148. ClinicalTrials.gov. Prospective phase II study of stereotactic magnetic resonance (MRI)-guided on-table adaptative radiation therapy (SMART) for patients with borderline or inoperable locally advanced pancreatic cancer, https://clinicaltrials.gov/ct2/show/NCT03621644 (accessed 6 April 2020).
  • 149. Dholakia AS, Kumar R, Raman SP, et al. Mapping patterns of local recurrence after pancreaticoduodenectomy for pancreatic adenocarcinoma: a new approach to adjuvant radiation field design. Int J Radiat Oncol Biol Phys 2013; 87: 1007–1015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150. Baine MJ, Sleightholm R, Lin C. Incidence and patterns of locoregional failure after stereotactic body radiation therapy for pancreatic adenocarcinoma. Pract Radiat Oncol 2019; 9: e29–e37. [DOI] [PubMed] [Google Scholar]
  • 151. ClinicalTrials.gov. Stereotactic body radiation therapy or conventionally fractionated concurrent chemotherapy and radiation therapy preoperatively for resectable or borderline resectable pancreatic adenocarcinoma, https://clinicaltrials.gov/ct2/show/NCT03704662 (accessed 6 April 2020).
  • 152. ClinicalTrials.gov. Phase III FOLFIRINOX (mFFX) +/- SBRT in locally advanced pancreatic cancer, https://clinicaltrials.gov/ct2/show/NCT01926197 (accessed 6 April 2020).
  • 153. Royal RE, Levy C, Turner K, et al. Phase 2 trial of single agent Ipilimumab (anti-CTLA-4) for locally advanced or metastatic pancreatic adenocarcinoma. J Immunother 2010; 33: 828–833. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154. Brahmer JR, Tykodi SS, Chow LQM, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med 2012; 366: 2455–2465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155. Aglietta M, Barone C, Sawyer MB, et al. A phase I dose escalation trial of tremelimumab (CP-675,206) in combination with gemcitabine in chemotherapy-naive patients with metastatic pancreatic cancer. Ann Oncol 2014; 25: 1750–1755. [DOI] [PubMed] [Google Scholar]
  • 156. O’Reilly EM, Oh DY, Dhani N, et al. Durvalumab with or without tremelimumab for patients with metastatic pancreatic ductal adenocarcinoma: a phase 2 randomized clinical trial. JAMA Oncol. Epub ahead of print 18 July 2019. DOI: 10.1001/jamaoncol.2019.1588. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157. Young K, Hughes DJ, Cunningham D, et al. Immunotherapy and pancreatic cancer: unique challenges and potential opportunities. Ther Adv Med Oncol 2018; 10: 1758835918816281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158. O’Hara MH, O’Reilly EM, Rosemarie M, et al. Abstract CT004: a phase Ib study of CD40 agonistic monoclonal antibody APX005M together with gemvitabine (Gem) and nab-paclitaxel (NP) with or without nivolumab (Nivo) in untreated metastatic ductal pancreatic adenocarcinoma (PDAC) patients. Presented at The American Association for Cancer Research 110th Annual Meeting 2019, 29 March – 3 April 2019, Atlanta. [Google Scholar]
  • 159. Nywening TM, Wang-Gillam A, Sanford DE, et al. Phase Ib study targeting tumour associated macrophages with CCR2 inhibition plus FOLFIRINOX in locally advanced and borderline resectable pancreatic cancer. Lancet Oncol 2016; 17: 651–662. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160. ClinicalTrials.gov. A trial of PEGPH20 in combination with Avelumab in chemotherapy resistant pancreatic cancer, https://clinicaltrials.gov/ct2/show/NCT03481920 (accessed 10 October 2019).
  • 161. ClinicalTrials.gov. A phase 1, open-label dose escalation first-in-human study to evaluate the tolerability, safety, maximum tolerated dose, preliminary clinical activity and pharmacokinetics of am0010 in patients with advanced solid tumors, https://clinicaltrials.gov/ct2/show/NCT02009449 (accessed 10 October 2019).
  • 162. Van Limbergen EJ, De Ruysscher DK, Olivo Pimentel V, et al. Combining radiotherapy with immunotherapy: the past, the present and the future. Br J Radiol 2017; 90: 20170157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163. Wang Y, Deng W, Li N, et al. Combining immunotherapy and radiotherapy for cancer treatment: current challenges and future directions. Front Pharmacol 2018; 9: 185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164. Gajiwala S, Torgeson A, Garrido-Laguna I, et al. Combination immunotherapy and radiation therapy strategies for pancreatic cancer – targeting multiple steps in the cancer immunity cycle. J Gastrointest Oncol 2018; 9: 1014–1026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165. Mitin T, Zietman AL. Promise and pitfalls of heavy-particle therapy. J Cin Oncol 2014; 32: 2855–2863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166. Ebner DK, Kamada T. The emerging role of carbon-ion radiotherapy. Front Oncol 2016; 6: 140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167. Terashima K, Demizu Y, Hashimoto N, et al. A phase I/II study of gemcitabine-concurrent proton radiotherapy for locally advanced pancreatic cancer without distant metastasis. Radiother Oncol 2012; 103: 25–31. [DOI] [PubMed] [Google Scholar]
  • 168. Sachsman S, Nichols RC, Morris CG, et al. Proton therapy and concomitant capecitabine for non-metastatic unresectable pancreatic adenocarcinoma. Int J Particle Ther 2014; 1: 692–701. [Google Scholar]
  • 169. Shinoto M, Yamada S, Terashima K, et al. ; Working Group for Pancreas Cancer. Carbon ion radiation therapy with concurrent gemcitabine for patients with locally advanced pancreatic cancer. Int J Radiat Oncol Biol Phys 2016; 95: 498–504. [DOI] [PubMed] [Google Scholar]
  • 170. Jethwa KR, Tryggestad EJ, Whitaker TJ, et al. Initial experience with intensity modulated proton therapy for intact, clinically localized pancreas cancer: clinical implementation, dosimetric analysis, acute treatment-related adverse events, and patient-reported outcomes. Adv Radiat Oncol 2018; 3: 314–321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171. Kawashiro S, Yamada S, Okamoto M, et al. Multi-institutional study of carbon-ion radiotherapy for locally advanced pancreatic cancer: Japan carbon-ion radiation oncology study group (J-CROS) study 1403 pancreas. Int J Radiat Oncol Biol Phys 2018; 101: 1212–1221. [DOI] [PubMed] [Google Scholar]
  • 172. Shinoto M, Terashima K, Suefuji H, et al. A single institutional experience of combined carbon-ion radiotherapy and chemotherapy for unresectable locally advanced pancreatic cancer. Radiother Oncol 2018; 129: 333–339. [DOI] [PubMed] [Google Scholar]
  • 173. Shinoto M, Yamada S, Yasuda S, et al. ; Working Group for Pancreas Cancer. Phase 1 trial of preoperative, short-course carbon-ion radiotherapy for patients with resectable pancreatic cancer. Cancer 2013; 119: 45–51. [DOI] [PubMed] [Google Scholar]
  • 174. Hong TS, Ryan DP, Borger DR, et al. A phase 1/2 and biomarker study of preoperative short course chemoradiation with proton beam therapy and capecitabine followed by early surgery for resectable pancreatic ductal adenocarcinoma. Int J Radiat Oncol Biol Phys 2014; 89: 830–838. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175. Thompson RF, Mayekar SU, Zhai H, et al. A dosimetric comparison of proton and photon therapy in unresectable cancers of the head of pancreas. Med Phys 2014; 41: 081711. [DOI] [PubMed] [Google Scholar]
  • 176. Houweling AC, Crama K, Visser J, et al. Comparing the dosimetric impact of interfractional anatomical changes in photon, proton and carbon ion radiotherapy for pancreatic cancer patients. Phys Med Biol 2017; 62: 3051–3064. [DOI] [PubMed] [Google Scholar]
  • 177. Takatori K, Terashima K, Yoshida R, et al. Upper gastrointestinal complications associated with gemcitabine-concurrent proton radiotherapy for inoperable pancreatic cancer. J Gastroenterol 2014; 49: 1074–1080. [DOI] [PubMed] [Google Scholar]
  • 178. Durante M, Orecchia R, Loeffler JS. Charged-particle therapy in cancer: clinical uses and future perspectives. Nat Rev Clin Oncol 2017; 14: 483–495. [DOI] [PubMed] [Google Scholar]
  • 179. Dolde K, Naumann P, Dávid C, et al. 4D dose calculation for pencil beam scanning proton therapy of pancreatic cancer using repeated 4DMRI datasets. Phys Med Biol 2018; 63: 165005. [DOI] [PubMed] [Google Scholar]
  • 180. Durante M, Tommasino F, Yamada S. Modeling combined chemotherapy and particle therapy for locally advanced pancreatic cancer. Front Oncol 2015; 5: 145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181. ClinicalTrials.gov. Trial of carbon ion versus photon radiotherapy for locally advanced, unresectable pancreatic cancer (CIPHER), https://clinicaltrials.gov/ct2/show/NCT03536182 (accessed 10 October 2019).

Articles from Therapeutic Advances in Medical Oncology are provided here courtesy of SAGE Publications

RESOURCES