Next Article in Journal
C-Reactive Protein Kinetic as a Potential Predictive and Prognostic Factor during Treatment with Checkpoint Inhibitors in R/M-HNSCC
Previous Article in Journal
Canine Cancer Diagnostics by X-ray Diffraction of Claws
Previous Article in Special Issue
Total Neoadjuvant Therapy for Rectal Cancer: Which Regimens to Use?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Total Neoadjuvant Therapy in Localized Pancreatic Cancer: Is More Better?

by
Rita Saúde-Conde
1,*,
Benjelloun El Ghali
2,
Julie Navez
3,
Christelle Bouchart
2,† and
Jean-Luc Van Laethem
1,†
1
Digestive Oncology Department, Hôpitaux Universitaires de Bruxelles (HUB), Université Libre de Bruxelles, 1070 Brussels, Belgium
2
Department of Radiation Oncology, Hôpitaux Universitaires de Bruxelles (HUB), Institut Jules Bordet, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
3
Department of Abdominal Surgery and Transplantation, Hôpitaux Universitaires de Bruxelles (HUB), Hopital Erasme, Université Libre de Bruxelles, 1070 Brussels, Belgium
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Cancers 2024, 16(13), 2423; https://doi.org/10.3390/cancers16132423
Submission received: 15 May 2024 / Revised: 24 June 2024 / Accepted: 24 June 2024 / Published: 30 June 2024
(This article belongs to the Special Issue Preoperative Chemoradiotherapy for Gastrointestinal Cancer)

Abstract

:

Simple Summary

Pancreatic cancer is challenging to treat due to late diagnosis and limited options. Surgery, the main treatment, often leads to poor long-term outcomes, prompting exploration of alternative approaches like neoadjuvant therapy (NAT) and total neoadjuvant therapy (TNT). While NAT aims to improve surgical success and overall survival, there is room for enhancement, leading to consideration of neoadjuvant strategies combining full-dose chemotherapy and radiotherapy in TNT. TNT, integrating chemotherapy and radiotherapy before surgery, could increase the likelihood of successful surgery and cure for locally advanced cases. This article explores different TNT strategies, categorized based on radiotherapy techniques, offering a thorough analysis of their effectiveness in borderline resectable and locally advanced pancreatic cancer. The central question remains: does more treatment before surgery always yield better results?

Abstract

Pancreatic ductal adenocarcinoma (PDAC) poses a significant challenge in oncology due to its advanced stage upon diagnosis and limited treatment options. Surgical resection, the primary curative approach, often results in poor long-term survival rates, leading to the exploration of alternative strategies like neoadjuvant therapy (NAT) and total neoadjuvant therapy (TNT). While NAT aims to enhance resectability and overall survival, there appears to be potential for improvement, prompting consideration of alternative neoadjuvant strategies integrating full-dose chemotherapy (CT) and radiotherapy (RT) in TNT approaches. TNT integrates chemotherapy and radiotherapy prior to surgery, potentially improving margin-negative resection rates and enabling curative resection for locally advanced cases. The lingering question: is more always better? This article categorizes TNT strategies into six main groups based on radiotherapy (RT) techniques: (1) conventional chemoradiotherapy (CRT), (2) the Dutch PREOPANC approach, (3) hypofractionated ablative intensity-modulated radiotherapy (HFA-IMRT), and stereotactic body radiotherapy (SBRT) techniques, which further divide into (4) non-ablative SBRT, (5) nearly ablative SBRT, and (6) adaptive ablative SBRT. A comprehensive analysis of the literature on TNT is provided for both borderline resectable pancreatic cancer (BRPC) and locally advanced pancreatic cancer (LAPC), with detailed sections for each.

1. Introduction

Pancreatic ductal adenocarcinoma (PDAC) poses a formidable challenge in oncology, marked by a grim prognosis primarily attributed to frequent distant metastases and/or advanced local growth precluding surgical intervention [1]. Pancreatic cancer (PC) ranks fourth in number of cancer deaths, with projections placing it at the second position by 2030 in the Western world [2]. Early detection is hindered by subtle and late-presenting symptoms, with close to half of patients already metastatic at diagnosis. Merely 20% of cases are immediately resectable, while approximately half of them fall into the category of “borderline resectable” (BR) and 1/3 are locally advanced (LA) and unresectable, according to the tumor’s relationship with adjacent vascular structures and the probability of obtaining a negative microscopic margin (R0) [2,3,4].
For all localized stages, surgical resection stands as the only potentially curable treatment, necessary to obtain long-term survival. Yet even in operated cases, the prognosis remains grim, with a 5-year overall survival of only 20%. While acknowledging that R0 status is one of the most important prognostic factors, it is worth noting that some patients with positive margins or margins < 1 mm survive without recurrence, highlighting the complexity of pancreatic cancer management [2,4]. As a standard practice, surgical intervention is commonly followed by adjuvant therapy, a strategy consistently proven to significantly improve survival rates. However, one should recognize that only half of the patients successfully complete the entire adjuvant therapy sequence [5,6,7]. As such, the conventional “surgery-first” approach has become less appealing, especially for individuals with BRPC and LAPC, where achieving a margin-negative resection is highly challenging [8]. Given the likelihood of micrometastatic disease at diagnosis, even in cases of localized disease, there has been emerging emphasis on the concept of neoadjuvant therapy (NAT) as a new therapeutic strategy to improve the probability of R0 resection and OS, in both resectable and BRPC. Similarly, a “sandwich” approach has gained attraction. This approach ensures the neoadjuvant administration of systemic therapy, minimizing the risk of missing a critical window for surgery and guaranteeing the delivery of systemic therapy, even in the presence of perioperative complications [9]. More recently, similarly to treatment strategies applied in rectal cancer, another concept called total neoadjuvant therapy (TNT) was developed by adding neoadjuvant radiotherapy (RT) to the treatment of BRPC and LAPC [10,11,12]. In TNT, the sequencing of chemotherapy (CT) and radiotherapy treatments has demonstrated an increased likelihood of patients achieving margin-negative and node-negative disease, coupled with higher rates of complete and major pathologic response which could lead to improved overall survival (OS) [5,13,14]. This novel approach in pancreatic cancer can mirror successful paradigms seen in rectal cancer, emphasizing the potential benefits of tailoring treatment sequences to enhance therapeutic outcomes and address challenges associated with completing postoperative adjuvant systemic therapy [15,16,17,18,19].
In parallel, efforts have been made by the American Joint Committee on Cancer (AJCC) and the American Hepato-Pancreato-Biliary Association (AHPBA), later adopted by the National Comprehensive Cancer Network (NCCN) in order to obtain a standardized definition of resectability [20,21,22,23]. While there is now a standardized definition of resectability criteria available, the current imperative lies in fostering a worldwide consensus allowing data comparison across clinical trials. In fact, many trials encompass a spectrum of pancreatic cancer patients, including and mixing those classified as resectable pancreatic cancer (RPC), BRPC, and even LAPC. Furthermore, it is paramount to prioritize the acquisition of a thorough understanding of the precise sequence, duration, and regimens of TNT that offer benefits to patients with BRPC and LAPC. In this review, we examine the existing evidence concerning the use of NAT for PDAC patients as well as an exploration of the advantages, drawbacks, and limitations of NAT. Thereafter, we delve into the available evidence related specifically to the different TNT strategies available for localized PDAC and compare their aspects regarding chemotherapy regimen/exposure/duration, RT techniques, and treatment sequences. Lastly, we will undertake a review of the assessment of treatment response and selection of patients in the era of precision medicine.

2. What Is Total Neoadjuvant Therapy?

TNT strategy is gaining popularity as it offers the potential advantage of increasing the likelihood of achieving negative margins as well as the potential to convert LAPC into candidates for curative oncological resection. TNT specifically aims to utilize a combination of CT and RT with or without concomitant chemotherapy before surgical exploration [5,24,25,26].
In this article, we divided the TNT strategies into six main categories according to the RT technique used: (1) conventional chemoradiotherapy (CRT) approach; (2) Dutch PREOPANC approach, (3) hypofractionated ablative intensity modulated radiotherapy (HFA-IMRT) strategy, and finally stereotaxic body radiotherapy (SBRT) techniques. The latter can be divided into the following: (4) non-ablative SBRT (biological effective doses (BED) 10 around 50–60 Gy), (5) nearly ablative SBRT (BED10 around 70–85 Gy), and finally (6) adaptive ablative SBRT approach (BED10 around 100 Gy). A comprehensive analysis of the existing literature on TNT was conducted for both BRPC and LAPC patients, detailed in distinct sections.

3. General Risks and Benefits of NAT and TNT

While NAT has gained increasing popularity, it is also associated with potential disadvantages. The risk of distant metastases development during NAT can be as high as 30% [27,28]. This rate aligns with the early disease recurrence observed in patients with RPC who undergo immediate surgery and can be regarded as analogous to an early recurrence (within 6 months) following surgical resection after NAT [29,30]. However, the risk of disease progression during NAT should be analyzed as part of selecting optimal surgery candidates [30]. Patients with PDAC encountering early distant metastases after undergoing upfront major pancreatectomy likely undergo surgery-related burdens without oncologic benefits [31]. On the other hand, delayed surgery, whether due to treatment toxicity or patient’s deterioration, could affect outcomes. Yet, studies suggest this delay in surgery may also allow for careful patient selection and preoperative preparations, such as pre-rehabilitation and nutritional optimization. Two retrospective cohort studies from the US National Cancer Database explored the impact of waiting time before surgery [32,33]. Mirkin et al. found that long-term survival remained unaffected by the waiting time [33]. Swords et al. observed lower mortality rates for patients with average and long waiting times for surgery. Furthermore, the 90-day mortality was lower in both groups compared to shorter waiting time for surgery [32].
On the other hand, postoperative complications may hinder the administration of adjuvant therapy associated with improved outcomes [34,35]. A retrospective study found that in the NAT setting, 83% of patients completed the entire treatment sequence, versus 58% in the surgery-first group. For the surgery-first group, the reasons for the failure to complete the entire sequence included early disease progression, major complications, and deterioration of performance status. Completing the entire sequence led to significantly longer overall survival (36.0 versus 11.0 months, p < 0.001) [36]. Additionally, NAT does not seem to increase the postoperative complication rate [37,38]. A systematic review found comparable rates of fistula and infection, as well as similar mortality [37]. Additional studies noted reduced specific complications such as pancreatic fistula and hemorrhage or gastroparesis after NAT [39,40]. The most plausible explanation for these observations is that NAT, particularly RT, may induce pancreatic fibrosis, leading to a decreased fistula incidence and lower complication risks [30,38]. Kim et al. studied specifically TNT in operable PDAC patients, analyzing 541 patients (58% BPRC) [8]. Of these, 16% received TNT, and 84% received standard neoadjuvant treatment (SNT). Treatment duration was longer in the TNT group (5.5 months) compared to SNT (4 months) (p < 0.01), with similar completion rates for intended therapy and subsequent surgery (p = 0.90). The study suggests that patients tolerating SNT might benefit from TNT without compromising surgical operability. Accordingly, the results from the randomized phase III PREOPANC-2 trial comparing NAT FOLFIRINOX versus TNT for RPC/BRPC showed no differences in resection rates (p = 0.69) [41].
Patients undergoing neoadjuvant chemotherapy may benefit from changing their chemotherapy regimen or adding radiotherapy before resection in case of early loco-regional progression or no relevant response [7,42]. TNT may facilitate such a therapeutic switch. Alva-Ruez et al. reported the Mayo Clinic’s experience with chemotherapy switching in 468 patients with BRPC and LAPC [42]. Following a median of six cycles of neoadjuvant chemotherapy, 30% underwent a switch due to progression, absence of an objective response, or toxicity. Of those, 72% were resected. Although acknowledging probable patient selection bias, this strategy presents an intriguing prospect for future trials comparing SNT vs. TNT [31].
Overall, the potential benefits of NAT and TNT appear to outweigh their drawbacks.

4. The Advent of TNT for BRPC Patients

While uncertainty remains, whereas neoadjuvant treatment should be given to RPC patients, especially following the results of the NORPACT-1 trial, the results from ongoing randomized trials are awaited [43,44]. On the other side, NCCN and ESMO guidelines recommend the use of NAT in the management of BRPC patients where the neoadjuvant approach appears to be more beneficial [45,46].

4.1. Use of Single Modality NAT

In a recent meta-analysis, the Dutch Pancreatic Cancer group compared upfront surgery with the use of a single NAT modality for both RPC and BRPC [47]. They analyzed 38 studies, involving 3884 patients, finding that the median overall survival (OS) was longer in the NAT group (19.2 months versus 12.8 months in the upfront-surgery group). The NAT group included the use of various chemotherapy or RT techniques, leading to heterogeneity that precluded subgroup analysis [47]. A separate patient-level meta-analysis specifically focused on neoadjuvant FOLFIRINOX in 313 BRPC patients, showing a resection rate of 67.8%, and an R0 resection rate of 83.9% (95% CI: 76.8–89.1) [48]. The median OS was 22.2 months, with a median progression-free survival (PFS) of 18.0 months [48].
To date, only two prospective randomized trials specifically explored the value of single modality NAT in patients with BRPC only [49,50]. A phase II Korean trial comparing preoperative versus postoperative CRT showed a higher resection rate (51.8% versus 26.1%) and 2-year survival rate (40.7% versus 26.1%; HR, 1.495; 95% CI, 0.66–3.36; p = 0.028), in favor of the NAT group [49]. The recurrence rate was notably high in both groups (around 88%), but the median OS was higher in the NAT group (21 versus 12 months). However, this trial was closed prematurely for efficacy following the first interim analysis, with only 58 enrolled patients [49]. In the randomized phase II ESPAC5 trial, 90 BRPC patients were assigned to receive either immediate surgery, 8 weeks of neoadjuvant gemcitabine/capecitabine, neoadjuvant FOLFIRINOX, or capecitabine-based CRT (total dose 50·4 Gy in 28 daily fractions) [50]. While resection rates did not significantly differ between immediate surgery and NAT groups (68% vs. 55%), R0 resection rates were indeed higher in the NAT groups (23% vs. 14%, p = 0.49). One-year OS rates showed significant differences, with higher rates in favor of NAT groups (39% for immediate surgery, 78% for gemcitabine plus capecitabine, 84% for FOLFIRINOX, and 60% for CRT, p = 0.0028) and DFS compared to immediate surgery (p = 0.016) [50].
Three additional randomized clinical trials (RCTs), including both BRPC and RPC patients, have also explored single modality NAT with distinct treatment regimens [51,52,53]. Caution is necessary when analyzing these trials due to the restricted number of patients in two of them, the limited accessibility of data in a third, and the variability in regimen treatments used across the trials. In 2015, Golcher et al. conducted a phase II RCT evaluating the role of NAT CRT (55.8 Gy in 31 fractions, gemcitabine/cisplatin-based) versus surgery first. However, the trial was prematurely closed due to poor patient enrolment (n = 73). The partial results showed no significant differences in R0 resection (48% vs. 52%, p = 0.81), postoperative complications, or survival in the intention to treat analysis [51]. In 2021, the phase II/III RCT NEPAFOX comparing adjuvant gemcitabine and perioperative FOLFIRINOX was also halted due to recruitment challenges after enrolling 40 RPC and BRPC patients [53]. Among the patients, 57.9% did not receive surgery due to progressive disease, death, toxicities, and pneumonia. Perioperative morbidity was higher in the surgery-first group (72.2%), but R0 resection rate was higher (72.2%) compared to 45.5% for both in the NAT group. Median OS was also favored in the surgery-first group (25.68 months vs. 10.03 months, HR 0.722, p = 0.4099) [53]. The Japanese phase III RCT Prep-02/JSAP-05 evaluated the role of two cycles of NAT using gemcitabine and S-1, followed by 6 months of adjuvant chemotherapy in 362 RPC and BRPC patients. While no differences were seen between R0 resection rates and morbidity of surgery (no statistical data were provided in the abstract), the median OS was higher in the NAT group compared with the upfront-surgery group (36.7 vs. 26.6 months, HR 0.73, 95%CI 0.55–0.94, p = 0.015) [54].
While these findings support an NAT strategy, they also indicate the possibility of improvement of the NAT sequences in order to allow for better outcomes for BRPC patients. This prompts consideration of alternative neoadjuvant strategies combining full-dose CT and RT in TNT approaches.

4.2. Use of TNT Strategies for BRPC

4.2.1. Conventional CRT with Induction or Consolidation Chemotherapy Prior to Surgery

Similarly, to a TNT approach often used in rectal cancer patients (OPRA trial), an approach that has been used in localized PDAC patients consists in a neoadjuvant conventional CRT (BED10 around 60 Gy) either preceded by induction chemotherapy or followed by consolidation chemotherapy [55].
Only two non-randomized phase II trials addressed the question of whether induction chemotherapy followed by CRT is beneficial specifically for BRPC patients [13,56]. First, Katz et al. presented the results of 22 patients with BRPC (intergroup criteria) who underwent an induction chemotherapy treatment of four cycles of FOLFIRINOX followed by CRT with concomitant capecitabine. The results showed a resection rate of 68% and an R0 resection rate of 93%, with 13% achieving a pathological complete response (pCR). The median OS was 21.7 months, indicating the feasibility of this TNT regimen despite notable toxicity associated with preoperative therapies (grade G ≥ 3: 64%) [13]. Second, Nagakawa et al. studied an induction chemotherapy treatment consisting of two cycles of gemcitabine followed by chemoradiotherapy with gemcitabine and S1 in 27 patients with BRPC (author’s own definition derived from NCCN). While in this study 48.1% of patients had ≥G3 adverse events (AEs) (mostly hematologic), a similar resection rate (70.3%) and R0 resection rate (94.7%) were found. Thirteen patients (68.4%) experienced distant metastasis at the initial site of recurrence following resection. Local recurrence occurred in just one of these patients (7.7%). The median OS was 22.4 months, with a corresponding 1-year survival rate of 81.3% [56]. The limited patient cohort and the lack of comparative analysis in both trials hinder the formulation of definitive conclusions in BPRC. Yet, retrospective studies encompassing a larger patient cohort, as outlined in Table 1, have shown encouraging results [6,57,58,59]. However, the use conventional CRT, associated with the delivery of low BED10 to the tumor and prolonged interruption of full-dose chemotherapy, may not be the fittest approach in PDAC, known to be radioresistant. Therefore, other TNT approaches were developed and studied.

4.2.2. The Dutch TNT Strategy: PREOPANC Regimen

The “PREOPANC approach” established by the Dutch Pancreatic Cancer Group consists in the administration of preoperative gemcitabine associated with hypofractionated CRT (36 Gy in 15 fractions; three cycles of gemcitabine 1000 mg/m2—second cycle concomitantly with CRT) [41,67,73]. The doses of gemcitabine and the RT scheme were established according to two phase I/II studies in order to use also full-dose gemcitabine during CRT [74,75].
The phase III RCT PREOPANC-1 (Table 1) compared the Dutch regimen versus upfront surgery in 246 RPC and BRPC (46%). Adjuvant gemcitabine was administered for six cycles in the upfront-surgery group and for four cycles in the CRT group. After long-term follow-up (59 months), the results highlighted a superior R0 resection rate (72% versus 43%, p  <  0.001), median loco-regional failure-free interval (13.4 months versus 31.2, p  =  0.004), and notably lower occurrences of pathologic lymph nodes (35% versus 82%, p  <  0.001) and perineural (45% versus 85%, p  <  0.001) and vascular invasion (36 versus 65%, p < 0.001) in favor of the TNT group. The median OS in the intention-to-treat population, which was the primary endpoint, was modestly improved for the CRT arm (15.7 months versus 14.3 months, p = 0.025), as well as distant metastasis-free survival (17.4 months versus 12.5 months, p = 0.070) and median DFS (8.1 versus 7.7 months, p = 0.09). Interestingly, the TNT group achieved a 5-year overall survival rate of 20.5%, whereas the upfront-surgery group attained a rate of 6.5%. For BRPC patients, the unstratified HR was 0.67 (95% CI 0.45–0.99), favoring TNT in OS, but did not reach statistical significance (p = 0.88). However, emerging multi-agent chemotherapy protocols like gemcitabine/nab-paclitaxel, FOLFIRINOX (FFX), and more recently NALIRIFOX have exhibited encouraging advancements compared to gemcitabine in metastatic, adjuvant, and increasingly in the neoadjuvant strategy as well [76,77,78]. Although FOLFIRINOX appears to be an efficacious option, uncertainty remains on its superiority compared to alternative multi-agent chemotherapy regimens [79,80,81,82]. As such, the phase III PREOPANC-2 trial (Table 1) compared the Dutch TNT regimen versus neoadjuvant FFX (eight cycles prior to surgery) [41]. Recently presented results from 375 randomized RPC and BRPC patients showed a median OS of 21.9 months in the FFX group versus 21.3 months in the TNT arm (HR 0.87; 95% CI 0.68–1.12, p = 0.28). Completion rates (p < 0.001) and resection rates were similar between both groups (77% in the FFX group and 75% in the TNT group, p = 0.69), as well as R0 resection rates (61% and 67% in the FFX and TNT group, respectively, p = 0.28). Serious adverse events (SAEs) were also similar (49% in the FFX group and 43% in the TNT arm, p = 0.26). While no differences were seen in OS, patients treated with the PREOPANC approach had a significantly higher pathologic response in lymph nodes (ypN0: 7% in the FFX group and 58% in the TNT arm; ypN2: 20% in the FFX group vs. 7% in the TNT arm, p < 0.01), hinting at the potential role of radiotherapy. Of note, BRPC patients were a minority in both groups (around 35%), suggesting the need for separate future studies to address each population. Furthermore, it is important to note that no adjuvant treatment was performed in the FFX arm contrary to the TNT arm (four cycles of adjuvant gemcitabine) and only 62% of the patients in the FFX arm completed the whole treatment, therefore leaving the question of potential further outcomes improvement in case of the use of a more optimized FOLFIRINOX arm.
Therefore, despite several interesting results with the Dutch TNT regimen, the use of gemcitabine monotherapy—the old chemotherapy standard for PDAC—also leaves room for further improvement of the TNT strategy.

4.2.3. Stereotactic Body Radiotherapy (SBRT)

SBRT enables the precise administration of elevated doses to tumors over a limited number of sessions (1–5), thereby minimizing dose exposure and toxicity to nearby organs at risk (OARs) [83]. Unlike conventional RT, SBRT avoids irradiating large volumes and does not include prophylactic irradiation of neighboring lymph node areas. With its shorter treatment duration (1 week versus 4–6 weeks), patients undergoing SBRT can promptly resume systemic therapy, minimizing interruptions to full-dose chemotherapy [4,84]. Furthermore, SBRT aims to enhance local control by delivering higher BED to tumors, addressing the significant proportion of PDAC patients who succumb to local progression alone [85]. Subsequently, there has been a surge in interest regarding the application of SBRT in localized PDAC patients. The incorporation of SBRT into a neoadjuvant strategy presents a seamless opportunity to deliver particularly high BED on tumor-vessel interfaces (TVIs), areas where tumors come into contact with blood vessels. By targeting precisely these regions, there is heightened potential to optimize oncological (R0) resection rates [4,86]. However, prospective data remain scarce [85].
SBRT techniques can be divided into non-ablative, nearly ablative, and ablative regimen.

Non-Ablative SBRT

Protective non-ablative SBRT schemes are mainly comprised between 25 and 35 Gy in five fractions, resulting in a maximal BED10 of 60 Gy, which falls far below the ablative doses that could be delivered with an SBRT technique [4]. If treatment safety is assured, the oncological efficacy of non-ablative SBRT is unsurprisingly lacking. The results of the main phase II trial and retrospective study are resumed in Table 2 [87,88,89,90,91,92,93,94]. This was illustrated with the results of the randomized phase II Alliance A021501 trial, comparing outcomes in BRPC patients who underwent induction FFX alone versus those who received TNT including FFX followed by non-ablative SBRT (33 Gy in five fractions with SIB up to 40 Gy at TVI or 25 Gy in five fractions) [95]. Initially, this trial encompassed three arms; however, following the findings of the LAP07 trial, the arm involving FFX followed by conventional CRT was terminated. The trial was halted after an interim analysis of 30 patients revealing a crossing of the futility boundary for R0 resection rates in the TNT arm (less than 11 patients among 30 underwent R0 resection). Regarding patients enrolled prior to the trial’s closure (70 patients in the FFX-alone arm and 56 in the TNT arm), the primary endpoint (18-month OS rate) was 67.9% versus 47.3%, favoring the FFX-alone arm [95]. However, the particularly unfavorable oncological results obtained in the TNT arm are to be somewhat mitigated as the data published results from underpowered arms with highly heterogenous RT schemes performed with up to 12.5% of palliative RT treatment (25 Gy in five fractions) administered in the TNT arm. In fact, the delivery of a BED10 > 60 Gy appears to be associated with improved OS and PFS, as indicated by multivariate analysis [4,96]. As such, hypofractionated SBRT techniques were modified in order to reach a (nearly) ablative dose to improve efficacity.

Nearly Ablative SBRT

In a recent observational study, Simoni et al. evaluated the use of SBRT (delivered in five consecutive daily fractions by administering 30 Gy to the planning target volume while simultaneously delivering a 50 Gy SIB to the TVI), after induction FFX or gemcitabine/nab-paclitaxel chemotherapy (median 12 cycles) in 59 patients (54.2% LAPC and 45.8% BRPC) [102]. Among the 58 patients who completed SBRT, 59.4% were resected. No ≥G3 AEs were reported. Comparing resected and unresected patients, the one- and two-year freedom from local progression rates were 85% and 80% versus 79.7% and 60.6%, respectively (p = 0.33). The median OS was 30.2 months, and the median PFS was 19 months, for the entire cohort. Notably, resected patients had superior 2-year OS rates (72.5% versus 49%, p = 0.012) and median PFS (13 months versus 5 months; p < 0.001) compared to unresected patients. Additionally, no survival differences were observed between BRPC and LAPC [102].
An interesting approach to delivering high-BED SBRT, without compromising the safety on non-MR-Linac systems, involves employing a full isotoxic dose prescription (IDP), which relies on OAR tolerance thresholds rather than tumor volume, as conventionally practiced. By safeguarding the OARs, the isotoxic high dose (iHD)-SBRT technique enables the attainment of the maximum feasible dose level, enhancing the probability of local tumor control while ensuring a safe achievement of BED10 ≥ 70 Gy [26,111]. Bouchart et al. presented feasibility and initial efficacy findings of a TNT sequence, comprising preoperative FFX for six cycles (or gemcitabine/nab-paclitaxel if intolerant or unresponsive) followed by iHD-SBRT (SIB up to 53 Gy at the TVI in five fractions) [26]. Out of 39 patients (21 BR and 18 LAPC), 87.2% completed the neoadjuvant sequence, with 55.9% undergoing oncological resection after iHD-SBRT. Grade 3 early and late gastrointestinal toxicities were minimal (<5%). With a median follow-up of 18.2 months, median OS and PFS were 24.5 and 15.6 months, respectively. Resected patients had significantly better OS and PFS than non-resected patients, and there were also no survival differences between BRPC and LAPC patients. The 1-year local control rate from SBRT was 74.1%, and loco-regional PFS was not reached for either group [26]. However, these studies included both BRPC and LAPC patients, and efforts should be made to study these groups separately. Following those promising results, the randomized comparative phase II trial STEREOPAC (NCT05083247) aims to assess the efficacy of adding iHD-SBRT (both nearly and ablative SBRT allowed) to preoperative FXX in the neoadjuvant treatment of BRPC patients only (Table 3) [112].

Ablative SBRT

The increasing accessibility of magnetic resonance linear accelerator (MR-linac) systems in prominent radiation therapy centers presents an elegant solution for delivering high-dose radiation therapy [4]. Stereotactic magnetic resonance (MR)-guided adaptive radiation therapy (SMART) allows the administration of ablative doses by providing real-time visualization and delineation of gastrointestinal structures, thereby adjusting for anatomic variations [106]. This technique is backed by dosimetric studies and a prospective phase I clinical trial, demonstrating that MR-guided adaptive SBRT increased the target volume dosing while protecting critical OARs [106,113,114].
In 2023, a prospective phase II trial, led by Parikh et al., included 136 patients with BRPC (56.6%) and LAPC (43.4%) to study the use of induction chemotherapy with mFFX (65.4%) or gemcitabine/nab-paclitaxel (16.9%) (mean of 5.1 months) followed by SMART (50Gy in five fractions, BED10 = 100 Gy) [108]. The study achieved its primary objective, which was to show that the incidence of acute G3 or higher gastrointestinal toxicity directly attributed to SMART would be less than 15.8%. The occurrence of acute ≥ G3 gastrointestinal toxicity, potentially or probably associated with SMART, was 8.8%. This included two postoperative deaths that were possibly linked to SMART in patients who underwent surgery. The resection rate was 32.4%. One-year overall survival was 56% in non-resected patients and 85% in resected patients [108]. Recently, more mature results showed a 2-year OS of 53.6% and a median OS of 22.7 months [115]. Results from studies evaluating ablative SBRT are summarized in Table 2 [106,107].

5. The Advent of TNT for LAPC Patients

LAPC patients are also a population target of choice for the use of TNT strategies in order to increase the chance of surgical and oncological resection, R0 resection rates, and survival outcomes. Similarly to BRPC patients, the previously described TNT approaches have also been studied in LAPC, as well as those including HFA-IMRT, and are described below.

5.1. TNT with Conventional CRT with Induction or Consolidation Chemotherapy Prior to Surgery

There are only two phase III RCTs on induction chemotherapy followed by CRT in LAPC patients [60,65].
Firstly, the LAP07 trial by the Groupe Coopérateur Multidisciplinaire en Oncologie (GERCOR) aimed to evaluate the benefit of CRT after 4 months of induction chemotherapy with gemcitabine with or without erlotinib [60]. CRT significantly reduced local progression rates (32% versus 46%, p = 0.030) and showed a trend towards better median PFS (9.9 versus 8.4 months, p = 0.060) and longer median time without retreatment (6.1 versus 3.7 months, p = 0.020). However, the primary endpoint was not achieved (median OS: 15.2 for CRT versus 16.5 months for chemotherapy alone, p = 0.830). Regarding RT quality assurance, 44% of CRT-treated patients experienced minor and 18% had major RT protocol deviations, with major deviations correlating with worse survival (median survival: 13.4 versus 17 months in case of minor deviations, p = 0.055) and with higher G ≥ 3 toxicities [116]. Since the LAP07 trial was designed in 2005, it used a gemcitabine regimen, as multi-agent chemotherapy was not yet introduced. Despite decreased, loco-regional progression, the TNT arm had a higher rate of metastatic progression (60% versus 44%, p = 0.040), indicating that CRT might confer additional survival benefits, with the use of chemotherapies offering better distant disease control [4]. It is worth mentioning that the resection rate in the LAP07 trial was notably low (7% after chemotherapy alone and 3% after CRT) due to the definitive nature of these treatments, with surgical exploration considered only in rare cases with significant response [60].
More recently, Fietkau et al. published the first results of the CONKO-007 phase III RCT comparing chemotherapy alone (6 months of gemcitabine or FFX) versus TNT with induction chemotherapy (3 months) followed by CRT (50.4 Gy in 28 fractions) with concomitant gemcitabine in LAPC patients [65]. The primary endpoint was initially OS since the beginning of induction chemotherapy, but due to the delayed accrual of patients, it was changed to R0 resection rate resulting in a reduced sample size of 525 patients. After induction chemotherapy, 22% were excluded due to progression or toxicity leading to the randomization of 335 patients with 121 of them resected. Resection rates for randomized patients were similar between two arms (35.9% and 36.3%). This TNT approach resulted in an improvement in negative circumferential resection margin (CRM) R0 resection and pCR rates (0 versus 6%, p = 0.0013), although it did not significantly differ for the global R0 resection rate (18 versus 25% for TNT, p = 0.1433). R1 resections occurred significantly more in the chemotherapy-alone arm (p = 0.0085). Nevertheless, with a median follow-up of 16 months, this effect on resectability did not translate into significant PFS and OS differences (HR for median PFS of 0.919 [95% CI 0.702–1.203], p = 0.540, and HR for median OS of 0.964 [95% CI 0.760–1.225], p = 0.766), whereas the PFS rate tended to be higher in the CRT arm after 2 years (17.5 months +/− 0.04% vs. 24.1 months +/− 0.04%) [65].
Three additional phase II trials aimed to explore the same question with globally disappointing results and are summarized in Table 1 [61,63,64]. Furthermore, two meta-analyses examining randomized trials failed to reveal a statistically significant discrepancy in OS between patients undergoing neoadjuvant chemotherapy alone versus TNT with CRT, except in a subgroup analysis involving consolidation CRT after a minimum of 3 months of induction CT [117,118].
An alternative strategy of TNT with CRT consists in a regimen of induction CRT followed by consolidation or maintenance chemotherapy. A phase III RCT aimed to compare gemcitabine alone with induction CRT (60 Gy in 30 fractions) with concomitant 5-FU/cisplatin followed by maintenance chemotherapy with gemcitabine in LAPC patients [66]. Second intention surgery was authorized if patients had a good response to treatment. As such, five patients (4.2%) underwent surgery (two in the CRT arm and three in the gemcitabine-alone arm). Overall, median OS was 13 months in the gemcitabine-alone arm and 11.1 months in the TNT strategy [66].
Results of selected and representative studies can be found in Table 1 [62,80].

5.2. TNT with Hyprofractionnated Ablative Intensity-Modulated Radiotherapy (HFA-IMRT)

Using hypofractionated ablative radiotherapy (HFA-RT) after induction chemotherapy presents an interesting dose-escalation option in order to elevate the BED10 delivered to the tumor while still using a technique close to the well-known conventional CRT. Krishnan et al. investigated 200 LAPC patients who underwent induction chemotherapy followed by either conventional CRT (50.4 Gy in 28 fractions with concomitant chemotherapy) or HFA-IMRT (using an SIB technique in 15 to 28 fractions to obtain a BED10 > 70 Gy, with concurrent chemotherapy) [70]. Only 47 patients with tumors more than 1 cm from the nearest gastrointestinal mucosa could receive HFA-IMRT. The study found that HFA-IMRT resulted in better OS (median: 17.8 vs. 15 months, p = 0.030; 3-year OS: 31% vs. 9%) and local control (median local–regional recurrence-free survival: 10.2 vs. 6.2 months, p = 0.050). No increased toxicity was noted, and high BED delivery was the only predictor of improved OS in multivariate analysis [70]. A prospective study by the Sloan Kettering group examined 136 LAPC patients who received definitive HFA-IMRT (delivering BED ≥ 100 Gy; either 75 Gy in 25 fractions or 67.5 Gy in 15 fractions). After a median follow-up of 12 months, the median OS and freedom from local progression (FFLP) were not reached. Remarkably, the 2-year OS and FFLP rates were 55% and 78%, respectively, with a favorable toxicity profile [71]. Following the encouraging results from a prospective cohort study evaluating the association of HFA-RT in LAPC patients, Reyngold et al. presented in 2023 the results from the phase II MAIBE trial studying 3–6 months (median 3.5 months) of modified FFX or gemcitabine/nab-paclitaxel followed by hypofractionated HFA-IMRT (67.5 Gy in 15 fractions or 75 Gy in 25 fractions with concurrent capecitabine) in 47 LAPC [68,72]. The resection rate was 26% with 58.3% of R0 resection. Two-year OS was 38.9% (95% CI, 21.9–55.6%), including 37.1% (18.5–55.8%) in non-surgical and 39.4% (7.0–72.1%) in surgical groups. The 90 days post-surgery mortality was 0%, and nine surgical AEs were recorded in six patients [68]. However, the phase II RCT SCALOP from the Oxford group, aiming for 2 × 2 randomization between an induction with gemcitabine/nab-paclitaxel +/− nelfinavir followed by conventional CRT or HFA-IMRT (60 Gy in 30 fractions with concomitant capecitabine) did not show a significant survival benefit in favor of the TNT arm (median OS: 15.6 versus 16.9 months for TNT, p = NS) despite a good tolerance of the dose-escalated CRT [69]. Results from studies evaluating HFA-IMRT can be found in Table 1.

5.3. TNT with SBRT

The only analysis pooling findings from 19 SBRT trials involving LAPC patients revealed a median OS of 17 months (ranging from 5.7 to 47 months), alongside a 1-year OS rate of 51.6% and a 1-year local control rate (LCR) of 72.3% [119]. Unfortunately, considerable heterogeneity existed among these studies, the majority of which were small-scale retrospective series [88,91,94,100,101]. Moreover, the dosage and fractionation regimens varied significantly, leaving the establishment of an optimal TNT protocol with fractionated SBRT ambiguous [4]. Results from studies evaluating SBRT in LAPC patients are summarized in Table 2.

5.3.1. Non-Ablative SBRT

In a meta-analysis conducted by Tchelebi et al., focusing on LAPC, SBRT (doses of ≥5 Gy per fraction) demonstrated a modest advantage in 2-year OS compared to conventional CRT (26.9 months versus 13.7 months, p = 0.004). However, differences in OS were not statistically significant at 1 year (53.7% versus 49.3%, p = 0.630) [120].
In 2015, a phase II trial evaluated a TNT approach consisting of three doses of gemcitabine followed by SBRT (33Gy in five fractions) in 49 LAPC patients [99]. Quality of life was assessed through the QLQ-PAN26 module before and at 4 weeks and 4 months after SBRT, revealing a notable improvement in pancreatic pain (p = 0.001) four weeks post-SBRT. The incidence rates of acute and late toxicities (primary endpoint), graded as ≥2, including gastritis, fistula, enteritis, or ulcer, were 2% and 11%, respectively. With a median follow-up of 13.9 months, median OS was 13.9 months, and freedom from local disease progression at one year was 78%. Four patients (8%) underwent margin-negative and lymph node-negative surgical resections [99]. Results from studies evaluating non-ablative SBRT in LAPC patients are summarized in Table 2 [100,101]. In light of these disappointing results, the delivery of higher BED10 to the tumor should be sought for LAPC, similarly to BRPC.

5.3.2. Nearly Ablative SBRT

In a large retrospective study using a large national database of cT2-4N0-1M0 pancreatic cancer patients, 631 LAPC patients received SBRT (40 Gy/5 fractions) after induction chemotherapy, with a resection rate of 9.2% (84% R0), similar to the rates seen in the 7819 patients treated with CRT (50.4 Gy/28 fractions) [105]. SBRT, however, was associated with a superior OS in the multivariate analysis (HR 0.84, 95% CI 0.75–0.93, p < 0.001) [105].
Those encouraging results were followed by two phase II trials studying SBRT following chemotherapy in LAPC patients [103,104]. Comito et al. analyzed the use of SBRT (45 Gy in six fractions) preceded by a gemcitabine-based chemotherapy in 71% of the 45 LAPC patients [104]. Median PFS and median OS were 8 months and 13 months, respectively [104]. In the LAPC-1 phase II trial, 50 LAPC patients were treated with eight cycles of induction FOLFIRINOX, followed by SBRT (40 Gy in five fractions) [103]. During FOLFIRINOX treatment, 30 G3 or 4 AEs were observed, and following SBRT, two (5%) G3 or four AEs were noted. Additionally, two (5%) patients experienced gastrointestinal bleeding-related deaths within three months post-SBRT. Thirty-nine (78%) patients received the SBRT treatment. The resection rate was 12% with a 100% R0 resection rate and 33% pCR. The 1-year OS and PFS were 64% (95% CI: 50–76%) and 34% (95% CI: 22–48%), respectively. Median OS was 17 months in patients receiving SBRT and 7 months in patients who did not receive SBRT [103]. Studies using nearly ablative SBRT after induction chemotherapy are summarized in Table 2.
Results from the Belgian ongoing phase II TORPEDO trial (B3002023000168) evaluating the role of SBRT (40 Gy in five fractions) in BRPC and LAPC patients are highly awaited (Table 3).

5.3.3. Ablative Adaptive SBRT

A multi-institutional retrospective study suggested that dose-escalated SMART might offer a survival benefit for patients with pancreatic cancer compared to more conventional MR-guided radiation treatments. In this study by Rudra et al., 44 patients with BRPC (23%) and LAPC (73%) received different radiation therapy regimens, including high-dose SBRT. The high-dose SBRT group was divided into two subgroups based on the BED: BED ≤ 70 Gy (40–55 Gy in 25–28 fractions or 30–35 Gy in five fractions) and BED > 70 Gy group (50–67.5 Gy in 10–15 fractions or 40–52 Gy in five fractions). The 2-year OS rate was 49% in the high-dose group versus 30% in the low-dose group (p = 0.030). However, it is noteworthy that only six patients had surgery post-RT, and high-dose RT was not an independent predictor of OS in multivariate analysis [109]. Additional results from other key retrospective studies using this technique are summarized in Table 2 [106,107,110].
Although the US LAP-ABLATE trial evaluating MRI-guided ablative SBRT in LAPC patients will not open, the results from LAPSTAR (NCT06272162) are highly anticipated (Table 3).

6. Evaluation of Treatment Response and Future Perspectives for Patient Selection in the Precision Medicine Era

6.1. Available Tools for the Evaluation of Treatment Response

During TNT, a proper evaluation of treatment response, usually performed through clinical, biochemical, radiologic, and/or metabolic features, is of outmost importance, considering the length of the treatment period and the possibility of therapy switching [42].
Carbohydrate antigen (CA) 19-9 stands as the only biomarker endorsed by the US FDA for monitoring therapy response in PDAC, playing a crucial role in clinical staging and treatment strategies for both localized and metastatic scenarios. Nevertheless, its utility is constrained to patients with a sialyl-Lewis A-positive genotype, encompassing roughly 90% of patients. Moreover, accurate assessment of CA 19-9 readings necessitates normal bilirubin levels, given that elevated CA 19-9 levels can also signify underlying inflammatory conditions [121]. For instance, in a retrospective series involving 131 patients initially presenting elevated CA19-9 (>35 U/dl) who underwent preoperative therapy and subsequent resection, the normalization of CA19-9 emerged as a robust prognostic marker for long-term survival rather than the magnitude of its reduction [122]. Another small study underscored the unlikelihood of achieving a major pathologic response in patients with persistently elevated CA19-9 after preoperative therapy. Notably, among 28 patients demonstrating a major pathologic response, 27 (96%) showed post-treatment CA19-9 within the normal range, despite 75% of them having elevated CA19-9 levels before treatment [123]. Furthermore, a recent systematic review encompassing 17 pertinent studies with comprehensive data on CA19-9 response during neoadjuvant therapy emphasized that achieving a post-neoadjuvant CA19-9 response exceeding 50% or achieving normalization was associated with improved overall survival [124].
While a consensus on resectability criteria at diagnosis remains elusive, the same challenge persists post-neoadjuvant treatment, with a specific hurdle emerging after TNT due to the fibrotic alterations induced by radiotherapy.
Multi-phase contrast-enhanced computed tomography (CT) currently stands as the most validated tool for staging PDAC. Adding a layer of intricacy, response to neoadjuvant therapy might not always be reflected by radiographic indicators [125]. In the neoadjuvant setting, preoperative CT re-staging may lack specificity in distinguishing residual viable tumors from post-treatment-induced changes (with no viable tumor) particularly near involved vessels, due to the dense fibrous stroma from pancreatic tumors. In addition, after chemoradiotherapy, cancer cells decrease or disappear leaving behind fibrotic tissue that cannot be distinguished from tumor tissue. Some studies suggest that preoperative therapy could lead to radiographic downstaging in unresectable PDAC, potentially improving resectability, akin to its effects like breast and rectal cancers. Yet, these findings are hindered by inconsistencies in therapeutic response metrics, resection criteria, surgical techniques, and staging definitions [125,126]. Katz et al. retrospectively studied 129 BR PDAC patients to assess how NAT affected tumor size reduction or stage of BR tumors, based on standardized criteria for the indication of resection (AHPBA and MD Anderson) and clinical and pathological response criteria (e.g., using RECIST criteria) [21,127,128]. Their findings showed minimal clinical downstaging according to AHPBA criteria (1%), with 19% showing disease progression, and 80% demonstrating no change. Results were even less favorable with MD Anderson criteria, with 1% showing downstaging, 21% progressing, and 78% remaining unchanged. However, intraoperative findings revealed a more positive chemotherapy response, as 66% of patients underwent resection, with 95% achieving R0 resection [125]. Histopathologic analysis of pancreatectomy specimens further revealed favorable grade III or IV responses in 17% of resected patients, suggesting a promising prognosis despite persistent anatomical relationships in CT scans [125,129]. Interestingly, while radiographic changes in primary tumor anatomy were rare, histopathological analysis indicated significant cytotoxic activity at the tumor–vessel interface, with 74% of patients showing >1 mm distance from the primary tumor to the inked superior mesenteric artery (SMA) margin in cases with negative SMA margins [125]. Notably, RECIST response did not correlate with OS [125]. Perri et al. examined responses to first-line chemotherapy (FOLFIRINOX or gemcitabine/nab-paclitaxel) in 485 treatment-naive patients with localized pancreatic cancer, finding higher rates of RECIST partial responses with FOLFIRINOX compared to gemcitabine/nab-paclitaxel (19% vs. 6%; p = 0.001) but no significant differences in median tumor volume change, local tumor downstaging rates, or CA19-9 levels (35). Similarly, in a recent study of 194 patients undergoing TNT, anatomical downstaging was uncommon (28%) and not linked to survival, but 94% achieved negative margins with R0 resection, while radiologic anatomical downstaging was uncommon (28%) and not correlated with survival, and 94% of patients achieved negative margins (R0 resection) [7]. Only three factors were independently associated with prolonged survival, including extended duration (≥6 cycles) chemotherapy, optimal post-chemotherapy CA19-9 response, and major pathologic response. Moreover, among 67 patients who underwent interval metabolic positron emission tomography (PET) imaging post-chemotherapy, complete metabolic response strongly correlated with major pathologic response [7].

6.2. Future Perspectives for Response Status and Patient Selection

Alternative imaging techniques, such as magnetic resonance (MR) imaging with specific sequences, show promising potential and are currently being studied in conjunction with metabolic imaging and conventional CT [130]. Although 18F-FDG PET is not standard in clinical practice, it was proven to be sensitive for initial TNM staging, treatment response, and recurrence detection [131,132,133]. Furthermore, 18F-FDG PET/CT imaging may predict treatment efficacy and clinical outcome for PDAC [134]. Some centers currently rely on post-chemotherapy metabolic responses, evaluated through PET/CT or PET/MRI as surrogate markers for treatment response assessment [135,136]. Reductions in SUVmax, SUVpeak, and MTV have been associated with improved overall survival [7,135,137]. PET/CT or PET/MRI-based metabolic response may offer greater sensitivity in assessing tumor response compared to traditional CT or MRI and warrants further investigation in clinical trials [137]. However, 18F-FDG PET/CT has limitations in PDAC due to variable detection of metastatic lymph nodes and potential false-positive results in cases of inflammation [138,139]. Additionally, promising results have emerged from the use of radiomics-based machine-learning models in diagnosing pancreatic cancer, demonstrating their potential to predict diagnoses earlier than human observers [140]. Radiomics might assist in patient selection for TNT by analyzing quantitative imaging features to predict individual responses to treatment. By assessing tumor characteristics, such as texture, heterogeneity, and vascularity, radiomics can identify patients likely to benefit from TNT, guiding personalized treatment decisions. Additionally, radiomics can help monitor treatment response over time, allowing for early adaptation of therapy if necessary. Ultimately, by providing insights into tumor biology and treatment response, radiomics enables more tailored and effective TNT strategies for pancreatic cancer patients.
Compared to other solid tumors, the tumor microenvironment (TME) of PDAC is uniquely complex and dynamic due to intensive interactions among its components. PDAC is characterized by intense stromal desmoplastic reactions surrounding cancer cells, creating an acidic and hypoxic microenvironment, with high interstitial pressure. This environment acts as a sanctuary, limiting drug penetration and impeding immune cells recruitment [141,142,143]. Cancer-associated fibroblasts (CAFs) are key players in the desmoplastic reaction, comprising up to 90% of the tumor tissue in carcinomas. Fibroblast activation protein (FAP), a type-II transmembrane serine protease, is prominently expressed in CAFs [144]. Given its widespread expression, FAP is an attractive target for radionuclide imaging with PET/CT [141,143,145,146]. FAP-targeted imaging has shown promising diagnostic sensitivity and specificity, with a low tracer uptake in non-tumoral tissues across various cancers, including PDAC [147]. Consequently, pancreatic cancer is expected to show intensive uptake of 68Ga-FAPI, encouraged by previous studies [148,149]. Biologically, 68Ga-FAPI-PET in PDAC is of special interest as it depicts tumor–stroma interaction, crucial for the tumorigenesis of PDAC, aspects not captured by morphologic or metabolic imaging modalities. Recently, Koerber et al. underscored the clinical impact of 68Ga-FAPI PET/CT imaging in 19 patients with PDAC (seven primary and 12 progressive/recurrent), highlighting its ability to detect new lesions or clarify ambiguous findings from standard CT scans. The group from Heidelberg found that biodistribution analyses revealed high FAPI uptake in primary PDAC, lymph nodes, and distant metastases, while healthy tissues exhibited minimal background activity, resulting in excellent tumor-to-background ratios [147,149,150]. The Heidelberg group also noted discrepancies between 18F-FDG PET and 68Ga-FAPI PET-based staging, often leading to upstaging with the latter, consistent with findings in other tumor types [150]. As such, 68Ga-FAPI PET may lead to significant changes in treatment decisions for PDAC when compared with 18F-FDG PET and is a key innovant player in improving PDAC management. A study addressing this specific question in managing PDAC patients treated with TNT is currently ongoing at our center.
Although some patients achieve pCR, more than half relapse after surgery [151].
Liquid biopsies enable the collection of repeated samples during neoadjuvant therapy and can help to monitor treatment response and to stratify patients to the most suitable treatment [31,152]. Notably, Yin et al. discovered the persistence of somatic mutations, circulating tumor cells (CTCs), and circulating tumor DNA (ctDNA) in individuals with pancreatic cancer who achieved pCR following neoadjuvant therapy. CTCs were found in five of six patients and ctDNA in 7 of 16 patients with pCR. This led to the introduction of a novel concept of molecular complete response (mCR), by amalgamating genomic analysis of resected specimens with liquid biopsy data [153]. In a study of 59 patients, preoperative ctDNA was detected in 69% of those directly undergoing surgery compared to 21% in patients receiving neoadjuvant treatment. The presence of preoperative ctDNA correlated with significantly lower recurrence-free survival (RFS) and overall survival (OS). Notably, patients with persistent ctDNA after neoadjuvant treatment all experienced relapse, with a median RFS of 5 months [154]. Monitoring KRAS mutant allele frequency (MAF) in exoDNA during NAT may also help with predicting the resectability of PDAC. Indeed, a study revealed that 71% of patients eligible for PDAC resection exhibited a decreased KRAS MAF, whereas in 94% of patients with unresectable PDAC post-NAT, KRAS MAF remained stable or increased [155]. Furthermore, Bernard et al. discovered that an increase in exoDNA after NAT was significantly associated with disease progression, suggesting that monitoring exoDNA during NAT using liquid biopsy provides predictive insight and prognostic information relevant to therapeutic stratification [156].
While personalized medicine in pancreatic cancer has made strides, it encounters persistent barriers hindering effective clinical application [30]. Presently, patients with BR and LAPC adhere to a standardized treatment strategy drawn from RCTs on neoadjuvant therapy, maintaining a one-size-fits-all paradigm [157,158,159,160]. However, substantial variability exists in survival and response to neoadjuvant therapy with FOLFIRINOX and gemcitabine/nab-paclitaxel. Despite FOLFIRINOX demonstrating higher RECIST partial response rates and subsequent pancreatectomy, overall survival remains comparable between regimens [161,162]. The absence of biomarkers for chemotherapy selection necessitates a transition toward more effective and tolerable regimens, guided by enhanced patient stratification. A notable breakthrough involves a patient-derived organoid library revealing gene expression signatures predictive of improved chemotherapy response [163]. This innovative approach holds promise for identifying genomic, transcriptomic, and therapeutic profiles, paving the way for tailored treatments that swiftly confer clinical benefits to each patient.
Pancreatic cancer patients may benefit from tumor molecular profiling and targeted therapies [164]. Transcriptomic signatures, such as GemPred, have been studied as predictive tools of response to adjuvant chemotherapy in PDAC [165]. Likewise, PAMG has been associated to progression under mFFX treatment (p < 0.001) [166]. Similarly, other gene expression signatures used to predict response to neoadjuvant CRT in PDAC patients have been investigated [167]. Predictive molecular signatures are required to forecast the response to TNT, enabling the molecular selection of patients. A retrospective analysis of the Know Your Tumor registry trial revealed that patients with actionable molecular alterations had improved outcomes with matched therapies [168]. However, only a small percentage (about 8%) of pancreatic cancer patients harbor potentially targetable genetic alterations [168]. Current treatment recommendations mainly apply to metastatic cases, but exploring these findings in earlier stages is warranted in future trials. ESMO suggests multi-gene sequencing for advanced pancreatic cancer patients in molecular screening programs, enabling access to an innovative drug [169]. Currently, there is limited evidence for specific neoadjuvant regimens other than FOLFIRINOX and gemcitabine/nab-paclitaxel. In the future, the utilization of molecular and mutational signatures could guide treatment decisions, influencing the selection between gemcitabine-based regimens, FOLFIRINOX, or radiotherapy.

7. Conclusions

The advent of TNT presents a promising avenue for both BRPC and LAPC patients, aiming to optimize outcomes through tailored radiotherapy and chemotherapy regimens. The Dutch PREOPANC regimen has shown superior R0 resection rates, highlighting the efficacy of integrating preoperative gemcitabine with hypofractionated CRT. Ongoing research, including the exploration of SBRT, offers potential for precise tumor targeting and improved survival rates. Ablative SBRT techniques, especially when combined with induction chemotherapy, demonstrate promise in enhancing overall survival and facilitating oncological resection. These innovative approaches underscore the evolving landscape of neoadjuvant therapy, offering hope for improved outcomes in pancreatic cancer management.
Current perioperative trials prioritize the assessment of optimal treatment regimens and sequences, often employing gemcitabine-abraxane or FOLFIRINOX for a minimum duration of 4 months with possibilities of switching and with a growing focus on the integration of SBRT, which offers the advantage of a very short course administration. Refining therapeutic protocols, especially by extending the duration of chemotherapy prior to surgery and incorporating refined SBRT techniques, is expected to lead to substantial advancements in pancreatic cancer management. The TNT strategy holds great potential, and conok the most favorable therapeutic outcomes.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Siegel, R.L.; Miller, K.D.; Goding Sauer, A.; Fedewa, S.A.; Butterly, L.F.; Anderson, J.C.; Cercek, A.; Smith, R.A.; Jemal, A. Colorectal Cancer Statistics, 2020. CA Cancer J. Clin. 2020, 70, 145–164. [Google Scholar] [CrossRef] [PubMed]
  2. Kleeff, J.; Korc, M.; Apte, M.; La Vecchia, C.; Johnson, C.D.; Biankin, A.V.; Neale, R.E.; Tempero, M.; Tuveson, D.A.; Hruban, R.H.; et al. Pancreatic Cancer. Nat. Rev. Dis. Primers 2016, 2, 16022. [Google Scholar] [CrossRef] [PubMed]
  3. Terlizzi, M.; Buscail, E.; Boussari, O.; Adgié, S.; Leduc, N.; Terrebonne, E.; Smith, D.; Blanc, J.F.; Lapuyade, B.; Laurent, C.; et al. Neoadjuvant Treatment for Borderline Resectable Pancreatic Adenocarcinoma Is Associated with Higher R0 Rate Compared to Upfront Surgery. Acta Oncol. 2021, 60, 1114–1121. [Google Scholar] [CrossRef] [PubMed]
  4. Bouchart, C.; Navez, J.; Closset, J.; Hendlisz, A.; Van Gestel, D.; Moretti, L.; Van Laethem, J.L. Novel Strategies Using Modern Radiotherapy to Improve Pancreatic Cancer Outcomes: Toward a New Standard? Ther. Adv. Med. Oncol. 2020, 12, 1758835920936093. [Google Scholar] [CrossRef]
  5. Murphy, J.E.; Wo, J.Y.; Ryan, D.P.; Clark, J.W.; Jiang, W.; Yeap, B.Y.; Drapek, L.C.; Ly, L.; Baglini, C.V.; Blaszkowsky, L.S.; et al. Total Neoadjuvant Therapy with FOLFIRINOX in Combination with Losartan Followed by Chemoradiotherapy for Locally Advanced Pancreatic Cancer: A Phase 2 Clinical Trial. JAMA Oncol. 2019, 5, 1020–1027. [Google Scholar] [CrossRef] [PubMed]
  6. Pietrasz, D.; Turrini, O.; Vendrely, V.; Simon, J.M.; Hentic, O.; Coriat, R.; Portales, F.; Le Roy, B.; Taieb, J.; Regenet, N.; 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. [Google Scholar] [CrossRef]
  7. Truty, M.J.; Kendrick, M.L.; Nagorney, D.M.; Smoot, R.L.; Cleary, S.P.; Graham, R.P.; Goenka, A.H.; Hallemeier, C.L.; Haddock, M.G.; Harmsen, W.S.; et al. Factors Predicting Response, Perioperative Outcomes, and Survival Following Total Neoadjuvant Therapy for Borderline/Locally Advanced Pancreatic Cancer. Ann. Surg. 2021, 273, 341–349. [Google Scholar] [CrossRef]
  8. Kim, R.Y.; Christians, K.K.; Aldakkak, M.; Clarke, C.N.; George, B.; Kamgar, M.; Khan, A.H.; Kulkarni, N.; Hall, W.A.; Erickson, B.A.; et al. Total Neoadjuvant Therapy for Operable Pancreatic Cancer. Ann. Surg. Oncol. 2021, 28, 2246–2256. [Google Scholar] [CrossRef] [PubMed]
  9. Rhim, A.D.; Mirek, E.T.; Aiello, N.M.; Maitra, A.; Bailey, J.M.; McAllister, F.; Reichert, M.; Beatty, G.L.; Rustgi, A.K.; Vonderheide, R.H.; et al. EMT and Dissemination Precede Pancreatic Tumor Formation. Cell 2012, 148, 349–361. [Google Scholar] [CrossRef]
  10. Deng, Y.; Chi, P.; Lan, P.; Wang, L.; Chen, W.; Cui, L.; Chen, D.; Cao, J.; Wei, H.; Peng, X.; et al. Modified FOLFOX6 with or Without Radiation Versus Fluorouracil and Leucovorin with Radiation in Neoadjuvant Treatment of Locally Advanced Rectal Cancer: Initial Results of the Chinese FOWARC Multicenter, Open-Label, Randomized Three-Arm Phase III Trial. J. Clin. Oncol. 2016, 34, 3300–3307. [Google Scholar] [CrossRef]
  11. Bahadoer, R.R.; Dijkstra, E.A.; van Etten, B.; Marijnen, C.A.M.; Putter, H.; Kranenbarg, E.M.-K.; Roodvoets, A.G.H.; Nagtegaal, I.D.; Beets-Tan, R.G.H.; Blomqvist, L.K.; et al. Short-Course Radiotherapy Followed by Chemotherapy before Total Mesorectal Excision (TME) versus Preoperative Chemoradiotherapy, TME, and Optional Adjuvant Chemotherapy in Locally Advanced Rectal Cancer (RAPIDO): A Randomised, Open-Label, Phase 3 Trial. Lancet Oncol. 2021, 22, 29–42. [Google Scholar] [CrossRef]
  12. Conroy, T.; Etienne, P.-L.; Rio, E.; Evesque, L.; Mesgouez-Nebout, N.; Vendrely, V.; Artignan, X.; Bouche, O.; Boileve, A.; Delaye, M.; et al. Total Neoadjuvant Therapy with MFOLFIRINOX versus Preoperative Chemoradiation in Patients with Locally Advanced Rectal Cancer: 7-Year Results of PRODIGE 23 Phase III Trial, a UNICANCER GI Trial. J. Clin. Oncol. 2023, 41, LBA3504. [Google Scholar] [CrossRef]
  13. Katz, M.H.G.; Shi, Q.; Ahmad, S.A.; Herman, J.M.; Marsh, R.D.W.; Collisson, E.; Schwartz, L.; Frankel, W.; Martin, R.; Conway, W.; 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. [Google Scholar] [CrossRef]
  14. Wittmann, D.; Hall, W.A.; Christians, K.K.; Barnes, C.A.; Jariwalla, N.R.; Aldakkak, M.; Clarke, C.N.; George, B.; Ritch, P.S.; Riese, M.; et al. Impact of Neoadjuvant Chemoradiation on Pathologic Response in Patients with Localized Pancreatic Cancer. Front. Oncol. 2020, 10, 460. [Google Scholar] [CrossRef]
  15. Pugalenthi, A.; Protic, M.; Gonen, M.; Kingham, T.P.; Angelica, M.I.D.; Dematteo, R.P.; Fong, Y.; Jarnagin, W.R.; Allen, P.J. Postoperative Complications and Overall Survival after Pancreaticoduodenectomy for Pancreatic Ductal Adenocarcinoma. J. Surg. Oncol. 2016, 113, 188–193. [Google Scholar] [CrossRef]
  16. Wu, W.; He, J.; Cameron, J.L.; Makary, M.; Soares, K.; Ahuja, N.; Rezaee, N.; Herman, J.; Zheng, L.; Laheru, D.; et al. The Impact of Postoperative Complications on the Administration of Adjuvant Therapy Following Pancreaticoduodenectomy for Adenocarcinoma. Ann. Surg. Oncol. 2014, 21, 2873–2881. [Google Scholar] [CrossRef]
  17. Wei, A.C.; Ou, F.S.; Shi, Q.; Carrero, X.; O’Reilly, E.M.; Meyerhardt, J.; Wolff, R.A.; Kindler, H.L.; Evans, D.B.; Deshpande, V.; et al. Perioperative Gemcitabine + Erlotinib Plus Pancreaticoduodenectomy for Resectable Pancreatic Adenocarcinoma: ACOSOG Z5041 (Alliance) Phase II Trial. Ann. Surg. Oncol. 2019, 26, 4489–4497. [Google Scholar] [CrossRef]
  18. Mackay, T.M.; Smits, F.J.; Roos, D.; Bonsing, B.A.; Bosscha, K.; Busch, O.R.; Creemers, G.J.; van Dam, R.M.; van Eijck, C.H.J.; Gerhards, M.F.; et al. The Risk of Not Receiving Adjuvant Chemotherapy after Resection of Pancreatic Ductal Adenocarcinoma: A Nationwide Analysis. HPB 2020, 22, 233–240. [Google Scholar] [CrossRef]
  19. Lubrano, J.; Bachelier, P.; Paye, F.; Le Treut, Y.P.; Chiche, L.; Sa-Cunha, A.; Turrini, O.; Menahem, B.; Launoy, G.; Delpero, J.R. Severe Postoperative Complications Decrease Overall and Disease Free Survival in Pancreatic Ductal Adenocarcinoma after Pancreaticoduodenectomy. Eur. J. Surg. Oncol. 2018, 44, 1078–1082. [Google Scholar] [CrossRef]
  20. Tempero, M.A.; Malafa, M.P.; Al-Hawary, M.; Asbun, H.; Bain, A.; Behrman, S.W.; Al Benson, B.; Binder, E.; Cardin, D.B.; Cha, C.; et al. Pancreatic Adenocarcinoma, Version 2.2017, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Cancer Netw. 2017, 15, 1028–1061. [Google Scholar] [CrossRef]
  21. Vauthey, J.N.; Dixon, E. AHPBA/SSO/SSAT Consensus Conference on Resectable and Borderline Resectable Pancreatic Cancer: Rationale and Overview of the Conference. Ann. Surg. Oncol. 2009, 16, 1725–1726. [Google Scholar] [CrossRef] [PubMed]
  22. Shin, D.W.; Kim, J. The American Joint Committee on Cancer 8th Edition Staging System for the Pancreatic Ductal Adenocarcinoma: Is It Better than the 7th Edition? Hepatobiliary Surg. Nutr. 2020, 9, 98. [Google Scholar] [CrossRef] [PubMed]
  23. Isaji, S.; Mizuno, S.; Windsor, J.A.; Bassi, C.; Fernández-del Castillo, C.; Hackert, T.; Hayasaki, A.; Katz, M.H.G.; Kim, S.W.; Kishiwada, M.; et al. International Consensus on Definition and Criteria of Borderline Resectable Pancreatic Ductal Adenocarcinoma 2017. Pancreatology 2018, 18, 2–11. [Google Scholar] [CrossRef] [PubMed]
  24. Tomasello, G.; Ghidini, M.; Ghidini, A.; Trevisan, F.; Celotti, A.; Russo, A.; Gambini, D.; Indini, A.; Rijavec, E.; Bareggi, C.; et al. Total Neoadjuvant Therapy for Initially Inoperable Pancreatic Cancer: A Systematic Review of Phase 2–3 Studies. Radiother. Oncol. 2021, 164, 13–19. [Google Scholar] [CrossRef] [PubMed]
  25. De Simoni, O.; Scarpa, M.; Soldà, C.; Bergamo, F.; Lonardi, S.; Fantin, A.; Pilati, P.; Gruppo, M. Could Total Neoadjuvant Therapy Followed by Surgical Resection Be the New Standard of Care in Pancreatic Cancer? A Systematic Review and Meta-Analysis. J. Clin. Med. 2022, 11, 812. [Google Scholar] [CrossRef] [PubMed]
  26. Bouchart, C.; Engelholm, J.L.; Closset, J.; Navez, J.; Loi, P.; Gökburun, Y.; De Grez, T.; Mans, L.; Hendlisz, A.; Bali, M.A.; et al. Isotoxic High-Dose Stereotactic Body Radiotherapy Integrated in a Total Multimodal Neoadjuvant Strategy for the Treatment of Localized Pancreatic Ductal Adenocarcinoma. Ther. Adv. Med. Oncol. 2021, 13, 17588359211045860. [Google Scholar] [CrossRef] [PubMed]
  27. Reni, M.; Balzano, G.; Zanon, S.; Zerbi, A.; Rimassa, L.; Castoldi, R.; Pinelli, D.; Mosconi, S.; Doglioni, C.; Chiaravalli, M.; et al. Safety and Efficacy of Preoperative or Postoperative Chemotherapy for Resectable Pancreatic Adenocarcinoma (PACT-15): A Randomised, Open-Label, Phase 2-3 Trial. Lancet Gastroenterol. Hepatol. 2018, 3, 413–423. [Google Scholar] [CrossRef] [PubMed]
  28. Tzeng, C.W.D.; Fleming, J.B.; Lee, J.E.; Xiao, L.; Pisters, P.W.T.; Vauthey, J.N.; Abdalla, E.K.; Wolff, R.A.; Varadhachary, G.R.; Fogelman, D.R.; et al. Defined Clinical Classifications Are Associated with Outcome of Patients with Anatomically Resectable Pancreatic Adenocarcinoma Treated with Neoadjuvant Therapy. Ann. Surg. Oncol. 2012, 19, 2045–2053. [Google Scholar] [CrossRef] [PubMed]
  29. Labori, K.J.; Katz, M.H.; Tzeng, C.W.; Bjørnbeth, B.A.; Cvancarova, M.; Edwin, B.; Kure, E.H.; Eide, T.J.; Dueland, S.; Buanes, T.; et al. Impact of Early Disease Progression and Surgical Complications on Adjuvant Chemotherapy Completion Rates and Survival in Patients Undergoing the Surgery First Approach for Resectable Pancreatic Ductal Adenocarcinoma—A Population-Based Cohort Study. Acta Oncol. 2016, 55, 265–277. [Google Scholar] [CrossRef]
  30. Lambert, A.; Schwarz, L.; Borbath, I.; Henry, A.; Van Laethem, J.L.; Malka, D.; Ducreux, M.; Conroy, T. An Update on Treatment Options for Pancreatic Adenocarcinoma. Ther. Adv. Med. Oncol. 2019, 11, 1758835919875568. [Google Scholar] [CrossRef]
  31. Labori, K.J. Short-Course or Total Neoadjuvant Chemotherapy in Resectable and Borderline Resectable Pancreatic Cancer—Current Status and Future Perspectives. Front. Surg. 2022, 9, 839339. [Google Scholar] [CrossRef] [PubMed]
  32. Swords, D.S.; Zhang, C.; Presson, A.P.; Firpo, M.A.; Mulvihill, S.J.; Scaife, C.L. Association of Time-to-Surgery with Outcomes in Clinical Stage I-II Pancreatic Adenocarcinoma Treated with Upfront Surgery. Surgery 2018, 163, 753–760. [Google Scholar] [CrossRef] [PubMed]
  33. Mirkin, K.A.; Hollenbeak, C.S.; Wong, J. Time to Surgery: A Misguided Quality Metric in Early Stage Pancreatic Cancer. J. Gastrointest. Surg. 2018, 22, 1365–1375. [Google Scholar] [CrossRef]
  34. Khuri, S.F.; Henderson, W.G.; DePalma, R.G.; Mosca, C.; Healey, N.A.; Kumbhani, D.J. Determinants of Long-Term Survival after Major Surgery and the Adverse Effect of Postoperative Complications. Ann. Surg. 2005, 242, 326–343. [Google Scholar] [CrossRef] [PubMed]
  35. Merkow, R.P.; Bilimoria, K.Y.; Tomlinson, J.S.; Paruch, J.L.; Fleming, J.B.; Talamonti, M.S.; Ko, C.Y.; Bentrem, D.J. Postoperative Complications Reduce Adjuvant Chemotherapy Use in Resectable Pancreatic Cancer. Ann. Surg. 2014, 260, 372–377. [Google Scholar] [CrossRef]
  36. Tzeng, C.W.D.; Cao, H.S.T.; Lee, J.E.; Pisters, P.W.T.; Varadhachary, G.R.; Wolff, R.A.; Abbruzzese, J.L.; Crane, C.H.; Evans, D.B.; Wang, H.; et al. Treatment Sequencing for Resectable Pancreatic Cancer: Influence of Early Metastases and Surgical Complications on Multimodality Therapy Completion and Survival. J. Gastrointest. Surg. 2014, 18, 16–25. [Google Scholar] [CrossRef] [PubMed]
  37. Verma, V.; Li, J.; Lin, C. Neoadjuvant Therapy for Pancreatic Cancer: Systematic Review of Postoperative Morbidity, Mortality, and Complications. Am. J. Clin. Oncol. 2016, 39, 302–313. [Google Scholar] [CrossRef] [PubMed]
  38. Navez, J.; Bouchart, C.; Mans, L.; Devos, S.; Loi, P.; Closset, J.; Van Laethem, J.L. Neoadjuvant Chemotherapy Associated with Isotoxic High-Dose Stereotactic Body Radiotherapy Does Not Increase Postoperative Complications after Pancreaticoduodenectomy for Nonmetastatic Pancreatic Cancer. J. Surg. Oncol. 2023, 128, 33–40. [Google Scholar] [CrossRef]
  39. Ferrone, C.R.; Marchegiani, G.; Hong, T.S.; Ryan, D.P.; Deshpande, V.; McDonnell, E.I.; Sabbatino, F.; Santos, D.D.; Allen, J.N.; Blaszkowsky, L.S.; 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. [Google Scholar] [CrossRef]
  40. Marchegiani, G.; Andrianello, S.; Nessi, C.; Sandini, M.; Maggino, L.; Malleo, G.; Paiella, S.; Polati, E.; Bassi, C.; Salvia, R. Neoadjuvant Therapy Versus Upfront Resection for Pancreatic Cancer: The Actual Spectrum and Clinical Burden of Postoperative Complications. Ann. Surg. Oncol. 2018, 25, 626–637. [Google Scholar] [CrossRef]
  41. Groot Koerkamp, B.; Janssen, Q.P.; van Dam, J.L.; Bonsing, B.A.; Bos, H.; Bosscha, K.P.; Haberkorn, B.C.M.; de Hingh, I.H.J.T.; Karsten, T.M.; Van der Kolk, M.B.; et al. LBA83 Neoadjuvant Chemotherapy with FOLFIRINOX versus Neoadjuvant Gemcitabine-Based Chemoradiotherapy for Borderline Resectable and Resectable Pancreatic Cancer (PREOPANC-2): A Multicenter Randomized Controlled Trial. Ann. Oncol. 2023, 34, S1323. [Google Scholar] [CrossRef]
  42. Alva-Ruiz, R.; Yohanathan, L.; Yonkus, J.A.; Abdelrahman, A.M.; Gregory, L.A.; Halfdanarson, T.R.; Mahipal, A.; McWilliams, R.R.; Ma, W.W.; Hallemeier, C.L.; et al. Neoadjuvant Chemotherapy Switch in Borderline Resectable/Locally Advanced Pancreatic Cancer. Ann. Surg. Oncol. 2022, 29, 1579–1591. [Google Scholar] [CrossRef] [PubMed]
  43. Labori, K.J.; Bratlie, S.O.; Andersson, B.; Angelsen, J.H.; Biörserud, C.; Björnsson, B.; Bringeland, E.A.; Elander, N.; Garresori, H.; Grønbech, J.E.; et al. Neoadjuvant FOLFIRINOX versus Upfront Surgery for Resectable Pancreatic Head Cancer (NORPACT-1): A Multicentre, Randomised, Phase 2 Trial. Lancet Gastroenterol. Hepatol. 2024, 9, 205–217. [Google Scholar] [CrossRef] [PubMed]
  44. van Dam, J.L.; Verkolf, E.M.M.; Dekker, E.N.; Bonsing, B.A.; Bratlie, S.O.; Brosens, L.A.A.; Busch, O.R.; van Driel, L.M.J.W.; van Eijck, C.H.J.; Feshtali, S.; et al. Perioperative or Adjuvant MFOLFIRINOX for Resectable Pancreatic Cancer (PREOPANC-3): Study Protocol for a Multicenter Randomized Controlled Trial. BMC Cancer 2023, 23, 1–8. [Google Scholar] [CrossRef]
  45. Conroy, T.; Pfeiffer, P.; Vilgrain, V.; Lamarca, A.; Seufferlein, T.; Hackert, T.; Golan, T.; Prager, G.; Haustermans, K.; Vogel, A.; et al. Pancreatic Cancer: ESMO Clinical Practice Guideline for Diagnosis, Treatment and Follow-up. Ann. Oncol. 2023, 34, 987–1002. [Google Scholar] [CrossRef]
  46. Lurie, R.H.; Cardin, D.B.; Gabriela Chiorean, E.; Hutchinson Cancer Center Jared Christensen, F.A.; Chung, V.; Czito, B.; Del Chiaro, M.; Donahue, T.R.; Dotan, E.; Fountzilas, C.; et al. NCCN Guidelines Version 1.2024 Pancreatic Adenocarcinoma Continue NCCN Guidelines Version 1.2024 Pancreatic Adenocarcinoma. 2023. Available online: https://www.nccn.org/professionals/physician_gls/pdf/pancreatic.pdf (accessed on 24 June 2024).
  47. Versteijne, E.; Vogel, J.A.; Besselink, M.G.; Busch, O.R.C.; Wilmink, J.W.; Daams, J.G.; van Eijck, C.H.J.; Groot Koerkamp, B.; Rasch, C.R.N.; van Tienhoven, G. Meta-Analysis Comparing Upfront Surgery with Neoadjuvant Treatment in Patients with Resectable or Borderline Resectable Pancreatic Cancer. Br. J. Surg. 2018, 105, 946–958. [Google Scholar] [CrossRef]
  48. Janssen, Q.P.; Buettner, S.; Suker, M.; Beumer, B.R.; Addeo, P.; Bachellier, P.; Bahary, N.; Bekaii-Saab, T.; Bali, M.A.; Besselink, M.G.; et al. Neoadjuvant FOLFIRINOX in Patients with Borderline Resectable Pancreatic Cancer: A Systematic Review and Patient-Level Meta-Analysis. J. Natl. Cancer Inst. 2019, 111, 782–794. [Google Scholar] [CrossRef] [PubMed]
  49. Jang, J.Y.; Han, Y.; Lee, H.; Kim, S.W.; Kwon, W.; Lee, K.H.; Oh, D.Y.; Chie, E.K.; Lee, J.M.; Heo, J.S.; 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. [Google Scholar] [CrossRef]
  50. Ghaneh, P.; Palmer, D.; Cicconi, S.; Jackson, R.; Halloran, C.M.; Rawcliffe, C.; Sripadam, R.; Mukherjee, S.; Soonawalla, Z.; Wadsley, J.; et al. Immediate Surgery Compared with Short-Course Neoadjuvant Gemcitabine plus Capecitabine, FOLFIRINOX, or Chemoradiotherapy in Patients with Borderline Resectable Pancreatic Cancer (ESPAC5): A Four-Arm, Multicentre, Randomised, Phase 2 Trial. Lancet Gastroenterol. Hepatol. 2023, 8, 157–168. [Google Scholar] [CrossRef]
  51. Golcher, H.; Brunner, T.B.; Witzigmann, H.; Marti, L.; Bechstein, W.O.; Bruns, C.; Jungnickel, H.; Schreiber, S.; Grabenbauer, G.G.; Meyer, T.; 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. [Google Scholar] [CrossRef]
  52. Motoi, F.; Kosuge, T.; Ueno, H.; Yamaue, H.; Satoi, S.; Sho, M.; Honda, G.; Matsumoto, I.; Wada, K.; Furuse, J.; et al. Randomized Phase II/III Trial of Neoadjuvant Chemotherapy with Gemcitabine and S-1 versus Upfront Surgery for Resectable Pancreatic Cancer (Prep-02/JSAP05). Jpn. J. Clin. Oncol. 2019, 49, 190–194. [Google Scholar] [CrossRef] [PubMed]
  53. Al-Batran, S.-E.; Reichart, A.; Bankstahl, U.S.; Pauligk, C.; Kraus, T.W.; Bechstein, W.O.; Trojan, J.; Behrend, M.; Potenberg, J.; Homann, N.; et al. Randomized Multicenter Phase II/III Study with Adjuvant Gemcitabine versus Neoadjuvant/Adjuvant FOLFIRINOX in Resectable Pancreatic Cancer: The NEPAFOX Trial. J. Clin. Oncol. 2021, 39, 406. [Google Scholar] [CrossRef]
  54. Unno, M.; Motoi, F.; Matsuyama, Y.; Satoi, S.; Matsumoto, I.; Aosasa, S.; Shirakawa, H.; Wada, K.; Fujii, T.; Yoshitomi, H.; 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, 189. [Google Scholar] [CrossRef]
  55. Garcia-Aguilar, J.; Patil, S.; Gollub, M.J.; Kim, J.K.; Yuval, J.B.; Thompson, H.M.; Verheij, F.S.; Omer, D.M.; Lee, M.; Dunne, R.F.; et al. Organ Preservation in Patients with Rectal Adenocarcinoma Treated with Total Neoadjuvant Therapy. J. Clin. Oncol. 2022, 40, 2546–2556. [Google Scholar] [CrossRef] [PubMed]
  56. Nagakawa, Y.; Hosokawa, Y.; Nakayama, H.; Sahara, Y.; Takishita, C.; Nakajima, T.; Hijikata, Y.; Kasuya, K.; Katsumata, K.; Tokuuye, K.; 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. [Google Scholar] [CrossRef] [PubMed]
  57. Dholakia, A.S.; Hacker-Prietz, A.; Wild, A.T.; Raman, S.P.; Wood, L.D.; Huang, P.; Laheru, D.A.; Zheng, L.; De Jesus-Acosta, A.; Le, D.T.; 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. [Google Scholar] [CrossRef] [PubMed]
  58. Auclin, E.; Marthey, L.; Abdallah, R.; Mas, L.; Francois, E.; Saint, A.; Cunha, A.S.; Vienot, A.; Lecomte, T.; Hautefeuille, V.; et al. Role of FOLFIRINOX and Chemoradiotherapy in Locally Advanced and Borderline Resectable Pancreatic Adenocarcinoma: Update of the AGEO Cohort. Br. J. Cancer 2021, 124, 1941–1948. [Google Scholar] [CrossRef] [PubMed]
  59. Wo, J.Y.; Niemierko, A.; Ryan, D.P.; Blaszkowsky, L.S.; Clark, J.W.; Kwak, E.L.; Lillemoe, K.D.; Drapek, L.N.; Zhu, A.X.; Allen, J.N.; 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. [Google Scholar] [CrossRef]
  60. Hammel, P.; Huguet, F.; Van Laethem, J.L.; Goldstein, D.; Glimelius, B.; Artru, P.; Borbath, I.; Bouché, O.; Shannon, J.; André, T.; et al. 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. [Google Scholar] [CrossRef]
  61. Sudo, K.; Hara, R.; Nakamura, K.; Kita, E.; Tsujimoto, A.; Yamaguchi, T. 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. [Google Scholar] [CrossRef]
  62. Huguet, F.; Hajj, C.; Winston, C.B.; Shi, W.; Zhang, Z.; Wu, A.J.; O’Reilly, E.M.; Reidy, D.L.; Allen, P.; Goodman, K.A. Chemotherapy and Intensity-Modulated Radiation Therapy for Locally Advanced Pancreatic Cancer Achieves a High Rate of R0 Resection. Acta Oncol. 2017, 56, 384–390. [Google Scholar] [CrossRef] [PubMed]
  63. Sherman, W.H.; Hecht, E.; Leung, D.; Chu, K. 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. [Google Scholar] [CrossRef]
  64. Ioka, T.; Furuse, J.; Fukutomi, A.; Mizusawa, J.; Nakamura, S.; Hiraoka, N.; Ito, Y.; Katayama, H.; Ueno, M.; Ikeda, M.; et al. Randomized phase II study of chemoradiotherapy with versus without induction chemotherapy for locally advanced pancreatic cancer: Japan Clinical Oncology Group trial, JCOG1106. Jpn. J. Clin. Oncol. 2021, 51, 235–243. [Google Scholar] [CrossRef] [PubMed]
  65. Fietkau, R.; Ghadimi, M.; Grützmann, R.; Wittel, U.A.; Jacobasch, L.; Uhl, W.; Croner, R.S.; Bechstein, W.O.; Neumann, U.P.; Waldschmidt, D.; et al. Randomized Phase III Trial of Induction Chemotherapy Followed by Chemoradiotherapy or Chemotherapy Alone for Nonresectable Locally Advanced Pancreatic Cancer: First Results of the CONKO-007 Trial. J. Clin. Oncol. 2022, 40, 4008. [Google Scholar] [CrossRef]
  66. Barhoumi, M.; Mornex, F.; Bonnetain, F.; Rougier, P.; Mariette, C.; Bouché, O.; Bosset, J.F.; Aparicio, T.; Mineur, L.; Azzedine, A.; et al. Cancer Du Pancréas Localement Évolué Non Resécable: Chimioradiothérapie d’induction Suivie de Chimiothérapie Par Gemcitabine Contre Chimiothérapie Exclusive Par Gemcitabine: Résultats Définitifs de l’étude de Phase III 2000–2001de La FFCD et de La SFRO. Cancer/Radiothérapie 2011, 15, 182–191. [Google Scholar] [CrossRef]
  67. Versteijne, E.; Van Dam, J.L.; Suker, M.; Janssen, Q.P.; Groothuis, K.; Akkermans-Vogelaar, J.M.; Besselink, M.G.; Bonsing, B.A.; Buijsen, J.; Busch, O.R.; et al. Neoadjuvant Chemoradiotherapy Versus Upfront Surgery for Resectable and Borderline Resectable Pancreatic Cancer: Long-Term Results of the Dutch Randomized PREOPANC Trial. J. Clin. Oncol. 2022, 40, 1220–1230. [Google Scholar] [CrossRef]
  68. Reyngold, M.; Alice, W.; O’Reilly, E.M.; D’Angelica, M.I.; Drebin, J.A.; Soares, K.; Kingham, T.P.; Balachandran, V.P.; Varghese, A.M.; Park, W.; et al. Phase II Trial of Maximal Ablative Irradiation Because of Encasement (MAIBE) for Patients with Potentially Resectable Locally Advanced Pancreatic Cancer. J. Clin. Oncol. 2023, 41, 710. [Google Scholar] [CrossRef]
  69. Mukherjee, S.; Qi, C.; Shaw, R.; Bridgewater, J.; Radhakrishna, G.; Patel, N.; Tranter, B.; Parsons, P.; Falk, S.; Wasan, H.; et al. SCALOP2:A Multicenter Randomized Trial of RT Dose Escalation and Nelfinavir in Pancreatic Cancer. Radiother. Oncol. 2022, 170, S77–S78. [Google Scholar] [CrossRef]
  70. Krishnan, S.; Chadha, A.S.; Suh, Y.; Chen, H.C.; Rao, A.; Das, P.; Minsky, B.D.; Mahmood, U.; Delclos, M.E.; Sawakuchi, G.O.; 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. [Google Scholar] [CrossRef]
  71. Reyngold, M.; O’Reilly, E.; Zinovoy, M.; Romesser, P.B.; Wu, A.J.; Hajj, C.; Cuaron, J.J.; Yorke, E.D.; Varghese, A.M.; Crane, C.H. Ablative RT Results in Excellent Local Control and Survival in Localized Pancreatic Cancer. Int. J. Radiat. Oncol. 2019, 105, S206. [Google Scholar] [CrossRef]
  72. Reyngold, M.; O’Reilly, E.M.; Varghese, A.M.; Fiasconaro, M.; Zinovoy, M.; Romesser, P.B.; Wu, A.; Hajj, C.; Cuaron, J.J.; Tuli, R.; et al. Association of Ablative Radiation Therapy with Survival Among Patients with Inoperable Pancreatic Cancer. JAMA Oncol. 2021, 7, 735–738. [Google Scholar] [CrossRef] [PubMed]
  73. Versteijne, E.; Lens, E.; van der Horst, A.; Bel, A.; Visser, J.; Punt, C.J.A.; Suker, M.; van Eijck, C.H.J.; van Tienhoven, G. Quality Assurance of the PREOPANC Trial (2012-003181-40) for Preoperative Radiochemotherapy in Pancreatic Cancer: The Dummy Run. Strahlenther. Onkol. 2017, 193, 630–638. [Google Scholar] [CrossRef] [PubMed]
  74. McGinn, C.J.; Zalupski, M.M.; Shureiqi, I.; Robertson, J.M.; Eckhauser, F.E.; Smith, D.C.; Brown, D.; Hejna, G.; Strawderman, M.; Normolle, D.; 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. [Google Scholar] [CrossRef]
  75. Small Jr, W.; Berlin, J.; Freedman, G.M.; Lawrence, T.; Talamonti, M.S.; Mulcahy, M.F.; Bapsi Chakravarthy, A.; Konski, A.A.; Zalupski, M.M.; Philip, P.A.; 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. [Google Scholar] [CrossRef]
  76. Conroy, T.; Desseigne, F.; Ychou, M.; Bouché, O.; Guimbaud, R.; Bécouarn, Y.; Adenis, A.; Raoul, J.-L.; Gourgou-Bourgade, S.; de la Fouchardière, C.; et al. FOLFIRINOX versus Gemcitabine for Metastatic Pancreatic Cancer. N. Engl. J. Med. 2011, 364, 1817–1825. [Google Scholar] [CrossRef] [PubMed]
  77. Goldstein, D.; El-Maraghi, R.H.; Hammel, P.; Heinemann, V.; Kunzmann, V.; Sastre, J.; Scheithauer, W.; Siena, S.; Tabernero, J.; Teixeira, L.; et al. Nab-Paclitaxel plus Gemcitabine for Metastatic Pancreatic Cancer: Long-Term Survival from a Phase III Trial. J. Natl. Cancer Inst. 2015, 107. [Google Scholar] [CrossRef] [PubMed]
  78. Wainberg, Z.A.; Melisi, D.; Macarulla, T.; Pazo Cid, R.; Chandana, S.R.; De La Fouchardière, C.; Dean, A.; Kiss, I.; Lee, W.J.; Goetze, T.O.; et al. NALIRIFOX versus Nab-Paclitaxel and Gemcitabine in Treatment-Naive Patients with Metastatic Pancreatic Ductal Adenocarcinoma (NAPOLI 3): A Randomised, Open-Label, Phase 3 Trial. Lancet 2023, 402, 1272–1281. [Google Scholar] [CrossRef]
  79. Suker, M.; Beumer, B.R.; Sadot, E.; Marthey, L.; Faris, J.E.; Mellon, E.A.; El-Rayes, B.F.; Wang-Gillam, A.; Lacy, J.; Hosein, P.J.; et al. FOLFIRINOX for Locally Advanced Pancreatic Cancer: A Systematic Review and Patient-Level Meta-Analysis. Lancet Oncol. 2016, 17, 801–810. [Google Scholar] [CrossRef] [PubMed]
  80. Hackert, T.; Sachsenmaier, M.; Hinz, U.; Schneider, L.; Michalski, C.W.; Springfeld, C.; Strobel, O.; Jäger, D.; Ulrich, A.; Büchler, M.W. Locally Advanced Pancreatic Cancer: Neoadjuvant Therapy with Folfirinox Results in Resectability in 60% of the Patients. Ann. Surg. 2016, 264, 457–461. [Google Scholar] [CrossRef]
  81. Dhir, M.; Zenati, M.S.; Hamad, A.; Singhi, A.D.; Bahary, N.; Hogg, M.E.; Zeh, H.J.; Zureikat, A.H. FOLFIRINOX Versus Gemcitabine/Nab-Paclitaxel for Neoadjuvant Treatment of Resectable and Borderline Resectable Pancreatic Head Adenocarcinoma. Ann. Surg. Oncol. 2018, 25, 1896–1903. [Google Scholar] [CrossRef]
  82. Sohal, D.; Duong, M.T.; Ahmad, S.A.; Gandhi, N.; Beg, M.S.; Wang-Gillam, A.; Wade, J.L.; Chiorean, E.G.; Guthrie, K.A.; Lowy, A.M.; et al. SWOG S1505: Results of Perioperative Chemotherapy (Peri-Op CTx) with Mfolfirinox versus Gemcitabine/Nab-Paclitaxel (Gem/NabP) for Resectable Pancreatic Ductal Adenocarcinoma (PDA). J. Clin. Oncol. 2020, 38, 4504. [Google Scholar] [CrossRef]
  83. Potters, L.; Kavanagh, B.; Galvin, J.M.; Hevezi, J.M.; Janjan, N.A.; Larson, D.A.; Mehta, M.P.; Ryu, S.; Steinberg, M.; Timmerman, R.; et al. 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. [Google Scholar] [CrossRef]
  84. Timmerman, R.D.; Herman, J.; Cho, L.C. Emergence of Stereotactic Body Radiation Therapy and Its Impact on Current and Future Clinical Practice. J. Clin. Oncol. 2014, 32, 2847–2854. [Google Scholar] [CrossRef] [PubMed]
  85. Kamel, R.; Dennis, K.; Doody, J.; Pantarotto, J. Ablative vs. Non-Ablative Radiotherapy in Palliating Locally Advanced Pancreatic Cancer: A Single Institution Experience and a Systematic Review of the Literature. Cancers 2023, 15, 3016. [Google Scholar] [CrossRef] [PubMed]
  86. Rwigema, J.C.M.; Parikh, S.D.; Heron, D.E.; Howell, M.; Zeh, H.; Moser, A.J.; Bahary, N.; Quinn, A.; Burton, S.A. Stereotactic Body Radiotherapy in the Treatment of Advanced Adenocarcinoma of the Pancreas. Am. J. Clin. Oncol. 2011, 34, 63–69. [Google Scholar] [CrossRef] [PubMed]
  87. Mellon, E.A.; Hoffe, S.E.; Springett, G.M.; Frakes, J.M.; Strom, T.J.; Hodul, P.J.; Malafa, M.P.; Chuong, M.D.; Shridhar, R. 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. [Google Scholar] [CrossRef] [PubMed]
  88. Moningi, S.; Dholakia, A.S.; Raman, S.P.; Blackford, A.; Cameron, J.L.; Le, D.T.; De Jesus-Acosta, A.M.C.; Hacker-Prietz, A.; Rosati, L.M.; Assadi, R.K.; et al. The Role of Stereotactic Body Radiation Therapy for Pancreatic Cancer: A Single-Institution Experience. Ann. Surg. Oncol. 2015, 22, 2352–2358. [Google Scholar] [CrossRef] [PubMed]
  89. Mellon, E.A.; Jin, W.H.; Frakes, J.M.; Centeno, B.A.; Strom, T.J.; Springett, G.M.; Malafa, M.P.; Shridhar, R.; Hodul, P.J.; Hoffe, S.E. 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. [Google Scholar] [CrossRef] [PubMed]
  90. Quan, K.; Sutera, P.; Xu, K.; Bernard, M.E.; Burton, S.A.; Wegner, R.E.; Zeh, H.; Bahary, N.; Stoller, R.; Heron, D.E. 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. [Google Scholar] [CrossRef]
  91. Gurka, M.K.; Kim, C.; He, A.R.; Charabaty, A.; Haddad, N.; Turocy, J.; Johnson, L.; Jackson, P.; Weiner, L.M.; Marshall, J.L.; et al. Stereotactic Body Radiation Therapy (SBRT) Combined with Chemotherapy for Unresected Pancreatic Adenocarcinoma. Am. J. Clin. Oncol. 2017, 40, 152–157. [Google Scholar] [CrossRef]
  92. Kharofa, J.; Mierzwa, M.; Olowokure, O.; Sussman, J.; Latif, T.; Gupta, A.; Xie, C.; Patel, S.; Esslinger, H.; McGill, B.; 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. Cancer Clin. Trials 2019, 42, 247–252. [Google Scholar] [CrossRef]
  93. Palta, M.; Czito, B.G.; Duffy, E.; Malicki, M.; Niedzwiecki, D.; Abbruzzese, J.L.; Uronis, H.E.; Blobe, G.C.; Blazer, D.G.; Willett, C. 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] [CrossRef]
  94. Jiang, W.; Haque, W.; Verma, V.; Butler, E.B.; Teh, B.S. Neoadjuvant Stereotactic Body Radiation Therapy for Nonmetastatic Pancreatic Adenocarcinoma. Acta Oncol. 2019, 58, 1259–1266. [Google Scholar] [CrossRef]
  95. Katz, M.H.G.; Shi, Q.; Meyers, J.P.; Herman, J.M.; Choung, M.; Wolpin, B.M.; Ahmad, S.; Marsh, R.d.W.; Schwartz, L.H.; Behr, S.; et al. Alliance A021501: Preoperative MFOLFIRINOX or MFOLFIRINOX plus Hypofractionated Radiation Therapy (RT) for Borderline Resectable (BR) Adenocarcinoma of the Pancreas. J. Clin. Oncol. 2021, 39, 377. [Google Scholar] [CrossRef]
  96. Zhu, X.; Shi, D.; Li, F.; Ju, X.; Cao, Y.; Shen, Y.; Cao, F.; Qing, S.; Fang, F.; Jia, Z.; 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. [Google Scholar] [CrossRef] [PubMed]
  97. Chuong, M.D.; Frakes, J.M.; Figura, N.; Hoffe, S.E.; Shridhar, R.; Mellon, E.A.; Hodul, P.J.; Malafa, M.P.; Springett, G.M.; Centeno, B.A. Histopathologic tumor response after induction chemotherapy and stereotactic body radiation therapy for borderline resectable pancreatic cancer. J. Gastrointest. Oncol. 2016, 7, 221–227. [Google Scholar] [CrossRef] [PubMed]
  98. Chuong, M.D.; Springett, G.M.; Freilich, J.M.; Park, C.K.; Weber, J.M.; Mellon, E.A.; Hodul, P.J.; Malafa, M.P.; Meredith, K.L.; Hoffe, S.E.; 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. [Google Scholar] [CrossRef]
  99. Herman, J.M.; Chang, D.T.; Goodman, K.A.; Dholakia, A.S.; Raman, S.P.; Hacker-Prietz, A.; Iacobuzio-Donahue, C.A.; Griffith, M.E.; Pawlik, T.M.; Pai, J.S.; 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. [Google Scholar] [CrossRef] [PubMed]
  100. Jumeau, R.; Delouya, G.; Roberge, D.; Donath, D.; Béliveau-Nadeau, D.; Campeau, M.P. Stereotactic Body Radiotherapy (SBRT) for Patients with Locally Advanced Pancreatic Cancer: A Single Center Experience. Dig. Liver Dis. 2018, 50, 396–400. [Google Scholar] [CrossRef]
  101. Jung, J.; Yoon, S.M.; Park, J.H.; Seo, D.W.; Lee, S.S.; Kim, M.H.; Lee, S.K.; Park, D.H.; Song, T.J.; Ryoo, B.Y.; et al. Stereotactic Body Radiation Therapy for Locally Advanced Pancreatic Cancer. PLoS ONE 2019, 14, e0214970. [Google Scholar] [CrossRef]
  102. Simoni, N.; Micera, R.; Paiella, S.; Guariglia, S.; Zivelonghi, E.; Malleo, G.; Rossi, G.; Addari, L.; Giuliani, T.; Pollini, T.; et al. Hypofractionated Stereotactic Body Radiation Therapy with Simultaneous Integrated Boost and Simultaneous Integrated Protection in Pancreatic Ductal Adenocarcinoma. Clin. Oncol. 2021, 33, e31–e38. [Google Scholar] [CrossRef] [PubMed]
  103. Suker, M.; Nuyttens, J.J.; Eskens, F.A.L.M.; Haberkorn, B.C.M.; Coene, P.P.L.O.; van der Harst, E.; Bonsing, B.A.; Vahrmeijer, A.L.; Mieog, J.S.D.; Jan Swijnenburg, R.; et al. Efficacy and Feasibility of Stereotactic Radiotherapy after Folfirinox in Patients with Locally Advanced Pancreatic Cancer (LAPC-1 Trial). EClinicalMedicine 2019, 17, 100200. [Google Scholar] [CrossRef] [PubMed]
  104. Comito, T.; Cozzi, L.; Clerici, E.; Franzese, C.; Tozzi, A.; Iftode, C.; Navarria, P.; D’Agostino, G.; Rimassa, L.; Carnaghi, C.; 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. [Google Scholar] [CrossRef] [PubMed]
  105. Zhong, J.; Patel, K.; Switchenko, J.; Cassidy, R.J.; Hall, W.A.; Gillespie, T.; Patel, P.R.; Kooby, D.; Landry, J. Outcomes for Patients with Locally Advanced Pancreatic Adenocarcinoma Treated with Stereotactic Body Radiation Therapy versus Conventionally Fractionated Radiation. Cancer 2017, 123, 3486–3493. [Google Scholar] [CrossRef] [PubMed]
  106. Hassanzadeh, C.; Rudra, S.; Bommireddy, A.; Hawkins, W.G.; Wang-Gillam, A.; Fields, R.C.; Cai, B.; Park, J.; Green, O.; Roach, M.; et al. Ablative Five-Fraction Stereotactic Body Radiation Therapy for Inoperable Pancreatic Cancer Using Online MR-Guided Adaptation. Adv. Radiat. Oncol. 2021, 6, 100506. [Google Scholar] [CrossRef]
  107. Chuong, M.D.; Herrera, R.; Kaiser, A.; Rubens, M.; Romaguera, T.; Alvarez, D.; Kotecha, R.; Hall, M.D.; McCulloch, J.; Ucar, A.; et al. Induction Chemotherapy and Ablative Stereotactic Magnetic Resonance Image-Guided Adaptive Radiation Therapy for Inoperable Pancreas Cancer. Front. Oncol. 2022, 12, 888462. [Google Scholar] [CrossRef] [PubMed]
  108. Parikh, P.J.; Lee, P.; Low, D.A.; Kim, J.; Mittauer, K.E.; Bassetti, M.F.; Glide-Hurst, C.K.; Raldow, A.C.; Yang, Y.; Portelance, L.; et al. A Multi-Institutional Phase 2 Trial of Ablative 5-Fraction Stereotactic Magnetic Resonance-Guided On-Table Adaptive Radiation Therapy for Borderline Resectable and Locally Advanced Pancreatic Cancer. Int. J. Radiat. Oncol. Biol. Phys. 2023, 117, 799–808. [Google Scholar] [CrossRef]
  109. Rudra, S.; Jiang, N.; Rosenberg, S.A.; Olsen, J.R.; Roach, M.C.; Wan, L.; Portelance, L.; Mellon, E.A.; Bruynzeel, A.; Lagerwaard, F.; et al. Using Adaptive Magnetic Resonance Image-Guided Radiation Therapy for Treatment of Inoperable Pancreatic Cancer. Cancer Med. 2019, 8, 2123–2132. [Google Scholar] [CrossRef]
  110. Tringale, K.R.; Tyagi, N.; Romesser, P.B.; Wu, A.; O’Reilly, E.M.; Varghese, A.M.; Scripes, P.G.; Khalil, D.N.; Park, W.; Yu, K.; et al. Stereotactic Ablative Radiation for Pancreatic Cancer on a 1.5 Telsa Magnetic Resonance-Linac System. Phys. Imaging Radiat. Oncol. 2022, 24, 88. [Google Scholar] [CrossRef]
  111. Manderlier, M.; Navez, J.; Hein, M.; Engelholm, J.L.; Closset, J.; Bali, M.A.; Van Gestel, D.; Moretti, L.; Van Laethem, J.L.; Bouchart, C. Isotoxic High-Dose Stereotactic Body Radiotherapy (IHD-SBRT) Versus Conventional Chemoradiotherapy for Localized Pancreatic Cancer: A Single Cancer Center Evaluation. Cancers 2022, 14, 5730. [Google Scholar] [CrossRef]
  112. Bouchart, C.; Navez, J.; Borbath, I.; Geboes, K.; Vandamme, T.; Closset, J.; Moretti, L.; Demetter, P.; Paesmans, M.; Van Laethem, J.L. Preoperative Treatment with MFOLFIRINOX or Gemcitabine/Nab-Paclitaxel +/− Isotoxic High-Dose Stereotactic Body Radiation Therapy (IHD-SBRT) for Borderline Resectable Pancreatic Adenocarcinoma (the STEREOPAC Trial): Study Protocol for a Randomised Comparative Multicenter Phase II Trial. BMC Cancer 2023, 23, 891. [Google Scholar]
  113. Henke, L.E.; Contreras, J.A.; Mazur, T.; Green, O.; Daniel, N.; Lashmett, H.; Senter, T.; Gach, H.M.; Ochoa, L.; Mutic, S.; et al. Delineation of a Cardiac Planning Organ-At-Risk Volume Using Real-Time Magnetic Resonance Imaging for Cardiac Protection in Thoracic and Breast Radiation Therapy. Pract. Radiat. Oncol. 2019, 9, e298–e306. [Google Scholar] [CrossRef]
  114. Rosenberg, S.A.; Henke, L.E.; Shaverdian, N.; Mittauer, K.; Wojcieszynski, A.P.; Hullett, C.R.; Kamrava, M.; Lamb, J.; Cao, M.; Green, O.L.; et al. A Multi-Institutional Experience of MR-Guided Liver Stereotactic Body Radiation Therapy. Adv. Radiat. Oncol. 2018, 4, 142–149. [Google Scholar] [CrossRef] [PubMed]
  115. Chuong, M.D.; Lee, P.; Low, D.A.; Kim, J.; Mittauer, K.E.; Bassetti, M.F.; Glide-Hurst, C.K.; Raldow, A.C.; Yang, Y.; Portelance, L.; et al. Stereotactic MR-Guided on-Table Adaptive Radiation Therapy (SMART) for Borderline Resectable and Locally Advanced Pancreatic Cancer: A Multi-Center, Open-Label Phase 2 Study. Radiother. Oncol. 2024, 191, 110064. [Google Scholar] [CrossRef]
  116. Giraud, P.; Racadot, S.; Vernerey, D.; Goldstein, D.; Glimelius, B.; Van Houtte, P.; Gubanski, M.; Spry, N.; Van Laethem, J.L.; Hammel, P.; et al. Investigation of Relation of Radiation Therapy Quality with Toxicity and Survival in LAP07 Phase 3 Trial for Locally Advanced Pancreatic Carcinoma. Int. J. Radiat. Oncol. Biol. Phys. 2021, 110, 993–1002. [Google Scholar] [CrossRef]
  117. Wang, C.; Liu, X.; Wang, X.; Wang, Y.; Cha, N. Effects of Chemoradiotherapy and Chemotherapy on Survival of Patients with Locally Advanced Pancreatic Cancer: A Meta-Analysis of Randomized Controlled Trials. Medicine 2018, 97, e12260. [Google Scholar] [CrossRef] [PubMed]
  118. Chang, J.S.; Chiu, Y.F.; Yu, J.C.; Chen, L.T.; Ch’ang, H.J. 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. [Google Scholar] [CrossRef] [PubMed]
  119. Petrelli, F.; Comito, T.; Ghidini, A.; Torri, V.; Scorsetti, M.; Barni, S. Stereotactic Body Radiation Therapy for Locally Advanced Pancreatic Cancer: A Systematic Review and Pooled Analysis of 19 Trials. Int. J. Radiat. Oncol. Biol. Phys. 2017, 97, 313–322. [Google Scholar] [CrossRef]
  120. Tchelebi, L.T.; Lehrer, E.J.; Trifiletti, D.M.; Sharma, N.K.; Gusani, N.J.; Crane, C.H.; Zaorsky, N.G. 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. [Google Scholar] [CrossRef]
  121. Perri, G.; Prakash, L.R.; Katz, M.H.G. Response to Preoperative Therapy in Localized Pancreatic Cancer. Front. Oncol. 2020, 10, 516. [Google Scholar] [CrossRef]
  122. Tsai, S.; George, B.; Wittmann, D.; Ritch, P.S.; Krepline, A.N.; Aldakkak, M.; Barnes, C.A.; Christians, K.K.; Dua, K.; Griffin, M.; et al. Importance of Normalization of CA19-9 Levels Following Neoadjuvant Therapy in Patients with Localized Pancreatic Cancer. Ann. Surg. 2020, 271, 740–747. [Google Scholar] [CrossRef]
  123. Perri, G.; Prakash, L.; Wang, H.; Bhosale, P.; Varadhachary, G.R.; Wolff, R.; Fogelman, D.; Overman, M.; Pant, S.; Javle, M.; et al. Radiographic and Serologic Predictors of Pathologic Major Response to Preoperative Therapy for Pancreatic Cancer. Ann. Surg. 2021, 273, 806–813. [Google Scholar] [CrossRef]
  124. Ye, C.; Sadula, A.; Ren, S.; Guo, X.; Yuan, M.; Yuan, C.; Xiu, D. The Prognostic Value of CA19-9 Response after Neoadjuvant Therapy in Patients with Pancreatic Cancer: A Systematic Review and Pooled Analysis. Cancer Chemother. Pharmacol. 2020, 86, 731–740. [Google Scholar] [CrossRef]
  125. Katz, M.H.G.; Fleming, J.B.; Bhosale, P.; Varadhachary, G.; Lee, J.E.; Wolff, R.; Wang, H.; Abbruzzese, J.; Pisters, P.W.T.; Vauthey, J.N.; et al. Response of Borderline Resectable Pancreatic Cancer to Neoadjuvant Therapy Is Not Reflected by Radiographic Indicators. Cancer 2012, 118, 5749–5756. [Google Scholar] [CrossRef]
  126. Katz, M.H.G.; Pisters, P.W.T.; Lee, J.E.; Fleming, J.B. Borderline Resectable Pancreatic Cancer: What Have We Learned and Where Do We Go from Here? Ann. Surg. Oncol. 2011, 18, 608–610. [Google Scholar] [CrossRef] [PubMed]
  127. Tempero, M.A.; Arnoletti, J.P.; Behrman, S.W.; Ben-Josef, E.; Benson, A.B.; Casper, E.S.; Cohen, S.J.; Czito, B.; Ellenhorn, J.D.I.; Hawkins, W.G.; et al. Pancreatic Adenocarcinoma, Version 2.2012: Featured Updates to the NCCN Guidelines. J. Natl. Compr. Cancer Netw. 2012, 10, 703–713. [Google Scholar] [CrossRef] [PubMed]
  128. Varadhachary, G.R.; Tamm, E.P.; Abbruzzese, J.L.; Xiong, H.Q.; Crane, C.H.; Wang, H.; Lee, J.E.; Pisters, P.W.T.; Evans, D.B.; Wolff, R.A. Borderline Resectable Pancreatic Cancer: Definitions, Management, and Role of Preoperative Therapy. Ann. Surg. Oncol. 2006, 13, 1035–1046. [Google Scholar] [CrossRef]
  129. Chatterjee, D.; Katz, M.H.; Rashid, A.; Varadhachary, G.R.; Wolff, R.A.; Wang, H.; Lee, J.E.; Pisters, P.W.T.; Vauthey, J.N.; Crane, C.; et al. Histologic Grading of the Extent of Residual Carcinoma Following Neoadjuvant Chemoradiation in Pancreatic Ductal Adenocarcinoma: A Predictor for Patient Outcome. Cancer 2012, 118, 3182–3190. [Google Scholar] [CrossRef] [PubMed]
  130. Bali, M.A.; Pullini, S.; Metens, T.; Absil, J.; Chao, S.L.; Marechal, R.; Matos, C.; Peerboccus, B.M.; Van Laethem, J.L. Assessment of Response to Chemotherapy in Pancreatic Ductal Adenocarcinoma: Comparison between Diffusion-Weighted MR Quantitative Parameters and RECIST. Eur. J. Radiol. 2018, 104, 49–57. [Google Scholar] [CrossRef]
  131. Yeh, R.; Dercle, L.; Garg, I.; Wang, Z.J.; Hough, D.M.; Goenka, A.H. The Role of 18F-FDG PET/CT and PET/MRI in Pancreatic Ductal Adenocarcinoma. Abdom. Radiol. 2018, 43, 415–434. [Google Scholar] [CrossRef]
  132. Yoshioka, M.; Sato, T.; Furuya, T.; Shibata, S.; Andoh, H.; Asanuma, Y.; Hatazawa, J.; Shimosegawa, E.; Koyama, K.; Yamamoto, Y. Role of Positron Emission Tomography with 2-Deoxy-2-[18F]Fluoro-D-Glucose in Evaluating the Effects of Arterial Infusion Chemotherapy and Radiotherapy on Pancreatic Cancer. J. Gastroenterol. 2004, 39, 50–55. [Google Scholar] [CrossRef]
  133. Sperti, C.; Pasquali, C.; Bissoli, S.; Chierichetti, F.; Liessi, G.; Pedrazzoli, S. Tumor Relapse after Pancreatic Cancer Resection Is Detected Earlier by 18-FDG PET than by CT. J. Gastrointest. Surg. 2010, 14, 131–140. [Google Scholar] [CrossRef] [PubMed]
  134. Wang, L.; Dong, P.; Shen, G.; Hou, S.; Zhang, Y.; Liu, X.; Tian, B. 18F-Fluorodeoxyglucose Positron Emission Tomography Predicts Treatment Efficacy and Clinical Outcome for Patients with Pancreatic Carcinoma: A Meta-Analysis. Pancreas 2019, 48, 996–1002. [Google Scholar] [CrossRef] [PubMed]
  135. Panda, A.; Garg, I.; Truty, M.J.; Kline, T.L.; Johnson, M.P.; Ehman, E.C.; Suman, G.; Anaam, D.A.; Kemp, B.J.; Johnson, G.B.; et al. Borderline Resectable and Locally Advanced Pancreatic Cancer: FDG PET/MRI and CT Tumor Metrics for Assessment of Pathologic Response to Neoadjuvant Therapy and Prediction of Survival. AJR. Am. J. Roentgenol. 2021, 217, 730–740. [Google Scholar] [CrossRef] [PubMed]
  136. Lee, W.; Oh, M.; Kim, J.S.; Park, Y.; Kwon, J.W.; Jun, E.; Song, K.B.; Lee, J.H.; Hwang, D.W.; Yoo, C.; et al. Metabolic Activity by FDG-PET/CT after Neoadjuvant Chemotherapy in Borderline Resectable and Locally Advanced Pancreatic Cancer and Association with Survival. Br. J. Surg. 2021, 109, 61–70. [Google Scholar] [CrossRef] [PubMed]
  137. Evangelista, L.; Zucchetta, P.; Moletta, L.; Serafini, S.; Cassarino, G.; Pegoraro, N.; Bergamo, F.; Sperti, C.; Cecchin, D. The Role of FDG PET/CT or PET/MRI in Assessing Response to Neoadjuvant Therapy for Patients with Borderline or Resectable Pancreatic Cancer: A Systematic Literature Review. Ann. Nucl. Med. 2021, 35, 767–776. [Google Scholar] [CrossRef] [PubMed]
  138. Kauhanen, S.P.; Komar, G.; Seppänen, M.P.; Dean, K.I.; Minn, H.R.; Kajander, S.A.; Rinta-Kiikka, I.; Alanen, K.; Borra, R.J.; Puolakkainen, P.A.; et al. A Prospective Diagnostic Accuracy Study of 18F-Fluorodeoxyglucose Positron Emission Tomography/Computed Tomography, Multidetector Row Computed Tomography, and Magnetic Resonance Imaging in Primary Diagnosis and Staging of Pancreatic Cancer. Ann. Surg. 2009, 250, 957–963. [Google Scholar] [CrossRef] [PubMed]
  139. Strobel, O.; Büchler, M.W. Pancreatic Cancer: FDG-PET Is Not Useful in Early Pancreatic Cancer Diagnosis. Nat. Rev. Gastroenterol. Hepatol. 2013, 10, 203–205. [Google Scholar] [CrossRef] [PubMed]
  140. Mukherjee, S.; Patra, A.; Khasawneh, H.; Korfiatis, P.; Rajamohan, N.; Suman, G.; Majumder, S.; Panda, A.; Johnson, M.P.; Larson, N.B.; et al. Radiomics-Based Machine-Learning Models Can Detect Pancreatic Cancer on Prediagnostic Computed Tomography Scans at a Substantial Lead Time Before Clinical Diagnosis. Gastroenterology 2022, 163, 1435–1446. [Google Scholar] [CrossRef]
  141. Whatcott, C.J.; Diep, C.H.; Jiang, P.; Watanabe, A.; Lobello, J.; Sima, C.; Hostetter, G.; Shepard, H.M.; Von Hoff, D.D.; Han, H. Desmoplasia in Primary Tumors and Metastatic Lesions of Pancreatic Cancer. Clin. Cancer Res. 2015, 21, 3561–3568. [Google Scholar] [CrossRef]
  142. Mei, L.; Du, W.; Ma, W.W. Targeting Stromal Microenvironment in Pancreatic Ductal Adenocarcinoma: Controversies and Promises. J. Gastrointest. Oncol. 2016, 7, 487. [Google Scholar] [CrossRef] [PubMed]
  143. Duluc, C.; Moatassim-Billah, S.; Chalabi-Dchar, M.; Perraud, A.; Samain, R.; Breibach, F.; Gayral, M.; Cordelier, P.; Delisle, M.-B.; Bousquet-Dubouch, M.-P.; et al. Pharmacological Targeting of the Protein Synthesis MTOR/4E-BP1 Pathway in Cancer-Associated Fibroblasts Abrogates Pancreatic Tumour Chemoresistance. EMBO Mol. Med. 2015, 7, 735. [Google Scholar] [CrossRef] [PubMed]
  144. Liu, F.; Qi, L.; Liu, B.; Liu, J.; Zhang, H.; Che, D.H.; Cao, J.Y.; Shen, J.; Geng, J.X.; Bi, Y.; et al. Fibroblast Activation Protein Overexpression and Clinical Implications in Solid Tumors: A Meta-Analysis. PLoS ONE 2015, 10, e0116683. [Google Scholar] [CrossRef] [PubMed]
  145. Moffitt, R.A.; Marayati, R.; Flate, E.L.; Volmar, K.E.; Loeza, S.G.H.; Hoadley, K.A.; Rashid, N.U.; Williams, L.A.; Eaton, S.C.; Chung, A.H.; et al. Virtual Microdissection Identifies Distinct Tumor- and Stroma-Specific Subtypes of Pancreatic Ductal Adenocarcinoma. Nat. Genet. 2015, 47, 1168–1178. [Google Scholar] [CrossRef] [PubMed]
  146. Bulle, A.; Lim, K.H. Beyond Just a Tight Fortress: Contribution of Stroma to Epithelial-Mesenchymal Transition in Pancreatic Cancer. Signal Transduct. Target. Ther. 2020, 5, 249. [Google Scholar] [CrossRef]
  147. Kratochwil, C.; Flechsig, P.; Lindner, T.; Abderrahim, L.; Altmann, A.; Mier, W.; Adeberg, S.; Rathke, H.; Röhrich, M.; Winter, H.; et al. 68Ga-FAPI PET/CT: Tracer Uptake in 28 Different Kinds of Cancer. J. Nucl. Med. 2019, 60, 801. [Google Scholar] [CrossRef] [PubMed]
  148. Pang, Y.; Zhao, L.; Shang, Q.; Meng, T.; Zhao, L.; Feng, L.; Wang, S.; Guo, P.; Wu, X.; Lin, Q.; et al. Positron Emission Tomography and Computed Tomography with [68Ga]Ga-Fibroblast Activation Protein Inhibitors Improves Tumor Detection and Staging in Patients with Pancreatic Cancer. Eur. J. Nucl. Med. Mol. Imaging 2021, 49, 1322–1337. [Google Scholar] [CrossRef] [PubMed]
  149. Rohrich, M.; Naumann, P.; Giesel, F.L.; Choyke, P.L.; Staudinger, F.; Wefers, A.; Liew, D.P.; Kratochwil, C.; Rathke, H.; Liermann, J.; et al. Impact of 68Ga-FAPI PET/CT Imaging on the Therapeutic Management of Primary and Recurrent Pancreatic Ductal Adenocarcinomas. J. Nucl. Med. 2021, 62, 779. [Google Scholar] [CrossRef] [PubMed]
  150. Chen, H.; Pang, Y.; Wu, J.; Zhao, L.; Hao, B.; Wu, J.; Wei, J.; Wu, S.; Zhao, L.; Luo, Z.; et al. Comparison of [68Ga]Ga-DOTA-FAPI-04 and [18F] FDG PET/CT for the Diagnosis of Primary and Metastatic Lesions in Patients with Various Types of Cancer. Eur. J. Nucl. Med. Mol. Imaging 2020, 47, 1820–1832. [Google Scholar] [CrossRef]
  151. Heredia-Soto, V.; Rodríguez-Salas, N.; Feliu, J. Liquid Biopsy in Pancreatic Cancer: Are We Ready to Apply It in the Clinical Practice? Cancers 2021, 13, 1986. [Google Scholar] [CrossRef]
  152. Stosic, K.; Senar, O.A.; Tarfouss, J.; Bouchart, C.; Navez, J.; Van Laethem, J.L.; Arsenijevic, T. A Comprehensive Review of the Potential Role of Liquid Biopsy as a Diagnostic, Prognostic, and Predictive Biomarker in Pancreatic Ductal Adenocarcinoma. Cells 2023, 13, 3. [Google Scholar] [CrossRef] [PubMed]
  153. Yin, L.; Pu, N.; Thompson, E.; Miao, Y.; Wolfgang, C.; Yu, J. Improved Assessment of Response Status in Patients with Pancreatic Cancer Treated with Neoadjuvant Therapy Using Somatic Mutations and Liquid Biopsy Analysis. Clin. Cancer Res. 2021, 27, 740–748. [Google Scholar] [CrossRef]
  154. Groot, V.P.; Mosier, S.; Javed, A.A.; Teinor, J.A.; Gemenetzis, G.; Ding, D.; Haley, L.M.; Yu, J.; Burkhart, R.A.; Hasanain, A.; et al. Circulating Tumor DNA as a Clinical Test in Resected Pancreatic Cancer. Clin. Cancer Res. 2019, 25, 4973–4984. [Google Scholar] [CrossRef]
  155. Sefrioui, D.; Blanchard, F.; Toure, E.; Basile, P.; Beaussire, L.; Dolfus, C.; Perdrix, A.; Paresy, M.; Antonietti, M.; Iwanicki-Caron, I.; et al. Diagnostic Value of CA19.9, Circulating Tumour DNA and Circulating Tumour Cells in Patients with Solid Pancreatic Tumours. Br. J. Cancer 2017, 117, 1017–1025. [Google Scholar] [CrossRef] [PubMed]
  156. Bernard, V.; Kim, D.U.; San Lucas, F.A.; Castillo, J.; Allenson, K.; Mulu, F.C.; Stephens, B.M.; Huang, J.; Semaan, A.; Guerrero, P.A.; et al. Circulating Nucleic Acids Are Associated with Outcomes of Patients with Pancreatic Cancer. Gastroenterology 2019, 156, 108–118.e4. [Google Scholar] [CrossRef]
  157. Neoptolemos, J.P.; Stocken, D.D.; Friess, H.; Bassi, C.; Dunn, J.A.; Hickey, H.; Beger, H.; Fernandez-Cruz, L.; Dervenis, C.; Lacaine, F.; et al. A Randomized Trial of Chemoradiotherapy and Chemotherapy after Resection of Pancreatic Cancer. N. Engl. J. Med. 2004, 350, 1200–1210. [Google Scholar] [CrossRef]
  158. Ghaneh, P.; Palmer, D.H.; Cicconi, S.; Halloran, C.; Psarelli, E.E.; Rawcliffe, C.L.; Sripadam, R.; Mukherjee, S.; Wadsley, J.; Al-Mukhtar, A.; et al. ESPAC-5F: Four-Arm, Prospective, Multicenter, International Randomized Phase II Trial of Immediate Surgery Compared with Neoadjuvant Gemcitabine plus Capecitabine (GEMCAP) or FOLFIRINOX or Chemoradiotherapy (CRT) in Patients with Borderline Resectable Pancreatic Cancer. J. Clin. Oncol. 2020, 38, 4505. [Google Scholar] [CrossRef]
  159. Versteijne, E.; Suker, M.; Groothuis, K.; Akkermans-Vogelaar, J.M.; Besselink, M.G.; Bonsing, B.A.; Buijsen, J.; Busch, O.R.; Creemers, G.J.M.; van Dam, R.M.; et al. 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. [Google Scholar] [CrossRef] [PubMed]
  160. Janssen, Q.P.; van Dam, J.L.; Bonsing, B.A.; Bos, H.; Bosscha, K.P.; Coene, P.P.L.O.; van Eijck, C.H.J.; de Hingh, I.H.J.T.; Karsten, T.M.; van der Kolk, M.B.; et al. Total Neoadjuvant FOLFIRINOX versus Neoadjuvant Gemcitabine-Based Chemoradiotherapy and Adjuvant Gemcitabine for Resectable and Borderline Resectable Pancreatic Cancer (PREOPANC-2 Trial): Study Protocol for a Nationwide Multicenter Randomized Controlled Trial. BMC Cancer 2021, 21, 300. [Google Scholar] [CrossRef]
  161. Perri, G.; Prakash, L.; Qiao, W.; Varadhachary, G.R.; Wolff, R.; Fogelman, D.; Overman, M.; Pant, S.; Javle, M.; Koay, E.J.; et al. Response and Survival Associated with First-Line FOLFIRINOX vs Gemcitabine and Nab-Paclitaxel Chemotherapy for Localized Pancreatic Ductal Adenocarcinoma. JAMA Surg. 2020, 155, 832–839. [Google Scholar] [CrossRef]
  162. Sohal, D.P.S.; Duong, M.; Ahmad, S.A.; Gandhi, N.S.; Beg, M.S.; Wang-Gillam, A.; Wade, J.L.; Chiorean, E.G.; Guthrie, K.A.; Lowy, A.M.; et al. Efficacy of Perioperative Chemotherapy for Resectable Pancreatic Adenocarcinoma: A Phase 2 Randomized Clinical Trial. JAMA Oncol. 2021, 7, 421–427. [Google Scholar] [CrossRef] [PubMed]
  163. Tiriac, H.; Belleau, P.; Engle, D.D.; Plenker, D.; Deschênes, A.; Somerville, T.D.D.; Froeling, F.E.M.; Burkhart, R.A.; Denroche, R.E.; Jang, G.H.; et al. Organoid Profiling Identifies Common Responders to Chemotherapy in Pancreatic Cancer. Cancer Discov. 2018, 8, 1112–1129. [Google Scholar] [CrossRef] [PubMed]
  164. Miyabayashi, K.; Nakagawa, H.; Koike, K. Molecular and Phenotypic Profiling for Precision Medicine in Pancreatic Cancer: Current Advances and Future Perspectives. Front. Oncol. 2021, 11, 682872. [Google Scholar] [CrossRef] [PubMed]
  165. Nicolle, R.; Bachet, J.-B.; Harlé, A.; Iovanna, J.; Hammel, P.; Rebours, V.; Turpin, A.; Ben Abdelghani, M.; Wei, A.; Mitry, E.; et al. Prediction of Adjuvant Gemcitabine Sensitivity in Resectable Pancreatic Adenocarcinoma Using the GemPred RNA Signature: An Ancillary Study of the PRODIGE-24/CCTG PA6 Clinical Trial. J. Clin. Oncol. 2024, 42, 1067–1076. [Google Scholar] [CrossRef]
  166. Nicolle, R.; Blum, Y.; Duconseil, P.; Vanbrugghe, C.; Brandone, N.; Poizat, F.; Roques, J.; Bigonnet, M.; Gayet, O.; Rubis, M.; et al. Establishment of a Pancreatic Adenocarcinoma Molecular Gradient (PAMG) That Predicts the Clinical Outcome of Pancreatic Cancer. EBioMedicine 2020, 57, 102858. [Google Scholar] [CrossRef] [PubMed]
  167. Nishiwada, S.; Sho, M.; Cui, Y.; Yamamura, K.; Akahori, T.; Nakagawa, K.; Nagai, M.; Nakamura, K.; Takagi, T.; Ikeda, N.; et al. A Gene Expression Signature for Predicting Response to Neoadjuvant Chemoradiotherapy in Pancreatic Ductal Adenocarcinoma. Int. J. Cancer 2021, 148, 769–779. [Google Scholar] [CrossRef] [PubMed]
  168. Pishvaian, M.J.; Blais, E.M.; Brody, J.R.; Lyons, E.; DeArbeloa, P.; Hendifar, A.; Mikhail, S.; Chung, V.; Sahai, V.; Sohal, D.P.S.; et al. Overall Survival in Patients with Pancreatic Cancer Receiving Matched Therapies Following Molecular Profiling: A Retrospective Analysis of the Know Your Tumor Registry Trial. Lancet Oncol. 2020, 21, 508–518. [Google Scholar] [CrossRef]
  169. Mosele, F.; Remon, J.; Mateo, J.; Westphalen, C.B.; Barlesi, F.; Lolkema, M.P.; Normanno, N.; Scarpa, A.; Robson, M.; Meric-Bernstam, F.; et al. Recommendations for the Use of Next-Generation Sequencing (NGS) for Patients with Metastatic Cancers: A Report from the ESMO Precision Medicine Working Group. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2020, 31, 1491–1505. [Google Scholar] [CrossRef]
Table 1. Main studies evaluating TNT in BRPC and LAPC.
Table 1. Main studies evaluating TNT in BRPC and LAPC.
StudyStudy DesignTypeNTreatmentResection (%)Survival/Progression
Neoadjuvant TreatmentAdj Cht (%)R RateR0 RateDFSPFSOS
Induction Cht → CRT or CRT → Consolidation Cht
Dholakia 2013 [57]RetrospBRPC50mFFX/Gem-based → CRT50 Gy/25 (Cap/Gem)NA5893NA13.4 M
NR/R: 16.7/5.9 M
(p < 0.001) *
17.2 M
NR/R: 22.9/13 M
(p < 0.001) *
Katz 2016 [13] (Alliance A021101)Phase IIBRPC22mFFX (4c) → CRT 50.4 Gy/28 (Cap)456893NANA21.7 M
Nagakawa 2017 [56]Phase IIBRPC27Gem (2c) → CRT 50.4 Gy/28 (Gem+S-1)94.77094.7NANA22.4 M
1 y OS 81.3%
2 y OS 33.9%
Wo 2018 [59]RetrospBRPC
LAPC
99FFX /FOLFOX/Gem based (4-8c) → CRT 58.8 Gy/28 (Cap/5FU/Gem)NA3787NANA18.1 M
Pietrasz 2019 [6] AGEO-FRENCHRetrospBRPC
LAPC
203FFX (6c)73.273.276.313.5 MNA35.5 M
FFX (6c) → CRT 54 Gy/30 (5-FU or Cap)41.241.289.2
(p = 0.017) *
17.7 M
(p = 0.121)
NA57.8 M
(p = 0.007) *
Auclin 2021 [58]
AGEO cohort
RetrospBRPC
LAPC
330FFX (7c) → CRT 50.4 Gy/28 (Cap)NA13.874.712.8 MNA21.4 M
Hammel 2016 [60] LAP07Phase III
Rando
LAPC449Gem +/− Erlotinib 4c → 2c CRTNA62.5NANA16.5 M
Gem +/− Erlotinib 4c → CRT 54 Gy/30NA3NANA15.2 M
Sudo 2017 [61]Phase IILAPC30S1+Gem → CRT 50.4 Gy/28 (S1)7010100NA12.7 M21.3 M
Huguet 2017 [62]RetrospLAPC134Gem-based/FFX → CRT 50.4 Gy/28 (Gem/Cap)NA1985NANA2 M
1 y OS 86%
2 y OS 48%
Sherman 2018 [63]Phase IILAPC45GTX → CRT 56 Gy/28 (Cap)NA8970R0 31 M
R1 14 M
NAR0: 37 M
R1: 29 M
Loka 2021 [64]Phase II
Rando
LAPC102S1 → CRT 50.4 Gy/28 (Gem)NANANANANA1 y OS 66.7%
2 y OS 36.9%
Gem 3c → CRT 50.4 Gy/28 (Gem)NANANANANA1 y O 69.3%
2 y OS 18.9%
Fietkau 2022 [65] CONKO-007Phase III
Rando
LAPC335Gem 6c/FFX (12c)NA35.918NA1 yOS 59.0%
2 yOS 17.5%
1 y OS 71.3%
2 y OS 32.5%
Gem (3c) /FFX (6c) → CRT 50.4 Gy/28 (Gem)NA36.325NA1 y OS 56.3%
2 y OS 24.1%
1 y OS 71.1%
2 y OS 34.8%
Barhoumi 2011 [66]Phase IIILAPC119CRT 60 Gy/30 (5FU+CDDP) → GemNANANANANA8.6 M
1 y OS 53%
Cht (Gem)13 M (p = 0.03) *
1 y OS%
PREOPANC
Versteijne 2022 [67] PREOPANC1Phase III
Rando
RPC
BRPC
246Immediate surgery5172437.7 MLFFI 13.4 M14.3 M
CRT: 36 Gy/15 (Gem)466172
(p < 0.001)
8.1 MLFFI 31.2 M
(p = 0.04)
15.7 M
Koerkamp 2023 [41] PREOPANC2Phase III
Rando
RPC
BRPC
375FFX (8c)NA77NANANA21.9 M
CRT: 36 Gy/15 (Gem)Yes75NANANA21.3 M
HFA-IMRT
Reyngold 2023 [68]ProspecBRPC47mFFX/Gem-based → CRT 75 Gy/25; 67.5/15 (Cap)NA2658.3NANA2 y OS 38.9%
2 y OS (NR/R):
37.1%/39.4%
Mukherjee 2022 [69]
SCALOP-2
Phase IILAPC186Gem/Nab-P+/−Nelfinavir → CRT 50.4 Gy/28 (Cap)NANANA1 yPFS
33.3%
1 yLRF 26.7%15.6 M
Gem/Nab-P+/−Nelfinavir → HFA 60 Gy/30 1 yPFS
23.9%
1 yLRF
15.2%
16.9 M
Krishnan 2016 [70]RetrospLAPC200FFX/Gem-based → CRT 50.4 Gy/28 (Cap/Gem)NANANANALR RFS
6.2 M
15 M
3 y OS 9%
FFX/Gem-based → HFA (BED > 70 Gy)LR RFS
10.2 M
17.8 M
3 y OS 31%
Reyngold 2019 [71]ProspecLAPC136CMT → RT 75 Gy/25; 67.5 Gy/15; SBRT 50 Gy/5NANANANA2 y FFLP 78%2 y OS 55%
Reyngold 2021 [72]ProspecLAPC119mFOLFIRINOX/Gem Based → RT 75 Gy/25; 67.5/15NANANANA6.3 M
1 yLRF 17.6%
2 yLRF 32.8%
18.2 M
Abbreviations: * statistically significant; 5-FU: 5-fluoracil; Adj: adjuvant; BRPC: borderline resectable pancreatic cancer; c: cycle; Cap: capecitabine; CDDP: cisplatin; Cht: chemotherapy; CRT: combined chemoradiotherapy; DFS: disease-free survival; FFLP: freedom from local progression; FFX: FOLFIRINOX; Gem: gemcitabine; GTX: gemcitabine, docetaxel, and capecitabine; HFA-IMRT: hypofractionated ablative intensity-modulated radiotherapy; LAPC: locally advanced pancreatic cancer; LFFI: loco-regional failure-free interval; LR: local–regional; LRF: local–regional failure; M: months; N: number; NA: not available; NR: non-resected; OS: overall survival; Prospec: prospective; PFS: progression-free survival; R: resected; Rand: randomized; RCP: resectable pancreatic cancer; RFS: recurrence-free survival; RT: radiotherapy; Retrosp: retrospective; TNT: total neoadjuvant treatment; Y: year.
Table 2. Main studies evaluating SBRT in BRPC and LAPC.
Table 2. Main studies evaluating SBRT in BRPC and LAPC.
StudyStudy DesignTypeNTreatmentResection (%)Survival/Progression
Neoadjuvant TreatmentAdj Cht (%)R RateR0 RateDFS/PFSLCOS
Non-Ablative Stereo (BED10 [50–60 Gy])
Chuong 2016 [97]RetrospBRPC36GTX → SBRT 35 Gy/5 (32.5–40 Gy)No88.697.2mPFS:14.9 MNA22.5 M
Chuong 2013 [98]RetrospBRPC
LAPC
73Gem (3c) → SBRT 25–30 Gy/5 (SIB TVI 35–50 Gy)NABRPC: 56BRPC: 97mPFS
BRPC 9.7 M
LAPC 9.8 M
1 yPFS
BRPC: 42.8%
LAPC: 41%
1 y LC (NR):
81%
BRPC 16.4 M
LAPC 15 M
1 y OS
BRPC: 72.2%
LAPC: 68.1%
Mellon 2015 [87]RetrospBRPC
LAPC
159Gem/mFFX → SBRT 28–30/5 (SIB TVI 50)NABRPC: 51
LAPC: 14
BRPC: 96
LAPC: 100
mPFS
BRPC 19.2 M
LAPC 15 M
NABRPC 19.2 M
LAPC 15 M
Moningi 2015 [88]RetrospBRPC
LAPC
88Gem/mFFX → SBRT 25–33 Gy/5NA2284mPFS 9.9 M
1 y PFS 41%
2 y PFS 11%
13.9 M
1 y LC: 61%
2 y LC: 14%
13.7 M
1 y OS 60%
2 y OS 15%
Mellon 2017 [89]RetrospBRPC
LAPC
222Gem/FFX (3c) → SBRT 25–30/5 (SIB TVI 50 Gy)NABRPC: 51
LAPC: 11
97.5NANA37.5 M
Quan 2018 [90]Phase IIBRPC
LAPC
35Gem/Cap 4c → SBRT 36 Gy/3NABRPC: 53
LAPC: 12.5
91.7NA1 y LC (R/NR) 80/44%BRPC 18.8 M
LAPC 28.3 M
Gurka 2017 [91]RetrospBRPC
LAPC
38Concurrent Gem based/mFOLFOX → SBRT 25–30 Gy/5NANANAmPFS 6.8 M6 M LC 82%12.3 M
Kharofa 2019 [92]Phase II
Rando
BRPC
RPC
18Gem or FFX 3c → SBRT 25 Gy/5 (SIB TVI 33 Gy)NA6792mPFS 1 M1 y LC 50%21 M
Palta 2018 [93]Phase IIBRPC
RPC
25Gem/Nab-P (2c) → SBRT 25 Gy/5NA6893NA1 y LC 77%24 M
Jiang 2019 [94]RetrospBRPC
LAPC
5828CMT + SBRT (5.7%)NANA84.9NANA32.1 M
CMT (55.5%)75.927.5 M
CMT + CRT (38.8)83.227.1 M
Katz 2021 [95]
Alliance A021501
Phase II
Rando
BRPC
LAPC
126mFFX 8c0.345888NANA29.8 M
18 M-OS 66.7%
mFFX (7c) → SBRT 25–33 Gy/5 (SIB TVI 40 Gy)0.24517417.1 M
18 M-OS 47.3%
Herman 2015 [99]Phase IILAPC49Gem → SBRT 33 Gy/5Yes 8100mPFS 7.9 M
1 y PFS 32%
2 y PFS 10%
1 y LC 78%13.9 M
1 y OS 59%
2 y OS 18%
Jumeau 2018 [100]RetrospLAPC17Gem/FFX (38%) → SBRT 30 Gy/5NANANANA6 M LC: 93%1 y LC 67%22 M
Jung 2019 [101]RetrospLAPC95Gem Based/mFFX → SBRT 24–36 (median 28 Gy/4)NA7.457mPFS 10.2 M
1 y PFS 42.9%
NA16.7 M
1 y OS 67.4%
Nearly Ablative Stereo (BED10 [70–85 Gy])
Bouchart 2021 [26]ObsBRPC
LAPC
39mFFX (6c) → SBRT 25–30/5 (SIB TVI 50 Gy)NA55.9NAmPFS 15.6 M
NR/R 7/24.1 M
p < 0.001
1 y LC 74.1% (SBRT)24.6 M
NR/R: 18.2/32.3 M
p = 0.02
Simoni 2021 [102]ObsBRPC
LAPC
59Gem-based/FFX → SBRT 25–30/5 (SIB TVI 50 Gy)NA59.4NAmPFS 10.7 M
NR/R 5/13 M
p < 0.001
1 y LC (NR/R) 79.7/85%
2 y LC (NR/R) 60.6/80%
19.1 M
2 y OS (NR/R) 49/72.5%
p = 0.012
Suker 2019 [103]
LAPC-1
Phase IILAPC50FFX → SBRT 40 Gy/5NA12100mDFS
SBRT 11 M
non-SBRT 3 M
mPFS 9 M
1 y PFS 34%
SBRT 20 M
non-SBRT 3 M
SBRT 1 M
non-SBRT 7 M
1 y OS: 64%
Comito 2017 [104]Phase IILAPC45Gem-based → SBRT 45 Gy/60.48NANAmPFS 8 MmFFLP: 26 M
2 y OS 90%
13 M
Zhong 2017 [105]RetrospLAPC7819CRT 50.4 Gy/28NA9.284NANA11.6 M
2 y OS 16.5%
631SBRT 40 Gy/510.89213.9 M
2 y OS 21.7% (p = 0.014) *
Ablative Stereo (BED10 [100 Gy])
Hassanzadeh 2021 [106]RetrospBRPC
LAPC
44FFX or Gem/Nab-paclitaxel or Gem alone → SBRT 50 Gy/543.29.150mDFS 21.3 M
1 y DFS 78%
2 y DFS 37.8%
mPFS 12.4 M
1 y PFS 52.3%
2 y PFS 13.9%
1 y LC: 84.3%
2 y LC: 59.3%
15.7 M
1 y OS 68.2%
2 y OS 37.9%
Chuong 2022 [107]RetrospBRPC
LAPC
62FFXor Gem/Nab-P → SBRT 50 Gy/5NA22.6NAmPFS 20 M
1 y PFS 88.4%
2 y PFS 40%
1 y LC 98.3%
2 y LC 87.7%
23 M
1 y OS 90.2%
2 y OS 45.5%
Parikh 2023 [108]Phase IIBRPC
LAPC
136FFX or Gem/Nab-P → SBRT 50 Gy/519.932.4NA1 y PFS (NR/R):
46%/82%
1 y LC (NR/R):
78%/93
1 y OS (NR/R):
56%/85%
Rudra 2019 [109]RetrospLAPC44BED < 70 Gy: CRT (40–45 Gy/25–28)/SBRT (30–35 Gy/5)NANANA18 M
FFDF 48%
2 y FFLF 57%2 y OS 30%
BED > 70 Gy: CRT (50–67.5 Gy/10–15)/SBRT (40–52 Gy/5)18 M
FFDF 24%
2 y FFLF 77%2 y OS 49%
(p = 0.03)
Tringale 2022 [110]RetrospLAPC30FFX → SBRT 50 Gy/5NANANAmPFS 10.1 M
1 y PFS 39.7%
NA1 y OS 96.4%
Abbreviations: * statistically significant; 5-FU: 5-fluoracil; Adj: adjuvant; BED: biologically effective dose; BRPC: borderline resectable pancreatic cancer; c: cycle; Cap: capecitabine; Cht: chemotherapy; CRT: combined chemoradiotherapy; DFS: disease-free survival; FFDF: freedom from distant failure; FFLF: freedom from local failure; FFLP: freedom from local progression; FFX: FOLFIRINOX; Gem: gemcitabine; GTX: gemcitabine, docetaxel and capecitabine; LAPC: locally advanced pancreatic cancer; LC: local–regional control; m: median; M: months; N: number; NA: not available; NR: non-resected; OS: overall survival; Prospec: prospective; PFS: progression-free survival; R: resected; Rand: randomized; RCP: resectable pancreatic cancer; RFS: recurrence-free survival; RT: radiotherapy; Retrosp: retrospective; SBRT: stereotactic body radiation therapy; Y: year.
Table 3. Main ongoing studies evaluating TNT in PDAC.
Table 3. Main ongoing studies evaluating TNT in PDAC.
Study
NCT
Study DesignRecruitment StatusTypeNTreatmentPrimary Endpoint
Study TreatmentAjd Cht
Bouchart STEREOPAC
NCT05083247
Phase II
Random
RecruitingBRPC2568c mFFX/12c Gem/Nab-PNAR0
DFS
6c mFFX or 9c Gem/Nab-P → SBRT 35–55 Gy/5NA
AGITG Masterplan
ACTRN12619000409178
Phase II
Random
RecruitingRPC
BRPC
LAPC
1206c mFFXYesLC 1 year
6c mFFX → SBRT 40 Gy/5
PANDAS PRODIGE 44
NCT02676349
Phase II
Random
Active
Not recruiting
BRPC130mFFXYesR0 rate
mFFX → CRT 50.4 Gy/28(Cap)
Span-C trial
NCT03505229
Phase IIRecruitingBRPC
LAPC
40Oxaliplatin-based regimen/Gem-based regimen → SBRT 30–45 Gy/5NALocal failure at 12 M
Wei
NCT05851924
Phase IIRecruitingBRPC
LAPC
60FFX →AD-RTNAEFS
TORPEDO
B3002023000168
Phase IIStartingBRPC
LAPC
NAFFX (6c) or Gem-Nab-P (3c)NANA
FXX (2c) + SBRT 40 Gy/5
Abbreviations: ablative dose radiotherapy; Adj: adjuvant; BRPC: borderline resectable pancreatic cancer; c: Cycle; Cht: chemotherapy; DFS: disease-free survival; FFX: FOLFIRINOX; Gem: gemcitabine; LC: loco-regional control; N: number; NA: not available; OS: overall survival; R0: R0 resection rate; Random: randomized; SBRT: stereotactic body radiation therapy.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Saúde-Conde, R.; El Ghali, B.; Navez, J.; Bouchart, C.; Van Laethem, J.-L. Total Neoadjuvant Therapy in Localized Pancreatic Cancer: Is More Better? Cancers 2024, 16, 2423. https://doi.org/10.3390/cancers16132423

AMA Style

Saúde-Conde R, El Ghali B, Navez J, Bouchart C, Van Laethem J-L. Total Neoadjuvant Therapy in Localized Pancreatic Cancer: Is More Better? Cancers. 2024; 16(13):2423. https://doi.org/10.3390/cancers16132423

Chicago/Turabian Style

Saúde-Conde, Rita, Benjelloun El Ghali, Julie Navez, Christelle Bouchart, and Jean-Luc Van Laethem. 2024. "Total Neoadjuvant Therapy in Localized Pancreatic Cancer: Is More Better?" Cancers 16, no. 13: 2423. https://doi.org/10.3390/cancers16132423

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop