Skip to main content

Selection of patients with pancreatic adenocarcinoma who may benefit from radiotherapy

Abstract

Despite combination chemotherapy demonstrating a positive effect on survival, the clinical outcomes of pancreatic adenocarcinoma (PDAC) remain poor. Radiotherapy was previously a component of the curative treatment of PDAC. Advances in imaging and computer sciences have enabled the prescription of higher dosage of radiation focused on tumours with minimal toxicity to normal tissue. However, the role of radiotherapy has not been established in the curative treatment of localized PDAC because of the conflicting results from large prospective trials. Most studies have demonstrated improved locoregional control but no survival benefit from additional chemoradiotherapy (CRT) in addition to chemotherapy for resectable, borderline or locally advanced PDAC. The improved locoregional control enabled by CRT does not cause extended survival because of rapid distant progression in a significant proportion of patients with PDAC. Several single-institute studies of prescribing intensive chemotherapy with modern ablative radiotherapy for locally advanced PDAC have demonstrated extended survival with an acceptable safety profile. In an analysis after long-term follow-up, the PREOPANC study demonstrated a survival benefit from neoadjuvant gemcitabine-based CRT in resected PDAC relative to upfront surgery followed by adjuvant gemcitabine only. These observations indicated that the role of radiotherapy in PDAC should be evaluated in a subgroup of patients without rapid distant progression because systemic therapy for PDAC remains underdeveloped. We reviewed critical imaging, tissue, liquid and clinical biomarkers to differentiate the heterogeneous biologic spectra of patients with PDAC to identify those who may benefit the most from local radiotherapy. Exclusion of patients with localised PDAC who develop distant progression in a short time and undergo extended upfront chemotherapy for over 4 months may enable the identification of a survival benefit of local radiotherapy. Though promising, the effectiveness of biomarkers must be validated in a multi-institutional prospective study of patients with PDAC receiving CRT or not receiving CRT.

Background

No evidence of survival benefit from radiotherapy as curative treatment for PDAC

Pancreatic ductal adenocarcinoma (PDAC) is one of the most severe malignancies among all solid tumours, with a 5-year survival rate of less than 10% [1, 2]. Most patients with PDAC present with locally advanced pancreatic cancer (LAPC) or metastatic disease that is not suitable for resection [3]. Chemotherapy, radiotherapy, and modern targeted, immunologic therapy exhibit limited efficacy in treating PDAC. Therefore, patients with PDAC usually experience rapid recurrence in the form of locally destructive diseases or distant metastasis [4, 5].

The development of combination chemotherapy consisting of (modified) leucovorin calcium (folinic acid), fluorouracil, irinotecan hydrochloride, oxaliplatin (FOLFIRINOX) [6, 7], and gemcitabine plus nab-paclitaxel (GEM-Nab) [8] has resulted in superior tumour response and survival compared with chemotherapy using single- agent GEM or 5-fluorouracil (5FU) in patients with metastatic or unresectable PDAC. Prospective randomized trials have demonstrated the overall survival (OS) benefit of adjuvant chemotherapy using FOLFIRINOX (54.4 vs. 35.0 months, p = 0.003) [9], GEM plus capecitabine (GEM-Cape; 28.0 vs. 25.5 months, p = 0.032) [10], or GEM plus nab-paclitaxel (41.8 vs. 37.7 months, p = 0.009) [11] compared with using single-agent GEM to treat resected PDAC. For borderline resectable PDAC, neoadjuavant chemotherapy achieves a higher R0 resection rate and survival than does upfront surgery [12,13,14]. A meta-analysis of seven trials with 938 patients revealed significantly improved OS using neoadjuvant therapy (29 vs. 19 months, p = 0.001), especially among patients with borderline resectable PDAC (p = 0.004) [15].

Unlike that of chemotherapy for PDAC, the efficacy of radiotherapy as an adjuvant or curative treatment for PDAC is limited. The results of the European Study Group for Pancreatic Cancer-1 (ESPAC-1) trial led to the omission of radiotherapy from most European adjuvant trials involving resectable PDAC [16]. We conducted a prospective randomised study to evaluate chemo-radiotherapy (CRT) with adjuvant 6-month GEM. The results indicated improved local control (loco-regional recurrence rate of GEM vs. GEM-CRT arms: 54.1% vs. 38.4%, p = 0.056) but no survival benefit (median OS of GEM vs. GEM-CRT: 23.5 vs. 21.5 months, p = 0.73 ) from administering additional CRT to patients with curatively resected PDAC [17]. The results of the Radiation Therapy Oncology Group (RTOG) 0848 study evaluating adjuvant CRT in resected PDAC after adjuvant GEM are highly anticipated [18]. However, the impact of RTOG 0848 may be less relevant because FOLFIRINOX and GEM-Cape have become the standard of care for adjuvant chemotherapy [9, 10]. For borderline resectable PDAC, the PREOPANC-1 study [13, 14] demonstrated long-term survival improvement (median OS: 15.7 vs. 14.3 months, p = 0.025; 5-year survival rate: 20.5% vs. 6.5%) with neoadjuvant GEM-based CRT and improved loco-regional control (p = 0.004) compared with adjuvant GEM alone. The ESPAC-5 [19] and A021501 [20] studies have demonstrated extended survival with neoadjuvant chemotherapy especially using FOLFIRINOX in ESPAC-5 (1-year survival rate: 84% vs. 39% for immediate surgery, p = 0.0028). Despite the high R0 resection and pathologic complete remission rate, neoadjuvant radiotherapy was not associated with favourable survival in either study. For LAPC, the LAP07 study [21] identified better local control (46% vs. 32%, p = 0.03) but no survival benefit (11.9 months vs. 13.6 months, p = 0.09) from the addition of CRT after induction GEM. These results conflict with the report from the Eastern Cooperative Oncology Group trial, which indicated a survival benefit from upfront GEM-based CRT compared with GEM alone (11.1 vs. 9.2 months, p = 0.017) [22]. The conflicting results of the randomized studies concerning borderline resectable and locally advanced PDAC imply a narrow therapeutic window associated with radiotherapy.

Reasons of continued evaluation of radiotherapy for curative PDAC treatment

The role of CRT has been questioned because of controversial clinical trial results. However, CRT remains under careful consideration for PDAC for several reasons: First, the survival outcomes of PDAC remain inferior compared to those of other solid tumours. Novel therapeutic options and modern techniques including stereotactic body radiotherapy (SBRT), magnetic resonance (MR) imaging guided radiotherapy and proton therapy enabled highly conformal and tolerable radiation to be given with solutions for respiratory motion and reduced toxicity to the gastrointestinal area [23, 24]. The Massachusetts General Hospital group demonstrated total neoadjuvant therapy with eight cycles of FOLFIRINOX and losartan, an inhibitor of thrombospondin-1 mediated activation of latent tumour growth factor β (TGFβ), followed by a short or long course of modern radiotherapy for 49 patients with LAPC resulted in a high rate of down-staging and R0 resection in 61% of patients, with a median progression-free survival (PFS) and OS of 17.5 and 31.4 months, respectively [25]. Ablative radiotherapy of 75 Gy in 25 fractions was administered to 119 patients with inoperable PDAC following multiagent induction chemotherapy at Memorial Sloan Kettering Cancer Center. The retrospective analysis revealed safe and durable local control with a median OS of 26.8 months [26]. These studies may influence and inspire current standard approaches. Second, the margin positivity rate and locoregional recurrence rate are high in PDAC, despite radical surgery and intensive systemic chemotherapy [9, 10, 27]. A rapid autopsy study indicated that one-third of patients with PDAC die from local destructive disease without widespread distant metastasis [28]. The efficacy of locoregional control and palliation by radiotherapy has been demonstrated in most studies of PDAC. Jolissaint et al. compared the clinical outcomes of patients with PDAC receiving ablative radiotherapy (n = 104) or surgical resection (n = 105). Despite a selection bias favouring the surgical group, the incidence of locoregional recurrence was similar (16% vs. 21%, p = 0.252) [29]. The excellent locoregional outcomes achieved using modern radiotherapy should be integrated into multimodality treatment of PDAC. Third, the survival benefit of CRT has been demonstrated after exclusion of patients with PDAC with early progression. In the PREOPANC study [14], a significant survival benefit was demonstrated for CRT after long term follow-up (p = 0.025). The steep initial slope of the survival curve, representing early progression, starts to bend and clearly separate from that of patients not receiving CRT after a year from diagnosis, indicating a small difference in median survival (1.4months; 15.7 vs. 14.3 months) between the groups; 5- year survival exhibited a 14% difference (20.5% vs. 6.5%). These results are consistent with the general consensus to prescribe CRT after initial systemic treatment. Accordingly, selecting patients with PDAC with low risk of early disease progression is crucial to translate local control using CRT into a survival benefit.

This review highlights the role of biomarkers in predicting patients with PDAC with low risk of early progression and who are thus suitable for being considered for subsequent radiotherapy with or without concomitant chemotherapy. A biomarker is a characteristic that is objectively measured and evaluated as an indicator of normal biological processes, pathogenic processes or pharmacologic responses to therapeutic intervention [30].

Potential biomarkers for identifying patients with PDAC suitable for radiotherapy

Imaging biomarkers

Radiomics, refers to the extraction and analysis of numerous quantitative features from medical images, and it has shown early promise in the analysis of imaging features and in prognostic modeling and outcome analysis [31]. The baseline imaging textural profile of the tumour microenvironment, including vascularity and oxygenation, and tumor heterogeneity was correlated with pathologic and clinical outcomes in resected PDAC (Table 1). Radiomic features derived from textural signals and groupings of pixels of baseline contrast-enhanced computed tomography (CT) in resectable PDAC were demonstrated to predict OS after surgery [32]. The signal intensity multiplied by the contour volume of pancreas was inversely associated with the pathologic lymph node category and correlated with the OS and PFS of patients with resected PDAC [33]. A seven-feature radiomic signature of a contrast-enhanced CT simulation scan could predict locoregional recurrence in patients with PDAC receiving SBRT [34]. Blood perfusion of tumor from CT scans was correlated with fractional tumour cell death in PDAC. The normalised area under the enhancement curve (nAUC) was correlated with OS and response to CRT patients with borderline resectable PDAC and LAPC [35]. These studies demonstrated baseline CT to be a potential tool for predicting the clinical outcomes of PDAC. If further validated, the signature could be used to help select patients who may benefit from neoadjuvant or adjuvant CRT.

Table 1 Studies of potential radiomic biomarkers for PDAC patients considering radiotherapy

CT imaging profiles after upfront chemotherapy for PDAC are associated with clinical outcomes. A more defined interface response of tumor post chemotherapy was associated with prolonged OS among patients with borderline resectable or locally advanced PDAC [36]. Four radiomic features from simulation CT scans were selected to construct a model to predict resectability in LAPC after neoadjuvant CRT [37]. Radiomic signatures indicating the relationship between tumours and key arteries from CT for radiotherapy treatment planning predicted local control, resectability and OS for borderline resectable and locally advanced PDAC cases after systemic chemotherapy [38, 39]. Patients’ longitudinal radiomic data progress throughout treatment (delta-radiomics) were able to help assess treatment response earlier and more reliably [40]. Yamamoto et al. established a logistic growth pattern of PDAC and defined the Local Advancement Index (LAI) to determine eventual primary tumour size and predict the number of metastases; a smaller LAI value indicates a larger metastatic burden. Radiotherapy after induction chemotherapy improved the survival of patients with larger LAI values [41]. The subgroup of patients with PDAC suitable for consolidative CRT after upfront or induction chemotherapy may be differentiated using potential radiomic parameters developed after chemotherapy.

Furthermore, diffusion-weighted MR quantitative metrics after chemotherapy were demonstrated to indicate response of patients with PDAC to chemotherapy [42]. Collagen molecular imaging using selective MR enhancement of fibrosis with CM-101, a type I collagen-targeted probe, revealed a robust fibrotic response after neoadjuvant therapy of FOLFIRINOX and correlated with improved survival in murine model of PDAC receiving CRT [43]. The preoperative uptake value of fluoro-deoxyglucose positron emission tomography (FDG-PET) and metabolic response to neoadjuvant therapy could predict the OS of patients with PDAC [44,45,46,47,48].

The ability of radiomic signatures to provide superior information for evidence-based clinical decision-making regarding PDAC is promising. To select patients who will benefit from radiotherapy, potential radiomic signatures should be explored in prospective clinical trials and validated through expansion of the available dataset, preferably in a multi-institutional study. Standardisation of radiomic signatures and imaging modalities to reduce inter-observer variability is also necessary.

Histopathologic, liquid and clinical biomarkers

Molecular classifications of PDAC based on genomic, transcriptomic, proteomic and epigenetic data have provided considerable insights into the molecular heterogeneity and aggressive biology of PDAC [49]. Several potential biomarkers have been demonstrated to enable differentiation of the failure patterns in patients with PDAC. (Table 2) SMAD4 gene status and expression have been highly correlated with radiosensitivity and the initial failure site of PDAC in clinical and preclinical studies [28, 50, 51]. In a phase II prospective study of 69 patients with LAPC, a local dominant pattern of progression was identified in patients with intact SAMD4 and not in those with SMAD4 loss (73% vs. 28%, p = 0.016) [52]. A retrospective study of 641 patients with resected PDAC demonstrated that inactivated SMAD4 was strongly associated with metastatic recurrence (hazard ratio (HR) = 4.28, 95% CI = 2.75–6.68). Improved survival with additional radiotherapy was observed only in patients with PDAC with SMAD4 expression (p = 0.002). The investigators concluded that patients with SMAD4 expression benefit more from intensive local control [53]. Whittle et al. further demonstrated that heterozygous mutation of SMAD4 attenuated the metastatic potential of PDAC and increased its proliferation. Loss of the heterozygosity of SMAD4 restored metastatic competency and further increased proliferation– a highly lethal combination. The authors further demonstrated that RUNX3 responded to and interacted with SMAD4 status to regulate the balance between cancer cell division and dissemination, and they suggested that RUNX3 and SMAD4 levels can be used together to inform clinical decision-making for resectable PDAC [54]. Krüppel-like factor 10 (KLF10), a TGFβ early-response gene, has been demonstrated by investigators, including us, to contribute to PDAC radiosensitivity, epithelial - mesenchymal transition, and cancer stemness and progression [55,56,57]. We evaluated potential biomarkers including SMAD4, RUNX3 and KLF10 in tumour tissues from 111 patients with resected PDAC randomised to adjuvant GEM with or without CRT [58]. Loss of both SMAD4 and KLF10 expression in patients with curatively resected PDAC was associated with rapid development of distant metastasis; those who expressed either SMAD4 or KLF10 had a significantly higher chances of benefiting from adjuvant CRT (for patients with KLF10 or SMAD4 expression: GEM–CRT vs. GEM: PFS ∞ vs. 19.8 months; p = 0.026; OS 33 vs. 23 months; p = 0.12) [58]. The tryptophan catabolic enzyme, indoleamine 2,3 dioxygenase-2 (IDO2) has been demonstrated to promote pancreatic tumourigenesis in preclinical studies [59]. An IDO2-deficient genotype correlates with improved PFS for patients with PDAC who received adjuvant radiotherapy (39.0 ± 6.3 vs. 74.1 ± 6.4 months, p = 0.023). Analysis of metabolic profiles from patients with resectable PDAC receiving neoadjuvant therapy demonstrated a significant difference in choline metabolism between those responding favourably and unfavourably. Lower levels of choline and phosphocholine correlated with a low recurrence rate among patients with PDAC receiving neoadjuvant CRT [60]. Genomic profiling using targeted gene sequencing for radiotherapy response prediction was evaluated among 88 patients with cancer receiving local tumour irradiation. Alterations of DNA repair pathways and mutations of CHEK2, MSH2 and NOTCH1 were associated with durable local control using radiotherapy [61]. A radiation sensitivity index (RSI) score for intrinsic tumour radiosensitivity derived from the expression of 10 specific genes (HDAC1, PKCb, RelA, c-Abl, STAT1, AR, Cdk1, c-Jun, SUMO1, and IRF1) and a linear regression algorithm modeled on the surviving fraction at 2 Gy (SF2) of 48 cancer cells were evaluated for 73 patients with PDAC receiving surgery with or without radiotherapy. Among high-risk patients, radiotherapy provided significantly improved survival among radio-sensitive patients compared with radio-resistant patients (p = 0.04). This difference was not observed among low-risk patients [62]. The RSI score was combined with the linear quadratic model to derive a genomic-adjusted radiation dose (GARD) by the same group of investigators to identify the optimum radiotherapy dose at a patient-specific molecular signature level. A high GARD value predicted a strong therapeutic effect of radiotherapy and greater time to first recurrence and OS. GARD independently predicted clinical outcomes for pancreatic cancer, and its use enabled the individualization of radiotherapy dose according to the tumour radiosensitivity [63, 64].

Table 2 Studies of potential tissue biomarker for PDAC patients considering radiotherapy

Several peripheral blood biomarkers have been demonstrated to determine survival or therapeutic response in PDAC (Table 3). Absolute monocyte count during CRT and changes in the lymphocyte-to-monocyte ratio correlated with OS and PFS among patients with LAPC treated with CRT [65]. The baseline neutrophil-to-lymphocyte ratio (NLR) and NLR dynamics during neoadjuvant chemotherapy were independently associated with pathologic response in resectable PDAC [66]. Despite not being specific to a cancerous condition and a lack of expression in 5 -10% of patients, CA19-9 is the most used tumour marker for monitoring therapy for PDAC. A decrease in the CA19-9 level after neoadjuvant therapy is correlated with improved OS and pathologic major response in PDAC [67,68,69]. We analyzed CA19-9 change during adjuvant chemotherapy among 125 patients with resected PDAC with or without adjuvant radiation. Significant correlations of CA19-9 response with initial failure at distant sites and OS were identified. However, neither postoperative CA19-9 level nor CA19-9 response were helpful in identifying patients who may experience a survival benefit from additional adjuvant CRT [70]. A retrospective analysis reported that a high level of carcinoembryonic antigen but not CA19-9 before neoadjuvant CRT was the most significant predictor of poor survival after surgery for PDAC [71]. Regarding other circulating biomarkers, baseline CC motif chemokine ligand 5 (CCL5) was identified as an independent prognostic biomarker for OS in patients with LAPC in the Selective Chemoradiation in Advanced Localised Pancreatic Cancer (SCALOP) study, which evaluated induction GEM-Cape and CRT [72]. A correlation between CCL5 levels and failure patterns was not identified. Increasing evidence indicates that microRNAs (miRNAs) may serve as diagnostic, predictive and prognostic biomarkers in various cancer entities, including PDAC. The expression of miRNAs was correlated with pancreatic cancer progression and radio-resistance [73]. A four-miRNA molecular signature (miR-29c, miR-125a, miR-155, and mR-200b) was developed to predict risk of locoregional recurrence and OS after PDAC resection. Using the miRNA risk score has potential for identifying patients with PDAC who are most likely to benefit from postoperative CRT [74]. Circulating tumor DNA (ctDNA) is released into the peripheral blood stream during cell death. The presence of ctDNA in patients with PDAC after neoadjuvant therapy indicates recurrence and poor survival [75, 76]. Circulating tumour cells that enter peripheral blood are thought to contribute to metastatic disease with worse survival [77]. In an analysis of the Surveillance, Epidemiology, and End Results database, patients with PDAC with a tumour location over the pancreatic head, stage II/III cancer, T4 cancer, N1 cancer, regional resection, or lymphadenectomy of ≥ 4 lymph nodes were demonstrated to benefit from adjuvant radiotherapy [78, 79]. Several studies have revealed that a combined analysis of radiomic features, clinical parameters, pathology score, and tissue/serum biomarkers improves the prognostic power of clinical outcomes in PDAC [32, 80].

Table 3 Studies of potential peripheral blood biomarkers for PDAC patients considering radiotherapy

Conclusions

Despite progress in surgical techniques and systemic therapy, the survival outcomes of patients with PDAC remain unsatisfactory. Radiotherapy was a central component of treatment for PDAC. The value of CRT to PDAC has been questioned because of conflicting results of clinical trials. Most studies have been criticised for low patient numbers, poor study design, inappropriate radiation doses or split-course regimens, and poor adherence to the radiation protocol [81,82,83]. However, several prospective trials have demonstrated the efficacy of modern radiation therapy, with an elevated dosage and reduced toxicity to the small bowel, exhibiting a satisfactory safety profile, local control, and prolonged survival for localised PDAC [25, 26]. In addition to the technical improvement of radiotherapy, the development of radiogenomics and the biology of radiotherapy for PDAC may help to optimise the integration of radiotherapy in multimodality PDAC treatment strategies. Because distant metastases are more effectively controlled through modern systemic therapy, local control of the primary site is increasingly critical for patients with PDAC with extended survival [23]. Advances in radiomic, tissue, or peripheral biomarkers may enable superior stratification of patients’ metastatic potential and prediction of those who would most likely benefit from enhanced locoregional therapy. However, studies evaluating the role of potential biomarkers have mostly been retrospective and have demonstrated correlations with survival but not failure patterns. Multi-institutional prospective clinical trials that validate candidate biomarkers in patients with PDAC receiving up-to-date systemic chemotherapy with or without modern radiotherapy are urgently required.

The role of radiotherapy in the curative treatment of PDAC remains unclear. In designing future clinical trials, the exclusion of patients with early distant progression by extended systemic therapy (≥ 4 months) and predictive biomarkers is reasonable. Local control using radiotherapy may yield a survival benefit, especially among patients with PDAC without early distant metastasis.

Data availability

Data sharing not applicable to this article as no datasets were generated or analyzed during the study.

Abbreviations

PDAC:

pancreatic adenocarcinoma

CRT:

chemoradiotherapy

LAPC:

locally advanced pancreatic cancer

FOLFIRINOX:

leucovorin calcium (folinic acid), fluorouracil, irinotecan hydrochloride, oxaliplatin

GEM-Nab:

gemcitabine plus nab-paclitaxel

5FU:

5-fluorouracil

OS:

overall survival

GEM-Cape:

gemcitabine plus capecitabine

SBRT:

stereotactic body radiotherapy

MRI:

magnetic resonance imaging

PFS:

progression-free survival

nAUC:

normalized area under the enhancement curve

LAI:

local advancement Index

FDG-PET:

fluoro-deoxygluocse positron emission tomography

KLF10:

krüppel-like factor 10

IDO2:

indoleamine 2,3 dioxygenase-2

RSI:

radiation sensitivity index

SF2:

surviving fraction at 2 Gy

GARD:

genomic-adjusted radiation dose

LMR:

lymphocyte to monocyte ratio

NLR:

neutrophil to lymphocyte ratio

CCL5:

c-c motif chemokine ligand 5

CTCs:

circulating tumor cells

SEER:

surveillance, epidemiology, and end results

References

  1. Kamisawa T, Wood LD, Itoi T, Takaori K. Pancreatic cancer. Lancet. 2016;388(10039):73–85.

    Article  CAS  PubMed  Google Scholar 

  2. Surveillance Epidemiology and End Results (SEER). (1975–2018). In: National Cancer Institute SRP CSB, editor. 2021.

  3. Maitra A, Hruban RH. Pancreatic cancer. Annu Rev Pathol. 2008;3:157–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Neesse A, Michl P, Frese KK, Feig C, Cook N, Jacobetz MA, et al. Stromal biology and therapy in pancreatic cancer. Gut. 2011;60(6):861–8.

    Article  PubMed  Google Scholar 

  5. Royal RE, Levy C, Turner K, Mathur A, Hughes M, Kammula US, et al. Phase 2 trial of single agent Ipilimumab (anti-CTLA-4) for locally advanced or metastatic pancreatic adenocarcinoma. J Immunother. 2010;33(8):828–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Conroy T, Desseigne F, Ychou M, Bouche O, Guimbaud R, Becouarn Y, et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med. 2011;364(19):1817–25.

    Article  CAS  PubMed  Google Scholar 

  7. Stein SM, James ES, Deng Y, Cong X, Kortmansky JS, Li J, et al. Final analysis of a phase II study of modified FOLFIRINOX in locally advanced and metastatic pancreatic cancer. Br J Cancer. 2016;114(7):737–43.

    Article  PubMed  Google Scholar 

  8. Von Hoff DD, Ervin T, Arena FP, Chiorean EG, Infante J, Moore M, et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med. 2013;369(18):1691–703.

    Article  Google Scholar 

  9. Conroy T, Hammel P, Hebbar M, Ben Abdelghani M, Wei AC, Raoul JL, et al. FOLFIRINOX or Gemcitabine as Adjuvant Therapy for Pancreatic Cancer. N Engl J Med. 2018;379(25):2395–406.

    Article  CAS  PubMed  Google Scholar 

  10. Neoptolemos JP, Palmer DH, Ghaneh P, Psarelli EE, Valle JW, Halloran CM, et al. Comparison of adjuvant gemcitabine and capecitabine with gemcitabine monotherapy in patients with resected pancreatic cancer (ESPAC-4): a multicentre, open-label, randomised, phase 3 trial. Lancet. 2017;389(10073):1011–24.

    Article  CAS  PubMed  Google Scholar 

  11. Tempero MA, Pelzer U, O’Reilly EM, Winter J, Oh DY, Li CP, et al. Adjuvant nab-paclitaxel + gemcitabine in Resected Pancreatic Ductal Adenocarcinoma: results from a randomized, Open-Label, phase III trial. J Clin Oncol. 2023;41(11):2007–19.

    Article  CAS  PubMed  Google Scholar 

  12. Ahmad SA, Duong M, Sohal DPS, Gandhi NS, Beg MS, Wang-Gillam A, et al. Surgical Outcome results from SWOG S1505: a Randomized Clinical Trial of mFOLFIRINOX Versus Gemcitabine/Nab-paclitaxel for Perioperative Treatment of Resectable Pancreatic Ductal Adenocarcinoma. Ann Surg. 2020;272(3):481–6.

    Article  PubMed  Google Scholar 

  13. Versteijne E, Suker M, Groothuis K, Akkermans-Vogelaar JM, Besselink MG, Bonsing BA, 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(16):1763–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Versteijne E, van Dam JL, Suker M, Janssen QP, Groothuis K, Akkermans-Vogelaar JM, 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(11):1220–30.

    Article  CAS  PubMed  Google Scholar 

  15. van Dam JL, Janssen QP, Besselink MG, Homs MYV, van Santvoort HC, van Tienhoven G, et al. Neoadjuvant therapy or upfront surgery for resectable and borderline resectable pancreatic cancer: a meta-analysis of randomised controlled trials. Eur J Cancer. 2022;160:140–9.

    Article  PubMed  Google Scholar 

  16. Neoptolemos JP, Stocken DD, Friess H, Bassi C, Dunn JA, Hickey H, et al. A randomized trial of chemoradiotherapy and chemotherapy after resection of pancreatic cancer. N Engl J Med. 2004;350(12):1200–10.

    Article  CAS  PubMed  Google Scholar 

  17. Chang HJ, Chiu YF, Chen JS, Li CP, Ho CL, Shyr YM et al. Randomized, phase III trial comparing adjuvant gemcitabine (gem) versus gem plus chemoradiation (CCRT) in curatively resected pancreatic ductal adenocarcinoma (PDAC): a Taiwan cooperative oncology group study. Ann Oncol 2018;29(viii210 Gastrointestinal tumors, non-colorectal):626PD.

  18. Tchelebi L, Winter K, Abrams RA, Safran H, Regine JWF, McNulty S, et al. Analysis of Radiation Therapy Quality Assurance in NRG Oncology RTOG 0848. Int J Radiat Oncol Biol Phys. 2021;111(3):76.

    Article  Google Scholar 

  19. Ghaneh P, Palmer D, Cicconi S, Jackson R, Halloran CM, Rawcliffe C, 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(2):157–68.

    Article  PubMed  Google Scholar 

  20. Katz MHG, Shi Q, Meyers J, Herman JM, Chuong M, Wolpin BM, et al. Efficacy of preoperative mFOLFIRINOX vs mFOLFIRINOX Plus Hypofractionated Radiotherapy for Borderline Resectable Adenocarcinoma of the Pancreas: the A021501 phase 2 Randomized Clinical Trial. JAMA Oncol. 2022;8(9):1263–70.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Hammel P, Huguet F, van Laethem JL, Goldstein D, Glimelius B, Artru P, 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(17):1844–53.

    Article  CAS  PubMed  Google Scholar 

  22. Loehrer PJ, Sr., Feng Y, Cardenes H, Wagner L, Brell JM, Cella D, et al. Gemcitabine alone versus gemcitabine plus radiotherapy in patients with locally advanced pancreatic cancer: an Eastern Cooperative Oncology Group trial. J Clin Oncol. 2011;29(31):4105–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Hall WA, Goodman KA. Radiation therapy for pancreatic adenocarcinoma, a treatment option that must be considered in the management of a devastating malignancy. Radiat Oncol. 2019;14(1):114.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Hall WA, Erickson B, Crane CH. Evolving concepts regarding Radiation Therapy for Pancreatic Cancer. Surg Oncol Clin N Am. 2021;30(4):719–30.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Murphy JE, Wo JY, Ryan DP, Clark JW, Jiang W, Yeap BY, 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(7):1020–7.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Reyngold M, O’Reilly EM, Varghese AM, Fiasconaro M, Zinovoy M, Romesser PB, et al. Association of Ablative Radiation Therapy with Survival among patients with inoperable pancreatic Cancer. JAMA Oncol. 2021;7(5):735–8.

    Article  PubMed  Google Scholar 

  27. Uesaka K, Boku N, Fukutomi A, Okamura Y, Konishi M, Matsumoto I, et al. Adjuvant chemotherapy of S-1 versus gemcitabine for resected pancreatic cancer: a phase 3, open-label, randomised, non-inferiority trial (JASPAC 01). Lancet. 2016;388(10041):248–57.

    Article  CAS  PubMed  Google Scholar 

  28. Iacobuzio-Donahue CA, Fu B, Yachida S, Luo M, Abe H, Henderson CM, et al. DPC4 gene status of the primary carcinoma correlates with patterns of failure in patients with pancreatic cancer. J Clin Oncol. 2009;27(11):1806–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Jolissaint JS, Reyngold M, Bassmann J, Seier KP, Gonen M, Varghese AM, et al. Local control and survival after induction chemotherapy and ablative Radiation Versus Resection for pancreatic ductal adenocarcinoma with vascular involvement. Ann Surg. 2021;274(6):894–901.

    Article  PubMed  Google Scholar 

  30. Winter JM, Yeo CJ, Brody JR. Diagnostic, prognostic, and predictive biomarkers in pancreatic cancer. J Surg Oncol. 2013;107(1):15–22.

    Article  PubMed  Google Scholar 

  31. Kumar V, Gu Y, Basu S, Berglund A, Eschrich SA, Schabath MB, et al. Radiomics: the process and the challenges. Magn Reson Imaging. 2012;30(9):1234–48.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Khalvati F, Zhang Y, Baig S, Lobo-Mueller EM, Karanicolas P, Gallinger S, et al. Prognostic value of CT Radiomic features in Resectable Pancreatic Ductal Adenocarcinoma. Sci Rep. 2019;9(1):5449.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Elsherif SB, Javadi S, Le O, Lamba N, Katz MHG, Tamm EP, et al. Baseline CT-based Radiomic features Aid Prediction of nodal positivity after Neoadjuvant Therapy in Pancreatic Cancer. Radiol Imaging Cancer. 2022;4(2):e210068.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Parr E, Du Q, Zhang C, Lin C, Kamal A, McAlister J et al. Radiomics-Based Outcome Prediction for Pancreatic Cancer following stereotactic body Radiotherapy. Cancers (Basel). 2020;12(4).

  35. Wang CX, Elganainy D, Zaid MM, Butner JD, Agrawal A, Nizzero S, et al. Mass Transport Model of Radiation Response: calibration and application to Chemoradiation for Pancreatic Cancer. Int J Radiat Oncol Biol Phys. 2022;114(1):163–72.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Koay EJ, Zaid M, Aliru M, Bagereka P, Van Wieren A, Rodriguez MJ, et al. Nab-Paclitaxel, Capecitabine, and Radiation Therapy after induction chemotherapy in treating patients with locally Advanced and Borderline Resectable Pancreatic Cancer: phase 1 trial and imaging-based Biomarker Validation. Int J Radiat Oncol Biol Phys. 2022;114(3):444–53.

    Article  PubMed  Google Scholar 

  37. Rossi G, Altabella L, Simoni N, Benetti G, Rossi R, Venezia M, et al. Computed tomography-based radiomic to predict resectability in locally advanced pancreatic cancer treated with chemotherapy and radiotherapy. World J Gastrointest Oncol. 2022;14(3):703–15.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Cozzi L, Comito T, Fogliata A, Franzese C, Franceschini D, Bonifacio C, et al. Computed tomography based radiomic signature as predictive of survival and local control after stereotactic body radiation therapy in pancreatic carcinoma. PLoS ONE. 2019;14(1):e0210758.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Cheng Z, Rosati LM, Chen L, Mian OY, Cao Y, Villafania M, et al. Improving prediction of surgical resectability over current staging guidelines in patients with pancreatic cancer who receive stereotactic body radiation therapy. Adv Radiat Oncol. 2018;3(4):601–10.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Nasief H, Hall W, Zheng C, Tsai S, Wang L, Erickson B, et al. Improving treatment response prediction for Chemoradiation Therapy of Pancreatic Cancer using a combination of Delta-Radiomics and the clinical biomarker CA19-9. Front Oncol. 2019;9:1464.

    Article  PubMed  Google Scholar 

  41. Yamamoto KN, Nakamura A, Liu LL, Stein S, Tramontano AC, Kartoun U, et al. Computational modeling of pancreatic cancer patients receiving FOLFIRINOX and gemcitabine-based therapies identifies optimum intervention strategies. PLoS ONE. 2019;14(4):e0215409.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Bali MA, Pullini S, Metens T, Absil J, Chao SL, Marechal R, et al. 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.

    Article  PubMed  Google Scholar 

  43. Erstad DJ, Sojoodi M, Taylor MS, Jordan VC, Farrar CT, Axtell AL, et al. Fibrotic response to Neoadjuvant Therapy predicts Survival in Pancreatic Cancer and is measurable with collagen-targeted molecular MRI. Clin Cancer Res. 2020;26(18):5007–18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Itchins M, Chua TC, Arena J, Jamieson NB, Nahm CB, O’Connell RL, et al. Evaluation of Fluorodeoxyglucose Positron Emission Tomography scanning in the Neoadjuvant Therapy paradigm in pancreatic ductal adenocarcinoma. Pancreas. 2020;49(2):224–9.

    Article  CAS  PubMed  Google Scholar 

  45. Abdelrahman AM, Goenka AH, Alva-Ruiz R, Yonkus JA, Leiting JL, Graham RP, et al. FDG-PET predicts neoadjuvant therapy response and survival in Borderline Resectable/Locally advanced pancreatic adenocarcinoma. J Natl Compr Canc Netw. 2022;20(9):1023–32. e3.

    Article  PubMed  Google Scholar 

  46. Panda A, Garg I, Truty MJ, Kline TL, Johnson MP, Ehman EC, 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(3):730–40.

    Article  PubMed  Google Scholar 

  47. Zimmermann C, Distler M, Jentsch C, Blum S, Folprecht G, Zophel K, et al. Evaluation of response using FDG-PET/CT and diffusion weighted MRI after radiochemotherapy of pancreatic cancer: a non-randomized, monocentric phase II clinical trial-PaCa-DD-041 (Eudra-CT 2009-011968-11). Strahlenther Onkol. 2021;197(1):19–26.

    Article  PubMed  Google Scholar 

  48. Sakane M, Tatsumi M, Hori M, Onishi H, Tsuboyama T, Nakamoto A, et al. Volumetric parameters of 2-deoxy-2-[18F]fluoro-d-glucose positron emission tomography/computed tomography can predict histopathologic treatment response after neoadjuvant chemoradiotherapy in pancreatic adenocarcinoma. Eur J Radiol. 2017;94:64–9.

    Article  PubMed  Google Scholar 

  49. Taherian M, Wang H, Wang H. Pancreatic ductal adenocarcinoma: Molecular Pathology and predictive biomarkers. Cells-Basel. 2022;11:19.

    Google Scholar 

  50. Wang F, Xia X, Yang C, Shen J, Mai J, Kim HC, et al. SMAD4 gene mutation renders pancreatic Cancer Resistance to Radiotherapy through Promotion of Autophagy. Clin Cancer Res. 2018;24(13):3176–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Hu B, Ma X, Huang R, Wu Z, Lu J, Guo Y, et al. Identification of key genes mutations Associated with the radiosensitivity by whole exome sequencing in pancreatic Cancer. Front Oncol. 2021;11:697308.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Crane CH, Varadhachary GR, Yordy JS, Staerkel GA, Javle MM, Safran H, et al. Phase II trial of cetuximab, gemcitabine, and oxaliplatin followed by chemoradiation with cetuximab for locally advanced (T4) pancreatic adenocarcinoma: correlation of Smad4(Dpc4) immunostaining with pattern of disease progression. J Clin Oncol. 2011;29(22):3037–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Shin SH, Kim HJ, Hwang DW, Lee JH, Song KB, Jun E, et al. The DPC4/SMAD4 genetic status determines recurrence patterns and treatment outcomes in resected pancreatic ductal adenocarcinoma: a prospective cohort study. Oncotarget. 2017;8(11):17945–59.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Whittle MC, Izeradjene K, Rani PG, Feng L, Carlson MA, DelGiorno KE, et al. RUNX3 controls a metastatic switch in pancreatic ductal adenocarcinoma. Cell. 2015;161(6):1345–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Tsai YC, Chen SL, Peng SL, Tsai YL, Chang ZM, Chang VH, et al. Upregulating sirtuin 6 ameliorates glycolysis, EMT and distant metastasis of pancreatic adenocarcinoma with kruppel-like factor 10 deficiency. Exp Mol Med. 2021;53(10):1623–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Chang VH, Tsai YC, Tsai YL, Peng SL, Chen SL, Chang TM, et al. Krupple-like factor 10 regulates radio-sensitivity of pancreatic cancer via UV radiation resistance-associated gene. Radiother Oncol. 2017;122(3):476–84.

    Article  CAS  PubMed  Google Scholar 

  57. Chang VH, Chu PY, Peng SL, Mao TL, Shan YS, Hsu CF, et al. Kruppel-like factor 10 expression as a prognostic indicator for pancreatic adenocarcinoma. Am J Pathol. 2012;181(2):423–30.

    Article  CAS  PubMed  Google Scholar 

  58. Pen SL, Shan YS, Hsiao CF, Liu TW, Chen JS, Ho CL, et al. High expression of kruppel-like factor 10 or Smad4 predicts clinical benefit of adjuvant chemoradiotherapy in curatively resected pancreatic adenocarcinoma: from a randomized phase III trial. Radiother Oncol. 2021;158:146–54.

    Article  CAS  PubMed  Google Scholar 

  59. Nevler A, Muller AJ, Sutanto-Ward E, DuHadaway JB, Nagatomo K, Londin E, et al. Host IDO2 gene status influences Tumor Progression and Radiotherapy Response in KRAS-Driven sporadic pancreatic cancers. Clin Cancer Res. 2019;25(2):724–34.

    Article  CAS  PubMed  Google Scholar 

  60. Wada Y, Okano K, Sato K, Sugimoto M, Shimomura A, Nagao M, et al. Tumor metabolic alterations after neoadjuvant chemoradiotherapy predict postoperative recurrence in patients with pancreatic cancer. Jpn J Clin Oncol. 2022;52(8):887–95.

    Article  PubMed  Google Scholar 

  61. Jang BS, Chang JH, Jeon SH, Song MG, Lee KH, Im SA, et al. Radiation Response Prediction Model based on Integrated clinical and genomic data analysis. Cancer Res Treat. 2022;54(2):383–95.

    Article  PubMed  Google Scholar 

  62. Strom T, Hoffe SE, Fulp W, Frakes J, Coppola D, Springett GM, et al. Radiosensitivity index predicts for survival with adjuvant radiation in resectable pancreatic cancer. Radiother Oncol. 2015;117(1):159–64.

    Article  PubMed  PubMed Central  Google Scholar 

  63. Scott JG, Berglund A, Schell MJ, Mihaylov I, Fulp WJ, Yue B, et al. A genome-based model for adjusting radiotherapy dose (GARD): a retrospective, cohort-based study. Lancet Oncol. 2017;18(2):202–11.

    Article  PubMed  Google Scholar 

  64. Scott JG, Sedor G, Ellsworth P, Scarborough JA, Ahmed KA, Oliver DE, et al. Pan-cancer prediction of radiotherapy benefit using genomic-adjusted radiation dose (GARD): a cohort-based pooled analysis. Lancet Oncol. 2021;22(9):1221–9.

    Article  PubMed  Google Scholar 

  65. Giacomelli I, Scartoni D, Mohammadi H, Regine WF, Chuong MD. Does lymphocyte-to-monocyte ratio before, during, or after definitive chemoradiation for locally advanced pancreatic cancer predict for clinical outcomes? J Gastrointest Oncol. 2017;8(4):721–7.

    Article  PubMed  PubMed Central  Google Scholar 

  66. de Castro Silva I, Bianchi A, Deshpande NU, Sharma P, Mehra S, Garrido VT et al. Neutrophil-mediated fibroblast-tumor cell il-6/stat-3 signaling underlies the association between neutrophil-to-lymphocyte ratio dynamics and chemotherapy response in localized pancreatic cancer: a hybrid clinical-preclinical study. Elife. 2022;11.

  67. Heger U, Sun H, Hinz U, Klaiber U, Tanaka M, Liu B, et al. Induction chemotherapy in pancreatic cancer: CA 19 – 9 may predict resectability and survival. HPB (Oxford). 2020;22(2):224–32.

    Article  PubMed  Google Scholar 

  68. Ye C, Sadula A, Ren S, Guo X, Yuan M, Yuan C, et al. 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(6):731–40.

    Article  CAS  PubMed  Google Scholar 

  69. Perri G, Prakash L, Wang H, Bhosale P, Varadhachary GR, Wolff R, et al. Radiographic and Serologic Predictors of pathologic major response to preoperative therapy for pancreatic Cancer. Ann Surg. 2021;273(4):806–13.

    Article  PubMed  Google Scholar 

  70. Chiu YF, Liu TW, Shan YS, Chen JS, Li CP, Ho CL et al. Carbohydrate antigen 19 – 9 response to initial adjuvant chemotherapy predicts survival and failure pattern of resected pancreatic adenocarcinoma but not patients suited for additional adjuvant chemoradiotherapy -- from a prospective randomised study. Int J Radiat Oncol Biol Phys. 2023.

  71. Kato H, Kishiwada M, Hayasaki A, Chipaila J, Maeda K, Noguchi D, et al. Role of serum Carcinoma Embryonic Antigen (CEA) level in localized pancreatic adenocarcinoma: CEA level before operation is a significant Prognostic Indicator in patients with locally advanced pancreatic Cancer treated with Neoadjuvant Therapy followed by Surgical Resection: a retrospective analysis. Ann Surg. 2022;275(5):e698–e707.

    Article  PubMed  Google Scholar 

  72. Willenbrock F, Cox CM, Parkes EE, Wilhelm-Benartzi CS, Abraham AG, Owens R, et al. Circulating biomarkers and outcomes from a randomised phase 2 trial of gemcitabine versus capecitabine-based chemoradiotherapy for pancreatic cancer. Br J Cancer. 2021;124(3):581–6.

    Article  CAS  PubMed  Google Scholar 

  73. Nguyen L, Schilling D, Dobiasch S, Raulefs S, Santiago Franco M, Buschmann D et al. The emerging role of miRNAs for the Radiation treatment of pancreatic Cancer. Cancers (Basel). 2020;12(12).

  74. Wolfe AR, Wald P, Webb A, Sebastian N, Walston S, Robb R, et al. A microRNA-based signature predicts local-regional failure and overall survival after pancreatic cancer resection. Oncotarget. 2020;11(10):913–23.

    Article  PubMed  PubMed Central  Google Scholar 

  75. 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(3):740–8.

    Article  CAS  PubMed  Google Scholar 

  76. Kitahata Y, Kawai M, Hirono S, Okada KI, Miyazawa M, Motobayashi H, et al. Circulating tumor DNA as a potential prognostic marker in patients with Borderline-Resectable Pancreatic Cancer undergoing Neoadjuvant Chemotherapy followed by pancreatectomy. Ann Surg Oncol. 2022;29(3):1596–605.

    Article  PubMed  Google Scholar 

  77. Wang Y, Yu X, Hartmann D, Zhou J. Circulating tumor cells in peripheral blood of pancreatic cancer patients and their prognostic role: a systematic review and meta-analysis. HPB (Oxford). 2020;22(5):660–9.

    Article  PubMed  Google Scholar 

  78. Wang D, Ge H, Tian M, Li C, Zhao L, Pei Q, et al. The Survival Effect of Radiotherapy on Stage IIB/III pancreatic Cancer undergone surgery in different age and Tumor Site Groups: a propensity scores matching analysis based on SEER Database. Front Oncol. 2022;12:799930.

    Article  PubMed  PubMed Central  Google Scholar 

  79. Luo Y. The role of radiotherapy for pancreatic malignancies: a population-based analysis of the SEER database. Clin Transl Oncol. 2022;24(1):76–83.

    Article  CAS  PubMed  Google Scholar 

  80. Attiyeh MA, Chakraborty J, Doussot A, Langdon-Embry L, Mainarich S, Gonen M, et al. Survival prediction in pancreatic ductal adenocarcinoma by quantitative computed tomography image analysis. Ann Surg Oncol. 2018;25(4):1034–42.

    Article  PubMed  PubMed Central  Google Scholar 

  81. Choti MA. Adjuvant therapy for pancreatic cancer–the debate continues. N Engl J Med. 2004;350(12):1249–51.

    Article  CAS  PubMed  Google Scholar 

  82. Koshy MC, Landry JC, Cavanaugh SX, Fuller CD, Willett CG, Abrams RA, et al. A challenge to the therapeutic nihilism of ESPAC-1. Int J Radiat Oncol Biol Phys. 2005;61(4):965–6.

    Article  PubMed  Google Scholar 

  83. Abrams RA, Winter KA, Regine WF, Safran H, Hoffman JP, Lustig R, et al. Failure to adhere to protocol specified radiation therapy guidelines was associated with decreased survival in RTOG 9704–a phase III trial of adjuvant chemotherapy and chemoradiotherapy for patients with resected adenocarcinoma of the pancreas. Int J Radiat Oncol Biol Phys. 2012;82(2):809–16.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by the grant NHRI CA-110 ~ 111-PP-14 from National Health Research Institutes, Taiwan.

Author information

Authors and Affiliations

Authors

Contributions

I.S.J reviewed the manuscript, performed research and collected data. H.J.C. conceived and designed the review; performed research and collected data; and wrote the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Hui Ju Ch’ang.

Ethics declarations

Competing interests

The authors declare no competing interests.

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Jan, IS., Ch’ang, H.J. Selection of patients with pancreatic adenocarcinoma who may benefit from radiotherapy. Radiat Oncol 18, 137 (2023). https://doi.org/10.1186/s13014-023-02328-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13014-023-02328-y

Keywords