Next Article in Journal
Incisional Hernia Following Open Pancreaticoduodenectomy: Incidence and Risk Factors at a Tertiary Care Centre
Previous Article in Journal
Are Canadian Women Prepared for the Transition to Primary HPV Testing in Cervical Screening? A National Survey of Knowledge, Attitudes, and Beliefs
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Can STEreotactic Body Radiation Therapy (SBRT) Improve the Prognosis of Unresectable Locally Advanced Pancreatic Cancer? Long-Term Clinical Outcomes, Toxicity and Prognostic Factors on 142 Patients (STEP Study)

by
Tiziana Comito
1,
Maria Massaro
1,*,
Maria Ausilia Teriaca
1,
Ciro Franzese
1,2,
Davide Franceschini
1,
Pierina Navarria
1,
Elena Clerici
1,
Luciana Di Cristina
1,
Anna Bertolini
1,
Stefano Tomatis
1,
Giacomo Reggiori
1,
Andrea Bresolin
1,
Silvia Bozzarelli
3,
Lorenza Rimassa
2,3,
Cristiana Bonifacio
4,
Silvia Carrara
5,
Armando Santoro
2,3,
Alessandro Zerbi
2,6 and
Marta Scorsetti
1,2
1
Radiotherapy and Radiosurgery Department, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, 20089 Milan, Italy
2
Department of Biomedical Sciences, Humanitas University, 20072 Milan, Italy
3
Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, 20089 Milan, Italy
4
Department of Radiology, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, 20089 Milan, Italy
5
Department of Gastroenterology, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, 20089 Milan, Italy
6
Department of Pancreatic Surgery, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, 20089 Milan, Italy
*
Author to whom correspondence should be addressed.
Curr. Oncol. 2023, 30(7), 7073-7088; https://doi.org/10.3390/curroncol30070513
Submission received: 31 May 2023 / Revised: 17 July 2023 / Accepted: 21 July 2023 / Published: 24 July 2023
(This article belongs to the Topic Advances in Radiotherapy and Prognosis)

Abstract

:
Aim: The gold standard of care for pancreatic adenocarcinoma is the integrated treatment of surgery and chemotherapy (ChT), but about 50% of patients present with unresectable disease. Our study evaluated the efficacy in terms of local control, survival and safety of stereotactic body radiation therapy (SBRT) in locally advanced pancreatic cancer (LAPC). Methods: A retrospective study (STEP study) analyzed patients with LAPC treated with a dose of 45 Gy in 6 fractions. Local control (LC), distant progression free survival (DPFS), overall survival (OS) and toxicity were analyzed according to the Kaplan-Meier method. Results: A total of 142 patients were evaluated. Seventy-six patients (53.5%) received induction ChT before SBRT. The median follow-up was 11 months. One-, 2- and 3-year LC rate was 81.9%, 69.1% and 58.5%. Median DPFS was 6.03 months; 1- and 2-year DPFS rate was 19.9% and 4.5%. Median OS was 11.6 months and 1-, 2- and 3-year OS rates were 45.4%, 16.1%, and 9.8%. At univariate analysis, performed by the log-rank test, age < 70 years (p = 0.037), pre-SBRT ChT (p = 0.004) and post-SBRT ChT (p = 0.019) were associated with better OS. No patients experienced G3 toxicity. Conclusion: SBRT represents an effective and safe therapeutic option in the multimodal treatment of patients with LAPC in terms of increased LC. When SBRT was sequentially integrated with ChT, the treatment proved to be promising in terms of OS as well.

1. Introduction

Pancreatic cancer (PC) has a poor prognosis, with a 5-year overall survival (OS) rate of about 10% [1]. Currently, PC represents the fourth leading cause of cancer-related deaths in Western countries [1,2] and it is expected to become the second leading cause of cancer-related deaths by 2030 [3].
Although surgery is the only potentially curative treatment, more than 50% of patients are diagnosed with unresectable PC caused by vascular involvement without distant metastasis, which is defined as locally advanced pancreatic cancer (LAPC) [4].
Pancreatic ductal adenocarcinoma (PDAC) is typically considered a systemic disease, as its natural history is dominated by the development of metastases. Therefore, chemotherapy (ChT) is the standard of care for LAPC, according to current guidelines [5]. At the same time, local progression significantly contributes to morbidity and mortality. Therefore, about a third of patients with PC die from complications related to local tumor progression in the absence of metastases, according to an analysis of autopsy series by Johns Hopkins Medical Institution [6]. Furthermore, Crane et al. showed that local tumor progression is the most common cause of death in patients alive after more than 16 months from diagnosis [7].
According to this evidence, the integration of chemotherapy and radiation treatment could represent a valid therapeutic option in the management of LAPC. Nevertheless, some retrospective studies have suggested that induction chemotherapy administered before concurrent chemoradiotherapy could improve the survival of patients with LAPC [8,9].
Conventional fractionated radiation therapy (CFRT) has been shown to improve local pain control and to maintain the stability of disease progression. Nevertheless, these clinical benefits of ChT-RT in terms of local pain control have been limited by the long courses of therapy, the significant toxicity caused by a lack of dose homogeneity and the concurrent approach, and the decreasing use of full-dose systemic therapy with ChT-RT [10].
In the last few years, stereotactic body radiation therapy (SBRT) has emerged as a viable alternative to ChT-RT in the treatment of LAPC. SBRT is a radiation technique that allows the delivery of an ablative dose to the target lesion and a minimal dose to adjacent critical structures in a short treatment time, with higher local control and an optimal toxicity profile, without affecting the sequential integration of full-dose systemic therapy.
However, the optimal radiation treatment modality and, particularly, the optimal radiation treatment timing for LAPC remain debated.
Among the reasons for these open questions, we should include the characteristics of the studies currently available. Therefore, the literature about SBRT in unresectable PC encompasses series composed of a limited number of patients treated with nonhomogeneous prescription RT doses, fractionations and chemotherapy regimens. According to major prospective and retrospective studies published between 2013 and 2021 (Table 1 and Table 2), which analyzed an average of 40 patients (range 11–63 patients), the median 1y-LC was 81%, and the median OS and 1y-OS rate were 13.8 months and 59%, respectively.
Parallel to the outcome data, the first prospective studies about SBRT in LAPC, which evaluated the treatment delivered in a single fraction [25,26,27], recorded a significant gastrointestinal toxicity rate.
Thereby, in our study, we have collected and analyzed data related to the delivery of SBRT in a large number of unresectable patients treated with homogeneous dose prescription, fractionation scheme, and RT technique, reporting the long-term clinical outcomes and toxicity.

2. Materials and Methods

2.1. Study Population

Unresectable patients with LAPC were identified from a prospectively collected mono-center database started with a prospective phase 2 study, which provided the 6-fraction SBRT in PC [14].
This retrospective monocentric study was approved by the Humanitas Ethical Review Board (n° 33/22_14 June 2022).
All procedures performed were in accordance with the ethical standards of the Institution and with the 1964 Helsinki Declaration. Informed consent was obtained from all treated patients. All patients were discussed in a multidisciplinary tumor board composed of a medical oncologist, a biliary–pancreatic surgeon, a radiologist, a gastroenterologist, a pathologist and a radiation oncologist.
The inclusion criteria were:
  • Age ≥ 18 years old
  • Minimum Karnofsky Performance Status of 70%
  • Histologically and/or radiologically proven locally advanced unresectable PC
  • Tumor diameter < 5 cm
  • Absence of nodal and metastatic disease
The exclusion criteria included:
  • Other malignancies diagnosed within 5 years
  • Previous abdominal SBRT
  • Gastric or duodenal obstruction
  • Nodal metastases
  • Metastatic disease
  • Concomitant ChT
Unresectable LAPC was defined according to the American Hepato–Pancreato–Biliary Association/Society of Surgical Oncology/Society for Surgery of the Alimentary Tract criteria [28].

2.2. Stereotactic Body Radiation Therapy

During the simulation phase, all patients were immobilized in a supine position with the arms above the head, using a thermoplastic body mask. A contrast-free and a triphasic contrast-enhanced computed tomography (CT) scan with a slice thickness of 3 mm was acquired for all patients. The 4-dimensional CT (4D-CT) imaging was performed in patients with respiratory excursions greater than 5 mm.
The Gross Tumor Volume (GTV) was defined by macroscopic disease. In selected patients, the simulation CT images were co-registered with the magnetic resonance images (MRI) and/or fluorodeoxyglucose-positron emission tomography (FDG-PET CT) to better identify the GTV. No elective lymph node irradiation was performed. The Clinical Target Volume (CTV), defined as GTV with no additional margins, was delineated on the venous phase of the CT scan.
In all patients who underwent a 4D-CT scan, an internal target volume (ITV) was defined as the volume of the CTV delineated by the breathing phases acquired during the 4D CT.
Another 6 mm isotropic expansion was generated from the ITV to obtain the Planning Target Volume (PTV).
The critical structures (organs at risk, OaR), including the stomach, duodenum, small bowel, kidneys, liver and spinal cord, were delineated.
Dose prescription was 45 Gy in 6 consecutive fractions (BED10 = 78.75 Gy) and the treatment was delivered with volumetric modulated arc therapy (VMAT) by RapidArc tecnique.
A required target coverage of D98% > 98% for the CTV. A maximum acceptable dose heterogeneity to the PTV of D98% > 95% and D2% < 107%.
Dose-volume constraints for the OaR were: spinal cord D1cc < 27 Gy, kidneys V15Gy < 30%, duodenum V36Gy < 1 cc, stomach V36Gy < 1 cc, small bowel V36Gy < 3 cc, liver (Vwhole liver–V21Gy) > 700 cc.
In the area of overlap between PTV and OaR, we delivered the maximum tolerated dose by OaR to minimize the dose inhomogeneity on PTV according to dose-volume constraints, as shown in Figure 1.
To reduce interfraction variability, all patients kept a fast of at least 3 h before treatment.
Before every treatment session, the setup of patients was verified daily by image guidance with Cone Beam CT (CBCT). Regarding patients’ setup, after an initial adjustment based on bone anatomy, the setup was then adjusted to match soft tissue structures, such as main blood vessels.

2.3. Response Evaluation

Patients were reassessed 8 weeks after SBRT and then every 3 months. Clinical evaluation, blood exams with CEA and CA 19-9, a contrast-enhanced CT scan were performed; patients undergoing staging FDG-PET CT before SBRT have performed a re-staging FDG-PET CT 4–6 months after treatment.
Response assessment was performed according to CA19-9 levels, Response Evaluation Criteria in Solid Tumors v1.1 (RECIST) [29], and PET Response Criteria in Solid Tumors (PERCIST) [30]. According to RECIST criteria, a Complete Response (CR) was defined as the disappearance of the target lesion; a Partial Response (PR) was defined as a decrease in at least 30% in the sum of the diameter of the target lesion, taking as reference the baseline sum diameter; a Progressive Disease (PD) was defined as an increase in at least 20% in the sum of the diameter of the target lesion, taking as reference the smallest sum on the study; Stable Disease (SD) was defined as less than 30% reduction in the tumor major diameter or less than 20% increase in tumor major diameter.
An example of dose distribution and radiological response after SBRT is reported in Figure 2.
We also evaluated the clinical response in patients presenting with pain before radiation therapy according to the Numerical Rating Scale (NRS) scoring system.
The toxicity profile was assessed according to the National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE) v5.0 [31].
Acute toxicity was defined as toxicity described within 90 days of the end of SBRT, while late toxicity was defined as occurring beyond 90 days.

2.4. Statistical Analysis

We performed the statistical analysis with the software STATA 14. The primary endpoint was local control (LC), which was defined as the time interval between the end of the SBRT and the first evidence of local progression (in-field), and patients were censored from the date of death. Secondary endpoints were distant progression free survival (DPFS), overall survival (OS), and toxicity profile. DPFS was defined as the time interval between the end of SBRT and the first finding of a distant progression (out-of-field). Overall survival (OS) was calculated from the end of the SBRT to the patient’s death or the last follow-up.
Local control, DPFS and OS were calculated according to the Kaplan-Meier method; the log-rank test was used for the univariate analysis to evaluate a correlation with prognostic factors such as age, tumor diameter, CTV, PTV, time between diagnosis and SBRT, ChT before SBRT and ChT after SBRT, and pre-SBRT and post-SBRT CA 19-9 values. A multivariable analysis of the endpoints with prognostic factors such as age, ChT before SBRT and ChT after SBRT was performed by Cox regression. A p value < 0.05 was considered statistically significant.

3. Results

Between January 2011 and June 2021, 142 patients with LAPC were treated in a single institution. Demographic, clinical, and treatment characteristics are shown in Table 3.
The median age was 71 years (41–91). Sixty-three (44%) patients were younger than 70 years and 79 (66%) patients were 70 years, of age or older than 70 years.
The histological diagnosis was not available in 15 patients (10.5%), who were very elderly and frail and not candidates for chemotherapy.
In 124 patients (87.3%), the CA 19.9 value was available before SBRT, with a median range of 102.75 U/mL (0.8–12.000). In 79 patients (63.7%), the CA 19.9 value was <300 U/mL, in 45 patients (36.3%), it was ≥300 U/mL, and their changes were not statistically significant.
Most of the tumors were located in the head (91 lesions, 64.1%), then in uncinate process (23 lesions, 16.2%), then in body (20 lesions,14.1%) and the others were located in tail (3, 2,1%) and isthmus (5, 3.5%).
Seventy-six patients (53.5%) underwent induction ChT that ended at least 2 weeks prior to SBRT. The number of ChT cycles ranged from 3 to 10. This variability was related to performance status (PS), age, comorbidities, patient compliance, tumor response, and toxicity. In 55 patients (72%), gemcitabine-based ChT was administered: 7 gemcitabine monotherapy, 21 gemcitabine oxaliplatin (GEMOX), 10 PEX-G (cisplatin, epirubicin, 5-fluorouracil, gemcitabine), 17 gemcitabine-nab paclitaxel, whereas 18 patients (23.7%) received folinic acid, 5-fluorouracil, irinotecan and oxaliplatin (FOLFIRINOX). In 42 patients (29.6%), ChT was also administered after SBRT. The median follow-up (mfup) was 11 months (range: 6–55 months). No patient had to discontinue radiation treatment.
Twelve patients (8.4%) were alive at the time of the analysis, and seven of them (5%) had no evidence of local or metastatic disease. After SBRT, four patients (3%) underwent surgery with negative margins.
Based on the NRS scoring system, 45 (31.6%) patients presented with pain before SBRT. In 14 (9.8%) patients, pain control after radiation therapy allowed the suspension of analgesics treatment; in 10 (7.04%) patients, analgesics were reduced.

3.1. Local Control

One-, 2-, and 3-year LC rates were 81.9% (95% CI 73.1–88.1), 69.1% (95% CI 54.7–79.8), and 58.5% (95% CI 39.5–73.4), respectively (Figure 3a). The median LC was not reached. According to RECIST criteria (Table 4), we observed one patient with a complete response (0.7%), 31 partial response cases (22%), 81 patients showed disease stability (57%), and 29 (20.3%) had a local progression. Pre-SBRT and post-SBRT FDG-PET CT were performed in 17 patients (12%); in three patients (2.1%) with stable disease on CT scan, FDG-PET CT showed a partial metabolic response, while in five patients (3.5%), with an uncertain persistence of disease on CT scan, FDG-PET CT reported a complete metabolic response, which was also confirmed at subsequent restaging CT scans. Among patients with local progression, seven patients (24.1%) presented isolated local progression of disease without distant metastases, and they underwent retreatment after a gap of one to three years from the end of SBRT. The radiation dose delivered was 20 or 25 or 35 Gy in five fractions, and RT was well tolerated in all retreated patients.

3.2. Distant Progression Free Survival

The median DPFS was 6.03 months. Eighty-four patients (59.1%) developed distant metastases. The sites of progression were the liver (31, 37%), peritoneum (13, 15.5%), lung (8, 9.5%), lymph nodes (4, 4.7%), bone (2, 2.3%) and other multiple sites (26, 31%). One- and 2-year DPFS rates were 19.9% (95% CI 13.4–27.2) and 4.5% (95% CI 1.6–9.7%), respectively (Figure 3b).
At progression, 42 patients (29.6%) underwent ChT. In cases of oligoprogression, RT was performed on the site of metastatic disease (14 patients, 10%): 10 patients underwent SBRT for liver metastasis (4, 3%), lung lesions (4, 3%) and lymph nodes (2, 1%). Four patients (3%) underwent symptomatic RT treatment for bone metastases.

3.3. Overall Survival

Median OS (mOS) was 11.6 months, and the 1-, 2- and 3-year OS rates were 45.4% (95% CI 37.0–53.5), 16.1% (95% CI 10.5–22.9), and 9.8% (95% CI 5.4–15.7), respectively (Figure 3c). At the univariate analysis (Table 5), age < 70 years (HR 1.46, 95% CI 1.02-2.08, p = 0.037), pre-SBRT ChT (HR 0.59, 95% CI 0.41–0.84, p = 0.004) and post-SBRT ChT (HR 0.63, 95% CI 0.43–0.92, p = 0.019) were significantly associated with OS. Specifically, mOS in patients treated with induction ChT was 14.3 months, compared to 8.9 months in patients treated with exclusive SBRT (Figure 4).
In multivariable analysis, we did not find statistically significant results. (Table S1, Supplementary Files).
Before SBRT, 124 patients (87.3%) presented higher CA 19-9 values, and the analysis was repeated 8 weeks after the end of SBRT. We found stability of CA 19-9 levels in 57 patients (46%), a reduction in 49 patients (39.5%), of whom 15 patients (12%) had a decline to normal values, and an increase in marker levels in 18 patients (14.5%) who have presented local or systemic progression of disease.

3.4. Toxicity

Sixty-six patients (46.5%) experienced mild or moderate acute toxicity: nausea G1 in 23 patients (35%) and G2 in 14 patients (21.3%). Fifteen patients (22.7%) have reported epigastric pain (G1-G2), treated with minor analgesic drugs. There was no acute and/or late G3 toxicity.
Three patients developed ulceration of the duodenal mucosa (G2) after 8, 13, and 21 months of SBRT. In two of these patients, we observed a concurrent pathological infiltration from primary pancreatic disease.

4. Discussion

The role of SBRT in the treatment of unresectable pancreatic cancer was investigated and introduced in the therapeutic flowchart as a promising treatment option [5,32,33].
Nevertheless, most of these experiences have been performed on small cohorts of patients with heterogeneous characteristics, in terms of clinical features, fractionation RT schemes, and chemotherapy regimens.
The first prospective studies about SBRT in LAPC evaluated the feasibility of the single fraction [25,26,27]. A promising local control of disease was achieved at the price of significant gastrointestinal toxicity and poor overall survival, so the current studies are directed to a fractionated RT scheme in order to maintain local control and reduce the risk of toxicity to adjacent normal tissues.
Currently, there is not a consensus about the best dose-for-fraction regimen; the available literature recommends the scheme of 5 fractions with a minimum total dose of 33 Gy [13,18,33,34], 35 Gy [35], and 40 Gy [15,36,37,38,39].
In our analysis, SBRT at a total dose of 45 Gy delivered in 6 fractions showed an effective local control rate of 69.1% at two years. Considering the mOS of 14.3 months when SBRT was sequentially integrated with ChT, our results suggested that about 70% of patients treated with SBRT will be able to spend their remaining lives without relevant symptoms related to both local disease progression, such as pain, jaundice, sick and difficult duodenal transit, or radiotherapy related toxicity. All patients, indeed, tolerated radiation therapy without several complications.
Considering the clinical outcomes and toxicity profiles of the main prospective and retrospective studies published between 2013 and 2021 (Table 1 and Table 2), an average of 40 patients (range 11–63 patients) were enrolled in the study, and the most commonly used fractionation regimen was that of 5 fractions with a median total dose of 37.5 Gy. The median 1 y-LC is comparable to that obtained in our analysis (81% vs 81.9%). The median-OS and 1 y-OS rate were lower in our study (13.8 months vs. 11.6 months and 59% vs. 45.4%, respectively). This disparity may refer to the unfavorable selection of patients for pancreatic SBRT. In our study, we analyzed a large cohort of 142 patients with unresectable LAPC, and 79 of them were elderly, with an average age ≥ 70 years. This data were consequently correlated to the lower rate of patients (53.5%) treated with induction ChT before the SBRT, unlike the six out of seven prospective studies previously cited, which included induction ChT in the therapeutic program for all analyzed patients. To confirm the role of induction ChT in terms of improved survival, in our study we achieved a statistically significant correlation between induction ChT administered before SBRT and OS (p = 0.004) and between ChT after SBRT and OS (p = 0.019). These results suggested the crucial role of the patients’ selection and the timing of multimodality treatment in LAPC.
Among the retrospective studies, we mentioned the experiences of the Department of Radiation Oncology in Tampa (Florida) and Baltimore (Maryland) [19]. They performed SBRT in patients affected by BRPC and LAPC after ChT with an RT regimen delivered in five consecutive daily fractions with median total radiation doses of 30 Gy to the tumor and a 40 Gy dose painted to tumor-vessel interfaces. Patients affected by LAPC were 49. The estimated median OS was 15.0 months for LAPC patients. Five of 21 (24%) LAPC patients receiving FOLFIRINOX ChT underwent R0 resection. For those not undergoing resection, one-year locoregional control was 78%, consistent with our local control outcome (81.9%).
Further confirmation of these suggestions may come from a prospective Phase II study involving induction chemotherapy and then SBRT for patients with LAPC (ClinicalTrials.gov Identifier: NCT03158779).
Another crucial issue related to SBRT in pancreatic cancer is the toxicity profile. In 2015, Brunner et al. [40] reviewed the literature on pancreatic SBRT published from 2004 to 2013 and showed that late toxicity G2 and G3 correlated highly with equivalent radiation dose and survival. This review confirmed that late toxicity ≥ G2 was substantial and predominantly gastrointestinal, reflecting the use of partly inadequate dose constraints for the duodenum and gastrointestinal tract organs. Since then, main prospective and retrospective published studies, as reported in Table 1 and Table 2, showed acute ≥ G3 and late ≥ G3 gastrointestinal toxicity less than 10%, according to the results of a published systematic review and pooled analysis [41].
Our report confirmed these results, with a G2 late toxicity rate <1% and no cases of ≥ G3 toxicity. These data were related, on the one hand, to the priority we give to dose constraints for OaRs in the treatment planning phase (Figure 1) and, on the other hand, to the accuracy of treatment delivery with daily CBCT.
Regarding the role of FDG PET-CT, although previous studies have demonstrated its prognostic value in other cancers, its role in PC is not yet clear. In 2014, the team of Herman and Koong analyzed the prognostic utility of FDG PET-CT in patients with LAPC treated with SBRT. In the analysis of 32 patients undergoing induction ChT and SBRT (33 Gy/5 fr), pre-SBRT tumor metabolic volume (p = 0.029) and total lesion glycolysis (p = 0.038), defined as tumor metabolic volume multiplied by the mean SUV, are potential predictive factors for OS in patients with LAPC [42]. In our study, the prognostic value of FDG PET-CT was not confirmed, but this data may have been conditioned by the limited number of patients (12%) subjected to this procedure during the simulation phase.
Summarizing the available literature about SBRT in unresectable PC, a wide variety of prescription RT doses, fractionations, and chemotherapy regimens were proven. The value of our study is based on the large number of analyzed patients, all treated with homogeneous dose prescription, fractionation scheme and RT technique. Therefore, the results of our study therefore reflect, compared to the data in the literature, our strengths, including above all a high and homogeneous dose. The limits of our analysis are represented by the retrospective nature, the heterogeneity in ChT regimes administered before or after SBRT and unfavorable patients’ selection, particularly in terms of age and comorbidities, which adversely affected the use of an integrated treatment of ChT and RT in about 50% of analyzed cases.
Despite this, in these elderly and frail patients, excluded from ChT and otherwise candidates for best supportive care only, SBRT has proven to be a valid therapeutic option in terms of LC, toxicity, and survival.

5. Conclusions

SBRT might represent an effective and safe therapeutic option in the multimodal treatment of patients affected by unresectable LAPC. Local control was sustained, and overall survival has proven to be superior when SBRT was sequentially integrated with chemotherapy. Given the retrospective nature of the study, future prospective studies are needed to identify the best treatment timing and therapeutic strategy in combination with systemic therapy to increase the survival of these patients.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/curroncol30070513/s1, Table S1: Multivariable Analysis.

Author Contributions

Conceptualization, T.C. and M.S.; methodology, T.C.; validation, S.B., L.R., C.B., S.C., A.S., A.Z. and M.S.; formal analysis, C.F.; investigation, T.C.; data curation, T.C.; writing—original draft preparation, T.C. and M.M.; visualization, L.D.C., A.B. (Anna Bertolini), M.A.T., D.F., E.C., S.T. and A.B. (Andrea Bresolin); supervision, P.N. and G.R.; project administration, M.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki and approved by the Institutional Review Board (or Ethics Committee) of IRCCS Humanitas Research Hospital (n° 33/22 and date of approval: 14 June 2022).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

Not applicable.

Conflicts of Interest

LR reports consulting fees from AstraZeneca, Basilea, Bayer, BMS, Eisai, Exelixis, Genenta, Hengrui, Incyte, Ipsen, IQVIA, Lilly, MSD, Nerviano Medical Sciences, Roche, Servier, Taiho Oncology, Zymeworks; lecture fees from AstraZeneca, Bayer, Eisai, Gilead, Incyte, Ipsen, Merck Serono, Roche, Sanofi, Servier; travel expenses from AstraZeneca; research grants (to Institution) from Agios, AstraZeneca, BeiGene, Eisai, Exelixis, Fibrogen, Incyte, Ipsen, Lilly, MSD, Nerviano Medical Sciences, Roche, Zymeworks.

References

  1. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer statistics. CA Cancer J. Clin. 2021, 71, 7–33. [Google Scholar] [CrossRef]
  2. Carioli, G.; Bertuccio, P.; Boffetta, P.; Levi, F.; La Vecchia, C.; Negri, E.; Malvezzi, M. European cancer mortality predictions for the year 2020 with a focus on prostate cancer. Ann. Oncol. 2020, 31, 650–658. [Google Scholar] [CrossRef] [PubMed]
  3. Rahib, L.; Smith, B.D.; Aizenberg, R.; Rosenzweig, A.B.; Fleshman, J.M.; Matrisian, L.M. Projecting cancer incidence and deaths to 2030: The unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 2014, 74, 2913–2921. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Mizrahi, J.D.; Surana, R.; Valle, J.W.; Shroff, R.T. Pancreatic cancer. Lancet 2020, 395, 2008–2020. [Google Scholar] [CrossRef] [PubMed]
  5. Balaban, E.P.; Mangu, P.B.; Khorana, A.A.; Shah, M.A.; Mukherjee, S.; Crane, C.H.; Javle, M.M.; Eads, J.R.; Allen, P.; Ko, A.H.; et al. Locally Advanced, Unresectable Pancreatic Cancer: American Society of Clinical Oncology Clinical Practice Guideline. J. Clin. Oncol. 2016, 34, 2654–2668. [Google Scholar] [CrossRef]
  6. Iacobuzio-Donahue, C.A.; Fu, B.; Yachida, S.; Luo, M.; Abe, H.; Henderson, C.M.; Vilardell, F.; Wang, Z.; Keller, J.W.; Banerjee, P.; et al. DPC4 Gene Status of the Primary Carcinoma Correlates with Patterns of Failure in Patients with Pancreatic Cancer. J. Clin. Oncol. 2009, 27, 1806–1813. [Google Scholar] [CrossRef] [Green Version]
  7. Crane, C.H.; Varadhachary, G.R.; Yordy, J.S.; Staerkel, G.A.; Javle, M.M.; Safran, H.; Haque, W.; Hobbs, B.D.; Krishnan, S.; Fleming, J.B.; 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, 3037–3043. [Google Scholar] [CrossRef] [Green Version]
  8. Huguet, F.; André, T.; Hammel, P.; Artru, P.; Balosso, J.; Selle, F.; Deniaud-Alexandre, E.; Ruszniewski, P.; Touboul, E.; Labianca, R.; et al. Impact of chemotherapy in locally advanced pancreatic adenocarcinoma in GERCOR phase II and III studies. J. Clin. Oncol. 2007, 25, 326–331. [Google Scholar] [CrossRef] [Green Version]
  9. Krishnan, S.; Rana, V.; Janjan, N.A.; Varadhachary, G.R.; Abbruzzese, J.L.; Das, P.; Delclos, M.E.; Gould, M.S.; Evans, D.B.; Wolff, R.A.; et al. Induction chemotherapy selects patients with locally advanced, unresectable pancreatic cancer for optimal benefit from consolidative chemoradiation therapy. Cancer 2007, 110, 47–55. [Google Scholar] [CrossRef]
  10. Herman, J.M.; Koong, A. Stereotactic Body Radiation Therapy: A New Standard Option for Pancreatic Cancer? J. Natl. Compr. Cancer Netw. 2014, 12, 1489–1493. [Google Scholar] [CrossRef] [Green Version]
  11. Gurka, M.K.; Collins, S.P.; Slack, R.; Tse, G.; Charabaty, A.; Ley, L.; Berzcel, L.; Lei, S.; Suy, S.; Haddad, N.; et al. Stereotactic body radiation therapy with concurrent full-dose gemcitabine for locally advanced pancreatic cancer: A pilot trial demonstrating safety. Radiat. Oncol. 2013, 8, 44. [Google Scholar] [CrossRef] [Green Version]
  12. Tozzi, A.; Comito, T.; Alongi, F.; Navarria, P.; Iftode, C.; Mancosu, P.; Reggiori, G.; Clerici, E.; Rimassa, L.; Zerbi, A.; et al. SBRT in unresectable advanced pancreatic cancer: Preliminary results of a mono-institutional experience. Radiat. Oncol. 2013, 8, 148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. 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.; Ba, J.S.P.; et al. Phase 2 multi-institutional trial evaluating gemcitabine and stereotactic body radiotherapy for patients with locally advanced unresectable pancreatic adenocarcinoma. Cancer 2014, 121, 1128–1137. [Google Scholar] [CrossRef] [PubMed]
  14. 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]
  15. Teriaca, M.; Loi, M.; Suker, M.; Eskens, F.; van Eijck, C.; Nuyttens, J. A phase II study of stereotactic radiotherapy after FOLFIRINOX for locally advanced pancreatic cancer (LAPC-1 trial): Long-term outcome. Radiother. Oncol. 2020, 155, 232–236. [Google Scholar] [CrossRef]
  16. Chen-Zhao, X.; Hernando, O.; López, M.; Sánchez, E.; Montero, A.; García-Aranda, M.; Ciérvide, R.; Valero, J.; Alonso, R.; Cárdenas-Rebollo, J.M.; et al. A prospective observational study of the clinical and pathological impact of stereotactic body radiotherapy (SBRT) as a neoadjuvant strategy of chemoradiation in pancreatic cancer. Clin. Transl. Oncol. 2020, 22, 1499–1505. [Google Scholar] [CrossRef]
  17. Zhu, X.; Cao, Y.; Lu, M.; Zhao, X.; Jiang, L.; Ye, Y.; Ju, X.; Zhang, H. Stereotactic body radiation therapy with sequential S-1 for patients with locally advanced pancreatic cancer and poor performance status: An open-label, single-arm, phase 2 trial. Radiother. Oncol. 2021, 162, 178–184. [Google Scholar] [CrossRef]
  18. 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] [Green Version]
  19. 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]
  20. Gurka, M.K.; Kim, C.B.; He, A.R.; Charabaty, A.; Haddad, N.; Turocy, J.B.; 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]
  21. Zhu, X.; Li, F.; Ju, X.; Cao, F.; Cao, Y.; Fang, F.; Qing, S.; Shen, Y.; Jia, Z.; Zhang, H. Prognostic role of stereotactic body radiation therapy for elderly patients with advanced and medically inoperable pancreatic cancer. Cancer Med. 2017, 6, 2263–2270. [Google Scholar] [CrossRef] [PubMed]
  22. Mazzola, R.; Fersino, S.; Aiello, D.; Gregucci, F.; Tebano, U.; Corradini, S.; Di Paola, G.; Cirillo, M.; Tondulli, L.; Ruffo, G.; et al. Linac-based stereotactic body radiation therapy for unresectable locally advanced pancreatic cancer: Risk-adapted dose prescription and image-guided delivery. Strahlenther. Onkol. 2018, 194, 835–842. [Google Scholar] [CrossRef]
  23. Toesca, D.A.; Ahmed, F.; Kashyap, M.; Baclay, J.R.M.; von Eyben, R.; Pollom, E.L.; Koong, A.C.; Chang, D.T. Intensified systemic therapy and stereotactic ablative radiotherapy dose for patients with unresectable pancreatic adenocarcinoma. Radiother. Oncol. 2020, 152, 63–69. [Google Scholar] [CrossRef] [PubMed]
  24. Shen, Z.-T.; Zhou, H.; Li, A.-M.; Ji, X.-Q.; Jiang, C.-C.; Yuan, X.; Li, B.; Zhu, X.-X.; Huang, G.-C. Clinical outcomes and prognostic factors of stereotactic body radiation therapy combined with gemcitabine plus capecitabine for locally advanced unresectable pancreatic cancer. J. Cancer Res. Clin. Oncol. 2019, 146, 417–428. [Google Scholar] [CrossRef] [PubMed]
  25. Koong, A.C.; Le, Q.T.; Ho, A.; Fong, B.; Fisher, G.; Cho, C.; Ford, J.; Poen, J.; Gibbs, I.C.; Mehta, V.K.; et al. Phase I study of stereotactic radiosurgery in patients with locally advanced pancreatic cancer. Int. J. Radiat. Oncol. 2004, 58, 1017–1021. [Google Scholar] [CrossRef]
  26. Schellenberg, D.; Goodman, K.A.; Lee, F.; Chang, S.; Kuo, T.; Ford, J.M.; Fisher, G.A.; Quon, A.; Desser, T.S.; Norton, J.; et al. Gemcitabine Chemotherapy and Single-Fraction Stereotactic Body Radiotherapy for Locally Advanced Pancreatic Cancer. Int. J. Radiat. Oncol. 2008, 72, 678–686. [Google Scholar] [CrossRef] [PubMed]
  27. Schellenberg, D.; Kim, J.; Christman-Skieller, C.; Chun, C.L.; Columbo, L.A.; Ford, J.M.; Fisher, G.A.; Kunz, P.L.; Van Dam, J.; Quon, A.; et al. Single-Fraction Stereotactic Body Radiation Therapy and Sequential Gemcitabine for the Treatment of Locally Advanced Pancreatic Cancer. Int. J. Radiat. Oncol. 2011, 81, 181–188. [Google Scholar] [CrossRef]
  28. Callery, M.P.; Chang, K.J.; Fishman, E.K.; Talamonti, M.S.; Traverso, L.W.; Linehan, D.C. Pretreatment Assessment of Resectable and Borderline Resectable Pancreatic Cancer: Expert Consensus Statement. Ann. Surg. Oncol. 2009, 16, 1727–1733. [Google Scholar] [CrossRef]
  29. Eisenhauer, E.A.; Therasse, P.; Bogaerts, J.; Schwartz, L.H.; Sargent, D.; Ford, R.; Dancey, J.; Arbuck, S.; Gwyther, S.; Mooney, M.; et al. New response evaluation criteria in solid tumours: Revised RECIST guideline (version 1.1). Eur. J. Cancer 2009, 45, 228–247. [Google Scholar] [CrossRef]
  30. Wahl, R.L.; Jacene, H.; Kasamon, Y.; Lodge, M.A. From RECIST to PERCIST: Evolving Considerations for PET Response Criteria in Solid Tumors. J. Nucl. Med. 2009, 50 (Suppl. 1), 122S–150S. [Google Scholar] [CrossRef] [Green Version]
  31. Common Terminology Criteria for Adverse Events (CTCAE) Version 5; US Department of Health and Human Services, National Institutes of Health, National Cancer Institute: Frederick, MD, USA, 2017.
  32. National Comprehensive Cancer Network, NCCN Guidelines, Version 2.2021, Pancreatic Adenocarcinoma. Available online: www.nccn.org/professionals/physician_gls/pdf/pancreatic.pdf (accessed on 30 May 2023).
  33. Palta, M.; Godfrey, D.; Goodman, K.A.; Hoffe, S.; Dawson, L.A.; Dessert, D.; Hall, W.A.; Herman, J.M.; Khorana, A.A.; Merchant, N.; et al. Radiation Therapy for Pancreatic Cancer: Executive Summary of an ASTRO Clinical Practice Guideline. Pract. Radiat. Oncol. 2019, 9, 322–332. [Google Scholar] [CrossRef]
  34. Pollom, E.L.; Alagappan, M.; von Eyben, R.; Kunz, P.L.; Fisher, G.A.; Ford, J.A.; Poultsides, G.A.; Visser, B.C.; Norton, J.A.; Kamaya, A.; et al. Single- versus Multifraction Stereotactic Body Radiation Therapy for Pancreatic Adenocarcinoma: Outcomes and Toxicity. Int. J. Radiat. Oncol. Biol. Phys. 2014, 90, 918–925. [Google Scholar] [CrossRef]
  35. Zhu, X.; Ju, X.; Cao, Y.; Shen, Y.; Cao, F.; Qing, S.; Fang, F.; Jia, Z.; Zhang, H. Patterns of Local Failure after Stereotactic Body Radiation Therapy and Sequential Chemotherapy as Initial Treatment for Pancreatic Cancer: Implications of Target Volume Design. Int. J. Radiat. Oncol. Biol. Phys. 2019, 104, 101–110. [Google Scholar] [CrossRef] [PubMed]
  36. Oar, A.; Lee, M.; Le, H.; Hruby, G.; Dalfsen, R.; Pryor, D.; Lee, D.; Chu, J.; Holloway, L.; Briggs, A.; et al. Australasian Gastrointestinal Trials Group (AGITG) and Trans-Tasman Radiation Oncology Group (TROG) Guidelines for Pancreatic Stereotactic Body Radiation Therapy (SBRT). Pract. Radiat. Oncol. 2019, 10, e136–e146. [Google Scholar] [CrossRef] [PubMed]
  37. Bernard, V.; Herman, J.M. Pancreas SBRT: Who, What, When, Where, and How. Pract. Radiat. Oncol. 2020, 10, 183–185. [Google Scholar] [CrossRef] [PubMed]
  38. Colbert, L.; Rebueno, N.; Moningi, S.; Beddar, S.; Sawakuchi, G.O.; Herman, J.M.; Koong, A.C.; Das, P.; Holliday, E.B.; Koay, E.J.; et al. Dose escalation for locally advanced pancreatic cancer: How high can we go? Adv. Radiat. Oncol. 2018, 3, 693–700. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Mazzarotto, R.; Simoni, N.; Guariglia, S.; Rossi, G.; Micera, R.; De Robertis, R.; Pierelli, A.; Zivelonghi, E.; Malleo, G.; Paiella, S.; et al. Dosimetric Feasibility Study of Dose Escalated Stereotactic Body Radiation Therapy (SBRT) in Locally Advanced Pancreatic Cancer (LAPC) Patients: It Is Time to Raise the Bar. Front. Oncol. 2020, 10, 940. [Google Scholar] [CrossRef]
  40. Brunner, T.B.; Nestle, U.; Grosu, A.-L.; Partridge, M. SBRT in pancreatic cancer: What is the therapeutic window? Radiother. Oncol. 2015, 114, 109–116. [Google Scholar] [CrossRef] [PubMed]
  41. 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]
  42. Dholakia, A.S.; Chaudhry, M.; Leal, J.P.; Chang, D.T.; Raman, S.P.; Hacker-Prietz, A.; Su, Z.; Pai, J.; Oteiza, K.E.; Griffith, M.E.; et al. Baseline Metabolic Tumor Volume and Total Lesion Glycolysis Are Associated with Survival Outcomes in Patients with Locally Advanced Pancreatic Cancer Receiving Stereotactic Body Radiation Therapy. Int. J. Radiat. Oncol. Biol. Phys. 2014, 89, 539–546. [Google Scholar] [CrossRef] [Green Version]
Figure 1. A prescription dose of 45 Gy in 6 fractions was delivered with a maximum acceptable dose heterogeneity to the PTV of D98% > 95% and D2% < 107%. In the area of overlap between PTV and OaR, we delivered the maximum tolerated dose by OaR. PRV: Planning Organs at Risk Volume; OAR: Organ at Risk; PTV = Planning Target Volume; ITV: Internal Target Volume; CTV = Clinical Target Volume.
Figure 1. A prescription dose of 45 Gy in 6 fractions was delivered with a maximum acceptable dose heterogeneity to the PTV of D98% > 95% and D2% < 107%. In the area of overlap between PTV and OaR, we delivered the maximum tolerated dose by OaR. PRV: Planning Organs at Risk Volume; OAR: Organ at Risk; PTV = Planning Target Volume; ITV: Internal Target Volume; CTV = Clinical Target Volume.
Curroncol 30 00513 g001
Figure 2. Example of dose distribution and radiological response after SBRT.
Figure 2. Example of dose distribution and radiological response after SBRT.
Curroncol 30 00513 g002
Figure 3. (a) Local control; (b) distant progression free survival; and (c) overall survival.
Figure 3. (a) Local control; (b) distant progression free survival; and (c) overall survival.
Curroncol 30 00513 g003aCurroncol 30 00513 g003b
Figure 4. Overall survival in patients treated with chemotherapy and SBRT (n = 76) and in patients treated with SBRT alone (n = 66). Specifically, pre-SBRT ChT (HR 0.59, 95% CI 0.41–0.84, p = 0.004) and post-SBRT ChT (HR 0.63, 95% CI 0.43–0.92, p = 0.019) were significantly associated with OS.
Figure 4. Overall survival in patients treated with chemotherapy and SBRT (n = 76) and in patients treated with SBRT alone (n = 66). Specifically, pre-SBRT ChT (HR 0.59, 95% CI 0.41–0.84, p = 0.004) and post-SBRT ChT (HR 0.63, 95% CI 0.43–0.92, p = 0.019) were significantly associated with OS.
Curroncol 30 00513 g004
Table 1. Prospective studies of SBRT in patients with BRPC/LAPC from 2013 to 2021.
Table 1. Prospective studies of SBRT in patients with BRPC/LAPC from 2013 to 2021.
AuthorPatientsClinical StageSBRT (Dose/Fraction)Pre-, Post-SBRT TherapymFU (Months)LCPFSOSGastrointestinal Toxicity ≥ G3 (%)
Gurka et al., 2013 [11]11LAPC25 Gy/5 frGemcitabine pre + post-SBRTNaNamPFS: 6.8 monthsmOS: 12.2 monthsAcute ≥ G3: 0%
Late ≥ G3: 0%
Tozzi et al., 2013 [12]30LAPC
Local Relapse
45 Gy/6 frMultiple ChT schemes pre + post-SBRT111y: 77%
2y: 75%
mPFS: 8 monthsmOS: 11 months
1y: 47%
Acute ≥ G3: 0%
Late ≥ G3: 0%
Herman et al., 2015 [13]49LAPC33 Gy/5 frGemcitabine pre + post-SBRT13.91y: 78%mPFS: 7.8 months
1y: 32%
2y: 10%
mOS: 13.9 months
1y: 59%
2y: 18%
Acute G3: 10.2%; G4: 2%
Late G3: 6.4%; G4: 2%
Comito et al., 2017 [14]45LAPC45 Gy/6 frMultiple ChT schemes
(71% pre-SBRT and 41% post-SBRT)
13.5mLC: 26 months
1y: 87%
2y: 87%
mPFS: 8 months
1y: 39%
2y: 15%
mOS: 13 months
1y: 59%
2y: 18%
Acute ≥ G3: 0%
Late ≥ G3: 0%
Teriaca et al., 2021 [15]39LAPC40 Gy/5 frFOLFIRINOX pre-SBRT (8 cycles) + surgery post-SBRT (14%)13mLC: 36.3 months
1y: 81%
3y: 53%
mPFS: 10.7 months
1y: 43%
3y: 15%
mOS: 18 months
1y: 77%
3y: 13%
Acute G3: 10.2%
Chen Zhao et al., 2020 [16]45LAPC (20)
BRPC (25)
40–62 Gy/5–10 frMultiple ChT schemes pre-SBRT + surgery post-SBRT (71.1%)14.71y: 95%1y: 72%
2y: 58%
mOS: 13.8 months
1y: 67%
2y: 36%
Acute ≥ G3: 0%
Late ≥ G3: 0%
Zhu et al., 2021 [17]63LAPC35-40Gy/5 frS-1 (6 cycles) post-SBRT15.8NamPFS: 10.1 months14.4 monthsAcute G3: 14.3%
Late G3: 4.8%
Na: not available.
Table 2. Retrospective studies of SBRT in patients with BRPC/LAPC from 2013 to 2021.
Table 2. Retrospective studies of SBRT in patients with BRPC/LAPC from 2013 to 2021.
AuthorPatientsClinical StageSBRT (Dose/Fraction)Pre-, Post-SBRT TherapymFU (Months)LCPFSOSGastrointestinal Toxicity ≥ G3 (%)
Moningi et al., 2015 [18]88LAPC (74)
BRPC (14)
25–33 Gy/5 frMultiple ChT schemes pre-SBRT (87.5%)13.1mLC: 13.9 months
1y: 61%
2y: 14%
mPFS: 9.9 months
1y: 41%
2y: 11%
mOS: 18.4 months
1y: 73%
2y: 24%
Acute G3: 3.4%
Late G3: 5.7%
Mellon et al., 2015 [19]159LAPC (49)
BRPC (110)
20–50 Gy/5 frMultiple ChT schemes pre-SBRT + surgery post-SBRT (38%)14mLC: 12.7 months
1y: 78%
NamOS: 18.1 monthsAcute ≥ G3: 7%
Late ≥ G3: 7%
Gurka et al., 2017 [20]38LAPC (28)
BRPC (6)
Unresectable for clinical comorbidities (4)
25–30 Gy/5 frMultiple ChT schemes pre-SBRTNaNamPFS: 9.2 monthsmOS: 14.3 monthsAcute G3: 5.2%
Late G3: 5.2%; G4: 2.6%; G5: 2.6%
Zhu et al., 2017 [21]417LAPC (218)
BRPC (105)
Metastatic (94)
30–46.8 Gy/5–8 frMultiple ChT schemes pre-SBRT + post-SBRT (11.2%)11mLC: 10 months
1y: 26.6%
mPFS: 8 months
1y: 18.2%
mOS: 10 months
1y: 35.5%
Acute G4: 0.5%
Late G3: 0%
Mazzola et al., 2018 [22]33LAPC42-45 Gy/6 frMultiple ChT schemes pre-SBRT (72%) + post-SBRT (60%) and surgery post-SBRT (18%)181y: 81%Na1y: 75%Acute ≥ G3: 0%
Late ≥ G3: 0%
Toesca et al., 2020 [23]149LAPC20–45Gy/3-6 frMultiple ChT schemes pre-SBRT151y: 86%Na16 monthsAcute ≥ G3: 13%
Late ≥ G3: 0%
Shen et al., 2020 [24]56LAPC40 Gy (30–50 Gy)/5 frGEM-CAP (2 cycles) and concurrent to RT (4 cycles)171y: 85%mPFS: 12 months
1y: 48.2%
2y: 14.3%
mOS: 19 months
1y: 82.1%
2y: 35.7%
Acute G3: 10.8%
Late G3: 5.4%
Na: not available.
Table 3. Patients’ and treatment’s characteristics (n = 142).
Table 3. Patients’ and treatment’s characteristics (n = 142).
Median age (range)71 years (41–91)
 <70 years 63 patients
 ≥70 years79 patients
Tumor location (number of patients, %)
 Head91 (64.1%)
 Uncinate process23 (16.2%)
 Body20 (14.1%)
 Tail3 (2.1%)
 Isthmus5 (3.5%)
CA19.9 before SBRT 124 (87.3%)
 <300 U/mL79 (63.7%)
 ≥300 U/mL45 (36.3%)
 Not available18 (12.7%)
 Median (range) [U/mL]102.75 (0.8–12.000)
CA19.9 after SBRT
Median value (range) [U/mL]35 (0.4–8594)
Median diameter (cm, range)3.7 (1.4–9.3)
Median volume (cc, range)
 CTV 31.6 (2.75–187)
 PTV71.3 (17.6–321)
Chemotherapy before SBRT
 Yes76 (53.5%)
 No66 (46.5%)
Chemotherapy scheme before SBRT
 Gemcitabine 7 (9.2%)
 FOLFIRINOX18 (23.7%)
 Gemcitabine + nab-paclitaxel17 (22.3%)
 GEMOX21 (27.6%)
 PEX-G10 (13.2%)
 Others3 (4%)
Chemotherapy after SBRT
 Yes42 (29.6%)
 No100 (70.4%)
Chemotherapy scheme after SBRT
 Capecitabine-based5 (12%)
 FOLFIRINOX6 (14.3%)
 Gemcitabine-based22 (52.4%)
 Irinotecan2 (4.7%)
 Others7 (16.6%)
FOLFIRINOX: folinic acid, fluorouracil, irinotecan, oxaliplatin; GEMOX: gemcitabine, oxaliplatin; PEX-G: cisplatin, epirubicin, 5-fluorouracil, gemcitabine.
Table 4. Response to treatment according to RECIST criteria.
Table 4. Response to treatment according to RECIST criteria.
Tumor ResponseNumber of Patients (%)
Complete Response1 (0.7%)
Partial Response31 (22%)
Stable Disease81 (57%)
Progression Disease29 (20.3%)
Table 5. Univariate analysis.
Table 5. Univariate analysis.
Local ControlOverall Survival
HR95%CIp ValueHR95%CIp Value
Age > 70 years1.110.53–2.320.7761.461.02–2.080.037
Tumor Diameter0.980.95–1.010.3461.000.99–1.020.400
CTV Volume0.990.98–1.010.8811.000.99–1.010.163
PTV Volume0.990.99–1.000.8651.000.99–1.000.075
Time from diagnosis to SBRT0.970.91–1.040.4870.980.96–1.010.460
CA19-9 before SBRT1.000.99–1.000.7401.000.99–1.000.417
CA19-9 after SBRT------
ChT before SBRT0.850.40–1.790.6750.590.41–0.840.004
ChT after SBRT1.710.81–3.570.1550.630.43–0.920.019
Local progression---0.900.60–1.370.655
Distant progression1.100.26–4.700.8900.890.45–1.770.758
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

Comito, T.; Massaro, M.; Teriaca, M.A.; Franzese, C.; Franceschini, D.; Navarria, P.; Clerici, E.; Di Cristina, L.; Bertolini, A.; Tomatis, S.; et al. Can STEreotactic Body Radiation Therapy (SBRT) Improve the Prognosis of Unresectable Locally Advanced Pancreatic Cancer? Long-Term Clinical Outcomes, Toxicity and Prognostic Factors on 142 Patients (STEP Study). Curr. Oncol. 2023, 30, 7073-7088. https://doi.org/10.3390/curroncol30070513

AMA Style

Comito T, Massaro M, Teriaca MA, Franzese C, Franceschini D, Navarria P, Clerici E, Di Cristina L, Bertolini A, Tomatis S, et al. Can STEreotactic Body Radiation Therapy (SBRT) Improve the Prognosis of Unresectable Locally Advanced Pancreatic Cancer? Long-Term Clinical Outcomes, Toxicity and Prognostic Factors on 142 Patients (STEP Study). Current Oncology. 2023; 30(7):7073-7088. https://doi.org/10.3390/curroncol30070513

Chicago/Turabian Style

Comito, Tiziana, Maria Massaro, Maria Ausilia Teriaca, Ciro Franzese, Davide Franceschini, Pierina Navarria, Elena Clerici, Luciana Di Cristina, Anna Bertolini, Stefano Tomatis, and et al. 2023. "Can STEreotactic Body Radiation Therapy (SBRT) Improve the Prognosis of Unresectable Locally Advanced Pancreatic Cancer? Long-Term Clinical Outcomes, Toxicity and Prognostic Factors on 142 Patients (STEP Study)" Current Oncology 30, no. 7: 7073-7088. https://doi.org/10.3390/curroncol30070513

Article Metrics

Back to TopTop