Next Article in Journal
Numerical Simulation on Thermoelectric Cooling of Core Power Devices in Air Conditioning
Next Article in Special Issue
First Study of a HEXITEC Detector for Secondary Particle Characterisation during Proton Beam Therapy
Previous Article in Journal
A Machine-Learning-Inspired Opinion Extraction Mechanism for Classifying Customer Reviews on Social Media
Previous Article in Special Issue
Characterization of Ultra-High-Dose Rate Electron Beams with ElectronFlash Linac
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

CyberKnife Ultra-Hypofractionated SBRT for Localized Prostate Cancer with Dose Escalation to the Dominant Intraprostatic Lesion: In Silico Planning Study

by
Giovanni Carlo Mazzola
1,2,†,
Maria Giulia Vincini
1,†,
Elena Rondi
3,
Giuseppe Ronci
3,
Sabrina Vigorito
3,
Mattia Zaffaroni
1,*,
Giulia Corrao
1,
Salvatore Gallo
4,
Dario Zerini
1,
Stefano Durante
1,
Francesco Alessandro Mistretta
5,
Stefano Luzzago
2,5,
Matteo Ferro
5,
Andrea Vavassori
1,
Federica Cattani
3,
Gennaro Musi
2,5,
Ottavio De Cobelli
2,5,
Giuseppe Petralia
2,6,
Roberto Orecchia
7,
Giulia Marvaso
1,2 and
Barbara Alicja Jereczek-Fossa
1,2
add Show full author list remove Hide full author list
1
Division of Radiation Oncology, European Institute of Oncology (IEO) IRCCS, 20141 Milan, Italy
2
Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
3
Unit of Medical Physics, European Institute of Oncology (IEO) IRCCS, 20141 Milan, Italy
4
Department of Physics “Aldo Pontremoli”, University of Milan, 20133 Milan, Italy
5
Division of Urology, European Institute of Oncology (IEO) IRCCS, 20141 Milan, Italy
6
Precision Imaging and Research Unit, Department of Medical Imaging and Radiation Sciences, European Institute of Oncology (IEO) IRCCS, 20141 Milan, Italy
7
Scientific Directorate, European Institute of Oncology (IEO) IRCCS, 20141 Milan, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Appl. Sci. 2023, 13(12), 7273; https://doi.org/10.3390/app13127273
Submission received: 4 May 2023 / Revised: 12 June 2023 / Accepted: 13 June 2023 / Published: 19 June 2023
(This article belongs to the Special Issue Medical Physics: Latest Advances and Prospects)

Abstract

:
The aim is to evaluate the feasibility of ultra-hypofractionated (UH) SBRT with CyberKnife® (CK) radiosurgery (Accuray Inc., Sunnyvale, California, USA) for localized prostate cancer (PCa) with a concomitant focal boost to the dominant intraprostatic lesion (DIL). Patients with intermediate/high-risk PCa, with at least one visible DIL on multi-parametric MRI, were included. For each, two CK-SBRT in silico plans were calculated using 95% and 85% isodose lines (CK-95%, CK-85%) and compared with the UH-DWA plan delivered with VERO®. All plans simulated a SIB prescription of 40 Gy to PTV-DIL and 36.25 Gy to the whole prostate (PTV-prostate) in five fractions every other day. Fifteen patients were considered. All plans reached the primary planning goal (D95% > 95%) and compliance with organs at risk (OARs) constraints. DVH metrics median values increased (p < 0.05) from UH-DWA to CK-85%. The conformity index of PTV-DIL was 1.00 for all techniques, while for PTV-prostate was 0.978, 0.984, and 0.991 for UH-DWA, CK-95%, and CK-85%, respectively. The CK-85% plans were able to reach a maximum dose of 47 Gy to the DIL while respecting OARs constraints. CK-SBRT plus a focal boost to the DIL for localized PCa appears to be feasible. These encouraging dosimetric results are to be confirmed in upcoming clinical trials such as the phase-II “PRO-SPEED” IEO trial.

1. Introduction

Prostate cancer (PCa) represents the second most common malignancy in men worldwide, with the highest prevalence in developed countries [1]. Patients affected by clinically localized PCa with an intermediate- and high-risk disease, according to National Comprehensive Cancer Network (NCCN) guidelines, can be considered eligible for several local treatment modalities, including external-beam radiotherapy (EBRT) or brachytherapy ± androgen deprivation therapy (ADT) and surgery [2].
Exploiting the radiobiological rationale of the low PCa a/b ratio, resulting in enhanced sensitivity of PCa to higher doses per fraction [3], several trials and metanalysis reported comparable rates of biochemical control, progression-free survival (PFS), overall survival (OS), and toxicity of moderate hypofractionated radiation therapy (RT) respect to conventional fractionation [4,5,6,7,8,9,10,11,12,13].
Given these premises, two non-inferiority Randomized Controlled trials (RCTs) explored the role of ultra-hypofractionated RT (UH-RT) (defined as more or equal to 6.25 Gy/fraction) compared to conventional RT. The HYPO-RT-PC trial confirmed, with 5 years of follow-up, the same profiles of failure-free survival and genitourinary (GU)/gastrointestinal (GI) toxicity for both UH-RT and standard fractionation [14]. Recently published 12 months of GU and GI toxicity profile results from the PACE-B trial corroborate the non-inferiority of UH-RT with respect to conventional and moderate hypofractionated RT [15].
Considering these findings, UH-RT represents a well-established treatment option for localized PCa in high-expertise centers, as suggested by the NCCN guidelines in 2023 [2].
Because PCa local recurrences after RT often originates from the primary tumor site [16,17], a focal boost to the dominant intraprostatic lesion (DIL), identified through multi-parametric magnetic resonance imaging (mpMRI), has been proposed in order to increase biochemical disease-free survival (bDFS) in intermediate-to-high risk PCa patients.
This hypothesis has been confirmed by the results of the FLAME trial [18], which showed that, in conventional fractionation schemes, a focal boost on the DIL improves oncological outcomes without any relevant additional toxicity. Furthermore, as seen from the initial results of the SPARC Trial [19], dose-escalation to the DIL could increase the probability of an undetectable level of prostate-specific antigen (PSA) (<0.1 ng/mL) at least in the first 18 months after treatment in patients receiving ADT.
In our center at the European Institute of Oncology (IEO) IRCCS, Milan, Italy, a high level of expertise has been achieved in the setting of PCa UH-RT, counting more than one thousand patients treated with very low treatment-related toxicities reported [20]. In addition, data for patients receiving UH-RT and simultaneous integrated boost (SIB) on the DIL at IEO has already been published, demonstrating the safety and effectiveness of this treatment [20,21,22].
Several SBRT techniques for UH-RT administration have been explored in order to increase patients’ compliance and safety. Interestingly, the recently published update at 2 years of the PACE-B trial [23] showed that CyberKnife (CK)-SBRT has significantly reduced Common Terminology Criteria for Adverse Events (CTCAE) GU toxicity than SBRT with conventional linacs. CK-SBRT radiosurgery has been successfully used to treat the whole prostate up to 40 Gy, with excellent rates of bDFS while maintaining low rectal and genitourinary toxicities [24,25,26,27,28,29].
Given the above, CK-SBRT could represent the “winning horse” in the race for the best ablative treatment for localized PCa, considering both patient compliance and treatment effectiveness.
Dose escalation to the DIL with CK LINAC starting from these promising toxicity results is expected to further improve oncological outcomes even in UH schedules. These improvements are particularly needed from intermediate-unfavorable to high-risk localized PCas, which are the risk categories more prone to develop an early systemic disease progression [30].
Nevertheless, robust data on focal dose escalation in this setting should be yet generated. The ongoing HYPO-FLAME trial [31] is likely to shed light on the effectiveness of the DIL boost in this setting.
The present in silico study has evaluated the feasibility of ultra-hypofractionated (UH)-SBRT with CK radiosurgery system (Accuray Inc., Sunnyvale, California, USA) for localized PCa with a concomitant focal boost to the DIL. Two different isodose prescription levels were tested, and dose distributions were compared to delivered UH-dynamic wave arc (DWA) plans in the intermediate-unfavorable/high-risk PCa setting.

2. Materials and Methods

2.1. Study Design

The plans of UH-DWA and CK were compared in a series of 15 patients treated with UH-DWA. For the purpose of the study, new CK plans were generated using available simulation-CT and contours.

2.2. Patients’ Cohort

Patients with localized PCa who underwent UH-DWA at our institution from October 2021 to March 2022 were considered for study inclusion. The eligibility criteria were the following:
(i)
Histologically confirmed adenocarcinoma of the prostate;
(ii)
Availability of a previous diagnostic multi-parametric magnetic resonance imaging (mpMRI);
(iii)
At least one visible intraprostatic lesion classified with a PI-RADS 4 score [32];
(iv)
A maximum of 2 total DILs and a prostate gland volume < 100 cm3 of intermediate and high-risk categories, according to the NCCN version 01.2023 and the cN0 and cM0 TNM classifications.
This retrospective study was approved by the ethical committee with UID number IEO 1872.

2.3. Region of Interest Definition

Contouring of region of interest was performed by an expert radiation oncologist using RayStation® TPS (RaySearch Laboratories, Stockholm, Sweden) after rigid registration of simulation CT (CTsim) with mpMRI-T2W imaging.
The clinical target volume (CTV) of the prostate (CTV-p) included the whole prostate and proximal 1 cm of seminal vesicles; CTV of the DIL (CTV-d) included the DIL visible volume on the diagnostic mpMRI. According to institutional guidelines, prostate PTV (PTV-p) was created by the expansion of CTV-p by 5 mm in all directions except 3 mm posteriorly; for the DIL PTV (PTV-d), an expansion of 3 mm in all directions was added to CTV-d [33].

2.4. Treatment Characteristics

Patients were treated with UH-RT delivered with the VERO® system (Vero, BrainLab AG, Feldkirchen, Germany, and Mitsubishi Heavy Industries, Ltd., Tokyo, Japan) and DWA technique. The dose prescription was 36.25 Gy to the PTV-p and 40 Gy to the PTV-d with a simultaneous integrated boost (SIB) technique in 5 fractions every other day. The dose prescription scheme is reported in Figure 1. Dose distribution was calculated with Collapsed Cone Convolution (CCC) calculation algorithm in Raystation (RaySearch Laboratories, Stockholm, Sweden) treatment planning system (TPS). The primary planning goal was to achieve, both for PTV-p and PTV-d, the dose of 95% of volume ≥ 95% of the prescription dose (D95% > 95%) and to reach organs at risk (OARs) constraint compliance according to Folkert and Timmerman [34] (Table 1). Secondary planning goals were D0.03 cm3 < 108% for PTV-d and D0.03 cm3 < 111% for PTV-p.

2.5. Data Analysis

For the purpose of the study, for each patient, two CK-SBRT plans with virtual fiducials tracking were generated in silico using Accuray Precision® TPS (Accuray Inc., Sunnyvale, California, USA). Two different isodose prescriptions lines were tested, 95% and 85% (CK-85% and CK-95). More in detail, the prescription isodose line can be defined as the ratio of prescription dose and maximum dose (i.e., prescribing to the 85% or 95% isodose lines means normalizing the prescription dose to the 85% or 95% isodose surface, respectively, that includes the target volume).
CTsim and contours, including PTV-p and PTV-d with the same margins, were imported from UH-DWA plans, and the same dose prescription was used for both CK plans.
Comparison between the three treatment approaches focused on the following: (i) PTV coverage metrics, (ii) homogeneity index (HI), (iii) conformity index (CI), and (iv) OARs dosimetric parameters. HI was calculated as (D2%–D98%)/D50%, while CI was calculated as V95%/volume. Beam on time was considered, as well. Median dose values across all patients and their respective interquartile ranges (IQR) were reported. Statistical analyses were performed using the Friedman test and the Wilcoxon signed-rank test to assess statistical differences between the techniques. The significance level of multiple comparisons was set as p < 0.05/3, determined by Bonferroni correction for the Friedman test, and as p < 0.05 for the Wilcoxon signed-rank test.

3. Results

A total of 15 patients with a median age of 75 years (IQR 72–77) are included in the present analysis. The median mpMRI prostate volume is 38 cm3 (IQR 30–52), and the median DIL volume is 1.4 cm3 (IQR 1–1.6). Only one patient has two visible DILs. Characteristics of the patients are summarized in Table 2.
When considering treatment time, beam-on time for in silico CK plans is considerably higher with a median of 27.0 min (IQR 26.0–29.5), as opposed to 6.8 min (IQR 5.6–7.3) median treatment time for UH-DWA.

3.1. Target Coverage

The results of the planning study for target coverage of the three selected techniques are shown in Table 3. All target volumes (DIL and PTV) could achieve the planning goals as expected. A visual representation of the target coverage for different parameters among the three techniques is depicted in Figure 2a,b.
Regarding PTV parameters, the comparison of median values of DVH metrics both for PTV-p and PTV-d show an increase (p < 0.05, Table 3) from UH-DWA to CK-95% to CK-85% in almost all considered indexes. The median CI of PTV-p is 1.00 for the three considered techniques, while a statistically significant increase (p < 0.05) is observed for PTV-p in CK-85% if compared with both CK-95% and UH-DWA (0.991, 0.984, and 0.978). An example of dose distribution can be found in Figure 3; the maximum dose points are all located in the DIL.

3.2. Organs at Risk

Dose escalation to the DIL does not significantly impact the dose received by the OARs (Table 4). In fact, although borderline statistically significant differences between the three planning techniques exist for the rectum, bladder, testis, and bowel cavity, all the considered parameters remain well below the constraints. A visual representation of the dose received by the considered OARs among the three techniques is depicted in Figure 2c.

4. Discussion

The present study showed that in silico 85% isodose CK treatment plans could generate a higher boost dose to the DIL (PTV-d median D95% = 40.58 Gy and D0.03 cm3 = 46.66 Gy) while maintaining a lower dose prescription to the prostate volume (PTV-p median D95% = 35.16 Gy and D0.03 cm3 = 41.63 Gy) with homogeneous dose distribution and respecting all the OARs constraints applied.
Technical advances in the field of RT in recent years have enabled the progressive implementation of UH-RT in various scenarios of localized PCa treatment. As a matter of fact, the use of hypofractionated schedules in PCa has provided sufficient evidence in terms of tumor control results, QoL, toxicity, and reduced treatment time and costs for the patient, and the recent publication of two randomized trials comparing the use of UH-RT versus conventional fractionation (HYPO-RT-PC [14] and PACE-B trials [15]) has been crucial in supporting its use. This in silico planning comparison study showed that UH-RT for localized PCa with a SIB to the macroscopic lesion with CK radiosurgery SBRT-based system, both with 95% and 85% isodose prescriptions is feasible. Dose-volume histograms for the 85% isodose prescription reported better dose coverage up to a maximum of 47 Gy to the DIL PTV while respecting all the OARs constraints previously applied in UH-DWA plans delivered with the VERO system.
The rationale of the present study is based on the recently published data of Tree et al. [23] in the update at 2 years of the PACE-B trial, which reported lower levels of GU and GI Radiation Therapy Oncology Group (RTOG) toxicity for CK vs. standard linac SBRT treatments (significantly lower for GU: 5.8% vs. 16.5%).
Also, in the “FLAME” trial, Kerkmeijer et al. [18] proved that focal boosting the DIL up to 95 Gy in a conventional radiotherapy scheme for 264 patients (vs. 271 patients in the arm without focal boost) improved bDFS and PFS without any relevant additional toxicity. In addition, the preliminary results reported by Marvaso et al. [20] in the “GIVE ME FIVE” trial reported very low acute RTOG GU and GI toxic (G2 4.69% and 1.56%, respectively, and only one patient with GU G3 at 6 months) when a dose escalation to 37.5 Gy in 5 fractions was applied as a SIB to the DIL with the VERO® system.
In this context, a high focal boost strategy to the DIL with SBRT delivery technique is required to improve tumor control while respecting organ at-risk dose constraints. The pattern of failure analysis by Groen et al. [35] demonstrated that a focal boost to the DIL decreased the rates of local failure (7 vs. 21 events) and the development of regional-distant metastasis (32 vs. 56 events).
Similar to our study, Tree et al. [36] confronted two methods of SBRT delivery, CK and VMAT. The dosing scheme applied was a higher prescription dose of 47.5 Gy while maintaining a 36.25 Gy to the whole prostate in five fractions with different PTV margins. Both CK and VMAT planning produced clinically acceptable plans, but certain OARs constraints have not been met for all plans. More violations (43 out of 75) were observed in VMAT planning with larger prostate margins (5 mm).
With the experience derived from the above-mentioned planning study, Nicholls et al. [19] reported in the “SPARC” trial the longest follow-up (56 months) of the first eight patients treated with CK SBRT (at 70% isodose prescription) with focal boost for intermediate and high-risk PCa. No biochemical recurrence was detected at the follow-up time, and early results suggest a 2-year bDFS between 95% and 100%, but grade 2+ acute GU and GI toxicities were 37.5%, while grade 2+ late GU and GI toxicities were 12.5% and 0%, respectively.
A previous study from Aluwini et al. [37] reported the early results of CK SBRT with a focal boost to the DIL in low- and intermediate-risk PCa. Fifty patients received 38 Gy to the prostate and 44 Gy to the MRI-visible tumor in four daily fractions. At a median follow-up of 23 months, the 2-year bDFS was 100%, similar to the oncological results achieved in the “SPARC” trial, but grade 2 acute GU and GI toxicities were 15% and 12%, respectively, while grade 2 late GU and GI toxicities were 10% and 3%, respectively. The acute G2 toxicity reported was slightly lower than those found in the SPARC trial, but probably because only 14 patients out of 50 in this cohort received the boost to the DIL. A summary of the published literature for PCa CK-SBRT plus DIL boost can be found in Table 5.
Finally, when considering the median beam-on time for CK plans was, as expected, considerably higher respect UH-DWA. It should, however, be considered that our median beam-on time for in silico CK plans was 27 min, greatly reduced in comparison with the median of 46 min reported by Tree and colleagues [36] For our cohort of patients treated with the VERO system, the use of a Foley catheter is not required and was not taken into consideration in this in silico planning study.
The primary limit of this dosimetric study is relative to its intrinsic nature of the study and consists of the lack of clinical outcomes. Nevertheless, the experience acquired thanks to this in silico planning study will be applied to the upcoming PRO-SPEED trial in our Institute. This trial aims to evaluate the effectiveness of CK-SBRT treatment on the whole prostate gland plus SIB to the DIL(s) in intermediate-unfavorable to high-risk PCa patients, both in terms of toxicity and oncological outcomes, compared with UH-DWA treatments in a real-world setting. Another relevant limit in the dosimetric analysis was the absence of the prostatic urethra contours. However, the prostatic urethra constraints used by Draulans et al. [25] (D0.035 cm3 < 42 Gy) or by Nicholls et al. [19] (V42 Gy < 40% and V45.6 Gy < 10%) could probably be respected in the majority of the plans analyzed with the three different techniques for the dosing scheme taken in consideration by our study. In most of the cases, the location of the DIL would be peripheral and far enough from the central position of the prostatic urethra, and, as a surrogate result, the higher median near-maximum D0.03 cm3 of the prostate volume was 41.64 Gy for the in silico CK 85% isodose prescription plans, which is lower enough to meet the prostatic urethra constraints previously reported.

5. Conclusions

This in silico planning comparison study showed that UH-SBRT with CK radiosurgery system plus a high focal boost to the DIL is feasible, both prescribing to 95% and 85% isodose lines. Few clinical trials with similar radiosurgery SBRT techniques have been published, and others are ongoing with promising results in terms of oncological outcome and treatment toxicity profile. These encouraging dosimetric results need to be confirmed by oncological outcomes and toxicity data in the upcoming mono-institution phase 2 “PRO-SPEED” IEO trial.

Author Contributions

Conceptualization, G.C.M., M.G.V., E.R., G.C. and G.M. (Giulia Marvaso); methodology, G.C.M., M.G.V., M.Z., E.R. and G.M. (Giulia Marvaso); formal analysis, M.G.V. and M.Z.; investigation, G.C.M. and E.R.; data curation, G.C.M., E.R., S.V., G.R., M.G.V. and M.Z.; writing—original draft preparation, G.C.M., M.G.V. and M.Z.; writing—review and editing, D.Z., S.D., F.A.M., S.L., M.F., O.D.C., G.P. all authors; visualization, G.C.M., M.G.V., M.Z. and S.G.; supervision, A.V., F.C., G.M. (Giulia Marvaso), R.O. and B.A.J.-F. All authors have read and agreed to the published version of the manuscript.

Funding

IEO: the European Institute of Oncology, is partially supported by the Italian Ministry of Health (with “Ricerca Corrente” and “5 × 1000” funds). The Division of Radiation Oncology of IEO received research funding from AIRC (Italian Association for Cancer Research) and Fondazione IEO-CCM (Istituto Europeo di Oncologia-Centro Cardiologico Monzino), all outside the current project. M.G.V. and G.C. were supported by a research fellowship from the “Associazione Italiana per la Ricerca sul Cancro—AIRC” entitled “Radioablation ± hormonotherapy for prostate cancer oligorecurrences (RADIOSA trial): potential of imaging and biology” registered at ClinicalTrials.gov NCT03940235. G.C.M. was supported by a research fellowship from the AIRC entitled “Single Fraction Preoperative Radiotherapy for Early Stage Breast Cancer”, registered at ClinicalTrials.gov NCT04679454.

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 IEO, European Institute of Oncology, IRCCS (UID number IEO 1872, 15 November 2022).

Informed Consent Statement

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

Data Availability Statement

The data presented in this study are available upon reasonable request from the corresponding author.

Conflicts of Interest

IEO, the European Institute of Oncology, IRCCS, is partially supported by Institutional grants from Accuray Inc. and Ion Beam Applications (IBA). B.A.J.-F. received speakers’ fees from Roche, Bayer, Janssen, Carl Zeiss, Ipsen, Accuray, Astellas, Elekta, and IBA Astra Zeneca (all outside the current project). The remaining authors declare no conflicts of interest that are relevant to the content of this article.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  2. Schaeffer, E.; Srinivas, S.; Adra, N.; An, Y.; Barocas, D.; Bitting, R.; Bryce, A.; Chapin, B.; Cheng, H.; D’Amico, A.V.; et al. NCCN Guidelines Version 1.2023 Prostate Cancer. 2022. Available online: https://www.nccn.org/home/ (accessed on 4 April 2023).
  3. Miralbell, R.; Roberts, S.A.; Zubizarreta, E.; Hendry, J.H. Dose-fractionation sensitivity of prostate cancer deduced from radiotherapy outcomes of 5,969 patients in seven international institutional datasets: α/β = 1.4 (0.9–2.2) Gy. Int. J. Radiat. Oncol. Biol. Phys. 2012, 82, e17–e24. [Google Scholar] [CrossRef]
  4. Catton, C.N.; Lukka, H.; Gu, C.-S.; Martin, J.M.; Supiot, S.; Chung, P.W.M.; Bauman, G.S.; Bahary, J.-P.; Ahmed, S.; Cheung, P.; et al. Randomized Trial of a Hypofractionated Radiation Regimen for the Treatment of Localized Prostate Cancer. J. Clin. Oncol. 2017, 35, 1884–1890. [Google Scholar] [CrossRef]
  5. Hoffman, K.E.; Voong, K.R.; Levy, L.B.; Allen, P.K.; Choi, S.; Schlembach, P.J.; Lee, A.K.; McGuire, S.E.; Nguyen, Q.; Pugh, T.J.; et al. Randomized Trial of Hypofractionated, Dose-Escalated, Intensity-Modulated Radiation Therapy (IMRT) Versus Conventionally Fractionated IMRT for Localized Prostate Cancer. J. Clin. Oncol. 2018, 36, 2943–2949. [Google Scholar] [CrossRef]
  6. Lee, W.R.; Dignam, J.J.; Amin, M.B.; Bruner, D.W.; Low, D.; Swanson, G.P.; Shah, A.B.; D’Souza, D.P.; Michalski, J.M.; Dayes, I.S.; et al. Randomized Phase III Noninferiority Study Comparing Two Radiotherapy Fractionation Schedules in Patients With Low-Risk Prostate Cancer. J. Clin. Oncol. 2016, 34, 2325–2332. [Google Scholar] [CrossRef]
  7. Dearnaley, D.; Syndikus, I.; Mossop, H.; Khoo, V.; Birtle, A.; Bloomfield, D.; Graham, J.; Kirkbride, P.; Logue, J.; Malik, Z.; et al. Conventional versus hypofractionated high-dose intensity-modulated radiotherapy for prostate cancer: 5-year outcomes of the randomised, non-inferiority, phase 3 CHHiP trial. Lancet Oncol. 2016, 17, 1047–1060. [Google Scholar] [CrossRef] [Green Version]
  8. Royce, T.J.; Lee, D.H.; Keum, N.; Permpalung, N.; Chiew, C.J.; Epstein, S.; Pluchino, K.M.; D’amico, A.V. Conventional Versus Hypofractionated Radiation Therapy for Localized Prostate Cancer: A Meta-analysis of Randomized Noninferiority Trials. Eur. Urol. Focus 2019, 5, 577–584. [Google Scholar] [CrossRef] [Green Version]
  9. Cao, L.; Yang, Y.-J.; Li, Z.; Wu, H.-F.; Yang, Z.-C.; Liu, S.-X.; Wang, P. Moderate hypofractionated radiotherapy is more effective and safe for localized prostate cancer patients: A meta-analysis. Oncotarget 2017, 8, 2647–2658. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Carvalho, Í.T.; Baccaglini, W.; Claros, O.R.; Chen, F.K.; Kayano, P.P.; Lemos, G.C.; Weltman, E.; Kuban, D.A.; Carneiro, A. Genitourinary and gastrointestinal toxicity among patients with localized prostate cancer treated with conventional versus moderately hypofractionated radiation therapy: Systematic review and meta-analysis. Acta Oncol. 2018, 57, 1003–1010. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. de Vries, K.C.; Wortel, R.C.; Oomen-de Hoop, E.; Heemsbergen, W.D.; Pos, F.J.; Incrocci, L. Hyprofractionated Versus Conventionally Fractionated Radiation Therapy for Patients with Intermediate- or High-Risk, Localized, Prostate Cancer: 7-Year Outcomes From the Randomized, Multicenter, Open-Label, Phase 3 HYPRO Trial. Int. J. Radiat. Oncol. Biol. Phys. 2020, 106, 108–115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Jackson, W.C.; Silva, J.; Hartman, H.E.; Dess, R.T.; Kishan, A.U.; Beeler, W.H.; Gharzai, L.A.; Jaworski, E.M.; Mehra, R.; Hearn, J.W.D.; et al. Stereotactic Body Radiation Therapy for Localized Prostate Cancer: A Systematic Review and Meta-Analysis of over 6000 Patients Treated on Prospective Studies. Int. J. Radiat. Oncol. Biol. Phys. 2019, 104, 778–789. [Google Scholar] [CrossRef] [Green Version]
  13. Kishan, A.U.; Dang, A.; Katz, A.J.; Mantz, C.A.; Collins, S.P.; Aghdam, N.; Chu, F.-I.; Kaplan, I.D.; Appelbaum, L.; Fuller, D.B.; et al. Long-term Outcomes of Stereotactic Body Radiotherapy for Low-Risk and Intermediate-Risk Prostate Cancer. JAMA Netw. Open. 2019, 2, e188006. [Google Scholar] [CrossRef] [Green Version]
  14. Widmark, A.; Gunnlaugsson, A.; Beckman, L.; Thellenberg-Karlsson, C.; Hoyer, M.; Lagerlund, M.; Kindblom, J.; Ginman, C.; Johansson, B.; Björnlinger, K.; et al. Ultra-hypofractionated versus conventionally fractionated radiotherapy for prostate cancer: 5-year outcomes of the HYPO-RT-PC randomised, non-inferiority, phase 3 trial. Lancet 2019, 394, 385–395. [Google Scholar] [CrossRef]
  15. Brand, D.H.; Tree, A.C.; Ostler, P.; Van Der Voet, H.; Loblaw, A.; Chu, W.; Ford, D.; Tolan, S.; Jain, S.; Martin, A.; et al. Intensity-modulated fractionated radiotherapy versus stereotactic body radiotherapy for prostate cancer (PACE-B): Acute toxicity findings from an international, randomised, open-label, phase 3, non-inferiority trial. Lancet Oncol. 2019, 20, 1531–1543. [Google Scholar] [CrossRef]
  16. Pucar, D.; Hricak, H.; Shukla-Dave, A.; Kuroiwa, K.; Drobnjak, M.; Eastham, J.; Scardino, P.T.; Zelefsky, M. Clinically significant prostate cancer local recurrence after radiation therapy occurs at the site of primary tumor: Magnetic resonance imaging and step-section pathology evidence. Int. J. Radiat. Oncol. Biol. Phys. 2007, 69, 62–69. [Google Scholar] [CrossRef]
  17. Cellini, N.; Morganti, A.G.; Mattiucci, G.C.; Valentini, V.; Leone, M.; Luzi, S.; Manfredi, R.; Dinapoli, N.; Digesu’, C.; Smaniotto, D. Analysis of intraprostatic failures in patients treated with hormonal therapy and radiotherapy: Implications for conformal therapy planning. Int. J. Radiat. Oncol. Biol. Phys. 2002, 53, 595–599. [Google Scholar] [CrossRef] [PubMed]
  18. Kerkmeijer, L.G.W.; Groen, V.H.; Pos, F.J.; Haustermans, K.; Monninkhof, E.M.; Smeenk, R.J.; Kunze-Busch, M.; de Boer, J.C.J.; Zijp, J.V.D.V.V.; van Vulpen, M.; et al. Focal Boost to the Intraprostatic Tumor in External Beam Radiotherapy for Patients With Localized Prostate Cancer: Results From the FLAME Randomized Phase III Trial. J. Clin. Oncol. 2021, 39, 787–796. [Google Scholar] [CrossRef] [PubMed]
  19. Nicholls, L.; Suh, Y.-E.; Chapman, E.; Henderson, D.; Jones, C.; Morrison, K.; Sohaib, A.; Taylor, H.; Tree, A.; van As, N. Stereotactic radiotherapy with focal boost for intermediate and high-risk prostate cancer: Initial results of the SPARC trial. Clin. Transl. Radiat. Oncol. 2020, 25, 88–93. [Google Scholar] [CrossRef] [PubMed]
  20. Marvaso, G.; Gugliandolo, S.G.; Bellerba, F.; Gandini, S.; Corrao, G.; Volpe, S.; Rojas, D.P.; Riva, G.; Zerini, D.; Pepa, M.; et al. Phase II prospective trial “Give Me Five” short-term high precision radiotherapy for early prostate cancer with simultaneous boost to the dominant intraprostatic lesion: The impact of toxicity on quality of life (AIRC IG-13218). Med. Oncol. 2020, 37, 74. [Google Scholar] [CrossRef]
  21. Marvaso, G.; Riva, G.; Ciardo, D.; Gandini, S.; Fodor, C.; Zerini, D.; Colangione, S.P.; Timon, G.; Comi, S.; Cambria, R.; et al. “Give me five” ultra-hypofractionated radiotherapy for localized prostate cancer: Non-invasive ablative approach. Med. Oncol. 2018, 35, 96. [Google Scholar] [CrossRef]
  22. Marvaso, G.; Ciardo, D.; Gandini, S.; Riva, G.; Frigo, E.; Volpe, S.; Fodor, C.; Zerini, D.; Rojas, D.P.; Comi, S.; et al. Comparison of Outcomes and Toxicity Between Extreme and Moderate Radiation Therapy Hypofractionation in Localized Prostate Cancer: A Propensity Score Analysis. Int. J. Radiat. Oncol. Biol. Phys. 2019, 105, 735–744. [Google Scholar] [CrossRef] [PubMed]
  23. Tree, A.C.; Ostler, P.; van der Voet, H.; Chu, W.; Loblaw, A.; Ford, D.; Tolan, S.; Jain, S.; Martin, A.; Staffurth, J.; et al. Intensity-modulated radiotherapy versus stereotactic body radiotherapy for prostate cancer (PACE-B): 2-year toxicity results from an open-label, randomised, phase 3, non-inferiority trial. Lancet Oncol. 2022, 23, 1308–1320. [Google Scholar] [CrossRef]
  24. Meier, R.M.; Bloch, D.A.; Cotrutz, C.; Beckman, A.C.; Henning, G.T.; Woodhouse, S.A.; Williamson, S.K.; Mohideen, N.; Dombrowski, J.J.; Hong, R.L.; et al. Multicenter Trial of Stereotactic Body Radiation Therapy for Low- and Intermediate-Risk Prostate Cancer: Survival and Toxicity Endpoints. Int. J. Radiat. Oncol. Biol. Phys. 2018, 102, 296–303. [Google Scholar] [CrossRef]
  25. Vuolukka, K.; Auvinen, P.; Tiainen, E.; Palmgren, J.-E.; Heikkilä, J.; Seppälä, J.; Aaltomaa, S.; Kataja, V. Stereotactic body radiotherapy for localized prostate cancer—5-year efficacy results. Radiat. Oncol. 2020, 15, 173. [Google Scholar] [CrossRef]
  26. King, C.R.; Brooks, J.D.; Gill, H.; Presti, J.C. Long-term outcomes from a prospective trial of stereotactic body radiotherapy for low-risk prostate cancer. Int. J. Radiat. Oncol. Biol. Phys. 2012, 82, 877–882. [Google Scholar] [CrossRef]
  27. Freeman, D.E.; King, C.R. Stereotactic body radiotherapy for low-risk prostate cancer: Five-year outcomes. Radiat. Oncol. 2011, 6, 3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Katz, A.J.; Santoro, M.; Diblasio, F.; Ashley, R. Stereotactic body radiotherapy for localized prostate cancer: Disease control and quality of life at 6 years. Radiat. Oncol. 2013, 8, 118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Fuller, D.B.; Naitoh, J.; Mardirossian, G. Virtual HDR CyberKnife SBRT for Localized Prostatic Carcinoma: 5-Year Disease-Free Survival and Toxicity Observations. Front. Oncol. 2014, 4, 321. [Google Scholar] [CrossRef] [Green Version]
  30. Molitoris, J.K.; Alexander, G.S.; Siddiqui, O.; Cohen, J.; Mishra, M.V.; Rana, Z. High-risk, recurrent and oligometastatic prostate cancer: Recent developments on the role of radiation. Curr. Opin. Oncol. 2021, 33, 238–243. [Google Scholar] [CrossRef]
  31. Draulans, C.; van der Heide, U.A.; Haustermans, K.; Pos, F.J.; Zyp, J.V.D.V.V.; De Boer, H.; Groen, V.H.; Monninkhof, E.M.; Smeenk, R.J.; Kunze-Busch, M.; et al. Primary endpoint analysis of the multicentre phase II hypo-FLAME trial for intermediate and high risk prostate cancer. Radiother. Oncol. 2020, 147, 92–98. [Google Scholar] [CrossRef]
  32. Miszczyk, M.; Rembak-Szynkiewicz, J.; Magrowski, Ł.; Stawiski, K.; Namysł-Kaletka, A.; Napieralska, A.; Kraszkiewicz, M.; Woźniak, G.; Stąpór-Fudzińska, M.; Głowacki, G.; et al. The Prognostic Value of PI-RADS Score in CyberKnife Ultra-Hypofractionated Radiotherapy for Localized Prostate Cancer. Cancers 2022, 14, 1613. [Google Scholar] [CrossRef] [PubMed]
  33. Timon, G.; Ciardo, D.; Bazani, A.; Garioni, M.; Maestri, D.; De Lorenzo, D.; Pansini, F.; Cambria, R.; Rondi, E.; Cattani, F.; et al. Rationale and Protocol of AIRC IG-13218, Short-Term Radiotherapy for Early Prostate Cancer with Concomitant Boost to the Dominant Lesion. Tumori 2016, 102, 536–540. [Google Scholar] [CrossRef] [PubMed]
  34. Folkert, M.R.; Timmerman, R.D. Stereotactic ablative body radiosurgery (SABR) or Stereotactic body radiation therapy (SBRT). Adv. Drug Deliv. Rev. 2017, 109, 3–14. [Google Scholar] [CrossRef]
  35. Groen, V.H.; Haustermans, K.; Pos, F.J.; Draulans, C.; Isebaert, S.; Monninkhof, E.M.; Smeenk, R.J.; Kunze-Busch, M.; de Boer, J.C.; Zijp, J.V.D.V.V.; et al. Patterns of Failure Following External Beam Radiotherapy with or without an Additional Focal Boost in the Randomized Controlled FLAME Trial for Localized Prostate Cancer. Eur. Urol. 2022, 82, 252–257. [Google Scholar] [CrossRef]
  36. Tree, A.; Jones, C.; Sohaib, A.; Khoo, V.; van As, N. Prostate stereotactic body radiotherapy with simultaneous integrated boost: Which is the best planning method? Radiat. Oncol. 2013, 8, 228. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Aluwini, S.; van Rooij, P.; Hoogeman, M.; Kirkels, W.; Kolkman-Deurloo, I.K.; Bangma, C. Stereotactic body radiotherapy with a focal boost to the MRI-visible tumor as monotherapy for low- and intermediate-risk prostate cancer: Early results. Radiat. Oncol. 2013, 8, 84. [Google Scholar] [CrossRef] [Green Version]
  38. Herrera, F.G.; Valerio, M.; Berthold, D.; Tawadros, T.; Meuwly, J.-Y.; Vallet, V.; Baumgartner, P.; Thierry, A.-C.; De Bari, B.; Jichlinski, P.; et al. 50-Gy Stereotactic Body Radiation Therapy to the Dominant Intraprostatic Nodule: Results From a Phase 1a/b Trial. Int. J. Radiat. Oncol. 2019, 103, 320–334. [Google Scholar] [CrossRef] [Green Version]
  39. Zhao, X.; Ye, Y.; Yu, H.; Jiang, L.; Cheng, C.; Guo, X.; Ju, X.; Zhu, X.; Zhang, H. Five-year outcomes of stereotactic body radiation therapy (SBRT) for prostate cancer: The largest experience in China. J. Cancer Res. Clin. Oncol. 2021, 147, 3557–3564. [Google Scholar] [CrossRef]
Figure 1. Ultra-hypofractionated stereotactic body radiation therapy dose prescription scheme.
Figure 1. Ultra-hypofractionated stereotactic body radiation therapy dose prescription scheme.
Applsci 13 07273 g001
Figure 2. Graphics comparison of median target coverage parameters for PTV-DIL (PTV-d) (Panel a), PTV-prostate (PTV-p) (Panel b), and organ at risks (OARs) for the three considered techniques (Panel c). The greyed-out part in Panel c denotes the histograms for the bladder.
Figure 2. Graphics comparison of median target coverage parameters for PTV-DIL (PTV-d) (Panel a), PTV-prostate (PTV-p) (Panel b), and organ at risks (OARs) for the three considered techniques (Panel c). The greyed-out part in Panel c denotes the histograms for the bladder.
Applsci 13 07273 g002
Figure 3. Dose distribution in a 78-year-old man with a dominant lesion in the left peripheral-apex side of the prostate for the three different techniques. Light Blue: 50% of PTV-p prescription dose [PD]; green: 95% of PTV-p PD; orange: 100% of PTV-p PD; red: 100% of PTV-d PD; and purple: 108% of PTV-d PD); R: right; L: left.
Figure 3. Dose distribution in a 78-year-old man with a dominant lesion in the left peripheral-apex side of the prostate for the three different techniques. Light Blue: 50% of PTV-p prescription dose [PD]; green: 95% of PTV-p PD; orange: 100% of PTV-p PD; red: 100% of PTV-d PD; and purple: 108% of PTV-d PD); R: right; L: left.
Applsci 13 07273 g003
Table 1. Summary of the OARs dose constraint objectives [34].
Table 1. Summary of the OARs dose constraint objectives [34].
Organ at RisksObjective
Rectum
V18 Gy<50%
V29 Gy<20%
V33 Gy<10%
V36.25 Gy<5%
Rectum Posterior Wall
D1cc<17 Gy
Anal Canal
Dmean<15 Gy
Bladder
V36.25 Gy<5%
V36.25 Gy<5 cc
V18 Gy<40%
Femoral Heads
V15 Gy<5%
Bowel Cavity
V30 Gy<1 cc
Dmean<5.4 Gy
V17 Gy<195 cc
Penile Bulb
V29 Gy<50%
Penis
V13 Gy<1 cc
Testis
D20%<2 Gy
Table 2. Summary of patients’ characteristics.
Table 2. Summary of patients’ characteristics.
CharacteristicValue (IQR)
Median Age (years)75 (72–77)
Median iPSA (ng/mL)6.37 (5.04–8.45)
Gleason Score
3 + 47
4 + 36
4 + 42
Median ISUP Score3 (2–4)
NCCN Risk Category
Intermedium Favorable7
Intermedium Unfavorable5
High3
AJCC Stage
T1c N06
T2 N08
T3a N01
ADT Prescription
LHRHa7
Bicalutamide 150 mg1
Radiological Data
Median mpRMN Prostate Volume (cm3)38 (30–52)
Median DIL Max Dimension (mm)11 (7–14)
PIRADS 4 Score12
PIRADS 5 Score4
ECE+1
DIL Location
Peripheral15
Transitional1
Volumes (cc) and Distances (mm)
Median DIL Distance to the Bladder Wall12.00 (10.90–16.35)
Median DIL Distance to the Rectum3.90 (3.05–6.40)
Median DIL Clinical Volume (CTV-d)1.40 (1.01–1.65)
Median Prostate Clinical Volume (CTV-p)51.30 (43.26–61.35)
Median DIL-Prostate Clinical Volume Ratio (%)2.27 (1.66–3.76)
Median DIL Planning Volume (PTV-d)4.67 (3.65–5.49)
Median Prostate Planning Volume (PTV-p)101.93 (81.40–115.39)
Median DIL-Prostate Planning Volume Ratio (%)4.58 (3.30–6.90)
Table 3. Comparison of median values of dose-volume histogram parameters for PTV of the prostate (PTV-p) and PTV of the DIL (PTV-d).
Table 3. Comparison of median values of dose-volume histogram parameters for PTV of the prostate (PTV-p) and PTV of the DIL (PTV-d).
PTV-d
(40 Gy)
UH-DWACyberKnife 95%CyberKnife 85%Overall (p-Value from Friedman Test) *UH-DWA vs. CK95%UH-DWA vs. CK85%CK95% vs. CK85%
D 0.03 cm3104.00%105.25%116.65%<0.00001p < 0.05p < 0.05p < 0.05
D98%97.00%98.20%100.57%0.00007p < 0.05p < 0.05p < 0.05
D95%97.80%98.77%101.45%0.00012p < 0.05p < 0.05p < 0.05
D2%103.40%104.82%115.77%<0.00001p < 0.05p < 0.05p < 0.05
D50% (Median)100.80%101.62%106.57%<0.00001p < 0.05p < 0.05p < 0.05
V100%68.50%78.70%98.50%0.00003p < 0.05p < 0.05p < 0.05
V110%0.00%0.00%26.10%0.00001p < 0.05p < 0.05p < 0.05
CI1.001.001.000.8052nsns**
HI0.060.060.150.00001nsp < 0.05p < 0.05
PTV-p
(36.25 Gy)
UH-DWACyberKnife 95%CyberKnife 85%Overall (p-value from Friedman Test) *UH-DWA vs. CK95%UH-DWA vs. CK85%CK95% vs. CK85%
D 0.03 cm3110.70%111.39%114.86%0.00005p < 0.05p < 0.05p < 0.05
D98%94.10%93.93%95.36%0.00019p < 0.05p < 0.05p < 0.05
D95%96.55%96.16%96.99%0.00516p < 0.05p < 0.05ns
D2%100.10%102.12%103.53%0.00061p < 0.05p < 0.05p < 0.05
D50% (Median)107.00%108.08%110.59%0.00003p < 0.05p < 0.05p < 0.05
V100%58.00%74.60%81.60%0.00041p < 0.05p < 0.05p < 0.05
V110%0.30%0.20%3.40%0.00232p < 0.05p < 0.05p < 0.05
CI0.980.980.990.00129nsp < 0.05p < 0.05
HI0.130.140.140.03122p < 0.05p < 0.05ns
* Significant p-value if <0.016 (Bonferroni correction). ** comparison was not possible because of tied values. List of abbreviations: CK = CyberKnife; UH-DWA = ultra-hypofractionated dynamic wave arc; ns = not significant.
Table 4. Comparison of median values of dose-volume histogram parameters for considered OARs.
Table 4. Comparison of median values of dose-volume histogram parameters for considered OARs.
Organs at RiskResults (Median Value)Overall (p-Value from Friedman Test) *UH-DWA vs. CK95%UH-DWA vs. CK85%CK95% vs. CK85%
RectumUH-DWACyberKnife 95%CyberKnife 85%
V18 Gy19.00%19.60%19.30%0.0043p < 0.05p < 0.05ns
V29 Gy6.80%6.70%6.20%0.86071nsnsns
V33 Gy3.10%2.40%2.50%0.08073p < 0.05nsp < 0.05
V36.25 Gy0.18%0.20%0.30%0.02431nsnsp < 0.05
Rectum Posterior Wall
D1 cm315.1016.3015.560.05689p < 0.05nsns
Bladder
V36.25 Gy0.50%1.00%1.10%0.0043p < 0.05p < 0.05p < 0.05
V18 Gy15.00%16.90%15.90%0.21944p < 0.05p < 0.05ns
Femoral Heads
V15 Gy (Right)0.01%0.80%0.30%0.53974nsnsns
V15 Gy (Left)0.00%0.00%0.00%0.38674ns****
Penile Bulb
V29 Gy2.20%0.75%1.80%0.95123nsnsns
Testis
D20%0.300.740.570.00004p < 0.05p < 0.05ns
Bowel Cavity
Dmean0.401.100.950.00004p < 0.05p < 0.05ns
Penis
V13 Gy0.300.670.680.08487nsnsns
* Significant p-value if < 0.016 (Bonferroni correction). ** comparison is not possible due to tied values. List of abbreviations: CK = CyberKnife; UH-DWA = ultra-hypofractionated dynamic wave arc; ns = not significant.
Table 5. Overview of published literature of CyberKnife UH-SBRT with or without concomitant DIL boost.
Table 5. Overview of published literature of CyberKnife UH-SBRT with or without concomitant DIL boost.
AuthorsRisk GroupPatient CohortDose per Fraction (Gy)Total FractionFollow-Up
(Months)
bRFSAcute G2+
Toxicity
Late G2+
Toxicity
ProstateSIBGUGIGUGI
Nicholls et al. [19], 2020Int., High87.259.5556100%37.5%37.5%12.5%0%
Meier et al. [24], 2018Low, Int.3098-56197.2%26%8.1%13.3%2%
Vuolukka et al. [25], 2020Low, Int., High2137.25-564100% (L) 87.5% (I) 80% (H)NANANANA
King et al. [26], 2012Low3047.25-53294%NANA8.5%2%
Freeman et al. [27], 2011Low417/7.25-56093%7%NA7%2.5%
Katz et al. [28], 2013Low, Int., High3047/7.25-56097% (L) 90% (I) 74% (H)4.6%3.6%9.9%4.5%
Fuller et al. [29], 2014Low, Int.799.5-460100% (L) 92% (I)10%0%15%1%
Aluwini et al. [37], 2013Low, Int.509.511423100%23%14%16%3%
Herrera et al. [38], 2018Int., High97.259–9.5–1053100%25%5%NANA
Zhao et al. [39], 2021Low, Int., High1337.5-56083.6%1.5%0.8%2.3%0%
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

Mazzola, G.C.; Vincini, M.G.; Rondi, E.; Ronci, G.; Vigorito, S.; Zaffaroni, M.; Corrao, G.; Gallo, S.; Zerini, D.; Durante, S.; et al. CyberKnife Ultra-Hypofractionated SBRT for Localized Prostate Cancer with Dose Escalation to the Dominant Intraprostatic Lesion: In Silico Planning Study. Appl. Sci. 2023, 13, 7273. https://doi.org/10.3390/app13127273

AMA Style

Mazzola GC, Vincini MG, Rondi E, Ronci G, Vigorito S, Zaffaroni M, Corrao G, Gallo S, Zerini D, Durante S, et al. CyberKnife Ultra-Hypofractionated SBRT for Localized Prostate Cancer with Dose Escalation to the Dominant Intraprostatic Lesion: In Silico Planning Study. Applied Sciences. 2023; 13(12):7273. https://doi.org/10.3390/app13127273

Chicago/Turabian Style

Mazzola, Giovanni Carlo, Maria Giulia Vincini, Elena Rondi, Giuseppe Ronci, Sabrina Vigorito, Mattia Zaffaroni, Giulia Corrao, Salvatore Gallo, Dario Zerini, Stefano Durante, and et al. 2023. "CyberKnife Ultra-Hypofractionated SBRT for Localized Prostate Cancer with Dose Escalation to the Dominant Intraprostatic Lesion: In Silico Planning Study" Applied Sciences 13, no. 12: 7273. https://doi.org/10.3390/app13127273

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

Article Metrics

Back to TopTop