Submit manuscript...
International Journal of
eISSN: 2574-8084

Radiology & Radiation Therapy

Research Article Volume 8 Issue 2

Role of magnetic resonance imaging in radiation therapy planning

Qing Xue, Sweet Ping Ng, , Farshad Foroudi

Radiation Oncology, Melbourne University, Australia

Correspondence: Farshad Foroudi, Radiation Oncology, Melbourne University, Australia, Tel 613-949-69797

Received: March 29, 2021 | Published: May 4, 2021

Citation: Xue Q, Ng SP, Foroudi F. Role of magnetic resonance imaging in radiation therapy planning. Int J Radiol Radiat Ther. 2021;8(2):65-74. DOI: 10.15406/ijrrt.2021.08.00296

Download PDF

Abstract

Computed tomography (CT) is essential in radiation treatment planning (RTP) since it provides information on electron density using tissue-specific attenuation coefficients, which are used to calculate the radiation dosage. It also provides lucid resolution of air-filled cavities, fat tissue and bony tissue.

Currently, MRI is frequently incorporated in RTP in conjunction with CT for contrast enhancement benefits for its superior anatomical soft tissue contrast that can visualize the extent of tumor soft tissue infiltration into normal tissues. The introduction of MRI has significantly reduced intra and inter-observer contour variability in a number of tumor sites.

MRI is a highly versatile modality with the availability of numerous sequences. Apart from anatomical information, MRI can provide functional information on tissue characteristics, and improves the accuracy of delivery which promotes dose escalation, as well as monitoring treatment and assess response.

Volume delineation is one of the earliest steps in RTP, meaning inaccuracies can induce error propagation throughout the treatment course. Adequate sequence selection is therefore a critical step. An ideal MR sequence would be accurate, resistant to artifacts, reproducible with high soft tissue resolution and low intra- and inter observer variability in order to optimize the consistency in imaging interpretation, dosimetry and ultimately better patient outcome.

This review will discuss important aspects of tumor delineation and sequence selection, followed by an introduction to major types of MRI sequences commonly used in RTP.

Keywords: radiotherapy planning, magnetic resonance imaging, MRI sequence selection

Introduction

Radiation Oncology is undoubtedly an important pillar in cancer treatment. With an estimated 60% of all cancer patients should have RT as part of their management plan.1 It is important to delineate tumor target and adjacent critical normal structures, also known as organs at risk (OAR) as accurately as possible. This will allow optimal dose coverage of the tumor and reduce dose to the surrounding healthy tissues.2

CT is essential in radiotherapy treatment planning (RTP) since it provides information on electron density using tissue specific attenuation values, which is essential in the radiation dose evaluation.2,3 It also exhibits advantages such as wide availability, minimal geometrical distortion and clear resolution of air-filled cavities and greater visibility of fatty and bony tissues.4 However, CT has poor soft tissue contrast, thus in certain circumstances MRI seems to be assistive in target delineation due to its superior anatomical soft tissue contrast.3 Consequently, MRI images are often fused and registered with CT images in contour segmentation process. The introduction of MRI has significantly reduced intra and inter-observer variability in contouring.5,6 In addition, MRI can provide functional information on tissue characteristics, which improves the accuracy of delivery and promotes dose escalation, as well as monitoring treatment and assess its response later.7-10

However, MRI is subjected to geometric distortions,2,11 and does not provide information on electron density, unless a pseudo-CT is derived from MR images for dosage evaluation.12 Another challenge for MRI is the long acquisition time making it susceptible to motion artifact, which may be reduced by ultrafast techniques.13 The introduction of a MR-guided RT (MRgRT) system eliminates the aforementioned limitations by having a high-temporal cine MRI on-board to capture the positions of the tumor and OAR in real-time. These 4D-MRI are becoming more commercially available, which ensures there will be incremental utilization of MRI into the field of RT.14 MRI is a highly versatile modality with numerous available sequences. Properties of tumors and their surrounding tissues differ; therefore, the ideal sequence for every site varies accordingly. Here, we present a narrative review on the utilization and selection of MR sequences for target delineation in RTP.

Target delineation and sequence selection

The primary goal of implementing MRI into RTP is to precisely delineate Gross Tumour Volume (GTV)/Clinical Target Volume (CTV)/Organs at Risk (OAR), this is especially essential in intensity modulated radiation therapy (IMRT)/stereotactic body radiotherapy (SBRT) delivery where steep dose gradients are present at the edge of targeted region. Target delineation is one of the earliest steps in RTP, thereby any inaccuracies can induce error propagation downstream. Adequate MR sequence selection is a critical step in achieving this goal. An ideal sequence should be accurate, reproducible with high soft tissue resolution and low intra- and interobserver variability in order to optimize the consistency in imaging interpretation.

The gold standard in evaluating the accuracy of tumor delineation is comparing image-derived GTV to the corresponding pathological specimen, preferably “whole-mounted”. This ensures the delineated GTV to represent the actual disease. However, tissue shrinkage and deformation during tissue processing and precise alignment of delineated tumour on MR with the pathological reference remains a challenge. Therefore, a robust and a standardized process of tissue preparation and registration methods are desirable, that remained a challenge till now. A surrogate way to address these uncertainties is to compare the volume delineated by multiple observers, with the presumption that the volume accepted by the majority represents the actual volume, or is comparable to the volume delineated on MRI with that of other imaging modalities. However, such studies investigate the inter-observer variability component, also demonstrate a lack of consistency in various aspects making sequence selection a real challenging decision.15

It is worth noting that while the sequence selection is crucial, several other factors need to be considered when implementing MRI into RTP, including patient positioning and immobilization, scan protocols, control for respiratory motion, peristaltic motion or OAR filling differences where relevant, geometric distortion correction and quality assurance program.2

MRI sequences

Fundamentally, the sequences can be either spin-echo- (SE) or gradient-echo-based (GE) with addition of other parameters. Basic SE sequence has a long acquisition time so it is susceptible to motion artifacts; however, it has high spatial resolution and provides the gold standard in intensity distortion. Turbo SE (TSE) is an SE-based sequence that can substantially accelerate scanning speed, and is therefore widely used in RTP. Other SE-based sequences include fluid-attenuation inversion recovery (FLAIR) and short tau inversion recovery (STIR) both are useful in brain imaging and capturing bone marrow changes, respectively. In comparison to SE, GE has a faster acquisition time, therefore has minimal motion artifact, but with a lower spatial resolution and is susceptible to field inhomogeneities-induced artifact. Such local field inhomogeneities can arise from metal implant-induced magnetic field change, thus should be avoided in patients with metallic prosthesis.4

Conventionally, the signal arising during the decay (relaxation) of proton magnetic moments after being excited by radio-waves within a strong magnetic field are exploited to generate images. The relative difference in tissue relaxation times defines T1 (longitudinal)- or T2 (transverse)-weighted images. However, if the properties that give rise to the parameters for T1w and T2w image generation do not differentiate tumor tissues from surrounding healthy tissue, these sequences will not be useful for tumor delineation such as T1w in prostate tumor.4,16

More sophisticated MRI techniques such as functional MRI focus on the biological status of the tumor. These sequences emphasize various intrinsic properties of the tissue, such as proton density, perfusion, diffusion and chemical composition. The highlighted properties, or “imaging biomarkers” can exist in tumor microstructure, differentiating them from surrounding healthy tissues, therefore can be used to complement anatomical information for target definition (including intra-tumor spatial heterogeneity), assess aggressiveness for dosage mapping, monitor treatment response and early prediction for treatment toxicity, re-occurrence and prognosis.17

As with bio-specimen biomarkers, the sensitivity and specificity of these sequences alters with the change in spatio-temporal profile of a tumor, most of the functional sequences are affected by multiple underlying biological processes. Therefore, sufficient biological and clinical validations are required before clinical implementation.

Diffusion weighted imaging (DWI)

DWI constructs image based on the mobility of water molecules in tissues, which can be quantitatively measured by applying a mono-exponential model, called apparent diffusion coefficient (ADC) value, or biexponentially as intravoxel incoherent motion (IVIM) to assess tissue perfusion and cell density concomitantly.18 The mobility of water molecules is affected by several factors, including cellular space structure, membrane permeability, interstitial viscosity and cellular density.19 These properties are valuable tools in identifying abnormal structures. For example, mobility of water is more restricted in hypercellular tissues such as malignant tumors and less restricted in glandular tissue, inflammation or tissues with significant necrosis.16 These correlations have been validated with histological findings.20

The primary use of DWI in RTP is to assist target delineation, particularly the gross tumor in supplement to the conventional sequences. In addition, DWI can be used in monitoring treatment response, since tumor tissue undergoes regression and necrosis in response to daily radiation treatment, which leads to increased mobility of water molecules, reduced DWI signal and increased ADC.2 DWI has also shown promising results in identifying the early radiation treatment response in patients with esophageal cancer, pancreatic cancer, locally advanced rectal cancer and cervical cancer.9,21

The limitation of DWI is its susceptibility to the geometric distortion, especially with single-shot spin-echo echo-planar imaging (SE-EPI), which is often used for DWI. Geometric distortion is more prominent when b-value is increased for a higher sensitivity.22 A reduction in distortion is desired not only for the delineation purposes, but also to achieve better anatomical overlap with images obtained using other sequences for validation. To reduce distortion, selection of b-values are critical, and multi-shot sequences, segmented EPI, TSE-based readouts or non-EPI sequences must be adopted.11

In addition, DWI is not very specific and its alteration may be affected by many factors such as reduced cellular density as a result of necrosis or apoptosis, altered vasculature or cellular membrane permeability.23 Nonetheless, DWI remains as one of the most studied functional sequence in MRI.

Dynamic contrast enhanced (DCE)

DCE MRI involves an intravenous injection of paramagnetic contrast agent, usually gadolinium-based. Sequential T1w images are captured as the contrast medium pass through vessels, changes could reflect overall and microvascular perfusion, permeability, and extracellular leakage space.24 Similar to DWI, DCE can be quantitatively measured based on pharmacokinetic modeling, deriving the parameters such as transfer coefficient (Ktrans) for permeability and perfusion and extravascular extracellular space volume (Ve) or rate constant (Kep). (24)

DCE can measure tissue oxygenation and has been shown to correlate with tumor response to radiation in sites including brain, head and neck (HNC), liver, prostate, and non-small cell lung cancers (NSCLC).7,8,25 This is based on the fact that hypoxia, a feature in cancer due to the imbalance between oxygen supply and consumption, is associated with tumor aggressiveness and radio-resistance.26 DCE is also useful in distinguishing recurrent tumor, that are highly vascularized with increased permeability from radiation necrosis.27

DCE has a high spatial resolution and is not susceptible to artifacts. However, T1 signal intensity and contrast agent concentration are not linearly correlated, which tends to cause inaccuracy in quantifying the hemodynamic parameters.28 The image acquired on DCE is multifactorial including tissue relaxation time, permeability, vascularity and hypoxia, it is therefore not specific. In addition, scan parameters such as flip angle, repetition time and pre-contrast signal can all have impact on images. Although these limitations might be surmounted by combining with other sequences, they still impose obstacles for comparison across centers.29

MR spectroscopy (MRS)

MRS depicts the metabolic status of tissues by detecting the radiofrequency signals generated by endogenous nuclei such as 1H, 31P, 13C, and 19F, then the relative compositions of tissue metabolites can be measured.24 In particular, diminished citrate level is observed in tumor tissue because of the change in metabolic pathway. Choline (Cho) is a metabolite produced during cell membrane biosynthesis, therefore a measure of cellular turnover. Elevated Cho may be observed in the presence of brain cancer. Similarly, creatine (Cr) that represents cellularity and metabolism may also be elevated.30 Other common metabolites include N-acetylaspartate (NAA), a representation of neural density hence an indicator of neuronal injury, and lipid and lactate for hypoxia.31

The analysis and interpretation of MRS data may require extra expertise which limits its clinical use. The artefact from air-tissue interface in sites like HNC may also have impact on the data quality and affect interpretation. In body regions such as brain, prostate and breast, although MRS has shown promise in diagnosis and treatment response monitoring, its clinical application is still limited by the relatively low signal strength and sensitivity.32

Other sequences

Blood-Oxygen-Level-Dependent (BOLD) sequence is popular in examining neural function, where deoxyhemoglobin causes the signal change.33 It might be useful in delineating critical neuronal structures, identifying tumor, or predicting response by detecting areas of hypoxia.34 Tumour-oxygen-level dependent (TOLD) is sensitive to the alteration in the amount of oxygen molecules and may be integrated with DCE for hypoxia assessment.35 MR elastography (MRE) can quantitatively measure the biomechanical properties such as elasticity and viscosity, and has been investigated in the context of breast cancer and liver fibrosis. Chemical exchange saturation transfer MRI (CEST) can quantify smaller macromolecules, such as glucose metabolism, which could be used to assess tumor aggressiveness and treatment response.36

Use of MR sequences in specific tumour sites

Brain: MRI is a part of routine target volume delineation and treatment response assessment in brain cancers,37 and is described as the “gold standard” in stereotactic radiosurgery (SRS) treatment for brain tumor by American Association of Physicists in Medicine (AAPM) Task Group (TG-101).38

The introduction of MRI allowed better tumor and OAR delineation, and significantly reduced intra- and interobserver variability.39 Currently, both European Organization for Research and Treatment of Cancer (EORTC) and US/Canadian Radiotherapy and Oncology Group (RTOG) recommend native and contrast enhanced T1w (T1CE) and thin sliced T2/FLAIR to be used in delineation of glioma in RT.40,41 The choice of particular sequences depends on the grade of glioma and whether there is an intact blood brain barrier (BBB). For example, T2/FLAIR is recommended for detection of low-grade glioma with an intact BBB for delineation, whereas T1CE is indicated for high grade glioma or to exclude possible high-grade transformation.42 The downside of conventional MRI techniques is that the use of single sequence may underestimate the distribution of malignant cells. Thus, T2/FLAIR abnormalities are taken into consideration especially in non-resected gliomas.37

Functional MRI is perhaps most extensively used in characterizing intracranial lesions. While T1CE alone only has 50% specificity in differentiating tumor progression from radio-necrosis.43 100% sensitivity and 83% specificity has been demonstrated by Bisdas et al in using Ktrans derived from DCE for glioma recurrence detection.8 DCE has also been shown to predict glioma with high potential of aggressiveness.44 In addition, a sub-form of DWI, diffusion tensor (DTI) has been shown to improve the delineation of HGG than T1w alone.45 Its quantification fractional anisotropy is helpful in delineating and sparing neural tracts.46,47 DTI also has the potential to predict the invasiveness of high-grade tumor and pattern of recurrence.48,49

Radio-labelled amino acid PET scan has been shown to be useful for the identification of tumor extent as there is a higher uptake in biologically active tumor tissue and low in normal brain,50,51 and has been recommended by the Response Assessment in Neuro-oncology (RANO), European Association for Neuro-Oncology (EANO) and ESTRO.52 MRI has a sensitivity of 96% in detecting tumor tissue, but only 53% specificity, when MRI and PET are combined, the sensitivity becomes 93%, and specificity raises to 94%.53 There is also less interobserver variability, especially at the skull base.54 Therefore, an approach with multiparametric MRI in combination with CT and amino acid PET appears to be the ideal way of target volume delineation. These modalities are still being experimented, but surely will be contributing heftily to RTP in the future.42

Head and neck: Radiation dose delivery is intrinsically challenging in the head and neck region due to the close proximity of critical normal structures to the tumor. Late treatment-related toxicities such as xerostomia and dysphagia are common and can significantly impact quality of life of the patients.55

Currently, the mainstream strategy is complementing CT with the soft tissue information from MRI.56 T2w fast spin echo (FSE) with fat suppression and post contrast T1w FSE with or without suppression are most commonly used sequences in both diagnostic and RTP.57 In nasopharyngeal cancer, T1CE can uncover up to 40% of patients with intracranial infiltration that is missed on CT alone and MRI is helpful in identifying subtle bony invasion and has less interobserver variability. In Sinonasal cancer, conventional sequences are helpful in identifying tumor spread identified as hyperintensity on T2w and less enhancement on T1CE.58 In oropharyngeal tumors, T1CE allowed better base of tongue tumor delineation, and T2w/FLAIR are thought to be ideal for parotid imaging.59 However, for laryngeal and hypo-pharyngeal tumors, MRI does not show superiority over CT when compared with pathological specimen.60,61

Functional sequences, in particular DWI, has been extensively examined in HNC. The studies mainly focus on identifying regions within the tumor for dosage escalation and personalize oncological therapy based on their response prediction.62,63 Several studies have highlighted the high specificity and sensitivity of DWI in detecting metastatic lymph nodes, where significantly lower ADC is observed. DWI is exceptionally useful in sub-centrimetric cervical lymph nodes, where the sensitivity is increased to 76% in comparison to 7% in conventional MRI. These findings hold true when compared with histopathological studies.64-66

DWI can also be used to predict treatment response after RT or chemoradiation therapy (CRT). Patients with low pre-treatment baseline ADC corresponds to better response.63,67 The early prediction of tumor response could potentially allow timely adaptation of RTP or further personalization of treatment dose, potentially dose de-escalation for the early responders. Several studies have demonstrated a significantly elevated ADC compare to pre-treatment baseline in the complete responders.63 This may be explained by a loss of tumor integrity from RT.57 In addition, DWI is insensitive to the acute inflammation and studies have shown that DWI demonstrates a higher positive predictive value than anatomical imaging in detecting early reoccurrence.68

Majority of the studies on the application of DCE in HNC focuses on prediction and early assessment of treatment response. A correlation of an increased or high overall Ktrans with good treatment outcome was suggested.69,70 The data on the use of functional MRI in HNC remained preliminary, especially for DCE and MRS. Hence; further detailed research is desired before their integration and incorporation into routine clinical practices.

Lungs: RT plays a significant role in lung cancer treatment, especially in inoperable non-small cell lung cancer (NSCLC) patients that constitutes the majority in this cohort.71 The introduction of SBRT escalated tumor control to 98% in early-stage NSCLC, with a 17% risk of grade 3–4 toxicity.72,73 The high dose per fraction can potentially cause significant toxicities hence particular attention on OARs is required to ensure safe treatment delivery. Patients with more centrally located tumor (< 2cm from main proximal bronchial tree) are traditionally excluded from SBRT treatment due to proximity of tumor to the central normal structures such as oesophagus, heart, spinal cord, brachial plexus, central airway, major vessels and chest wall.72,74

The current workflow utilizes 4DCT and F-18-FDG PET for tumor delineation.75 Inter-fractional shifts from primary tumor and vertebrae ranges from 5-7 mm but can be as high as 3 cm on CT.76 CT produces larger uncertainties of up to a few centimeters for spinal cord and oesophagus owing to its low tissue contrast.75 For PET scan, the low spatial resolution of 5-7mm means tumors of less than 4 mm may not be detected.77

MRI can differentiate lung tumor from other pathologies such as lung collapse, consolidation or effusion better than CT or PET.16 There are two major challenges encountered when choosing sequences, to reduce the poor signal-to-noise ratio (SNR) due to low tissue density of lung parenchyma and respiration and cardiac motion induced artefacts.78 Sequences have been investigated to quantify lung motion include fast low-angle shot (FLASH) and true fast imaging with steady-state precession (TrueFISP). The former acquires 3 images per second, and the later acquires 10 with a sub-optimal signal-to-noise ratio. Both of them generated the images of diagnostic quality.79 Kumar et al.80 reviewed 30 articles concerning anatomical detection, MRI-based motion analysis and functional imaging for lung tumors and pulmonary nodules. Various sequences had been explored deeply, including volumetric interpolated breath-hold (VIBE), a T1w 3D spoiled GRE sequence, T1w and T2w TSE, half-Fourier acquisition single-shot TSE and inverse-recovery (IR) sequences. However, it still remains a challenge to optimally balance acquisition time, signal-to noise ratio and susceptibility to motion artifacts. They suggested that although anatomical tumor infiltration and mediastinal lymph nodes could be demonstrated by conventional sequences, their use in radiation oncology for GTV delineation warrants further investigation for possible tumor deformation during respiration.

Bainbridge et al. reviewed the inter-observer variability and commented that the addition of PET to CT reduced inter-observer variation from 1.0 mm to 0.4 mm.74,75 Adding MR sequences to CT and PET did not result in further improvement in interobserver variability, although this might be related to the lack of experiences in interpreting MRI contouring.78

As in other locations, studies have been investigating the use of functional MRI in lung cancer. In animal models DCE was able to differentiate radiation induced pneumonitis and fibrosis.81 DWI can identify critical OAR sub-structures such as brachial plexus in Pancoast tumor allowing careful design of RT plans to reduce dose to the plexus.82 DWI also shows comparable capability to PET in demonstrating malignant lymph nodes with higher specificity whenever there is inflammation.83 Similar to FDG-PET, DWI was able to differentiate tumor from consolidation.84 Unfortunately, DCE was not shown to be useful in predicting tumor response with consistency in lung cancer. Other imaging such as hyper-polarised 3He and 129Xe have been studied for delineating OAR by identifying healthy well-ventilated regions of the lung in advanced NSCLC with a promising result.85,86

Pancreas: Less than 20% of patients with pancreatic cancer present with resectable disease.87 In patients with inoperable disease, definitive conventional CRT serves as consolidation therapy following chemotherapy.88,89 Although CRT leads to the improved local control, GI toxicity was observed with no additional benefits in overall survival compared to chemotherapy alone.90,91 The introduction of SBRT has since substantially reduced OAR toxicity and may improve overall survival.88,92,93

Despite of the controversial role of RT in pancreatic cancer, current imaging strategies utilize contrast enhanced CT complemented with PET/CT for tumor delineation.88 Studies comparing GTV delineated with different imaging modalities with surgical specimen have shown mixed results and no optimal imaging modalities were identified.94-99 One study recommended T1w for pancreatic GTV visualization and T2w to assess the extent of pancreatic tumor when it compressed pancreatic or common bile duct.94 Previous study showed DWI was able to achieve a sensitivity of 0.86 and a specificity of 0.91 in detecting pancreatic tumor,100 however, as it was difficult to differentiate pancreatic cancer from pancreatitis or normal tissue using DWI, it is not recommended for tumor contouring.94 A small study demonstrated ADC increases in post-treatment and correlates with the degree of response.99

Cervical: The recommended definitive treatment for locally advanced cervical cancer involves concomitant EBRT and chemotherapy, followed by brachytherapy.101,102 MRI has been shown to be able to discriminate GTV from adjacent normal uterine tissue and surrounding OARs, namely the bladder, rectum, bowel and vagina.103 MRI is recognized by RTOG and GEC ESTRO as the most precise and reliable modality in gynecological tumor delineation.104-106 MRI also allowed dose escalation with 10-20% survival gains with reduced RT related GI and urinary morbidity.106

In particular, T2w sequences gives a better contrast at the boundary of the tumor than T1w, whereas ADC gives a smaller GTV than T2w.107 Conflicting results were achieved when comparing GTV delineated with T2w to that of surgical specimens.108,109 The addition of ADC increased the contrast and lead to less inter-observer variability, despite its lower anatomical resolution.110 ADC also uncovers up to 16% of suspicious regions that is absent on T2w. Therefore, rather than mapping individually, a combination of T2w and ADC may be a better solution.107 Nevertheless, the latest GEC ESTRO guidelines recommend a contouring strategy using T2w sequence as the gold standard for tumor delineation in image guided adaptive brachytherapy (IGABT), whereas T1CE and DWI are complementary and optional.105 One study has demonstrated that the three different targets (GTV, high-risk clinical tumor volume and intermediate-risk clinical target volume) as defined by GEC ESTRO exhibit significantly different ADC, their ADC remained stable during the course of brachytherapy, therefore it has the potential to be beneficial for better delineation in the grey zone on T2w.111 However, DWI is susceptible to geometric distortions and artifact induced by titanium applicator, therefore its use in brachytherapy may be limited in certain scenario.112

In patients treated with neoadjuvant CRT, post-treatment skewness of the ADC histogram and percentage change in ADC predicts and favorable response.21,113

Prostate: It is well established that EBRT is the standard and effective treatment for localized prostate cancer.114,115 Both ASTRO and National Comprehensive Cancer Network (NCCN) guidelines suggest that the introduction of IGRT is beneficial in improving treatment outcome and reducing OAR toxicity.116

MRI is superior to CT in depicting the prostate capsule and the intra-prostatic heterogeneity.117 It generates less inter-observer variability and less toxicity to OAR while maintaining comparable tumor control rates.118-120 These benefits have also been suggested by ACR Appropriateness Criteria.121 In addition, hip prosthesis in patient would produce less image distortion in MRI compared to CT.122

The ESTRO guidelines conclude that T2w MRI is the best modality in differentiating the prostate and the peri-prostatic structures.117 Studies show that the utilization of MRI resulted in a smaller CTV of about 10-35% than CT alone.6,123 MRI is useful in delineating the apex and the base, where a higher incidence of tumor occurs.5,124 CT-MRI delineation leads to significantly lower urinary frequency and urinary retention toxicity scores than CT-only delineation, although no significant difference in overall urinary or rectal toxicity were observed.125

Apart from evaluating extra-capsular and seminal vesicle invasion, a key advantage of MRI in RTP is the ability to visualize intra-prostatic lesions.126 Prostate cancer is known to be heterogeneous and multifocal. This property renders the healthy tissue to be susceptible to toxicity if the whole gland is targeted. In addition, local recurrences often occur at the original site of dominant intra-prostatic lesion.127-129 Therefore, a focal boost to regions of high tumor burden is logically beneficial, and may be visualized with the help of DCE.25 Several studies demonstrated that focal dose escalation can be achieved by delineating prostate with MRI without causing more toxicity to rectal walls, and has good clinical feasibility.10,130-132

Sequences including T2w, DWI, DCE and MRS have been investigated extensively for prostate cancer.9,125,133 A multi-parametric (mpMRI) approach combining T2w, DWI and DCE yields the highest sensitivity in intra-prostatic tumor nodule detection than each modality alone when compared to whole mount histology.134 However, PI-RADS2.1v and several other studies suggest that mpMRI underestimate tumor volume and extent compared to histology.135-138

In addition, ADC can be used to identify the aggressiveness of the lesions that are most likely to benefit from dosage escalation. Studies have also shown that there is marked increase in ADC after treatment, which positively correlates with good outcomes.9

Rectum: Patients with locally advanced rectal tumor are recommended to have neoadjuvant RT prior to surgical resection.139 There are currently no guidelines available on the choice of MR sequences for tumor delineation. As T2w images is known to be the gold standard for staging because it captures the three layers of rectal wall as well as the mesorectal fascia relatively well, it is therefore adapted to tumor delineation, though it is recommended that it be performed in RT treatment position.4

There are few studies investigated the use of MRI in GTV delineation. To date most of them are investigating inter and intra-observer variation in target delineation. There is little data on the correlation of MRI contouring with pathology specimen. This is mainly because in neoadjuvant therapy the planning process followed by CRT will cause tumor regression and deformation.140 In contouring studies, T2w yields better inter-observer consistency with a larger tumor volume than DWI, and the addition of DWI to T2w can increase sensitivity from 82-84% to 93-95%.140,141

DWI has also been studied for assessing the response to neoadjuvant CRT. Similar to HNC, a lower pre-treatment ADC in rectal tumor corresponded to improved treatment response.142,143 This may be explained by less necrosis in tissues with low ADC, which is correlated to better tissue perfusion, higher oxygen concentration and a non-acidic microenvironment, therefore more susceptibility to treatment. Some studies suggest that responding lesions exhibit an increase in ADC during and after CRT, in contrast to the non-responding lesions, which remain stable to pre-treatment value,143-146 while others proposed that DWI volumetry, rather than ADC value is more response-predictive.147,148

Conclusion

In summary, over the past decades, there has been a huge advance in imaging technologies in radiation treatment for cancer. The advantage of better soft tissue contrast and lack of ionizing radiation made MR an attractive imaging modality for RTP and frequent treatment response assessment during treatment Conventional sequences, such as T1w and T2w images allow good anatomical delineation in the majority of tumor sites. However, various functional sequences focusing on water diffusion, perfusion, and chemical properties may contribute extra layers of value. They may be used complementary to anatomical information, providing biological information of the tumor and therefore allow assessment of tumor aggressiveness, early response and recurrence prediction and post-treatment monitoring. Although the use of MRI is still in infancy in some tumor sites, it is a promising tool that is worth exploring in the era of MRgRT, with the aim of further treatment and dose personalization for patients.

Acknowledgments

None.

Conflicts of interest

Author declares there are no conflicts of interest.

References

  1. Citrin DE. Recent developments in radiotherapy. N Engl J Med. 2017;377(22):2200–2201.
  2. Chandarana H, Wang H, Tijssen RHN, et al. Emerging role of MRI in radiation therapy. J Magn Reson Imaging. 2018;48(6):1468–1478.
  3. Das IJ, McGee KP, Tyagi N, et al. Role and future of MRI in radiation oncology. Br J Radiol. 2019;92(1094):20180505.
  4. Dirix P, Haustermans K, Vandecaveye V. The value of magnetic resonance imaging for radiotherapy planning. Semin Radiat Oncol. 2014;24(3):151–159.
  5. Rasch C, Barillot I, Remeijer P, et al. Definition of the prostate in CT and MRI: a multi–observer study. Int J Radiat Oncol Biol Phys. 1999;43(1):57–66.
  6. Hentschel B, Oehler W, Strauss D, et al. Definition of the CTV prostate in CT and MRI by using CT–MRI image fusion in IMRT planning for prostate cancer. Strahlenther Onkol. 2011;187(3):183–190.
  7. Tao X, Wang L, Hui Z, et al. DCE–MRI perfusion and permeability parameters as predictors of tumor response to CCRT in patients with locally advanced NSCLC. Sci Rep. 2016;6:35569.
  8. Bisdas S, Naegele T, Ritz R, et al. Distinguishing recurrent high–grade gliomas from radiation injury: a pilot study using dynamic contrast–enhanced MR imaging. Acad Radiol. 2011;18(5):575–583.
  9. Liu L, Wu N, Ouyang H, et al. Diffusion–weighted MRI in early assessment of tumour response to radiotherapy in high–risk prostate cancer. Br J Radiol. 2014;87(1043):20140359.
  10. Lips IM, van der Heide UA, Haustermans K, et al. Single blind randomized phase III trial to investigate the benefit of a focal lesion ablative microboost in prostate cancer (FLAME–trial): study protocol for a randomized controlled trial. Trials. 2011;12:255.
  11. Datta A, Aznar MC, Dubec M, et al. Delivering functional imaging on the MRI–Linac: current challenges and potential solutions. Clin Oncol (R Coll Radiol). 2018;30(11):702–710.
  12. Greer PB, Dowling JA, Lambert JA, et al. A magnetic resonance imaging–based workflow for planning radiation therapy for prostate cancer. Med J Aust. 2011;194(4):S24–S27.
  13. Barth M, Breuer F, Koopmans PJ, et al. Simultaneous multislice (SMS) imaging techniques. Magn Reson Med. 2016;75(1):63–81.
  14. Hehakaya C, Van der Voort van Zyp JR, Lagendijk JJW, et al. Problems and pomises of introducing the magnetic resonance imaging linear accelerator into routine care: the case of prostate cancer. Front Oncol. 2020;10:1741.
  15. Vinod SK, Jameson MG, Min M, et al. Uncertainties in volume delineation in radiation oncology: a systematic review and recommendations for future studies. Radiother Oncol. 2016;121(2):169–179.
  16. Metcalfe P, Liney GP, Holloway L, et al. The potential for an enhanced role for MRI in radiation–therapy treatment planning. Technol Cancer Res Treat. 2013;12(5):429–446.
  17. García–Figueiras R, Baleato–González S, Padhani AR, et al. How clinical imaging can assess cancer biology. Insights Imaging. 2019;10(1):28.
  18. Jones KM, Michel KA, Bankson JA, et al. Emerging magnetic resonance imaging technologies for radiation therapy planning and response assessment. Int J Radiat Oncol Biol Phys. 2018;101(5):1046–1056.
  19. Le Bihan D. Apparent diffusion coefficient and beyond: what diffusion MR imaging can tell us about tissue structure. Radiology. 2013;268(2):318–322.
  20. Xu J, Humphrey PA, Kibel AS, et al. Magnetic resonance diffusion characteristics of histologically defined prostate cancer in humans. Magn Reson Med. 2009;61(4):842–850.
  21. Barbaro B, Vitale R, Valentini V, et al. Diffusion–weighted magnetic resonance imaging in monitoring rectal cancer response to neoadjuvant chemoradiotherapy. Int J Radiat Oncol Biol Phys. 2012;83(2):594–599.
  22. Embleton KV, Haroon HA, Morris DM, et al. Distortion correction for diffusion–weighted MRI tractography and fMRI in the temporal lobes. Hum Brain Mapp. 2010;31(10):1570–1587.
  23. Driessen JP, Caldas–Magalhaes J, Janssen LM, et al. Diffusion–weighted MR imaging in laryngeal and hypopharyngeal carcinoma: association between apparent diffusion coefficient and histologic findings. Radiology. 2014;272(2):456–463.
  24. Desar IM, van Herpen CM, van Laarhoven HW, et al. Beyond RECIST: molecular and functional imaging techniques for evaluation of response to targeted therapy. Cancer Treat Rev. 2009;35(4):309–321.
  25. Puech P, Potiron E, Lemaitre L, et al. Dynamic contrast–enhanced–magnetic resonance imaging evaluation of intraprostatic prostate cancer: correlation with radical prostatectomy specimens. Urology. 2009;74(5):1094–1099.
  26. Overgaard J. Hypoxic radiosensitization: adored and ignored. J Clin Oncol. 2007;25(26):4066–4074.
  27. Larsen VA, Simonsen HJ, Law I, et al. Evaluation of dynamic contrast–enhanced T1–weighted perfusion MRI in the differentiation of tumor recurrence from radiation necrosis. Neuroradiology. 2013;55(3):361–369.
  28. Beaton L, Bandula S, Gaze MN, et al. How rapid advances in imaging are defining the future of precision radiation oncology. Br J Cancer. 2019;120(8):779–790.
  29. Kim H. Variability in quantitative DCE–MRI: sources and solutions. J Nat Sci. 2018;4(1):e484.
  30. Weybright P, Sundgren PC, Maly P, et al. Differentiation between brain tumor recurrence and radiation injury using MR spectroscopy. AJR Am J Roentgenol. 2005;185(6):1471–1476.
  31. Poptani H, Gupta RK, Roy R, et al. Characterization of intracranial mass lesions with in vivo proton MR spectroscopy. AJNR Am J Neuroradiol. 1995;16(8):1593–603.
  32. Horská A, Barker PB. Imaging of brain tumors: MR spectroscopy and metabolic imaging. Neuroimaging Clin N Am. 2010;20(3):293–310.
  33. Hammond EM, Asselin MC, Forster D, et al. The meaning, measurement and modification of hypoxia in the laboratory and the clinic. Clin Oncol (R Coll Radiol). 2014;26(5):277–88.
  34. Wu GY, Suo ST, Lu Q, et al. The value of blood oxygenation level–dependent (BOLD) MR imaging in differentiation of renal solid mass and grading of renal cell carcinoma (RCC): analysis based on the largest cross–sectional area versus the entire whole tumour. PLoS One. 2015;10(4):e0123431.
  35. Young IR, Clarke GJ, Bailes DR, et al. Enhancement of relaxation rate with paramagnetic contrast agents in NMR imaging. J Comput Tomogr. 1981;5(6):543–547.
  36. Mehrabian H, Myrehaug S, Soliman H, et al. Evaluation of Glioblastoma Response to Therapy With Chemical Exchange Saturation Transfer. Int J Radiat Oncol Biol Phys. 2018;101(3):713–723.
  37. Popp I, Weber WA, Combs SE, et al. Neuroimaging for Radiation Therapy of Brain Tumors. Topics in Magnetic Resonance Imaging. 2019;28(2):63–71.
  38. Benedict SH, Yenice KM, Followill D, et al. Stereotactic body radiation therapy: the report of AAPM Task Group 101. Med Phys. 2010;37(8):4078–101.
  39. Heesters MA, Wijrdeman HK, Struikmans H, et al. Brain tumor delineation based on CT and MR imaging. Implications for radiotherapy treatment planning. Strahlenther Onkol. 1993;169(12):729–733.
  40. Niyazi M, Brada M, Chalmers AJ, et al. ESTRO–ACROP guideline "target delineation of glioblastomas". Radiother Oncol. 2016;118(1):35–42.
  41. Colman H, Berkey BA, Maor MH, et al. Phase II Radiation Therapy Oncology Group trial of conventional radiation therapy followed by treatment with recombinant interferon–beta for supratentorial glioblastoma: results of RTOG 9710. Int J Radiat Oncol Biol Phys. 2006;66(3):818–824.
  42. Whitfield GA, Kennedy SR, Djoukhadar IK, et al. Imaging and target volume delineation in glioma. Clinical Oncology (Royal College of Radiologists). 2014;26(7):364–376.
  43. Rachinger W, Goetz C, Pöpperl G, et al. Positron emission tomography with O–(2–[18F]fluoroethyl)–l–tyrosine versus magnetic resonance imaging in the diagnosis of recurrent gliomas. Neurosurgery. 2005;57(3):505–511.
  44. Datta NR, David R, Gupta RK, et al. Implications of contrast–enhanced CT–based and MRI–based target volume delineations in radiotherapy treatment planning for brain tumors. J Cancer Res Ther. 2008;4(1):9–13.
  45. Mazzara GP, Velthuizen RP, Pearlman JL, et al. Brain tumor target volume determination for radiation treatment planning through automated MRI segmentation. Int J Radiat Oncol Biol Phys. 2004;59(1):300–312.
  46. Price SJ, Peña A, Burnet NG, et al. Tissue signature characterisation of diffusion tensor abnormalities in cerebral gliomas. Eur Radiol. 2004;14(10):1909–17.
  47. Price SJ, Jena R, Burnet NG, et al. Improved delineation of glioma margins and regions of infiltration with the use of diffusion tensor imaging: an image–guided biopsy study. AJNR Am J Neuroradiol. 2006;27(9):1969–1974.
  48. Jena R, Price SJ, Baker C, et al. Diffusion tensor imaging: possible implications for radiotherapy treatment planning of patients with high–grade glioma. Clin Oncol (R Coll Radiol). 2005;17(8):581–590.
  49. Price SJ, Jena R, Burnet NG, et al. Predicting patterns of glioma recurrence using diffusion tensor imaging. Eur Radiol. 2007;17(7):1675–1684.
  50. Grosu AL, Weber WA, Riedel E, et al. L–(methyl–11C) methionine positron emission tomography for target delineation in resected high–grade gliomas before radiotherapy. Int J Radiat Oncol Biol Phys. 2005;63(1):64–74.
  51. Oehlke O, Mix M, Graf E, et al. Amino–acid PET versus MRI guided re–irradiation in patients with recurrent glioblastoma multiforme (GLIAA) – protocol of a randomized phase II trial (NOA 10/ARO 2013–1). BMC Cancer. 2016;16(1):769.
  52. Albert NL, Weller M, Suchorska B, et al. Response Assessment in Neuro–Oncology working group and European Association for Neuro–Oncology recommendations for the clinical use of PET imaging in gliomas. Neuro Oncol. 2016;18(9):1199–1208.
  53. Pauleit D, Floeth F, Hamacher K, et al. O–(2–[18F]fluoroethyl)–L–tyrosine PET combined with MRI improves the diagnostic assessment of cerebral gliomas. Brain. 2005;128(Pt 3):678–687.
  54. Astner ST, Bundschuh RA, Beer AJ, et al. Assessment of tumor volumes in skull base glomus tumors using Gluc–Lys[(18)F]–TOCA positron emission tomography. Int J Radiat Oncol Biol Phys. 2009;73(4):1135–1140.
  55. Huang TL, Chien CY, Tsai WL, et al. Long–term late toxicities and quality of life for survivors of nasopharyngeal carcinoma treated with intensity–modulated radiotherapy versus non–intensity–modulated radiotherapy. Head Neck. 2016;38 Suppl 1:E1026–1032.
  56. Lee AW, Ng WT, Pan JJ, et al. International guideline for the delineation of the clinical target volumes (CTV) for nasopharyngeal carcinoma. Radiother Oncol. 2018;126(1):25–36.
  57. Yuan J, Lo G, King AD. Functional magnetic resonance imaging techniques and their development for radiation therapy planning and monitoring in the head and neck cancers. Quantitative imaging in medicine and surgery. 2016;6(4):430–448.
  58. Chung NN, Ting LL, Hsu WC, et al. Impact of magnetic resonance imaging versus CT on nasopharyngeal carcinoma: primary tumor target delineation for radiotherapy. Head Neck. 2004;26(3):241–246.
  59. Ahmed M, Schmidt M, Sohaib A, et al. The value of magnetic resonance imaging in target volume delineation of base of tongue tumours––a study using flexible surface coils. Radiother Oncol. 2010;94(2):161–167.
  60. Geets X, Daisne JF, Arcangeli S, et al. Inter–observer variability in the delineation of pharyngo–laryngeal tumor, parotid glands and cervical spinal cord: comparison between CT–scan and MRI. Radiother Oncol. 2005;77(1):25–31.
  61. Jager EA, Ligtenberg H, Caldas–Magalhaes J, et al. Validated guidelines for tumor delineation on magnetic resonance imaging for laryngeal and hypopharyngeal cancer. Acta Oncol. 2016;55(11):1305–1312.
  62. Hatakenaka M, Nakamura K, Yabuuchi H, et al. Pretreatment apparent diffusion coefficient of the primary lesion correlates with local failure in head–and–neck cancer treated with chemoradiotherapy or radiotherapy. Int J Radiat Oncol Biol Phys. 2011;81(2):339–345.
  63. Kim S, Loevner L, Quon H, et al. Diffusion–weighted magnetic resonance imaging for predicting and detecting early response to chemoradiation therapy of squamous cell carcinomas of the head and neck. Clin Cancer Res. 2009;15(3):986–994.
  64. Vandecaveye V, De Keyzer F, Vander Poorten V, et al. Head and neck squamous cell carcinoma: value of diffusion–weighted MR imaging for nodal staging. Radiology. 2009;251(1):134–146.
  65. Abdel Razek AA, Soliman NY, Elkhamary S, et al. Role of diffusion–weighted MR imaging in cervical lymphadenopathy. Eur Radiol. 2006;16(7):1468–1477.
  66. de Bondt RB, Hoeberigs MC, Nelemans PJ, et al. Diagnostic accuracy and additional value of diffusion–weighted imaging for discrimination of malignant cervical lymph nodes in head and neck squamous cell carcinoma. Neuroradiology. 2009;51(3):183–192.
  67. Brenet E, Barbe C, Hoeffel C, et al. Predictive value of early post–treatment diffusion–weighted mri for recurrence or tumor progression of head and neck squamous cell carcinoma treated with chemo–radiotherapy. Cancers (Basel). 2020;12(5).
  68. Vandecaveye V, Dirix P, De Keyzer F, et al. Diffusion–weighted magnetic resonance imaging early after chemoradiotherapy to monitor treatment response in head–and–neck squamous cell carcinoma. Int J Radiat Oncol Biol Phys. 2012;82(3):1098–107.
  69. Shukla–Dave A, Lee NY, Jansen JF, et al. Dynamic contrast–enhanced magnetic resonance imaging as a predictor of outcome in head–and–neck squamous cell carcinoma patients with nodal metastases. Int J Radiat Oncol Biol Phys. 2012;82(5):1837–1844.
  70. Kim S, Loevner LA, Quon H, et al. Prediction of response to chemoradiation therapy in squamous cell carcinomas of the head and neck using dynamic contrast–enhanced MR imaging. AJNR Am J Neuroradiol. 2010;31(2):262–268.
  71. Molina JR, Yang P, Cassivi SD, Schild SE, Adjei AA. Non–small cell lung cancer: epidemiology, risk factors, treatment, and survivorship. Mayo Clin Proc. 2008;83(5):584–594.
  72. Timmerman R, Paulus R, Galvin J, et al. Stereotactic body radiation therapy for inoperable early stage lung cancer. Jama. 2010;303(11):1070–106.
  73. Timmerman RD, Paulus R, Pass HI, et al. Stereotactic body radiation therapy for operable early–stage lung cancer: findings from the nrg oncology rtog 0618 Trial. JAMA Oncol. 2018;4(9):1263–1266.
  74. Bainbridge H, Salem A, Tijssen RHN, et al. Magnetic resonance imaging in precision radiation therapy for lung cancer. Transl Lung Cancer Res. 2017;6(6):689–707.
  75. Steenbakkers RJ, Duppen JC, Fitton I, et al. Reduction of observer variation using matched CT–PET for lung cancer delineation: a three–dimensional analysis. Int J Radiat Oncol Biol Phys. 2006;64(2):435–448.
  76. Bhatt AD, El–Ghamry MN, Dunlap NE, et al. Tumor volume change with stereotactic body radiotherapy (SBRT) for early–stage lung cancer: evaluating the potential for adaptive SBRT. Am J Clin Oncol. 2015;38(1):41–46.
  77. De Ruysscher D, Belderbos J, Reymen B, et al. State of the Art Radiation Therapy for Lung Cancer 2012: A Glimpse of the Future. Clinical Lung Cancer. 2013;14(2):89–95.
  78. Karki K, Saraiya S, Hugo GD, et al. Variabilities of magnetic resonance imaging–, computed tomography–, and positron emission tomography–computed tomography–based tumor and lymph node delineations for lung cancer radiation therapy planning. Int J Radiat Oncol Biol Phys. 2017;99(1):80–89.
  79. Plathow C, Klopp M, Fink C, et al. Quantitative analysis of lung and tumour mobility: comparison of two time–resolved MRI sequences. Br J Radiol. 2005;78(933):836–840.
  80. Kumar S, Liney G, Rai R, et al. Magnetic resonance imaging in lung: a review of its potential for radiotherapy. Br J Radiol. 2016;89(1060):20150431.
  81. Zou Y, Zhang M, Wang Q, et al. Quantitative investigation of solitary pulmonary nodules: dynamic contrast–enhanced MRI and histopathologic analysis. AJR Am J Roentgenol. 2008;191(1):252–259.
  82. Andreou A, Sohaib A, Collins DJ, et al. Diffusion–weighted MR neurography for the assessment of brachial plexopathy in oncological practice. Cancer Imaging. 2015;15(1):6.
  83. Mori T, Nomori H, Ikeda K, et al. Diffusion–weighted magnetic resonance imaging for diagnosing malignant pulmonary nodules/masses: comparison with positron emission tomography. J Thorac Oncol. 2008;3(4):358–364.
  84. Yang RM, Li L, Wei XH, et al. Differentiation of central lung cancer from atelectasis: comparison of diffusion–weighted MRI with PET/CT. PLoS One. 2013;8(4):e60279.
  85. Ireland RH, Bragg CM, McJury M, et al. Feasibility of image registration and intensity–modulated radiotherapy planning with hyperpolarized helium–3 magnetic resonance imaging for non–small–cell lung cancer. Int J Radiat Oncol Biol Phys. 2007;68(1):273–281.
  86. Qing K, Mugler J, Chen Q. WE–FG–206–07: Assessing the lung function of patients with non–small cell lung cancer using hyperpolarized xenon–129 Dissolved–Phase MRI2016. 3832.
  87. Gillen S, Schuster T, Meyer Zum Büschenfelde C, et al. Preoperative/neoadjuvant therapy in pancreatic cancer: a systematic review and meta–analysis of response and resection percentages. PLoS Med. 2010;7(4):e1000267.
  88. Panje C, Andratschke N, Brunner TB, et al. Stereotactic body radiotherapy for renal cell cancer and pancreatic cancer : literature review and practice recommendations of the degro working group on stereotactic radiotherapy. Strahlenther Onkol. 2016;192(12):875–885.
  89. Palta M, Godfrey D, Goodman KA, et al. Radiation therapy for pancreatic cancer: executive summary of an ASTRO clinical practice guideline. Pract Radiat Oncol. 2019;9(5):322–332.
  90. Loehrer PJ, Sr Feng Y, Cardenes H, et al. Gemcitabine alone versus gemcitabine plus radiotherapy in patients with locally advanced pancreatic cancer: an eastern cooperative oncology group trial. J Clin Oncol. 2011;29(31):4105–4112.
  91. Chauffert B, Mornex F, Bonnetain F, et al. Phase III trial comparing intensive induction chemoradiotherapy (60 Gy, infusional 5–FU and intermittent cisplatin) followed by maintenance gemcitabine with gemcitabine alone for locally advanced unresectable pancreatic cancer. Definitive results of the 2000–01 FFCD/SFRO study. Ann Oncol. 2008;19(9):1592–
  92. Arcelli A, Buwenge M, Macchia G, et al. Stereotactic body radiotherapy vs conventionally fractionated chemoradiation in locally advanced pancreatic cancer: A multicenter case–control study (PAULA–1). Cancer Med. 2020;9(21):7879–
  93. Tchelebi LT, Lehrer EJ, Trifiletti DM, et al. Conventionally fractionated radiation therapy versus stereotactic body radiation therapy for locally advanced pancreatic cancer (CRiSP): An international systematic review and meta–analysis. Cancer. 2020;126(10):2120–21
  94. Heerkens HD, Hall WA, Li XA, et al. Recommendations for MRI–based contouring of gross tumor volume and organs at risk for radiation therapy of pancreatic cancer. Pract Radiat Oncol. 2017;7(2):126–1
  95. Gurney–Champion OJ, Versteijne E, Van der Horst A, et al. Addition of MRI for CT–based pancreatic tumor delineation: a feasibility study. Acta Oncol. 2017;56(7):923–9
  96. Dalah E, Moraru I, Paulson E, et al. Variability of target and normal structure delineation using multimodality imaging for radiation therapy of pancreatic cancer. Int J Radiat Oncol Biol Phys. 2014;89(3):633–6
  97. Arvold ND, Niemierko A, Mamon HJ, et al. Pancreatic cancer tumor size on CT scan versus pathologic specimen: implications for radiation treatment planning. Int J Radiat Oncol Biol Phys. 2011;80(5):1383–13
  98. Hall WA, Mikell JL, Mittal P, et al. Tumor size on abdominal MRI versus pathologic specimen in resected pancreatic adenocarcinoma: implications for radiation treatment planning. Int J Radiat Oncol Biol Phys. 2013;86(1):102–10
  99. Dalah E, Erickson B, Oshima K, et al. Correlation of ADC With Pathological Treatment Response for Radiation Therapy of Pancreatic Cancer. Transl Oncol. 2018;11(2):391–39
  100. Wu LM, Hu JN, Hua J, et al. Diagnostic value of diffusion–weighted magnetic resonance imaging compared with fluorodeoxyglucose positron emission tomography/computed tomography for pancreatic malignancy: a meta–analysis using a hierarchical regression model. J Gastroenterol Hepatol. 2012;27(6):1027–10
  101. Eifel PJ, Winter K, Morris M, et al. Pelvic irradiation with concurrent chemotherapy versus pelvic and para–aortic irradiation for high–risk cervical cancer: an update of radiation therapy oncology group trial (RTOG) 90–01. J Clin Oncol. 2004;22(5):872–8
  102. Chino J, Annunziata CM, Beriwal S, et al. Radiation Therapy for Cervical Cancer: Executive Summary of an ASTRO Clinical Practice Guideline. Pract Radiat Oncol. 2020;10(4):220–2
  103. Dimopoulos JC, Schard G, Berger D, et al. Systematic evaluation of MRI findings in different stages of treatment of cervical cancer: potential of MRI on delineation of target, pathoanatomic structures, and organs at risk. Int J Radiat Oncol Biol Phys. 2006;64(5):1380–
  104. Gay HA, Barthold HJ, O Meara E, et al. Pelvic normal tissue contouring guidelines for radiation therapy: a Radiation Therapy Oncology Group consensus panel atlas. Int J Radiat Oncol Biol Phys. 2012;83(3):e353–e3
  105. Dimopoulos JC, Petrow P, Tanderup K, et al. Recommendations from Gynaecological (GYN) GEC–ESTRO Working Group (IV): Basic principles and parameters for MR imaging within the frame of image based adaptive cervix cancer brachytherapy. Radiother Oncol. 2012;103(1):113–1
  106. Pötter R, Haie–Meder C, Van Limbergen E, et al. Recommendations from gynaecological (GYN) GEC ESTRO working group (II): concepts and terms in 3D image–based treatment planning in cervix cancer brachytherapy–3D dose volume parameters and aspects of 3D image–based anatomy, radiation physics, radiobiology. Radiother Oncol. 2006;78(1):67–77.
  107. Song Y, Erickson B, Chen X, et al. Appropriate magnetic resonance imaging techniques for gross tumor volume delineation in external beam radiation therapy of locally advanced cervical cancer. Oncotarget. 2018;9(11):10100–
  108. Van de Schoot AJ, De Boer P, Buist MR, et al. Quantification of delineation errors of the gross tumor volume on magnetic resonance imaging in uterine cervical cancer using pathology data and deformation correction. Acta Oncol. 2015;54(2):224–2
  109. Zhang Y, Hu J, Li J, et al. Comparison of imaging–based gross tumor volume and pathological volume determined by whole–mount serial sections in primary cervical cancer. Onco Targets Ther. 2013;6:917–9
  110. Exner M, Kuhn A, Stumpp P, et al. Value of diffusion–weighted MRI in diagnosis of uterine cervical cancer: a prospective study evaluating the benefits of DWI compared to conventional MR sequences in a 3T environment. Acta Radiol. 2016;57(7):869–8
  111. Haack S, Pedersen EM, Jespersen SN, et al. Apparent diffusion coefficients in GEC ESTRO target volumes for image guided adaptive brachytherapy of locally advanced cervical cancer. Acta Oncol. 2010;49(7):978–9
  112. Sullivan T, Yacoub JH, Harkenrider MM, et al. Providing MR Imaging for Cervical Cancer Brachytherapy: Lessons for Radiologists. Radiographics. 2018;38(3):932–9
  113. Bowen SR, Yuh WTC, Hippe DS, et al. Tumor radiomic heterogeneity: Multiparametric functional imaging to characterize variability and predict response following cervical cancer radiation therapy. J Magn Reson Imaging. 2018;47(5):1388–13
  114. Hamdy FC, Donovan JL, Lane JA, et al. 10–Year Outcomes after Monitoring, Surgery, or Radiotherapy for Localized Prostate Cancer. N Engl J Med. 2016;375(15):1415–
  115. Morgan SC, Hoffman K, Loblaw DA, et al. Hypofractionated Radiation Therapy for Localized Prostate Cancer: Executive Summary of an ASTRO, ASCO, and AUA Evidence–Based Guideline. Pract Radiat Oncol. 2018;8(6):354–3
  116. Henderson DR, Tree AC, Harrington KJ, et al. Dosimetric Implications of Computerised Tomography–Only versus Magnetic Resonance–Fusion Contouring in Stereotactic Body Radiotherapy for Prostate Cancer. Medicines (Basel). 2018;5(2):32.
  117. Salembier C, Villeirs G, De Bari B, et al. ESTRO ACROP consensus guideline on CT– and MRI–based target volume delineation for primary radiation therapy of localized prostate cancer. Radiother Oncol. 2018;127(1):49–
  118. Pathmanathan AU, Schmidt MA, Brand DH, et al. Improving fiducial and prostate capsule visualization for radiotherapy planning using MRI. J Appl Clin Med Phys. 2019;20(3):27–
  119. Doemer A, Chetty IJ, Glide–Hurst C, et al. Evaluating organ delineation, dose calculation and daily localization in an open–MRI simulation workflow for prostate cancer patients. Radiat Oncol. 2015;10:37.
  120. Pathmanathan AU, McNair HA, Schmidt MA, et al. Comparison of prostate delineation on multimodality imaging for MR–guided radiotherapy. Br J Radiol. 2019;92(1095):20180948.
  121. Nguyen PL, Aizer A, Assimos DG, et al. ACR Appropriateness Criteria® Definitive External–Beam Irradiation in stage T1 and T2 prostate cancer. Am J Clin Oncol. 2014;37(3):278–88.
  122. Rosewall T, Kong V, Vesprini D, et al. Prostate delineation using CT and MRI for radiotherapy patients with bilateral hip prostheses. Radiother Oncol. 2009;90(3):325–330.
  123. Roach M,3d, Faillace–Akazawa P, Malfatti C, et al. Prostate volumes defined by magnetic resonance imaging and computerized tomographic scans for three–dimensional conformal radiotherapy. Int J Radiat Oncol Biol Phys. 1996;35(5):1011–1018.
  124. Wachter S, Wachter–Gerstner N, Bock T, et al. Interobserver comparison of CT and MRI–based prostate apex definition. Clinical relevance for conformal radiotherapy treatment planning. Strahlenther Onkol. 2002;178(5):263–268.
  125. Sander L, Langkilde NC, Holmberg M, et al. MRI target delineation may reduce long–term toxicity after prostate radiotherapy. Acta Oncol. 2014;53(6):809–814.
  126. De Meerleer G, Villeirs G, Bral S, et al. The magnetic resonance detected intraprostatic lesion in prostate cancer: planning and delivery of intensity–modulated radiotherapy. Radiother Oncol. 2005;75(3):325–333.
  127. Pucar D, Hricak H, Shukla–Dave A, et al. 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(1):62–69.
  128. Arrayeh E, Westphalen AC, Kurhanewicz J, et al. Does local recurrence of prostate cancer after radiation therapy occur at the site of primary tumor? Results of a longitudinal MRI and MRSI study. Int J Radiat Oncol Biol Phys. 2012;82(5):e787–793.
  129. Cellini N, Morganti AG, Mattiucci GC, et al. 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(3):595–599.
  130. Singh AK, Guion P, Sears–Crouse N, et al. Simultaneous integrated boost of biopsy proven, MRI defined dominant intra–prostatic lesions to 95 Gray with IMRT: early results of a phase I NCI study. Radiat Oncol. 2007;2:36.
  131. Fonteyne V, Villeirs G, Speleers B, et al. Intensity–modulated radiotherapy as primary therapy for prostate cancer: report on acute toxicity after dose escalation with simultaneous integrated boost to intraprostatic lesion. Int J Radiat Oncol Biol Phys. 2008;72(3):799–807.
  132. Ippolito E, Mantini G, Morganti AG, et al. Intensity–modulated radiotherapy with simultaneous integrated boost to dominant intraprostatic lesion: preliminary report on toxicity. Am J Clin Oncol. 2012;35(2):158–162.
  133. Haider MA, Chung P, Sweet J, et al. Dynamic contrast–enhanced magnetic resonance imaging for localization of recurrent prostate cancer after external beam radiotherapy. Int J Radiat Oncol Biol Phys. 2008;70(2):425–430.
  134. Isebaert S, Van den Bergh L, Haustermans K, et al. Multiparametric MRI for prostate cancer localization in correlation to whole–mount histopathology. J Magn Reson Imaging. 2013;37(6):1392–1401.
  135. Baco E, Ukimura O, Rud E, et al. Magnetic resonance imaging–transectal ultrasound image–fusion biopsies accurately characterize the index tumor: correlation with step–sectioned radical prostatectomy specimens in 135 patients. Eur Urol. 2015;67(4):787–794.
  136. Radtke JP, Schwab C, Wolf MB, et al. Multiparametric Magnetic Resonance Imaging (MRI) and MRI–Transrectal Ultrasound Fusion Biopsy for Index Tumor Detection: Correlation with Radical Prostatectomy Specimen. Eur Urol. 2016;70(5):846–853.
  137. Rud E, Klotz D, Rennesund K, et al. Detection of the index tumour and tumour volume in prostate cancer using T2–weighted and diffusion–weighted magnetic resonance imaging (MRI) alone. BJU Int. 2014;114(6b):E32–E42.
  138. Barrett T, Rajesh A, Rosenkrantz AB, et al. PI–RADS version 2.1: one small step for prostate MRI. Clin Radiol. 2019;74(11):841–852.
  139. Feeney G, Sehgal R, Sheehan M, et al. Neoadjuvant radiotherapy for rectal cancer management. World J Gastroenterol. 2019;25(33):4850–4869.
  140. Burbach JP, Kleijnen JP, Reerink O, et al. Inter–observer agreement of MRI–based tumor delineation for preoperative radiotherapy boost in locally advanced rectal cancer. Radiother Oncol. 2016;118(2):399–407.
  141. Regini F, Gourtsoyianni S, Cardoso De Melo R, et al. Rectal tumour volume (GTV) delineation using T2–weighted and diffusion–weighted MRI: Implications for radiotherapy planning. Eur J Radiol. 2014;83(5):768–72.
  142. Lambrecht M, Vandecaveye V, De Keyzer F, et al. Value of diffusion–weighted magnetic resonance imaging for prediction and early assessment of response to neoadjuvant radiochemotherapy in rectal cancer: preliminary results. Int J Radiat Oncol Biol Phys. 2012;82(2):863–870.
  143. Intven M, Reerink O, Philippens ME. Diffusion–weighted MRI in locally advanced rectal cancer : pathological response prediction after neo–adjuvant radiochemotherapy. Strahlenther Onkol. 2013;189(2):117–122.
  144. Song I, Kim SH, Lee SJ, et al. Value of diffusion–weighted imaging in the detection of viable tumour after neoadjuvant chemoradiation therapy in patients with locally advanced rectal cancer: comparison with T2 weighted and PET/CT imaging. Br J Radiol. 2012;85(1013):577–586.
  145. Jiménez de Los Santos ME, Reyes–Pérez JA, Sandoval–Nava RM, et al. The apparent diffusion coefficient is a useful biomarker in predicting treatment response in patients with locally advanced rectal cancer. Acta Radiol Open. 2020;9(9):2058460120957295.
  146. Shaverdian N, Yang Y, Hu P, et al. Feasibility evaluation of diffusion–weighted imaging using an integrated MRI–radiotherapy system for response assessment to neoadjuvant therapy in rectal cancer. Br J Radiol. 2017;90(1071):20160739.
  147. Bostel T, Dreher C, Wollschläger D, et al. Exploring MR regression patterns in rectal cancer during neoadjuvant radiochemotherapy with daily T2– and diffusion–weighted MRI. Radiat Oncol. 2020;15(1):171.
  148. Curvo–Semedo L, Lambregts DM, Maas M, et al. Rectal cancer: assessment of complete response to preoperative combined radiation therapy with chemotherapy––conventional MR volumetry versus diffusion–weighted MR imaging. 2011;260(3):734–743.
Creative Commons Attribution License

©2021 Xue, et al. This is an open access article distributed under the terms of the, which permits unrestricted use, distribution, and build upon your work non-commercially.