Next Article in Journal
The Health Impacts of Better Access to Axicabtagene Ciloleucel: The Case of Spain
Previous Article in Journal
Prospective Study of Non-Contrast, Abbreviated MRI for Hepatocellular Carcinoma Surveillance in Patients with Suboptimal Hepatic Visualisation on Ultrasound
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Harnessing the Power of Radiotherapy for Lung Cancer: A Narrative Review of the Evolving Role of Magnetic Resonance Imaging Guidance

by
Sarah Hsin Cheng
1,
Shao-Yun Lee
2 and
Hsin-Hua Lee
3,4,5,6,*
1
Department of Clinical Education and Training, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
2
Department of Medical Education, Taichung Veterans General Hospital, Taichung 407, Taiwan
3
Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
4
Ph.D. Program in Environmental and Occupational Medicine, Kaohsiung Medical University and National Health Research Institutes, Kaohsiung 807, Taiwan
5
Department of Radiation Oncology, Faculty of Medicine, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
6
Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
*
Author to whom correspondence should be addressed.
Cancers 2024, 16(15), 2710; https://doi.org/10.3390/cancers16152710
Submission received: 27 June 2024 / Revised: 22 July 2024 / Accepted: 29 July 2024 / Published: 30 July 2024

Abstract

:

Simple Summary

MR-Linac is a novel magnetic resonance imaging (MRI)-guided radiotherapy (IGRT) that combines MRI with a linear accelerator (Linac). Although radiation therapy (RT) for lung cancer has traditionally been managed with a computed tomography (CT)-based workflow, an MR-Linac-based workflow would be able to address the many limitations of current practice. This narrative review summarizes the latest developments in MR-Linac lung cancer treatment, as well as its boundaries. Future research directions are also highlighted.

Abstract

Compared with computed tomography (CT), magnetic resonance imaging (MRI) traditionally plays a very limited role in lung cancer management, although there is plenty of room for improvement in the current CT-based workflow, for example, in structures such as the brachial plexus and chest wall invasion, which are difficult to visualize with CT alone. Furthermore, in the treatment of high-risk tumors such as ultracentral lung cancer, treatment-associated toxicity currently still outweighs its benefits. The advent of MR-Linac, an MRI-guided radiotherapy (RT) that combines MRI with a linear accelerator, could potentially address these limitations. Compared with CT-based technologies, MR-Linac could offer superior soft tissue visualization, daily adaptive capability, real-time target tracking, and an early assessment of treatment response. Clinically, it could be especially advantageous in the treatment of central/ultracentral lung cancer, early-stage lung cancer, and locally advanced lung cancer. Increasing demands for stereotactic body radiotherapy (SBRT) for lung cancer have led to MR-Linac adoption in some cancer centers. In this review, a broad overview of the latest research on imaging-guided radiotherapy (IGRT) with MR-Linac for lung cancer management is provided, and development pertaining to artificial intelligence is also highlighted. New avenues of research are also discussed.

1. Introduction

Lung cancer is one of the most common cancers in the world and a leading cause of cancer death for both sexes [1]. Lung cancer has two main subtypes: small-cell lung cancer (SCLC) and non-small-cell lung cancer (NSCLC). Despite great strides having been made in lung cancer treatment over the past few decades, outcomes still appear dismal. One major treatment modality is radiotherapy (RT), which is used with curative intent for early-stage NSCLC, in combination treatment with systemic therapy during advanced stages, and in palliative treatment in the terminal stage. As imaging technology advances and demands for more precise RT increase, magnetic resonance imaging (MRI)-guided radiation therapy (MRgRT) has been introduced.
The most prominent example of MRgRT is MR-Linac, which combines an MRI scanner with a linear accelerator (Linac). There are currently two clinically operable MR-Linac systems that are commonly used [2]: the 0.35 tesla (T) ViewRay MRIdian and the 1.5T Elekta Unity MR-Linac [3]. Although MR-Linac’s high capital investment still limits its accessibility, its utilization is rapidly expanding around the globe [4,5]. From 2014 to 2020, 0.35T MR-Linac usage in the United States steadily increased, with an annual growth rate of 59.6%, and a large proportion of that growth was seen in on-table adaptive planning and ultrahypofractionation dose scheduling (UHfx), which includes stereotactic body radiotherapy (SBRT) or the so-called stereotactic ablative body radiotherapy (SABR) [4]. A comparable pattern of usage was also reported in an analysis of both European and Asian usage of the 0.35T MR-Linac from 2015 to 2020 [5]. In both analyses, lung cancer was among the most treated tumor types, along with abdominal and pelvic tumors [4,5].
Our institutional experience also reflects a similar pattern of usage. We are a medical center located in a metropolis in southwestern Taiwan. A 0.35 T MR-Linac has been commissioned since 2020, and from June 2020 to December 2024, 594 treatment courses were completed, over half of which were SBRT. Lung cancer was among the common malignancies to be treated, and as demand for lung cancer SBRT continues to grow [6], so will MR-Linac use in lung cancer. This paper provides a broad overview of the latest in MR-Linac lung cancer treatment while also highlighting new avenues of research in the area.

2. A Brief Overview of the Linear Accelerator’s Development

Figure 1 presents an illustrated timeline of major milestones in the development of Linacs. In 1928, Wideröe proposed the concept of a Linac in a paper titled “On a New Principle for the Production of Higher Voltages” [7]. Although the proposal turned out to be impractical for clinical application, it remained a forerunner of modern Linacs. It was not until 1953 that the first clinical Linac was installed and patients began being treated at Hammersmith Hospital in London [7]. In the subsequent years, various modifications were made to clinical Linacs. For example, the concept of conformational RT and the multileaf collimator (MLC) was first developed by Takahashi in Japan in the 1960s [7]; nevertheless, owing to difficulties in operating the control system, the MLC’s adoption elsewhere would have to wait until later years. By the 1970s, the Linac had established itself, with both the low-energy and multimodality systems being widely available from a number of manufacturers [7]. In the 1990s, David Jaffray developed cone-beam CT (CBCT), which push-started the concept of imaging-guided radiotherapy (IGRT) [8], and around the same time, adaptive RT was first proposed by Yen et al., which we discuss in detail in later sections [3,7]. Although the development of these techniques has mitigated side effects to some extent, there are inadequacies in managing dose distribution [9]. Finally, in 2004 and 2005, Raaymakers et al. [10] and Raaijmakers et al. [11] published the first papers on the feasibility of combining MRI with a Linac, initiating the development of MR-Linac [3]. As mentioned earlier, ViewRay MRIdian and Elekta Unity are two widely adopted systems, and the third system, Aurora RT, which utilizes a 0.5T magnet and 6MV Linac system, recently received U.S. Food and Drug Administration (FDA) approval in 2022 [3]. It boasts a large bore space, thereby allowing for the iso-centric positioning of large tumors [12]. In addition, a prototype is being developed in Australia, with a machine designed to combine a 1.0 T magnet with open-bore MRI and a 6MV Linac [13], and due to its open-bore design, the Australian prototype is versatile and can direct beams at different directions. This version is set to undergo a feasibility and safety study (ACTRN12621000418875).

3. Advantages of MRgRT in Lung Cancer Treatment

Figure 2 provides a brief overview of the current RT workflow for lung cancer and its various limitations. Thoracic MRI traditionally plays a limited role in lung cancer treatment. This is because, compared with CT, thoracic MRI tends to contain a poor signal-to-noise ratio secondary to intrafractional respiratory and cardiac motions, as well as low tissue density of the lung parenchyma [14,15]. Various guidelines, including those of the National Institute of Clinical Excellence (NICE), the National Comprehensive Cancer Network (NCCN), and the American College of Chest Physicians (ACCP) therefore recommend MRI use only during the evaluation of suspected chest wall invasions or superior sulcus tumors [16,17,18], with the only other major MRI application in lung cancer management being for the detection of brain metastasis [14]. However, MRgRT has much to offer and could potentially revolutionize lung cancer therapy. In the following section, we discuss the advantages of MRgRT over the current CT-based workflow. A summary of MRgRT advantages can also be found in Figure 2.

3.1. Superior Soft Tissue Visualization

Compared with CT imaging, an MR scanner has an inherently superior soft tissue visualization capability [14], which has the benefit of increasing delineation precision around targets and organs at risk (OARs) that have traditionally been challenging to visualize with CT alone, even with the use of a CT OAR atlas. Examples of such structures include the brachial plexus, esophagus, heart, and spinal cord. For these structures, an approach combining CT and MRI is already being recommended [19]. In particular, MRI could be useful in reducing cardiac exposure to irradiation.
The latter has become a topic of growing interest in lung RT ever since the landmark Radiation Therapy Oncology Group (RTOG) 0617 trial demonstrated that radiation exposure to the heart correlates with poor post-RT survival [20], with subsequent analyses finding differences in radiosensitivity among different cardiac substructures. The idea of cardiac substructure constraint was thus proposed; however, clinically, the mean heart dose is still the most commonly considered factor in lung SBRT planning. MRgRT could help cardiac substructure sparing become feasible [21]. Van der Pol et al. conducted a small study of 34 patients with stage II to IV lung cancer treated with SBRT [21]. For each subject, both cardiac-sparing and non-cardiac-sparing MR-Linac treatment plans were created and compared. It was found that 47% of the patients needed cardiac sparing and that a successful cardiac-sparing MR-Linac treatment plan could be created for 62.5% of these patients without compromising doses to the tumor or other OARs.
Similarly, in terms of target delineation, centrally located primary lung tumors and metastatic lymph nodes are also challenging for CT to visualize alone, as we discuss in detail in later paragraphs. To date, studies in pelvic, central nervous system, and head and neck cancers have demonstrated smaller and more accurate target contouring with the addition of MRI than with CT-based delineations alone [22,23,24], while similar studies are sparse in lung cancers due to the absence of suitable thoracic MRI sequences [14,25]. Despite this, a clinical need for a more precise delineation still exists in lung RT, particularly in cases with tumors invading the mediastinum, abutting parenchymal lung changes, or invading major arteries [14]; to date, an accurate assessment of disease extent in such situations remains challenging. Although the addition of an 18F-labeled fluoro-2-deoxyglucose positron emission tomography (F-18-FDG PET) scan to CT has been found to reduce inter-observer variations in challenging cases, the spatial resolution of a PET scan, along with the healthy and cancerous tissue contrast, is still inferior to that of MRI [14,26]. Therefore, the International MR-Linac Consortium, which is a collaboration between researchers and commercial partners to introduce MR-Linac, is working on refining MRI sequencing for IGRT [27]. Additionally, two recent small studies both demonstrated T1-weighted DIXON sequences to be the preferred MRI sequence for thorax imaging and contouring in healthy non-patient volunteers due to its image contrast resembling that of planning CT, which makes image interpretation and subsequent registration more straightforward [25,28]. Naturally, larger studies with patient volunteers are needed to validate these results, and the optimization of MRI sequences for lung IGRT is a prerequisite to carrying out any subsequent high-quality studies regarding MRgRT in lung cancer management.
Thus far, studies in MR-based planning have shown that diffusion-weighted MRI (DW-MRI)-based gross tumor volume (GTV) correlates with CT-based GTV plans in lung RT [29]; however, there is a paucity of clinician experience in target delineation with lung MRI, as reflected by studies on inter-observer delineation variability described below. Karki et al. compared inter-observer variations between MRI and PET-CT-based GTV, with MRI appearing to have an acceptable yet greater inter-observer variability than PET-CT [26]. In another recent study that evaluated the inter- and intraobserver variability of MR- versus PET-CT-based GTV for lung cancer SBRT, greater inter-observer variability was also found between MR-based GTV plans, despite significant intraobserver agreement [30]. Both teams of investigators attributed the inter-observer variation to a lack of observer experience with MR-based lung GTV delineation. As a growing number of MR-Linac systems become available globally, this variability might reduce as more clinicians gain experience with the MR-based delineation of lung tumors.
As for position verification, the current clinical standard is CBCT imaging with online target matching to decrease positional error and ensure consistency in the dose delivered [14]. Compared with traditional fan-beam CT, CBCT induces significantly less radiation exposure, especially since the development of the low-dose CBCT protocol [31]. Nevertheless, a CBCT image is also subject to higher degrees of scatter and artifact, rendering it inadmissible for subsequent adaptive planning; thus, the generation of synthetic CT (sCT) using an artificial intelligence (AI) model was developed to address this issue [31]. Unfortunately, CBCT-generated sCT has the same limitation as planning CT, i.e., poor soft tissue visualizations, and MRgRT could be advantageous in this regard. Current AI technologies are already able to generate sCT from MR images of the thorax with a good dose accuracy and image quality, without inter-scanner variability or radiation exposure [32]; additionally, MR-derived sCT has demonstrated good overall agreement with conventional CT in lung cases [33].

3.2. Daily Adaptive Capability

MR-Linac is a major advance in IGRT because of its adaptive capabilities [34]. Thus, any discussion on MR-Linac requires a brief introduction to adaptive radiotherapy (ART). ART was first proposed by Yan et al. at the turn of the century [35] as a means of simply refining the planning target volume (PTV) to account for uncertainties during delivery [34]. Over the years, ART has evolved to use advanced imaging systems to visualize the anatomical and functional changes in patients as they progress through their treatment, and modifications are made accordingly. The ART concept believes that a baseline simulation is just a snapshot of our body at the time of the simulation, and regardless of how precise it is, throughout the treatment course, changes in the patient’s body inevitably occur. These include tumor shrinkage, as well as normal day-to-day changes in body anatomy, such as bowel peristalsis and bladder filling. In short, the treatment plan devised on the baseline simulation will likely not fit the patient on the day of treatment, and continuous usage of the outdated simulation plan will cause subsequent problems, such as suboptimal dosing and radiation toxicity to healthy tissues. In a recent phase II clinical trial, the LARTIA study, the benefit of ART was demonstrated [36]. LARTIA showed that ART in the form of weekly replanning CT scans led not only to a low rate of marginal failure but also to a low rate of toxicity at a median follow-up of 20.5 months. Although a definitive conclusion will have to wait until larger phase III studies, this result provides impetus for further ART research in lung cancer.
In general, the scientific community supports the concept of ART, and over the years, ART has become more sophisticated, with MR-Linac being its latest breakthrough. Lamb et al. reported that there were nine centers equipped with onboard MRgRT in 2017 [37] and published a technical report on the process of ART. Currently, there are two commonly seen classes of ART technologies [38]: (1) offline ART, where verification and adaptive planning are scheduled between fractions, which means that the adaptive process can take days, and (2) online ART, where verification and adaptive planning are carried out immediately before treatment delivery, with the time between the adaptive process only taking minutes (Figure 2). The Adaptive Radiation Therapy Physician Guidelines were published in 2022 with recommendations from experts from the USA, UK, and Korea [39]. The third type of ART, real-time ART, can reduce the adaptive process to seconds, although this is rare in clinical practice and falls outside the scope of our discussion.
Prior to the advent of MR-based Linac systems, only offline ART was available clinically; thus, MR-Linac was revolutionary in RT because it enables online ART. Compared with the LATRIA study that utilized weekly adaptive CT scans, MR-Linac has the capability to offer rapid and automated online plan adaptation prior to treatment delivery each day. With MR-Linac’s daily adaptive capability, there is a possibility to realize the full potential of adaptive planning while minimizing concomitant radiation exposure [14]. Indeed, a recent study by Kang et al. supported this view by demonstrating that lung SBRT with MR-Linac versus a CT-guided modality could decrease the radiation field without lowering the local control rate and survival rates at 1 year [40].

3.3. Real-Time Target Tracking

Prior to MR-Linac, the only option for real-time tracking was the application of internal fiducial markers [14], which required the fiducial marker to be positioned at or near its target. A kilovoltage imaging system was then used to image the marker. In patients with locally advanced diseases and requiring multiple fiducial markers, or in patients with protracted treatment courses, the practice could cause significant radiation exposure [14]. However, MR-Linac systems could also offer intrafractional motion detection, yet without additional ionizing radiation.
For thoracic imaging and RT, a major source of intrafractional motion is respiratory motion. Currently, respiratory motions are accommodated through the expansion of the clinical target volume (CTV)–planning target volume (PTV) margin [41]. It is important to note that, although 4D-CT could account for structural changes with time, it does not account for real-time daily motion management. This is because 4D-CT is conducted at simulation, where the machine studies tumor motion through a patient’s respiratory cycle and calculates an internal target volume (ITV) based on the union of all tumor positions during the imaging process; however, 4D-CT does not account for the changes that occur during RT delivery. In fact, a study using real-time MRI found that ITV size often varies more than previously considered, suggesting significantly higher inter- and intrafractional variations in breathing amplitude than accounted for during the current ITV-based planning process [42]. The real-time anatomical and dosimetric information can enable intrafractional plan adaptation, ensuring the intended daily dose is delivered. This approach may allow for smaller PTV margins and further reduce the dose to healthy tissues [43].
While the current literature shows that ITV-based SBRT is adequate with a tolerable toxicity profile for most patients, there is still much room for improvement, especially in cases where minimizing the treatment margin is crucial [42]. This includes patients with central lung tumor(s) and patients with erratic breathing patterns; for these populations, a more precise treatment option with real-time target tracking that minimizes intrafractional variation is necessary.
Currently, the 1.5T MR-Linac utilizes MLC tracking, for which studies have demonstrated technical feasibility and improved dose accuracy [44,45]. However, the 0.35 T system uses automatically gated beam delivery with cine MRI; this is often coupled with patient self-sustained breath-holds (BHs) [46], which can be monitored via real-time imaging, though this is still subject to patient compliance and individual respiratory function. To address this issue, the Active Breathing Coordinator (ABC) was invented to improve lung volume reproducibility, thereby lowering intrafractional variation [47]. In radical NSCLC RT specifically, the ABC has been shown to reduce the treatment margin [48]. The ABC has been used in SBRT with conventional Linacs for many years. Recently, the first MR-ABC protocol was established by Kaza et al., and their results showed that the ABC, in lung cancer MRI, could lead to good lung volume reproducibility [49]. This means that the ABC could soon be used for gating purposes on MR-Linac to further reduce uncertainties in BH techniques. Alternatively, an AI algorithm has also been developed that could effectively predict tumor motion in 0.35 T MR-Linac machines, enabling the future implementation of MLC tracking [46].

3.4. Early Assessment of Treatment Response

In the current workflow, there is no early assessment of treatment response (Figure 2). However, in an MR-based workflow, in addition to the conventional qualitative MRI, clinicians will be able to use quantitative MRI (qMRI) to quantitatively map out a tumor’s biological information, thereby evaluating changes early in the treatment in a more timely manner to predict responses. Various qMR techniques and associated non-invasive imaging biomarkers have been identified, with two of the most investigated MR techniques being diffusion-weighted imagining (DWI) and dynamic contrast-enhanced (DCE) MRI [14,50]. Table 1 contains a list of the qMR techniques that have been investigated in the context of lung cancer.
DWI MRI calculates the apparent diffusion coefficient (ADC), which is associated with the magnitude of diffusion within a tissue [50]. When cells die and the cell density reduces, diffusion increases, and an increase in the ADC is observed. It has been reported that the ADC value increases during chemoradiation in NSCLC [53]. However, compared with cancer of other organs, where a greater increase in the ADC value might predict responders [50], lung cancer studies are scant due to the traditionally limited role of thoracic MRI, and of those few studies available, the results are mixed [50,54]. There is even a small study that suggests that the ADC value increase at 1 month is associated with local failure in NSCLC [55]. However, importantly, researchers have shown that the ADC biomarker is able to predict responses before morphological changes can be seen [54]. This has important implications for regimen design, as patients may be able to switch or terminate anticancer drugs based on early biological marker changes, thereby avoiding unnecessary systemic toxicity or delayed time in receiving more effective treatment options [54]. In fact, ADC data have been found to improve the performance of the NSCLC survival prediction AI model [63], further substantiating the importance of the ADC as a biomarker for treatment response.
In contrast, DCE-MRI measures tissue perfusion and capillary permeability [50]; thus, within the tumor microenvironment, where an abundance of abnormal microvasculature can be found, there tends to be an enhancement in DCE-MRI. Although the kinetic parameters of capillary permeability have been linked to tumor response to RT in some cancers [64,65,66], the data for lung cancer remain scarce, and of the limited studies, DCE-MRI seems promising for the prediction of treatment response following concurrent chemoradiation (CCRT) in NSCLC [58] and stereotactic radiosurgery (SRS) for metastatic brain tumors from primary lung cancer [59].
Further investigation is still needed. A major downside to the DCE MRI technique is the need for contrast agents, which come with their own risk, and as a result, options for non-contrast-enhanced MRI methods are being explored [67]. Oxygen-enhanced MRI is also being explored for the detection of regional hypoxia in NSCLC [62], as this might predict tumor regions refractory to RT, and additional treatment strategies might therefore be employed. As various qMRI techniques are being explored, it is also important to validate the techniques against one another. For example, it has been found that, although both intravoxel incoherent motion (IVIM) and DCE-MRI could be used to measure metastatic brain tumor blood volume post-SRS, their measurements do not correlate. In a comparative study by Kapadia et al., it was shown that while DCE-MRI showed a reduction in tumor blood volume, IVIM showed the opposite, or an increase in blood volume [68]. Therefore, until qMRI techniques have been well validated, the interpretation of single qMRI techniques should be performed with caution, with multiple parameters being considered when necessary. In short, numerous qMRI techniques have potential as biomarkers for early treatment response, although more data on lung cancer are still required.

3.5. Combining Biological Targeting with Conventional ART

As discussed, the images produced by the above qMRI techniques are called biological images. They reveal various biological data, ranging from metabolic to functional data that reflect a tumor’s biological heterogeneity. These data can then be used to establish a dose distribution, which was validated to be different from that of conventional anatomical imaging [69,70]. These biological image-based dose distributions then allow clinicians to visualize tumor radiosensitivity [71], which is invaluable for treatment response prediction, as previously discussed, while also helping to reduce toxicity and preserve function. An example is the use of functional information for normal lung avoidance. Quantitative gas MR has been developed, and it could allow us to directly visualize pulmonary ventilation and perfusion [72]. Areas of pre-existing pulmonary dysfunction could be identified, and by incorporating functional information into RT planning, we could perform accurate dose escalation to areas of low pulmonary function while decreasing radiation exposure to areas of highly functioning normal lung tissue [73]. Although single-photon emission computed tomography (SPECT) and PET-CT could also visualize lung function, gas MR has the added advantage of a superior spatial and temporal resolution without ionizing radiation exposure [72]. Unfortunately, the only gas MR-based functional lung avoidance RT clinical trial in recent years was terminated due to insufficient accrual (NCT02002052).
Just as conventional ART emphasizes morphological changes during RT, the biological heterogeneity of a tumor is also dynamic, making it vital that biological images also undergo adaptive planning, which has become known as biological image-guided adaptive RT (BIGART) [50]. Before MRgRT, CT-based BIGART had already demonstrated feasibility and tolerability in clinical trials [74,75]. Unfortunately, it is still difficult to realize in practice due to logistical challenges [50]. For example, because planning CT and functional images are not acquired at the same time, it is difficult to compare the images; thus, the validity of image comparison and the accuracy of image registration are called into question [73]. In contrast, MR-Linac has the potential to perform daily biological imaging and adaptive planning concurrently, which fully realizes the potential of BIGART [50]. However, before BIGART can become a clinical routine, many requirements still need to be fulfilled. For example, Van Houdt et al. believe that issues such as investigating daily changes in qMRI values, choosing the most suitable qMRI techniques as biomarkers, and standardizing qMRI protocols all need to be dealt with first [50], and for each of the above steps, large, multicenter clinical trials are needed for validation.

3.6. AI and Machine Learning

AI, especially its subset, machine learning (ML), is revolutionizing radiology by enhancing image analysis and reducing diagnostic errors [76]. AI is transforming radiology by optimizing workflows and improving non-interpretative tasks [77]. Integrated with Natural Language Processing (NLP), AI automates the triage of imaging studies, prioritizing urgent cases through electronic health records and expediting patient triage, reporting, and follow-up management [78]. Deep learning (DL), an ML subset, improves report consistency and clarity, enhancing radiology service quality. DL accelerates MRI scanning, harmonizing efficiency and quality, with commensurate progress witnessed in CT and PET image reconstruction.
Understanding the interrelationships between AI, ML, and DL helps to conceptualize each subfield’s contribution and progression within the broader AI narrative [76]. AI provides the foundation, ML enhances AI’s potential by enabling machines to learn from data, and DL further extends these capabilities with neural networks that decipher complex data patterns [76]. Despite AI’s potential, only 30% of radiologists used it clinically by 2021, with many skeptical [79]. Effective AI integration requires supportive infrastructure and workflow redefinition. It may help reduce errors and radiologist burnout [80].
The integration of AI, such as automation into the RT planning pathway, will allow ART to become a reality in clinical practice [81]. In one study, AI-generated data were favored over conventional MRI planning data by the radiation oncologist in the study [82]. Accordingly, we believe that the development of AI-driven ART could speed up the MRgRT workflow and overcome a major barrier to MRgRT implementation.

4. MRgRT Clinical Application

Since most MR-Linac systems for lung cancers are used in SBRT treatment [4,5], the following discussion focuses on SBRT. Table 2 contains all ongoing clinical trials involving MR-Linac at the time of the writing of this manuscript.

4.1. Central and Ultracentral Lung Tumor

Despite advances in lung cancer treatment with SBRT, treatment for central (CLT) and ultracentral (ULT) lung tumors remains a challenge due to significant toxicity. Here, we adopted the Nordic-HILUS trial’s definition, where a central tumor is defined as a tumor within 2 cm of the proximal bronchial trees, while ULT is defined as touching or within 1 cm of the mediastinum, esophagus, and proximal bronchial tree [83]. Such proximity to vulnerable mediastinal structures, coupled with respiratory and cardiac motions, makes the grade 5 toxicity risk with SBRT as high as 15.4% [83]; consequently, the American Society of Radiation Oncology (ASTRO) deems the toxicity risk to negate any treatment benefit and therefore discourages SBRT to ULTs [84]. Here, MRgRT’s strength in gated dose delivery and daily plan adaptation may be useful in mitigating the toxicities. In fact, Ligtenberg et al. demonstrated that clinicians could reduce CLT treatment toxicities on MR-Linac by lowering the OAR doses of ITV-based plans with mid-position-based planning, a method based on the time-weighted mean position of the tumor [85]. Furthermore, CLT dose accumulation studies have shown that, at 1 cm of the PTV, the OAR dose of online adaptive MRgRT is not significantly different from that of non-adaptive gated proton therapy or even online adaptive proton therapy [86]. Given their similar toxicity profiles, MRgRT presents as a more attractive treatment option than proton therapy due to its lower structural investment.
Current clinical trial results support the use of stereotactic MR-guided adaptive radiation therapy (SMART) in treating CLTs/ULTs. Regnery et al. performed a small prospective cohort study that showed that SMART in ULT treatment was able to protect the OARs while maintaining adequate PTV coverage, and in the subsequent long-term follow-up, they found no statistically significant difference in 2-year OS, PFS, or local progression between the ULT and non-ULT groups [87]. However, significantly more toxicities of ≥ grade 2 occurred in the ULT group, two of which were grade 3 esophagitis and bronchial bleeding, but most patients experienced mild toxicities only. The results showed SMART to be a potentially safe and effective treatment for ULT, although ULT remains a high-risk target area [87].
In contrast, Sandoval et al. also treated ULT and CLT with SMART [88]. At 1 year, OS was 82%, the local control rate was 87%, and PFS was 54%, with most patients experiencing grade 1 and 2 toxicities, while only two patients experienced grade 3 toxicities. Finazzi et al. also reported a promising 1-year local control rate (95.6%) and low high-grade toxicity risks (30% for ≥ grade 2 and 8% for ≥ grade 3 toxicities) in their trial of high-risk lung tumor patients treated with SMART [89], where the inclusion criteria were not limited to patients with central tumors but also encompassed patients with previous thoracic RT histories and interstitial lung diseases. Despite optimistic results, data from larger clinical trials are still needed before definitive conclusions can be drawn. Currently, a prospective phase I dose escalation trial, MAGELLAN, is underway [90]. It will enroll primary and secondary ULT patients with tumor size ≤ 5 cm to be treated with a 0.35 T MR-Linac. Dose escalation will begin at 10 × 5.5 Gy and reach a maximum of 10 × 6.5 Gy under the guidance of a time-to-event continual reassessment method. MAGELLAN will establish the safe dose for UCT treatment with SMART, which will guide further research in the area.

4.2. Early-Stage Lung Cancer

In RT, the current standard treatment for early-stage inoperable NSCLC is SBRT, with two methods of SBRT delivery available: over multiple fractions or a single fraction. Owing to concerns about missing the tumor target and toxicity to OARs when administering treatment in one large single dose, single-fraction delivery is not popular among clinicians [91], and while internal fiducial markers could potentially be employed to increase accuracy, issues with radiation exposure, as discussed earlier, still exist. Nevertheless, there is no clinical evidence showing that single-fraction SBRT is inferior to its multiple-fraction counterpart. In fact, two randomized phase 2 trials showed that there are no short-term survival or toxicity differences between single- and multiple-fraction treatments in early-stage NSCLC [92,93]. At a five-year follow-up, there still appeared to be similar toxicity rates, tumor control rates, and median survival times [93]. However, the quality of life measurement, such as social functioning and dyspnea, appeared to be better in the single-fraction arm treatment in one clinical trial [92]. In addition, single-fraction SBRT might potentially be more immunogenic than multiple-fraction SBRT due to a potentially greater lymphocyte-sparing effect, although further clinical evidence is pending [94].
During the COVID-19 pandemic, there was a renewed interest in single-fraction SBRT due to the need to reduce patient exposure to the hospital environment [94]. At present, however, with MR-Linac’s real-tracking ability, single-fraction SBRT could become a standard, although problems with its implementation, especially its potentially lengthy treatment time, still need to be resolved. Finazzi et al. recently reported their institutional experience with treating lung tumors with single-fraction, gated breath-hold (BH) SBRT on a 0.35T MR-Linac [95]. Of the 10 patients treated, a median in-room procedural time of 120 min and a median treatment time of 39 min were reported, which is significantly longer than the 15 min treatment time required for flattening-filter-free (FFF) volumetric-modulated arc therapy (VMAT), a faster and more conventional method of performing SBRT [96]. The total procedural time included two breath-hold (BH) 3D MR scans, with one at the start of the treatment day, which was immediately followed by plan editing and adaptation; then, the patients underwent a second scan during mid-treatment with optional readaptation and a break if needed. Their final analysis revealed that the resulting BH patterns among the patients were variable and ultimately found to have no influence on GTV coverage with MR-Linac’s real-time tracking. Although the on-table adaptation improved PTV coverage, it had no influence on GTV doses. Finazzi et al.’s result raised concerns over the increased treatment time and labor-intensive treatment procedure [95].
In comparison, Chuong et al. reported another case of peripheral early-stage NSCLC treated with mid-inspiration BH single-fraction SBRT on MR-Linac [97]; however, their reported procedure time was 40 min, while the treatment time was only 17 min. This shorter treatment time was attributed to a few modifications made to the method of Finazzi et al. Firstly, they eliminated on-table adaptation as they deemed it to be clinically unnecessary for peripheral tumors and Finazzi et al. reported it to have unclear clinical benefits. Secondly, they utilized visual biofeedback respiratory gating, which involved a monitor in the treatment room that showed the patient real-time cine-MR imaging with the target position and treatment boundary. By watching the monitor, the patient was able to time their breath-holding according to the target location within the boundary. Visual biofeedback was so effective in achieving the correct BH position that mid-treatment breaks were eliminated from the protocol. The clinical usefulness of visual biofeedback was also recently corroborated by a Korean study [98]. The small study of 15 patients demonstrated that a similarly concepted visual guidance patient-controlled (VG-PC) respiratory-gated MR-IGRT was able to reduce the total treatment time by 37.6% compared with free-breathing. Our institutional protocol includes on-table adaptation with or without breath-holding. Moreover, our data suggest that procedural optimization over time will significantly reduce the MR-Linac total in-room treatment time.

4.3. Locally Advanced (LA)/Oligo-Progressive Lung Cancer

The current standard of treatment for LA NSCLC is platinum-based CCRT, followed by consolidation with immunotherapy, although the prognosis remains poor [99]. Despite major advances in lung cancer treatment, such as immunotherapy, relatively few clinical studies have been conducted in LA NSCLC patient groups compared with metastatic lung cancer patient groups. For example, in the study of the immune checkpoint inhibitor (ICI) and SBRT synergy, no clinical trials specifically for LA NSCLC were completed, and only one small phase II study is currently underway (NCT03589547); consequently, effective treatment for this patient group is urgently required. When MRgRT was first introduced, most studies focused on treating central or ultracentral lung tumors, as previously discussed, but now there is a push toward using MR-Linac for the definitive treatment of LA lung cancer.
In LA-NSCLC, MR-Linac may be proven advantageous for this population due to its powerful soft tissue imaging capability. As discussed earlier, PET scans and CT are not omnipotent, and in cases where there are local invasions into surrounding soft tissues such as the mediastinum or the chest wall, MRI could offer clearer imaging for delineation than CT or FDG-PET [16,17,18]. Even in nodal staging for LA patients, several published studies, including meta-analyses and systemic reviews, agree that DW-MRI has better specificity than CT or FDG-PET [100,101]. Recently, a study that compared two MRI sequences with CBCT on their ability to identify lymph node invasion in LA-NSCLC patients was published, where both MRI sequences (T2w Turbo Spin-Echo and T1w DIXON water-only images) were found to have significantly better image quality and generated more observer confidence than CBCT, despite there being no significant inter-observer variability among the imaging modalities [102].
The development of RT for lung cancer has always been limited by a balance between effective RT dose escalation and normal lung tissue sparing [15]. Retrospective planning studies have shown that the use of MR-Linac for RT treatment could decrease the PTV margin and increase OAR sparing, thereby achieving isotoxic dose escalation and the widening of the therapeutic index [103]. However, the implementation of definitive CCRT using MRgRT for LA lung cancers is labor-intensive and significantly increases treatment time. A standardized protocol has not yet been set up and validated, and only case reports detailing individual institutional workflows have been published.
Recently, a technical report was published by Eze et al. that details a new clinical pathway for a node-positive LA-NSCLC patient with severely compromised pulmonary function and reserve [104], where a 0.35 T MR-Linac was used to deliver a total dose of 48.9 Gy/16 daily fractions with real-time continuous tracking by a 2D MRI during treatment. The treatment was well tolerated, with no grade 2 or above toxicities, and a 3-month follow-up CT scan showed partial remission per RESIST 1.1 criteria. La Rosa et al. also published a case of a stage IV NSCLC patient with oligo-progression and lymph node invasion who was treated with MRgRT [105]. As a result of online ART, the cumulative dose to OARs was reduced by 11.34% (esophagus), 4.2% (proximal bronchial tree), and 5.62% (trachea). These reports demonstrate the clinical feasibility of MR guidance, but both protocols still await validation in the form of large clinical trials.
The current clinical trial NCT03916419 is a single-institution, single-arm, phase II study with a safety lead-in, with the aim of investigating the safety and efficacy of MRgRT in curative-intent CCRT treatment for inoperable LA-NSCLC patients. Its primary end points are the number of participants with dose-limiting toxicities, and local and regional control rates. The study is set for completion in 2025. Both trials will add valuable information to the integration of MRgRT into the LA-NSCLC curative-intent treatment workflow.

5. Challenges for MRgRT in Lung Cancer Treatment

Despite its numerous advantages over current CT-based technologies, many limitations related to MRgRT exist. One such challenge is the potential effect of the Lorentz force. During photon irradiation, electrons deposit energy along their pathway, but when these electrons encounter a magnetic field, their path may be deflected by the Lorentz force, which then alters the dose distribution. This is especially significant in areas of heterogenous tissue density such as skin–air or lung–tissue interface regions [11,106]. The Lorentz force at these sites can cause electrons that have already exited the tissue to return to its surface in a phenomenon called the electron return effect (ERE), and in lung RT, ERE can potentially lead to lung tissue toxicities [11,107]. Fortunately, research so far has not found the dose alteration to be clinically significant [44,107]. Moreover, lung RT usually employs low-field imaging for better imaging quality; thus, the ERE is much lower than in high-field imaging [108].
Clinically, there are several methods for MLC calibration or quality assurance [109]. Some challenges related to the MRgRT workflow include the development of a reliable and consistent quality assurance and quality control procedure that is specific to MRgRT [110]. In addition, there is a relative contraindication to treat patients with metallic implants or cardiac devices. Yang et al. recently reported safely treating four patients with a cardiac pacemaker using a 1.5T MR-Linac [111]; similarly, Keesman et al. also published their MRgRT workflow for three prostate cancer patients with a metallic hip prosthesis [112], although both protocols await confirmation through larger studies. Substantial capital investment and training for team members also impede MR-Linac’s widespread use, and finally, the low throughput and the time-consuming adaptation process are also major drawbacks of this novel technology. Most importantly, for individuals that have experienced episodes of claustrophobia during an MRI scan, MRgRT may not be suitable.
Moreover, significant debate remains regarding the prolonged treatment times. Such limitations can impact patient comfort, compliance, and overall treatment efficiency. In our experience, from 2020 to 2023, for non-single-fraction BH SMART, the median in-room procedural time was 37 min, with a median treatment time of 15 min. As discussed in Section 4.2, visual biofeedback respiratory gating was studied by Chuong et al. [97] and reported with good efficiency, and it was examined in another Korean study [98] in which the total treatment time was reduced by 37.6% compared with free-breathing.
Some researchers have taken things further by shortening the entire workflow to one day. A one-step-shop (OSS) service is a procedure well known in the palliative setting that condenses consultation, simulation, and RT delivery all in one day [113] with the goal of shortening overall treatment time, thus improving patient convenience and ameliorating patient discomfort. This is especially important for countries such as the United States and Canada, where patients often must travel great distances to receive RT; therefore, shortening treatment to one day means that time and costs for traveling and lodging could be significantly reduced, further improving the patient adherence rate.
In the past, Brunenberg et al. demonstrated the feasibility of OSS-palliative RT with an MR-based diagnostic protocol and MR simulation [113], and recently, Palacios et al. applied this procedure to single-fraction SMART for small tumors of early-stage lung cancer [114]. The reported median procedural time was 6.6 h, and with procedural optimization, the treatment could be completed in half a day. Good patient satisfaction was reported, with highly accurate gated treatment delivery, as demonstrated by the above 78–100% GTV coverage and 94.4–100% corresponding PTV values. Taken together, these studies demonstrate that MRgRT is capable of performing single-fraction SBRT with high precision, but plenty of room remains for discussing the exact workflow and implementation.
However, as we previously discussed, research efforts are being made to ameliorate these problems. The PUMA trial (NCT05237453) is a prospective, multicenter phase I trial in Germany that will test the clinical feasibility of the two MR-Linac systems in CCRT treatment for LA-NSCLC patients [115]. It will use conventionally fractionated RT with dose escalation and weekly online adaptation. Their primary end points include the successful completion of online-adapted fractions and on-table time. Additionally, AI development and procedural optimization, as team members become more familiar with the new workflow, can significantly increase throughput over time. For example, the manual delineation of OARs represents one of the most time-consuming processes of MRgRT, and AI technologies that can perform both left and right lung auto-segmentations with high accuracy have already been developed [82].

6. Conclusions

MR-Linac is a transformative technology that offers significant advantages over the current CT-based systems in treating lung cancer patients. Its superior soft tissue contrast, daily adaptive capability, and real-time image tracking set it apart. Furthermore, MR-Linac enables the integration of biological and anatomical images into the adaptive planning process, enhancing its clinical utility in treating early-stage lung cancer and locally advanced non-small-cell lung cancer (LA-NSCLC).
Despite its impressive benefits, MR-Linac does have limitations. As the demand for lung cancer SBRT grows, the adoption of MR-Linac in lung cancer treatment will become more prevalent. Clinicians will gain more experience and data, leading to improved outcomes. To ensure that expediency does not compromise treatment quality or patient safety, future research should focus on optimizing the workflow procedure. AI will play a crucial role in this advancement, enhancing the precision and efficiency of lung cancer treatment with MR-Linac.

Author Contributions

Conceptualization, H.-H.L.; data curation, S.-Y.L., S.H.C. and H.-H.L.; investigation, S.-Y.L., S.H.C. and H.-H.L.; project administration, H.-H.L.; writing—original draft preparation, S.H.C., S.-Y.L. and H.-H.L.; writing—review and editing, H.-H.L. and S.H.C.; visualization, S.-Y.L.; supervision, H.-H.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within the article.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Bray, F.; Laversanne, M.; Sung, H.; Ferlay, J.; Siegel, R.L.; Soerjomataram, I.; Jemal, A. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2024, 74, 229–263. [Google Scholar] [CrossRef]
  2. Wu, T.C.; Smith, L.M.; Woolf, D.; Faivre-Finn, C.; Lee, P. Exploring the Advantages and Challenges of MR-Guided Radiotherapy in Non-Small-Cell Lung Cancer: Who are the Optimal Candidates? Semin. Radiat. Oncol. 2024, 34, 56–63. [Google Scholar] [CrossRef]
  3. Rammohan, N.; Randall, J.W.; Yadav, P. History of Technological Advancements towards MR-Linac: The Future of Image-Guided Radiotherapy. J. Clin. Med. 2022, 11, 4730. [Google Scholar] [CrossRef]
  4. Chuong, M.D.; Clark, M.A.; Henke, L.E.; Kishan, A.U.; Portelance, L.; Parikh, P.J.; Bassetti, M.F.; Nagar, H.; Rosenberg, S.A.; Mehta, M.P.; et al. Patterns of utilization and clinical adoption of 0.35 Tesla MR-guided radiation therapy in the United States—Understanding the transition to adaptive, ultra-hypofractionated treatments. Clin. Transl. Radiat. Oncol. 2023, 38, 161–168. [Google Scholar] [CrossRef]
  5. Slotman, B.J.; Clark, M.A.; Özyar, E.; Kim, M.; Itami, J.; Tallet, A.; Debus, J.; Pfeffer, R.; Gentile, P.; Hama, Y.; et al. Clinical adoption patterns of 0.35 Tesla MR-guided radiation therapy in Europe and Asia. Radiat. Oncol. 2022, 17, 146. [Google Scholar] [CrossRef]
  6. Vlaskou Badra, E.; Baumgartl, M.; Fabiano, S.; Jongen, A.; Guckenberger, M. Stereotactic radiotherapy for early stage non-small cell lung cancer: Current standards and ongoing research. Transl. Lung Cancer Res. 2021, 10, 1930–1949. [Google Scholar] [CrossRef]
  7. Thwaites, D.I.; Tuohy, J.B. Back to the future: The history and development of the clinical linear accelerator. Phys. Med. Biol. 2006, 51, R343–R362. [Google Scholar] [CrossRef]
  8. Jaffray, D.A.; Siewerdsen, J.H. Cone-beam computed tomography with a flat-panel imager: Initial performance characterization. Med. Phys. 2000, 27, 1311–1323. [Google Scholar] [CrossRef]
  9. Zhou, R.; Zhao, D.; Beeraka, N.M.; Wang, X.; Lu, P.; Song, R.; Chen, K.; Liu, J. Novel Implications of Nanoparticle-Enhanced Radiotherapy and Brachytherapy: Z-Effect and Tumor Hypoxia. Metabolites 2022, 12, 943. [Google Scholar] [CrossRef]
  10. Raaymakers, B.W.; Raaijmakers, A.J.; Kotte, A.N.; Jette, D.; Lagendijk, J.J. Integrating a MRI scanner with a 6 MV radiotherapy accelerator: Dose deposition in a transverse magnetic field. Phys. Med. Biol. 2004, 49, 4109–4118. [Google Scholar] [CrossRef]
  11. Raaijmakers, A.J.; Raaymakers, B.W.; Lagendijk, J.J. Integrating a MRI scanner with a 6 MV radiotherapy accelerator: Dose increase at tissue-air interfaces in a lateral magnetic field due to returning electrons. Phys. Med. Biol. 2005, 50, 1363–1376. [Google Scholar] [CrossRef] [PubMed]
  12. Ladbury, C.; Amini, A.; Schwer, A.; Liu, A.; Williams, T.; Lee, P. Clinical Applications of Magnetic Resonance-Guided Radiotherapy: A Narrative Review. Cancers 2023, 15, 2916. [Google Scholar] [CrossRef]
  13. Keall, P.J.; Barton, M.; Crozier, S. The Australian magnetic resonance imaging-linac program. Semin. Radiat. Oncol. 2014, 24, 203–206. [Google Scholar] [CrossRef] [PubMed]
  14. Bainbridge, H.; Salem, A.; Tijssen, R.H.N.; Dubec, M.; Wetscherek, A.; Van Es, C.; Belderbos, J.; Faivre-Finn, C.; McDonald, F. Magnetic resonance imaging in precision radiation therapy for lung cancer. Transl. Lung Cancer Res. 2017, 6, 689–707. [Google Scholar] [CrossRef] [PubMed]
  15. Giaj-Levra, N.; Borghetti, P.; Bruni, A.; Ciammella, P.; Cuccia, F.; Fozza, A.; Franceschini, D.; Scotti, V.; Vagge, S.; Alongi, F. Current radiotherapy techniques in NSCLC: Challenges and potential solutions. Expert Rev. Anticancer Ther. 2020, 20, 387–402. [Google Scholar] [CrossRef] [PubMed]
  16. National Comprehensive Cancer Network. Non-Small Cell Lung Cancer (Version 2. 2024). Available online: https://www.nccn.org/professionals/physician_gls/pdf/nscl.pdf (accessed on 17 February 2024).
  17. National Institute for Health and Care Excellence. Lung Cancer: Diagnosis and Management. Available online: https://www.nice.org.uk/guidance/ng122 (accessed on 17 February 2024).
  18. Kozower, B.D.; Larner, J.M.; Detterbeck, F.C.; Jones, D.R. Special treatment issues in non-small cell lung cancer: Diagnosis and management of lung cancer, 3rd ed: American College of Chest Physicians evidence-based clinical practice guidelines. CHEST 2013, 143, e369S–e399S. [Google Scholar] [PubMed]
  19. Kong, F.M.; Ritter, T.; Quint, D.J.; Senan, S.; Gaspar, L.E.; Komaki, R.U.; Hurkmans, C.W.; Timmerman, R.; Bezjak, A.; Bradley, J.D.; et al. Consideration of dose limits for organs at risk of thoracic radiotherapy: Atlas for lung, proximal bronchial tree, esophagus, spinal cord, ribs, and brachial plexus. Int. J. Radiat. Oncol. Biol. Phys. 2011, 81, 1442–1457. [Google Scholar] [CrossRef] [PubMed]
  20. Bradley, J.D.; Paulus, R.; Komaki, R.; Masters, G.; Blumenschein, G.; Schild, S.; Bogart, J.; Hu, C.; Forster, K.; Magliocco, A.; et al. Standard-dose versus high-dose conformal radiotherapy with concurrent and consolidation carboplatin plus paclitaxel with or without cetuximab for patients with stage IIIA or IIIB non-small-cell lung cancer (RTOG 0617): A randomised, two-by-two factorial phase 3 study. Lancet Oncol. 2015, 16, 187–199. [Google Scholar] [CrossRef] [PubMed]
  21. van der Pol, L.H.G.; Hackett, S.L.; Hoesein, F.; Snoeren, L.M.W.; Pomp, J.; Raaymakers, B.W.; Verhoeff, J.J.C.; Fast, M.F. On the feasibility of cardiac substructure sparing in magnetic resonance imaging guided stereotactic lung radiotherapy. Med. Phys. 2023, 50, 397–409. [Google Scholar] [CrossRef] [PubMed]
  22. Chuter, R.; Prestwich, R.; Bird, D.; Scarsbrook, A.; Sykes, J.; Wilson, D.; Speight, R. The use of deformable image registration to integrate diagnostic MRI into the radiotherapy planning pathway for head and neck cancer. Radiother. Oncol. 2017, 122, 229–235. [Google Scholar] [CrossRef] [PubMed]
  23. Rasch, C.; Barillot, I.; Remeijer, P.; Touw, A.; van Herk, M.; Lebesque, J.V. Definition of the prostate in CT and MRI: A multi-observer study. Int. J. Radiat. Oncol. Biol. Phys. 1999, 43, 57–66. [Google Scholar] [CrossRef]
  24. Aoyama, H.; Shirato, H.; Nishioka, T.; Hashimoto, S.; Tsuchiya, K.; Kagei, K.; Onimaru, R.; Watanabe, Y.; Miyasaka, K. Magnetic resonance imaging system for three-dimensional conformal radiotherapy and its impact on gross tumor volume delineation of central nervous system tumors. Int. J. Radiat. Oncol. Biol. Phys. 2001, 50, 821–827. [Google Scholar] [CrossRef] [PubMed]
  25. Whiteside, L.; McDaid, L.; Hales, R.B.; Rodgers, J.; Dubec, M.; Huddart, R.A.; Choudhury, A.; Eccles, C.L. To see or not to see: Evaluation of magnetic resonance imaging sequences for use in MR Linac-based radiotherapy treatment. J. Med. Imaging Radiat. Sci. 2022, 53, 362–373. [Google Scholar] [CrossRef]
  26. Karki, K.; Saraiya, S.; Hugo, G.D.; Mukhopadhyay, N.; Jan, N.; Schuster, J.; Schutzer, M.; Fahrner, L.; Groves, R.; Olsen, K.M.; 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, 80–89. [Google Scholar] [CrossRef] [PubMed]
  27. Choudhury, A.; Budgell, G.; MacKay, R.; Falk, S.; Faivre-Finn, C.; Dubec, M.; van Herk, M.; McWilliam, A. The Future of Image-guided Radiotherapy. Clin. Oncol. (R Coll. Radiol.) 2017, 29, 662–666. [Google Scholar] [CrossRef] [PubMed]
  28. Eccles, C.L.; Adair Smith, G.; Bower, L.; Hafeez, S.; Herbert, T.; Hunt, A.; McNair, H.A.; Ofuya, M.; Oelfke, U.; Nill, S.; et al. Magnetic resonance imaging sequence evaluation of an MR Linac system; early clinical experience. Tech. Innov. Patient Support. Radiat. Oncol. 2019, 12, 56–63. [Google Scholar] [CrossRef] [PubMed]
  29. Fleckenstein, J.; Jelden, M.; Kremp, S.; Jagoda, P.; Stroeder, J.; Khreish, F.; Ezziddin, S.; Buecker, A.; Rübe, C.; Schneider, G.K. The Impact of Diffusion-Weighted MRI on the Definition of Gross Tumor Volume in Radiotherapy of Non-Small-Cell Lung Cancer. PLoS ONE 2016, 11, e0162816. [Google Scholar] [CrossRef] [PubMed]
  30. Wee, C.W.; An, H.J.; Kang, H.C.; Kim, H.J.; Wu, H.G. Variability of Gross Tumor Volume Delineation for Stereotactic Body Radiotherapy of the Lung With Tri-(60)Co Magnetic Resonance Image-Guided Radiotherapy System (ViewRay): A Comparative Study With Magnetic Resonance- and Computed Tomography-Based Target Delineation. Technol. Cancer Res. Treat. 2018, 17, 1533033818787383. [Google Scholar] [CrossRef] [PubMed]
  31. Gao, L.; Xie, K.; Wu, X.; Lu, Z.; Li, C.; Sun, J.; Lin, T.; Sui, J.; Ni, X. Generating synthetic CT from low-dose cone-beam CT by using generative adversarial networks for adaptive radiotherapy. Radiat. Oncol. 2021, 16, 202. [Google Scholar] [CrossRef] [PubMed]
  32. Lenkowicz, J.; Votta, C.; Nardini, M.; Quaranta, F.; Catucci, F.; Boldrini, L.; Vagni, M.; Menna, S.; Placidi, L.; Romano, A.; et al. A deep learning approach to generate synthetic CT in low field MR-guided radiotherapy for lung cases. Radiother. Oncol. 2022, 176, 31–38. [Google Scholar] [CrossRef] [PubMed]
  33. Freedman, J.N.; Bainbridge, H.E.; Nill, S.; Collins, D.J.; Kachelrieß, M.; Leach, M.O.; McDonald, F.; Oelfke, U.; Wetscherek, A. Synthetic 4D-CT of the thorax for treatment plan adaptation on MR-guided radiotherapy systems. Phys. Med. Biol. 2019, 64, 115005. [Google Scholar] [CrossRef] [PubMed]
  34. Brock, K.K. Adaptive Radiotherapy: Moving Into the Future. Semin. Radiat. Oncol. 2019, 29, 181–184. [Google Scholar] [CrossRef] [PubMed]
  35. Yan, D.; Vicini, F.; Wong, J.; Martinez, A. Adaptive radiation therapy. Phys. Med. Biol. 1997, 42, 123–132. [Google Scholar] [CrossRef] [PubMed]
  36. Ramella, S.; Fiore, M.; Silipigni, S.; Zappa, M.C.; Jaus, M.; Alberti, A.M.; Matteucci, P.; Molfese, E.; Cornacchione, P.; Greco, C.; et al. Local Control and Toxicity of Adaptive Radiotherapy Using Weekly CT Imaging: Results from the LARTIA Trial in Stage III NSCLC. J. Thorac. Oncol. 2017, 12, 1122–1130. [Google Scholar] [CrossRef] [PubMed]
  37. Lamb, J.; Cao, M.; Kishan, A.; Agazaryan, N.; Thomas, D.H.; Shaverdian, N.; Yang, Y.; Ray, S.; Low, D.A.; Raldow, A.; et al. Online Adaptive Radiation Therapy: Implementation of a New Process of Care. Cureus 2017, 9, e1618. [Google Scholar] [CrossRef] [PubMed]
  38. Liu, H.; Schaal, D.; Curry, H.; Clark, R.; Magliari, A.; Kupelian, P.; Khuntia, D.; Beriwal, S. Review of cone beam computed tomography based online adaptive radiotherapy: Current trend and future direction. Radiat. Oncol. 2023, 18, 144. [Google Scholar] [CrossRef] [PubMed]
  39. Kim, H.; Lee, P.; Tree, A.C.; Chuong, M.D.; Raldow, A.C.; Kishan, A.U.; Fuller, C.D.; Rosenberg, S.A.; Hall, W.A.; Chie, E.K.; et al. Adaptive Radiation Therapy Physician Guidelines: Recommendations From an Expert Users’ Panel. Pract. Radiat. Oncol. 2022, 12, e355–e362. [Google Scholar] [CrossRef] [PubMed]
  40. Kang, H.J.; Kwak, Y.K.; Kim, M.; Lee, S.J. Application of real-time MRI-guided linear accelerator in stereotactic ablative body radiotherapy for non-small cell lung cancer: One step forward to precise targeting. J. Cancer Res. Clin. Oncol. 2022, 148, 3215–3223. [Google Scholar] [CrossRef] [PubMed]
  41. Ng, J.; Gregucci, F.; Pennell, R.T.; Nagar, H.; Golden, E.B.; Knisely, J.P.S.; Sanfilippo, N.J.; Formenti, S.C. MRI-LINAC: A transformative technology in radiation oncology. Front. Oncol. 2023, 13, 1117874. [Google Scholar] [CrossRef]
  42. Thomas, D.H.; Santhanam, A.; Kishan, A.U.; Cao, M.; Lamb, J.; Min, Y.; O’Connell, D.; Yang, Y.; Agazaryan, N.; Lee, P.; et al. Initial clinical observations of intra- and interfractional motion variation in MR-guided lung SBRT. Br. J. Radiol. 2018, 91, 20170522. [Google Scholar] [CrossRef] [PubMed]
  43. Brown, S.; Banfill, K.; Aznar, M.C.; Whitehurst, P.; Faivre Finn, C. The evolving role of radiotherapy in non-small cell lung cancer. Br. J. Radiol. 2019, 92, 20190524. [Google Scholar] [CrossRef] [PubMed]
  44. Menten, M.J.; Fast, M.F.; Nill, S.; Kamerling, C.P.; McDonald, F.; Oelfke, U. Lung stereotactic body radiotherapy with an MR-linac—Quantifying the impact of the magnetic field and real-time tumor tracking. Radiother. Oncol. 2016, 119, 461–466. [Google Scholar] [CrossRef] [PubMed]
  45. Glitzner, M.; Woodhead, P.L.; Borman, P.T.S.; Lagendijk, J.J.W.; Raaymakers, B.W. Technical note: MLC-tracking performance on the Elekta unity MRI-linac. Phys. Med. Biol. 2019, 64, 15NT02. [Google Scholar] [CrossRef] [PubMed]
  46. Lombardo, E.; Rabe, M.; Xiong, Y.; Nierer, L.; Cusumano, D.; Placidi, L.; Boldrini, L.; Corradini, S.; Niyazi, M.; Belka, C.; et al. Offline and online LSTM networks for respiratory motion prediction in MR-guided radiotherapy. Phys. Med. Biol. 2022, 67, 095006. [Google Scholar] [CrossRef]
  47. Kaza, E.; Dunlop, A.; Panek, R.; Collins, D.J.; Orton, M.; Symonds-Tayler, R.; McQuaid, D.; Scurr, E.; Hansen, V.; Leach, M.O. Lung volume reproducibility under ABC control and self-sustained breath-holding. J. Appl. Clin. Med. Phys. 2017, 18, 154–162. [Google Scholar] [CrossRef] [PubMed]
  48. Brock, J.; McNair, H.A.; Panakis, N.; Symonds-Tayler, R.; Evans, P.M.; Brada, M. The use of the Active Breathing Coordinator throughout radical non-small-cell lung cancer (NSCLC) radiotherapy. Int. J. Radiat. Oncol. Biol. Phys. 2011, 81, 369–375. [Google Scholar] [CrossRef] [PubMed]
  49. Kaza, E.; Symonds-Tayler, R.; Collins, D.J.; McDonald, F.; McNair, H.A.; Scurr, E.; Koh, D.-M.; Leach, M.O. First MRI application of an active breathing coordinator. Phys. Med. Biol. 2015, 60, 1681–1696. [Google Scholar] [CrossRef] [PubMed]
  50. van Houdt, P.J.; Yang, Y.; van der Heide, U.A. Quantitative Magnetic Resonance Imaging for Biological Image-Guided Adaptive Radiotherapy. Front. Oncol. 2020, 10, 615643. [Google Scholar] [CrossRef] [PubMed]
  51. Shintani, T.; Matsuo, Y.; Iizuka, Y.; Mitsuyoshi, T.; Umeoka, S.; Nakamoto, Y.; Mizowaki, T.; Togashi, K.; Hiraoka, M. Assessment of treatment response after lung stereotactic body radiotherapy using diffusion weighted magnetic resonance imaging and positron emission tomography: A pilot study. Eur. J. Radiol. 2017, 92, 58–63. [Google Scholar] [CrossRef]
  52. Chang, Q.; Wu, N.; Ouyang, H.; Huang, Y. Diffusion-weighted magnetic resonance imaging of lung cancer at 3.0 T: A preliminary study on monitoring diffusion changes during chemoradiation therapy. Clin. Imaging 2012, 36, 98–103. [Google Scholar] [CrossRef] [PubMed]
  53. Weiss, E.; Ford, J.C.; Olsen, K.M.; Karki, K.; Saraiya, S.; Groves, R.; Hugo, G.D. Apparent diffusion coefficient (ADC) change on repeated diffusion-weighted magnetic resonance imaging during radiochemotherapy for non-small cell lung cancer: A pilot study. Lung Cancer 2016, 96, 113–119. [Google Scholar] [CrossRef] [PubMed]
  54. Yabuuchi, H.; Hatakenaka, M.; Takayama, K.; Matsuo, Y.; Sunami, S.; Kamitani, T.; Jinnouchi, M.; Sakai, S.; Nakanishi, Y.; Honda, H. Non-small cell lung cancer: Detection of early response to chemotherapy by using contrast-enhanced dynamic and diffusion-weighted MR imaging. Radiology 2011, 261, 598–604. [Google Scholar] [CrossRef] [PubMed]
  55. Sampath, S.; Rahmanuddin, S.; Sahoo, P.; Frankel, P.; Boswell, S.; Wong, J.; Rotter, A.; Rockne, R.; Wong, J.; Park, J.M. Change in Apparent Diffusion Coefficient Is Associated With Local Failure After Stereotactic Body Radiation Therapy for Non-Small Cell Lung Cancer: A Prospective Clinical Trial. Int. J. Radiat. Oncol. Biol. Phys. 2019, 105, 659–663. [Google Scholar] [CrossRef] [PubMed]
  56. Sun, Y.S.; Cui, Y.; Tang, L.; Qi, L.P.; Wang, N.; Zhang, X.Y.; Cao, K.; Zhang, X.P. Early evaluation of cancer response by a new functional biomarker: Apparent diffusion coefficient. AJR Am. J. Roentgenol. 2011, 197, W23–W29. [Google Scholar] [CrossRef] [PubMed]
  57. Seki, S.; Fujisawa, Y.; Yui, M.; Kishida, Y.; Koyama, H.; Ohyu, S.; Sugihara, N.; Yoshikawa, T.; Ohno, Y. Dynamic Contrast-enhanced Area-detector CT vs Dynamic Contrast-enhanced Perfusion MRI vs FDG-PET/CT: Comparison of Utility for Quantitative Therapeutic Outcome Prediction for NSCLC Patients Undergoing Chemoradiotherapy. Magn. Reson. Med. Sci. 2020, 19, 29–39. [Google Scholar] [CrossRef]
  58. Tao, X.; Wang, L.; Hui, Z.; Liu, L.; Ye, F.; Song, Y.; Tang, Y.; Men, Y.; Lambrou, T.; Su, 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. [Google Scholar] [CrossRef] [PubMed]
  59. Mehrabian, H.; Desmond, K.L.; Chavez, S.; Bailey, C.; Rola, R.; Sahgal, A.; Czarnota, G.J.; Soliman, H.; Martel, A.L.; Stanisz, G.J. Water Exchange Rate Constant as a Biomarker of Treatment Efficacy in Patients With Brain Metastases Undergoing Stereotactic Radiosurgery. Int. J. Radiat. Oncol. Biol. Phys. 2017, 98, 47–55. [Google Scholar] [CrossRef]
  60. Desmond, K.L.; Mehrabian, H.; Chavez, S.; Sahgal, A.; Soliman, H.; Rola, R.; Stanisz, G.J. Chemical exchange saturation transfer for predicting response to stereotactic radiosurgery in human brain metastasis. Magn. Reson. Med. 2017, 78, 1110–1120. [Google Scholar] [CrossRef] [PubMed]
  61. Gutsche, R.; Lohmann, P.; Hoevels, M.; Ruess, D.; Galldiks, N.; Visser-Vandewalle, V.; Treuer, H.; Ruge, M.; Kocher, M. Radiomics outperforms semantic features for prediction of response to stereotactic radiosurgery in brain metastases. Radiother. Oncol. 2022, 166, 37–43. [Google Scholar] [CrossRef] [PubMed]
  62. Salem, A.; Little, R.A.; Latif, A.; Featherstone, A.K.; Babur, M.; Peset, I.; Cheung, S.; Watson, Y.; Tessyman, V.; Mistry, H.; et al. Oxygen-Enhanced MRI Is Feasible, Repeatable, and Detects Radiotherapy-induced Change in Hypoxia in Xenograft Models and in Patients with Non-Small Cell Lung Cancer. Clin. Cancer Res. 2019, 25, 3818–3829. [Google Scholar] [CrossRef]
  63. Moon, J.W.; Yang, E.; Kim, J.H.; Kwon, O.J.; Park, M.A.-O.; Yi, C.A. Predicting Non-Small-Cell Lung Cancer Survival after Curative Surgery via Deep Learning of Diffusion MRI. Diagnostics 2023, 13, 2555. [Google Scholar] [CrossRef] [PubMed]
  64. Mayr, N.A.; Yuh, W.T.; Magnotta, V.A.; Ehrhardt, J.C.; Wheeler, J.A.; Sorosky, J.I.; Davis, C.S.; Wen, B.-C.; Martin, D.D.; Pelsang, R.E.; et al. Tumor perfusion studies using fast magnetic resonance imaging technique in advanced cervical cancer: A new noninvasive predictive assay. Int. J. Radiat. Oncol. Biol. Phys. 1996, 36, 623–633. [Google Scholar] [CrossRef] [PubMed]
  65. Baer, A.H.; Hoff, B.A.; Srinivasan, A.; Galbán, C.J.; Mukherji, S.K. Feasibility analysis of the parametric response map as an early predictor of treatment efficacy in head and neck cancer. AJNR Am. J. Neuroradiol. 2015, 36, 757–762. [Google Scholar] [CrossRef] [PubMed]
  66. Xie, T.; Ye, Z.; Pang, P.; Shao, G. Quantitative Multiparametric MRI May Augment the Response to Radiotherapy in Mid-Treatment Assessment of Patients with Esophageal Carcinoma. Oncol. Res. Treat. 2019, 42, 326–333. [Google Scholar] [CrossRef] [PubMed]
  67. Klaar, R.; Rabe, M.; Gaass, T.; Schneider, M.J.; Benlala, I.; Eze, C.; Corradini, S.; Belka, C.; Landry, G.; Kurz, C.; et al. Ventilation and perfusion MRI at a 0.35 T MR-Linac: Feasibility and reproducibility study. Radiat. Oncol. 2023, 18, 58. [Google Scholar] [CrossRef] [PubMed]
  68. Kapadia, A.; Mehrabian, H.; Conklin, J.; Symons, S.P.; Maralani, P.J.; Stanisz, G.J.; Sahgal, A.; Soliman, H.; Heyn, C.C. Temporal evolution of perfusion parameters in brain metastases treated with stereotactic radiosurgery: Comparison of intravoxel incoherent motion and dynamic contrast enhanced MRI. J. Neurooncol. 2017, 135, 119–127. [Google Scholar] [CrossRef] [PubMed]
  69. Siva, S.; Thomas, R.; Callahan, J.; Hardcastle, N.; Pham, D.; Kron, T.; Hicks, R.J.; MacManus, M.P.; Ball, D.L.; Hofman, M.S. High-resolution pulmonary ventilation and perfusion PET/CT allows for functionally adapted intensity modulated radiotherapy in lung cancer. Radiother. Oncol. 2015, 115, 157–162. [Google Scholar] [CrossRef] [PubMed]
  70. Mahasittiwat, P.; Yuan, S.; Xie, C.; Ritter, T.; Cao, Y.; Haken, R.K.T.; Kong, F.-M.S. Metabolic Tumor Volume on PET Reduced More than Gross Tumor Volume on CT during Radiotherapy in Patients with Non-Small Cell Lung Cancer Treated with 3DCRT or SBRT. J. Radiat. Oncol. 2013, 2, 191–202. [Google Scholar] [CrossRef] [PubMed]
  71. Ling, C.; Humm, J.; Larson, S.; Amols, H.; Fuks, Z.; Leibel, S.; Koutcher, J.A. Towards multidimensional radiotherapy (MD-CRT): Biological imaging and biological conformality. Int. J. Radiat. Oncol. Biol. Phys. 2000, 47, 551–560. [Google Scholar] [CrossRef] [PubMed]
  72. Foo, C.T.; Langton, D.; Thompson, B.R.; Thien, F. Functional lung imaging using novel and emerging MRI techniques. Front. Med. 2023, 10, 1060940. [Google Scholar] [CrossRef] [PubMed]
  73. Ireland, R.H.; Tahir, B.A.; Wild, J.M.; Lee, C.E.; Hatton, M.Q. Functional Image-guided Radiotherapy Planning for Normal Lung Avoidance. Clin. Oncol. (R Coll. Radiol.) 2016, 28, 695–707. [Google Scholar] [CrossRef] [PubMed]
  74. Yamamoto, T.; Kabus, S.; Bal, M.; Keall, P.J.; Moran, A.; Wright, C.; Benedict, S.H.; Holland, D.; Mahaffey, N.; Qi, L.; et al. Four-Dimensional Computed Tomography Ventilation Image-Guided Lung Functional Avoidance Radiation Therapy: A Single-Arm Prospective Pilot Clinical Trial. Int. J. Radiat. Oncol. Biol. Phys. 2023, 115, 1144–1154. [Google Scholar] [CrossRef] [PubMed]
  75. Porter, E.A.-O.; Myziuk, N.A.-O.; Quinn, T.J.; Lozano, D.; Peterson, A.B.; Quach, D.A.-O.; Siddiqui, Z.A.-O.; Guerrero, T.A.-O. Synthetic pulmonary perfusion images from 4DCT for functional avoidance using deep learning. Phys. Med. Biol. 2021, 66, 175005. [Google Scholar] [CrossRef] [PubMed]
  76. Najjar, R. Redefining Radiology: A Review of Artificial Intelligence Integration in Medical Imaging. Diagnostics 2023, 13, 2760. [Google Scholar] [CrossRef]
  77. Koh, D.-M.; Papanikolaou, N.; Bick, U.; Illing, R.; Kahn, C.E.; Kalpathi-Cramer, J.; Matos, C.; Martí-Bonmatí, L.; Miles, A.; Mun, S.K.; et al. Artificial intelligence and machine learning in cancer imaging. Commun. Med. 2022, 2, 133. [Google Scholar] [CrossRef] [PubMed]
  78. Dikici, E.; Bigelow, M.; Prevedello, L.M.; White, R.D.; Erdal, B.S. Integrating AI into radiology workflow: Levels of research, production, and feedback maturity. J. Med. Imaging 2020, 7, 016502. [Google Scholar] [CrossRef] [PubMed]
  79. Allen, B.; Agarwal, S.; Coombs, L.; Wald, C.; Dreyer, K. 2020 ACR Data Science Institute Artificial Intelligence Survey. J. Am. Coll. Radiol. 2021, 18, 1153–1159. [Google Scholar] [CrossRef] [PubMed]
  80. Hosny, A.; Parmar, C.; Quackenbush, J.; Schwartz, L.H.; Aerts, H. Artificial intelligence in radiology. Nat. Rev. Cancer 2018, 18, 500–510. [Google Scholar] [CrossRef] [PubMed]
  81. Beaton, L.; Bandula, S.; Gaze, M.N.; Sharma, R.A. How rapid advances in imaging are defining the future of precision radiation oncology. Br. J. Cancer 2019, 120, 779–790. [Google Scholar] [CrossRef] [PubMed]
  82. Ribeiro, M.F.; Marschner, S.; Kawula, M.; Rabe, M.; Corradini, S.; Belka, C.; Riboldi, M.; Landry, G.; Kurz, C. Deep learning based automatic segmentation of organs-at-risk for 0.35 T MRgRT of lung tumors. Radiat. Oncol. 2023, 18, 135. [Google Scholar] [CrossRef] [PubMed]
  83. Lindberg, K.; Grozman, V.; Karlsson, K.; Lindberg, S.; Lax, I.; Wersäll, P.; Persson, G.F.; Josipovic, M.; Khalil, A.A.; Moeller, D.S.; et al. The HILUS-Trial-a Prospective Nordic Multicenter Phase 2 Study of Ultracentral Lung Tumors Treated With Stereotactic Body Radiotherapy. J. Thorac. Oncol. 2021, 16, 1200–1210. [Google Scholar] [CrossRef] [PubMed]
  84. Simone, C.B., 2nd; Bogart, J.A.; Cabrera, A.R.; Daly, M.E.; DeNunzio, N.J.; Detterbeck, F.; Faivre-Finn, C.; Gatschet, N.; Gore, E.; Jabbour, S.K.; et al. Radiation Therapy for Small Cell Lung Cancer: An ASTRO Clinical Practice Guideline. Pract. Radiat. Oncol. 2020, 10, 158–173. [Google Scholar] [CrossRef] [PubMed]
  85. Ligtenberg, H.; Hackett, S.L.; Merckel, L.G.; Snoeren, L.; Kontaxis, C.; Zachiu, C.; Bol, G.H.; Verhoeff, J.J.C.; Fast, M.F. Towards mid-position based Stereotactic Body Radiation Therapy using online magnetic resonance imaging guidance for central lung tumours. Phys. Imaging Radiat. Oncol. 2022, 23, 24–31. [Google Scholar] [CrossRef] [PubMed]
  86. Rabe, M.; Palacios, M.A.; van Sörnsen de Koste, J.R.; Eze, C.; Hillbrand, M.; Belka, C.; Landry, G.; Senan, S.; Kurz, C. Comparison of MR-guided radiotherapy accumulated doses for central lung tumors with non-adaptive and online adaptive proton therapy. Med. Phys. 2023, 50, 2625–2636. [Google Scholar] [CrossRef] [PubMed]
  87. Regnery, S.; Katsigiannopulos, E.; Hoegen, P.; Weykamp, F.; Sandrini, E.; Held, T.; Deng, M.; Eichkorn, T.; Buchele, C.; Rippke, C.; et al. To fly or not to fly: Stereotactic MR-guided adaptive radiotherapy effectively treats ultracentral lung tumors with favorable long-term outcomes. Lung Cancer 2023, 179, 107175. [Google Scholar] [CrossRef] [PubMed]
  88. Sandoval, M.L.; Sim, A.J.; Bryant, J.M.; Bhandari, M.; Wuthrick, E.J.; Perez, B.A.; Dilling, T.J.; Redler, G.; Andreozzi, J.; Nardella, L.; et al. Magnetic Resonance-Guided Stereotactic Body Radiation Therapy/Hypofractionated Radiation therapy for Metastatic and Primary Central and Ultracentral Lung Lesions. JTO Clin. Res. Rep. 2023, 4, 100488. [Google Scholar] [CrossRef] [PubMed]
  89. Finazzi, T.; Haasbeek, C.J.A.; Spoelstra, F.O.B.; Palacios, M.A.; Admiraal, M.A.; Bruynzeel, A.M.E.; Slotman, B.J.; Lagerwaard, F.J.; Senan, S. Clinical Outcomes of Stereotactic MR-Guided Adaptive Radiation Therapy for High-Risk Lung Tumors. Int. J. Radiat. Oncol. Biol. Phys. 2020, 107, 270–278. [Google Scholar] [CrossRef] [PubMed]
  90. Regnery, S.; Ristau, J.; Weykamp, F.; Hoegen, P.; Sprengel, S.D.; Paul, K.M.; Buchele, C.; Klüter, S.; Rippke, C.; Renkamp, C.K.; et al. Magnetic resonance guided adaptive stereotactic body radiotherapy for lung tumors in ultracentral location: The MAGELLAN trial (ARO 2021-3). Radiat. Oncol. 2022, 17, 102. [Google Scholar] [CrossRef] [PubMed]
  91. Daly, M.E.; Perks, J.R.; Chen, A.M. Patterns-of-care for thoracic stereotactic body radiotherapy among practicing radiation oncologists in the United States. J. Thorac. Oncol. 2013, 8, 202–207. [Google Scholar] [CrossRef] [PubMed]
  92. Singh, A.K.; Gomez-Suescun, J.A.; Stephans, K.L.; Bogart, J.A.; Hermann, G.M.; Tian, L.; Groman, A.; Videtic, G.M. One Versus Three Fractions of Stereotactic Body Radiation Therapy for Peripheral Stage I to II Non-Small Cell Lung Cancer: A Randomized, Multi-Institution, Phase 2 Trial. Int. J. Radiat. Oncol. Biol. Phys. 2019, 105, 752–759. [Google Scholar] [CrossRef] [PubMed]
  93. Videtic, G.M.; Paulus, R.; Singh, A.K.; Chang, J.Y.; Parker, W.; Olivier, K.R.; Timmerman, R.D.; Komaki, R.R.; Urbanic, J.J.; Stephans, K.L.; et al. Long-term Follow-Up on NRG Oncology RTOG 0915 (NCCTG N0927): A Randomized Phase 2 Study Comparing 2 Stereotactic Body Radiation Therapy Schedules for Medically Inoperable Patients With Stage I Peripheral Non-Small Cell Lung Cancer. Int. J. Radiat. Oncol. Biol. Phys. 2019, 103, 1077–1084. [Google Scholar] [CrossRef] [PubMed]
  94. Ng, S.S.W.; Ning, M.S.; Lee, P.; McMahon, R.A.; Siva, S.; Chuong, M.D. Single-Fraction Stereotactic Body Radiation Therapy: A Paradigm During the Coronavirus Disease 2019 (COVID-19) Pandemic and Beyond? Adv. Radiat. Oncol. 2020, 5, 761–773. [Google Scholar] [CrossRef] [PubMed]
  95. Finazzi, T.; van Sörnsen de Koste, J.R.; Palacios, M.A.; Spoelstra, F.O.B.; Slotman, B.J.; Haasbeek, C.J.A.; Senan, S. Delivery of magnetic resonance-guided single-fraction stereotactic lung radiotherapy. Phys. Imaging Radiat. Oncol. 2020, 14, 17–23. [Google Scholar] [CrossRef]
  96. Pokhrel, D.; Sanford, L.; Dhanireddy, B.; Molloy, J.; Randall, M.; McGarry, R.C. Flattening filter free VMAT for a stereotactic, single-dose of 30 Gy to lung lesion in a 15-min treatment slot. J. Appl. Clin. Med. Phys. 2020, 21, 6–12. [Google Scholar] [CrossRef] [PubMed]
  97. Chuong, M.D.; Kotecha, R.; Mehta, M.P.; Adamson, S.; Romaguera, T.; Hall, M.D.; Alvarez, D.; Gutierrez, A.N.; Mishra, V.; De Zarraga, F.; et al. Case report of visual biofeedback-driven, magnetic resonance-guided single-fraction SABR in breath hold for early stage non-small-cell lung cancer. Med. Dosim. 2021, 46, 247–252. [Google Scholar] [CrossRef] [PubMed]
  98. Choun, H.J.; Kim, J.I.; Choi, C.H.; Jung, S.; Jin, H.; Wu, H.G.; Chie, E.K.; Park, J.M. Performance evaluation of a visual guidance patient-controlled respiratory gating system for respiratory-gated magnetic-resonance image-guided radiation therapy. Phys. Eng. Sci. Med. 2022, 45, 809–816. [Google Scholar] [CrossRef] [PubMed]
  99. Kumar, S.S.; Higgins, K.A.; McGarry, R.C. Emerging Therapies for Stage III Non-Small Cell Lung Cancer: Stereotactic Body Radiation Therapy and Immunotherapy. Front. Oncol. 2017, 7, 197. [Google Scholar] [CrossRef] [PubMed]
  100. Zhang, Y.; Qin, Q.; Li, B.; Wang, J.; Zhang, K. Magnetic resonance imaging for N staging in non-small cell lung cancer: A systematic review and meta-analysis. Thorac. Cancer 2015, 6, 123–132. [Google Scholar] [CrossRef]
  101. Wu, L.-M.; Xu, J.-R.; Gu, H.-Y.; Hua, J.; Chen, J.; Zhang, W.; Haacke, E.M.; Hu, J. Preoperative mediastinal and hilar nodal staging with diffusion-weighted magnetic resonance imaging and fluorodeoxyglucose positron emission tomography/computed tomography in patients with non-small-cell lung cancer: Which is better? J. Surg. Res. 2012, 178, 304–314. [Google Scholar] [CrossRef]
  102. Dubec, M.; Brown, S.; Chuter, R.; Hales, R.; Whiteside, L.; Rodgers, J.; Parker, J.; Eccles, C.L.; van Herk, M.; Faivre-Finn, C.; et al. MRI and CBCT for lymph node identification and registration in patients with NSCLC undergoing radical radiotherapy. Radiother. Oncol. 2021, 159, 112–118. [Google Scholar] [CrossRef] [PubMed]
  103. Bainbridge, H.E.; Menten, M.J.; Fast, M.F.; Nill, S.; Oelfke, U.; McDonald, F. Treating locally advanced lung cancer with a 1.5T MR-Linac—Effects of the magnetic field and irradiation geometry on conventionally fractionated and isotoxic dose-escalated radiotherapy. Radiother. Oncol. 2017, 125, 280–285. [Google Scholar] [CrossRef] [PubMed]
  104. Eze, C.; Lombardo, E.; Nierer, L.; Xiong, Y.; Niyazi, M.; Belka, C.; Manapov, F.; Corradini, S. MR-guided radiotherapy in node-positive non-small cell lung cancer and severely limited pulmonary reserve: A report proposing a new clinical pathway for the management of high-risk patients. Radiat. Oncol. 2022, 17, 43. [Google Scholar] [CrossRef] [PubMed]
  105. La Rosa, A.; Mittauer, K.E.; Chuong, M.D.; Hall, M.D.; Kutuk, T.; Bassiri, N.; McCulloch, J.; Alvarez, D.; Herrera, R.; Gutierrez, A.N.; et al. Accelerated hypofractionated magnetic resonance-guided adaptive radiotherapy for oligoprogressive non-small cell lung cancer. Med. Dosim. 2023, 48, 238–244. [Google Scholar] [CrossRef] [PubMed]
  106. Oborn, B.M.; Metcalfe, P.E.; Butson, M.J.; Rosenfeld, A.B. Monte Carlo characterization of skin doses in 6 MV transverse field MRI-linac systems: Effect of field size, surface orientation, magnetic field strength, and exit bolus. Med. Phys. 2010, 37, 5208–5217. [Google Scholar] [CrossRef]
  107. Huang, C.Y.; Yang, B.; Lam, W.W.; Geng, H.; Cheung, K.Y.; Yu, S.K. Magnetic field induced dose effects in radiation therapy using MR-linacs. Med. Phys. 2023, 50, 3623–3636. [Google Scholar] [CrossRef] [PubMed]
  108. Das, I.J.; Yadav, P.; Mittal, B.B. Emergence of MR-Linac in Radiation Oncology: Successes and Challenges of Riding on the MRgRT Bandwagon. J. Clin. Med. 2022, 11, 5136. [Google Scholar] [CrossRef] [PubMed]
  109. Ma, Y.; Mou, X.; Beeraka, N.M.; Guo, Y.; Liu, J.; Dai, J.; Fan, R. Machine Log File and Calibration Errors-based Patient-specific Quality Assurance (QA) for Volumetric Modulated Arc Therapy (VMAT). Curr. Pharm. Des. 2023, 29, 2738–2751. [Google Scholar] [CrossRef] [PubMed]
  110. Murgić, J.; Gregov, M.; Mrčela, I.; Budanec, M.; Krengli, M.; Fröbe, A.; Franco, P. Mri-guided radiotherapy for prostate cancer: A new paradigm. Acta Clin. Croat. 2022, 61, 65–70. [Google Scholar] [CrossRef] [PubMed]
  111. Yang, B.; Yuan, J.; Cheung, K.Y.; Huang, C.Y.; Poon, D.M.C.; Yu, S.K. Magnetic Resonance-Guided Radiation Therapy of Patients With Cardiovascular Implantable Electronic Device on a 1.5 T Magnetic Resonance-Linac. Pract. Radiat. Oncol. 2022, 12, e56–e61. [Google Scholar] [CrossRef] [PubMed]
  112. Keesman, R.; van der Bijl, E.; Janssen, T.M.; Vijlbrief, T.; Pos, F.J.; van der Heide, U.A. Clinical workflow for treating patients with a metallic hip prosthesis using magnetic resonance imaging-guided radiotherapy. Phys. Imaging Radiat. Oncol. 2020, 15, 85–90. [Google Scholar] [CrossRef] [PubMed]
  113. Brunenberg, E.J.L.; Kusters, J.M.A.M.; Van Kollenburg, P.G.M.; Braam, P.M. SP-0469: The development of a one-stop-shop palliative radiotherapy treatment using MR and CBCT. Radiother. Oncol. 2014, 111, S183–S184. [Google Scholar] [CrossRef]
  114. Palacios, M.A.; Verheijen, S.; Schneiders, F.L.; Bohoudi, O.; Slotman, B.J.; Lagerwaard, F.J.; Senan, S. Same-day consultation, simulation and lung Stereotactic Ablative Radiotherapy delivery on a Magnetic Resonance-linac. Phys. Imaging Radiat. Oncol. 2022, 24, 76–81. [Google Scholar] [CrossRef] [PubMed]
  115. Regnery, S.; de Colle, C.; Eze, C.; Corradini, S.; Thieke, C.; Sedlaczek, O.; Schlemmer, H.P.; Dinkel, J.; Seith, F.; Kopp-Schneider, A.; et al. Pulmonary magnetic resonance-guided online adaptive radiotherapy of locally advanced: The PUMA trial. Radiat. Oncol. 2023, 18, 74. [Google Scholar] [CrossRef] [PubMed]
Figure 1. A historical timeline of linear accelerator development and emerging trends in radiation therapy. MLC: multileaf collimators; IGRT: image-guided radiation therapy; CBCT: cone-beam CT; RT: radiotherapy; MRI: magnetic resonance imaging; FDA: Food and Drug Administration.
Figure 1. A historical timeline of linear accelerator development and emerging trends in radiation therapy. MLC: multileaf collimators; IGRT: image-guided radiation therapy; CBCT: cone-beam CT; RT: radiotherapy; MRI: magnetic resonance imaging; FDA: Food and Drug Administration.
Cancers 16 02710 g001
Figure 2. Current radiotherapy workflow, its limitations, and MR-Linac advantages. RT: radiotherapy; MRI: magnetic resonance imaging; CT: computed tomography; PET: positron emission tomography; 4D-CT: 4-dimension computed tomography; CBCT: cone-beam computed tomography.
Figure 2. Current radiotherapy workflow, its limitations, and MR-Linac advantages. RT: radiotherapy; MRI: magnetic resonance imaging; CT: computed tomography; PET: positron emission tomography; 4D-CT: 4-dimension computed tomography; CBCT: cone-beam computed tomography.
Cancers 16 02710 g002
Table 1. Overview of studies on qMRI metrics as predictor of lung cancer treatment response.
Table 1. Overview of studies on qMRI metrics as predictor of lung cancer treatment response.
Study, Publication YearMRI TechniqueqMRI MetricLung Cancer TypeNumber of PatientsFollow-up Time Points
Shintani, T., et al., 2017 [51]DWIADC, SUVmaxNSCLC14Pre, 3, 6, 9, 12 months
Chang, Q., et al., 2012 [52]DWIADCadvanced lung carcinoma14at regular intervals until the date of death
Weiss, E., et al., 2016 [53]DWIADCAdenocarcinoma (4)
SCC (6)
10Pre, 3, 6 weeks
Yabuuchi, H., et al., 2011 [54]DWIADCNSCLC28Before and after the first course of chemotherapy
Sampath, S., et al., 2019 [55]DWIADCNSCLC13Pre, 1 month
Sun, Y. S., et al., 2011 [56]DWIADCLung cancer21Pre, 1, 3, 6 weeks
Seki, S., et al., 2020 [57]DCE-MRIPulmonary arterial perfusion
Systemic arterial perfusion
Total perfusion
Adenocarcinoma (35)
SCC (7)
LCLC (1)
43Pre, every 6 months post-treatment
Tao, X., et al., 2016 [58]DCE-MRIBF, BV, MTT,
Ktrans, Kep, Ve, Vp
NSCLC36Pre, 1 month
Mehrabian, H., et al., 2017 [59]DCE-MRIkIE, kep,
M0,I, M0,E, M0,V
Primary lung cancer9Pre, 1 week, 1 month
Desmond, K. L., et al., 2017 [60]CESTAPTw, AREX,
Lorentzian peak properties
Brain metastases from primary lung cancer25Pre, 1 week, 1 month
Gutsche, R., et al., 2022 [61]RadiomicsLocal textural featuresBrain metastases from NSCLC80Pre, 180 days post-treatment
Salem, A., et al., 2019 [62]OE-MRIperfused Oxy-RNSCLC23Pre, post-treatment
Papers were searched on PubMed with search terms “early response” and “radiotherapy” as well as MRI techniques and qMRI metrics mentioned in the table above. Only studies in humans and published in English were included. Reference list of the included papers was checked to identify other relevant papers. DWI, diffusion weight imaging; ADC, apparent diffusion coefficient; SUVmax, maximum standardized uptake value; NSCLC, non-small-cell lung cancer; Pre, pretreatment; SCC, squamous cell carcinoma; DCE-MRI, dynamic contrast-enhanced MRI; LCLC, large cell lung carcinoma; BF, Blood Flow; BV, blood volume; MTT, Mean Transit Time; Ktrans, endothelial transfer constant; Kep, reflux rate; Ve, fractional extravascular extracellular space volume; Vp, fractional plasma volume; kIE, extracellular water exchange rate constant; kep, efflux rate constant; M0,I, M0,E, M0,V, water compartment volume fractions; IVIM, intravoxel incoherent motion; f, perfusion fraction; CEST, chemical exchange saturation transfer; APTw, amide proton transfer weighted images; AREX, apparent exchange-dependent relaxation; OE-MRI, oxygen-enhanced MRI; perfused Oxy-R, oxygen-refractory signals in perfused tissue.
Table 2. Ongoing trials of magnetic resonance imaging-guided radiation therapy in treatment of lung cancer.
Table 2. Ongoing trials of magnetic resonance imaging-guided radiation therapy in treatment of lung cancer.
NCT Number (Registration Year)Study TypeTumor TypeRT RegimenCombined TherapyTrial Design (Arms)Primary OutcomeNotes
NCT03048760
(2017)
ProspectiveStage I-III NSCLC or SCLCN/ANilMRI scanMeasure differences between target and OAR volumes contoured on PET, CT, and MRI imagesTo evaluate the feasibility of MRI for the delineation of OAR and target volumes in lung cancer patients
NCT05237453
(2022)
ProspectiveLocally advanced NSCLCMR-guided ARTNilExperimental: MR-guided ARTClinical feasibilityTo demonstrate the feasibility of MR-guided online ART for locally advanced NSCLC
NCT03916419
(2019)
Phase 2Inoperable stage IIB, IIIA, and select IIIB and IIIC NSCLC60 Gy/15 FrPaclitaxel + Carboplatin + DurvalumabChemoradiation + DurvalumabSafety lead-in only: number of participants with dose-limiting toxicities
Phase II only: local control rate; regional control rate
To test the feasibility and outcomes of MR-guided hypofractionated ART with concurrent chemotherapy and consolidation Durvalumab for inoperable stage IIB, IIIA, and select IIIB and IIIC NSCLC
NCT04925583
(2021)
Phase 1Ultracentral-located lung tumorSBRTNilLevel 0: 10 Fr × 5.0 Gy
Level 1: 10 Fr × 5.5 Gy
Level 2: 10 Fr × 6.0 Gy
Level 3: 10 Fr × 6.5 Gy
Dose-limiting toxicityTo identify the maximum tolerated dose of MR-guided SBRT of ultracentral lung tumors
NCT05354596
(2022)
Phase II Ultracentrally located lung tumorsSBRTNilMR-Linacs with daily MR-guided plan adaptationToxicity: cumulative CTCAE grade ≥ 4 SABR-related toxicity (6, 12, 24, 60 months)To evaluate the feasibility and safety of daily adaptive MR-Linac-based SBRT in ultracentrally located lung tumors (primary, oligo-metastatic, or oligo-progressive)
NCT05903430
(2023)
Prospective cohortCentrally located lung cancerSABRNilNot mentioned>85% success in delivery and completion of SABR to patients recruited on protocolTo determine if the investigators are able to deliver highly focused, intense radiation to tumors in the abdominal region or chest cavity whilst limiting the dose to OAR using a high-field-strength MR-Linac
NCT04789486
(2021)
Phase 1
Phase 2
Centrally located lung tumorsSMARTAGuIXPhase 1: AGUIX + SMART
Phase 2: AGUIX + SMART; SMART only
Phase 1: MTD
Phase 2: compare local control at 12 months of MTD
To help determine the safety and efficacy of gadolinium-based nanoparticle, AGuIX, used in conjunction with SMART in the treatment of pancreatic cancer and lung tumors
NCT04075305
(2019)
Prospective cohortCancer patients receiving treatment and/or imaging on an MR-Linac machineNot mentionedNilNot mentionedPFS; survival; DFS (3, 6, 24 months)
Patient-reported health-related quality of life and tumor-specific quality of life; acute toxicity in CTCAE (3, 6, 12, 24 months)
Clinical tumor response; pathological tumor response (2 years)
To facilitate the evidence-based introduction of MR-Linac into clinical practice
NCT04946019
(2021)
Phase 2Brain metastases from NSCLC30 Gy/5 FrNilExperimental: MR-Linac-guided adaptive FSRT1-year intracranial PFSTo determine the efficacy and safety of MR-Linac-guided adaptive FSRT in patients with brain metastases in NSCLC
NSCLC, non-small-cell lung cancer; SCLC, small cell lung cancer; Gy, gray; Fr, fraction; MRI, magnetic resonance imaging; OAR, organ at risk; PET, positron emission tomography; CT, computed tomography; ART, adaptive radiotherapy; SBRT, stereotactic body radiation therapy; SABR, stereotactic ablative radiotherapy; CTCAE, common toxicity criteria for adverse events; MTD, maximum tolerated dose; AGuIX, Activation and Guidance of Irradiation X; SMART, stereotactic MR-guided adaptive radiation therapy; PFS, progression-free survival; DFS, disease-free survival; FSRT, fractionated stereotactic radiotherapy.
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

Cheng, S.H.; Lee, S.-Y.; Lee, H.-H. Harnessing the Power of Radiotherapy for Lung Cancer: A Narrative Review of the Evolving Role of Magnetic Resonance Imaging Guidance. Cancers 2024, 16, 2710. https://doi.org/10.3390/cancers16152710

AMA Style

Cheng SH, Lee S-Y, Lee H-H. Harnessing the Power of Radiotherapy for Lung Cancer: A Narrative Review of the Evolving Role of Magnetic Resonance Imaging Guidance. Cancers. 2024; 16(15):2710. https://doi.org/10.3390/cancers16152710

Chicago/Turabian Style

Cheng, Sarah Hsin, Shao-Yun Lee, and Hsin-Hua Lee. 2024. "Harnessing the Power of Radiotherapy for Lung Cancer: A Narrative Review of the Evolving Role of Magnetic Resonance Imaging Guidance" Cancers 16, no. 15: 2710. https://doi.org/10.3390/cancers16152710

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