1,596
Views
6
CrossRef citations to date
0
Altmetric
EDITORIAL

Rethink radiotherapy – BIGART 2017

, , , , , , , ORCID Icon & show all

Introduction

The present issue of Acta Oncologica contains studies presented at the 15th Acta Oncologica Symposium, held in Aarhus, Denmark on June 13–16, 2017. The symposium was dedicated to aspects of Biology-Guided Adaptive Radiotherapy (BiGART) and attracted more than 200 physicians, physicists, radiobiologists and other scientists with an active interest in this specific area.

Acta Oncologica has sponsored scientific symposia since the late 80s. The aim of these meetings has been to focus on oncological issues of emerging interest, preferably with a multidisciplinary and multi-professional approach. The current series of Acta Oncologica meetings with focus on radiotherapy (RT) have encompassed stereotactic body RT (SBRT) [Citation1], image-guided RT [Citation2], particle RT [Citation3] and BiGART [Citation4–6], all reflecting development in RT during the last decennium within the fields of treatment planning [Citation7–12], (biological) imaging [Citation13–33], adoptive RT, [Citation34–44] particle therapy [Citation45–50], clinical outcome [Citation51–63], quality assurance [Citation64,Citation65], and other important factors [Citation66–72].

The aim of BiGART is treatment adaptation in time and space based on biological and anatomical features, maximizing the therapeutic ratio for each individual patient. Key topics for the BiGART2017 conference included:

  • Biology of tumors and normal tissue to guide patient selection, target volumes and dose prescription in photon and particle therapy.

  • Functional imaging of tumors and normal tissues with functional imaging techniques based on MRI and PET, and the use of such images for dose painting and normal tissue avoidance in photon and particle therapy.

  • Treatment planning and delivery challenges in adaptation of photon and particle therapy based on changes in tumor and normal tissue biology, anatomy and/or function.

  • Clinical outcome of biology-guided and adaptive RT and particle therapy.

Particle therapy and ENLIGHT

The meeting was held back-to-back with the annual meeting of ENLIGHT, The European Network for LIGht ion Hadron Therapy. Thus, there was more emphasis on particle therapy-related issues than in previous BiGART meetings. The meetings provided a good opportunity for interaction between scientists and clinicians within the rapidly emerging field of ion beam therapy. Particle therapy needs to be well aligned with other radiation techniques as well as with general developments in cancer research and patient care. The combined strengths of European initiatives including BiGART, ENLIGHT, ESTRO and the European Particle Therapy Network, is required to promote clinical and research collaboration between the rapidly increasing number of European particle therapy centers and to ensure that particle therapy becomes integrated into overall radiation oncology community.

With the establishment of more and more particle (proton) centers [Citation45,Citation73–79], it has increasingly become apparent that the classical RBE estimate of 1.1 for protons can be a dubious number [Citation80–83]. The original data and the consequent compilation of these into a 1.1 global value have become a dogma, which deserves to be scrutinized. Not only is there a wide spectrum of RBE values, but as the presentations at the BIGART 2017 indicate, this variation is also related to different early and late morbidity endpoints and not least to the fractionation pattern [Citation81]. The use of a RBE of 1.1 is far too simple and there is a crucial need to derive more exact values, which can be used in future dose planning.

The BIGART 2017 symposium clearly identified this problem and pointed to the need for an international collaboration with the aim to clarify in detail, variations in RBE according to tissue types and dose per fraction. Such activity is planned within the framework of the European Particle Network, and should urgently be implemented. This demands a multicenter collaboration with ample access to animal experiments and experimental particle beams – and this may prove to be more difficult than optimistically expected.

Not only is the RBE under challenge [Citation81,Citation84] but so is the homogeneous expression of this factor throughout the entire spread out Bragg peak. The reasons for this being the increase in LET at the back of the peak, which is coherent with a decrease of the proton dose [Citation85–87]. The balance between a higher RBE value (due to increased LET) counter acted by the decrease in proton dose makes the real biological dose at the backside uncertain [Citation87]. There are both clinical and experimental evidence for more pronounced late effects than anticipated [Citation73,Citation84,Citation87], and the particle therapy world must clearly acknowledge this risk and counter act it until the problem is both qualitatively and quantitatively identified and included in the biological dose algorithms for treatment planning.

On top of this, comes the issue of whether proton irradiation, in fact, have a different quantitative cellular effect [Citation88], together with an increasing awareness of the importance of radiogenomics for the probability of developing various early and late morbidities [Citation79,Citation89–91]. This should be explored in detail and taken into consideration, in relation to the individual patient’s risk of morbidity. Since many centers plan to give the treatment according to the model-based approach [Citation92–95], it should be acknowledged that such risk models cannot be fully implemented without taking the aforementioned RBE, LET and genomic variations into consideration.

Despite the promising aspects, particle therapy and especially proton treatment, is still an issue where an understanding and clarification of the underlying radiobiology in especially normal tissues is far from fully described or understood. Before the magnitude of this is integrated into the coming treatment plans, we are probably unable to get the full benefit of these advanced treatment options.

Like in conventional photon-based RT, there is also a continuous development of delivery techniques for treatment with proton and ion beams. Spot-scanning delivery techniques are now mature for widespread clinical implementation, although challenges still remain, in particular, related to its use at tumor sites influenced by intra-fractional motion. At BiGART 2017, a number of interesting scientific and technological developments to further improve and exploit particle therapy delivery were addressed, such as laser-based accelerators [Citation96] as well as so-called grid therapy, with protons [Citation97].

The relatively high building and equipment costs are major challenges for the further adoption of particle therapy. Current facilities are based on cyclotrons or synchrotrons, requiring large building space. However, there is considerable interest in developing more compact facilities. One interesting avenue of research explores the use of laser-accelerated particles/protons [Citation98–101], which is reviewed in this issue by Karsch et al. [Citation96]. This principle is based on the ionizing effects of high-intensity laser pulses, causing ions to be accelerated out of the laser target foil. As they discuss in their review, important steps forward have been taken documenting the potential of this technology, in particular for protons, however, much work still remains to reach clinical maturity, including obtaining a stable beam of sufficient energy.

Grid therapy, or spatially fractionated RT, is currently attracting increasing interest although it dates back more than 100 years, to the work of the German radiologist Köhler [Citation102] who also explored this principle clinically using the low-energy X-rays available at that time. The grid therapy principle implies irradiating the tumor by an array or grid of narrow beam-lets (pencil beams), usually from one beam direction. Grid therapy has been explored initially with X-rays/photons [Citation103–106] as well as with synchrotron radiation [Citation107], and later also with particle therapy beams [Citation108]. It has been shown that tumor control can be achieved despite the rather inhomogeneous target dose distributions and that normal tissue effects can be reduced (exploiting the normal tissue volume effects). Henry et al. performed Monte Carlo simulations of n interlacing cross-firing grids of narrow (1–3 mm) proton beams, and achieved a much more homogenous dose throughout the target volume while keeping the grid therapy patterns in the normal tissues [Citation97]. For tumor sites with favorable locations, this idea seems to have a considerable potential.

In treatment planning for particle therapy, the issue of range uncertainty is a major concern, and methods to decrease this uncertainty are sought. Increasing the accuracy of stopping power determination in tissue can be obtained by the use of dual-energy CT [Citation109], and several presentations at BIGART 2017 addressed this. While range uncertainty remains an important issue, the problem in daily practice may not be so large as it is highly dependent on beam angles. A higher number of beams results in higher robustness concerning range uncertainties in treatment plans.

Given that there is still a significant range uncertainty, this needs to be included in the planning process, and this is most commonly done through robust optimization, which often also includes robustness toward geometrical uncertainties [Citation110–114]. While robust optimization is mostly directed toward range uncertainty and geometrical optimization, the patients’ changing anatomy during a course of treatment is an unsolved problem in particle RT. Better robustness can be obtained through a variety of techniques including primarily simpler field arrangements and robust optimization algorithms. Most noteworthy was the introduction of robustness toward anatomical changes through model prediction of the changing anatomy between treatment fractions. The changing anatomy between fractions is potentially very detrimental to dose coverage and is presently not addressed.

Emerging technologies and treatment principles

The BIGART 2017 contributions on image guidance and motion management mainly focused on particle therapy. Compared to photon RT, scanning beam proton therapy is much more impacted by organ motion and anatomical changes and has a much larger discrepancy between needed and commonly available in-room imaging. While proton pencil beam scanning dose distributions may look excellent in the treatment planning system, there will often be larger differences than for photon RT between the planned and actually delivered doses and thoracic and abdominal intensity-modulated proton therapy (IMPT) has still not matured into routine clinical practice. As pointed out at the conference, this calls for a comprehensive framework comprising the joined forces of robust treatment planning with robustness evaluation, in-room motion monitoring with dose evaluation and possibly adaptive replanning if the conditions for the plan robustness are not fulfilled at treatment.

If treatment evaluation by a full dose reconstruction is not possible, simpler geometrical predictors for the dose degradation with anatomy changes may be used [Citation115,Citation116]. The monitoring should ideally be performed throughout the treatment fraction for observation of intra-fraction baseline drifts [Citation117,Citation118] or – in the case of breath-hold treatments – variations in the breath-hold level [Citation119–121].

Tools to decide on individualized motion management techniques, such as treatment in breath-hold or in free breathing with or without gating, repainting strategy, must be implemented. Robust planning strategies that aim at sufficient target dose coverage for a range of likely patient positions, internal patient anatomies and proton ranges in the patient inherently result in more doses to the normal tissue. The level of proton plan robustness will thus impact the balance between photon and proton treatments in a model-based patient selection approach. A meaningful model-based patient selection, therefore, requires a level of proton plan robustness that ensures comparable confidence in the target dose coverage for both photons and protons in the comprehensive picture that includes motion management, in-room imaging and plan adaptation. Just like the model-based selection will change over time as more clinical data become available for improved morbidity modeling, it will also change as we gain a better understanding of the relationship between the planned doses used for decision making and the actually delivered doses.

While in-room imaging by on-board cone-beam CT has been standard in photon RT for a decade, CBCT has barely entered the proton treatment room. On the other hand, a massive growth to catch up with photon image guidance is expected for proton therapy over the next few years. Furthermore, exciting modality-specific technologies are under development for in vivo range verification in particle therapy by detection of the ultrasound signal or nuclear emissions that are generated in the patient by the ion beam. Another emerging imaging technique presented at BIGART 2017 is proton radiography for image-guidance in small animal experiments [Citation122], and a few contributions explored the RBE uncertainties of proton therapy in the context of 4D or motion-including modeling [Citation123,Citation124].

Models which predict morbidity after RT are currently most often based on risk factors related to classical patient characteristics and to treatment-related parameters. Typical examples of factors which may influence the development of side effects are age, comorbidities, chemotherapy and dose parameters. Identification of dosimetric risk factors can be used to build dose-response models [Citation125]. These models can be used to set clinically relevant priorities during treatment planning [Citation126–129]. Furthermore, identification of genetic factors which can improve the prediction of radiosensitivity is a research field receiving increasing attention. The hope is to further facilitate individualization of RT according to the risk of side effects. Identification of genomic predictors has shown to be a complicated field with contradictory findings [Citation130], many underpowered studies and lack of validation studies and convincing associations have required large amounts of data [Citation89,Citation131,Citation132]. During the BIGART 2017 meeting, a novel approach to identification of genomic predictors was discussed by J. Deasy, New York. The idea is to apply machine learning to make a prediction in samples with a larger number of predictors than events [Citation133]. The methodology includes the building of predictive models which are validated in independent patient samples. Further support of hypotheses may be established through identification of biological processes related to the genomic findings.

Assessment of the burden of side effects after RT is a complicated matter. The burden of symptoms and the related impact on quality of life [Citation134] is associated with both extend of bother as well as development and persistence over time. Actuarial statistics has been recommended for evaluation of radiation induced toxicities [Citation135–137], although this approach is limited by not taking into account the development of a given symptom after its first occurrence. Actuarial statistics is appropriate for irreversible symptoms but is less suited for evaluation of symptoms which fluctuate or heal over time. K. Kirchheiner, Vienna, demonstrated a novel statistical method to identify patients with late, persistent, substantial and treatment-related symptoms (LAPERS). This was done by taking into account baseline status as well as a progression over time as well as the duration of symptoms. The hypothesis is that LAPERS may identify those patients who are experiencing the most pronounced burden of persisting symptoms.

With the introduction of radiomics, use of images in RT is truly undergoing ‘rethinking’. The advanced analysis of images by quantitative feature extraction is still in early days, and validation studies to ensure the stability of features over for instance different scanners and different patient groups are still necessary and ongoing. However, at BIGART 2017, a range of radiomics applications were presented for several clinical sites, and for several imaging modalities (CT, CBCT, MR and PET) used throughout the course of RT. Prediction of recurrent disease was reported based on radiomics analysis for pretreatment CT versus PET images for head and neck cancer patients [Citation138], adding to previous studies on CT radiomics markers for both head and neck and lung cancers [Citation139,Citation140]. Cone-beam CT images were used for the longitudinal investigation to detect changes in features during RT in lung cancer patients, for potential decision support in treatment adaptation [Citation141,Citation142]. Overall, the results of radiomics analysis so far are immature and preliminary, and it remains to be seen whether this search for biological information in images will prove to be successful.

Image-based adaptation to anatomical changes during the course of RT has gradually become a clinical reality for many tumor sites [Citation143–157]. Practical guidelines on how to facilitate the clinical implementation of adaptive RT were presented with examples from rectal and cervix cancer. In locally advanced non-small cell lung cancer (NSCLC), a study showed how adaptive RT can result in dosimetric gain and treatment outcome prediction [Citation158]. Tumor shrinkage adaptive RT allowed significant organ at risk (OAR) dose reduction without compromising initial CTV coverage. The improved outcome was observed for patients with higher tumor shrinkage gradient. A study showed reduced acute and late toxicity rates in bladder cancer. A brain MRI study showed that ultra-early ADC footprint can predict response to RT, and thus facilitate individualization [Citation159]. Finally, a study of adaptation based on off-line CBCT dose reconstruction showed only marginal parotid sparing, when compared to daily CBCT image review by the radiation therapists.

Current indications and treatment principles for RT

The BIGART 2017 Symposium ended with a challenging look at the current indications and treatment principles for RT of some major tumor sites. That these are under continuous development is clearly demonstrated in the role of RT of breast cancer. This is by far the most prominent indication for (adjuvant) RT with more than half a million patients treated in Europe each year, and with a 5-year prevalence of more than 2 million women. Therefore, both the benefit of the treatment and the risk of side effects must be very clear. With an increase in the number of patients diagnosed after the screening, small tumors are becoming more frequent and at the same time, changed population demographics results in an increasing number of elderly women [Citation160,Citation161]. Together with a more intensive use of adjuvant systemic therapy, this has brought us into a situation where the benefit and risk profiles are both relatively small, but still, due to the large incidence and prevalence, the burden of treatment is relatively large.

The indication for RT is primarily to avoid local recurrence. This is successfully achieved, but maybe because the true incidence of recurrence is on a rapid decline [Citation162,Citation163], which for some low-risk patient groups, make the indications for RT at all questionable [Citation163,Citation164]. On the other hand, there is still a prominent enhanced risk in the very young patients [Citation161], which cannot be explained by differences in intrinsic subtypes or presence of BRCA mutations [Citation165], but still is an issue with an unsolved etiology. In more advanced cases, we still see an overall survival benefit of a magnitude of 2% (if patients have their internal mammary nodes includes in the radiation field) [Citation166], but the price for such activity may be an enhanced risk of cardiac morbidity and radiation induced lung cancer [Citation167–169], not least if the patient is a current smoker [Citation164].

The status of breast irradiation is influenced by many factors, which affect both overall outcome and morbidity, and the mutual pros and cons may call for revised visions on the importance of breast RT. In general, it seems the current risk of morbidity is larger than anticipated and the overall benefits smaller (or overlapping with that produced with systemic therapy). Therefore, a more individual consideration of the risks and benefits must be applied to secure an optimal individual balance [Citation79,Citation89,Citation164,Citation165,Citation170–174], and we must probably initiate trials to identify the indications for breast RT. In fact, such trials are already emerging.

Since this is happening on an ever changing epidemiological and demographic background, and where the number of events are relatively small (but in a large patient population), this can only be revealed in international collaborative studies. Fortunately, this activity has already started, not least based on the Skagen initiative, which has resulted in several collaborative protocols [Citation175].

Head and neck cancer is the classical tumor site for studying radiobiological based treatment optimization. Squamous cell carcinomas do typically exhibit the characteristics with a clear dose-response relationship, the influence of volume, dose per fraction, overall treatment time and hypoxia [Citation79,Citation176,Citation177]. In addition, individual prognostic factors are also clearly identifiable in this patient cohort [Citation178,Citation179]. In recent years, the scenario has changed dramatically with the almost epidemic increase in the number of human papillomavirus (HPV) positive oropharyngeal cancers [Citation180]. This has two implications, one being the fact that HPV positive tumors are found to be much more radiosensitive [Citation181] and consequently, these patients have a superior prognostic outcome which is further supported by reduced risk from smoking and comorbidity [Citation180,Citation182]. In turn, this has resulted in various attempts to reduce the treatment burden in HPV positive patients, either by descalation of the radiation dose or in small tumors to exchange RT with trans-oral surgery. Several studies are underway or under discussion to deal with this issue, but no conclusive results are available. Until this has happened, dose reduction in this patient cohort should only take place within the framework of controlled clinical trials. The other implication is linked to the treatment of the ‘traditional’ head and neck cancer patients, which are HPV negative and typically have a tobacco-related etiology with a consequential enhanced risk of late morbidity and comorbidity [Citation182–187]. Such patients, which demand the most challenging treatment, unfortunately also are the ones who have major difficulties in tolerating it. Therefore, they may be candidates for more refined radiation techniques with particle beam therapy, based on risk selecting strategies and/or the recurrence risk would be challenged based on individual risk parameters, focusing on especially hypoxia, tumor stem cells, immunological parameters or other individual molecular genomic parameters [Citation79,Citation178,Citation188,Citation189].

To comfort this plentiful scenario of treatment options, we are moving toward multicenter matrix protocols addressing both modified therapy with e.g. hypoxic radiosensitizers and predictive selection based on genetic profiles, radiogenomics or biological imaging. A challenge persists in identifying the risk of failures relative to intratumoral variations in tumor resistance (e.g. hypoxic imaging) [Citation190–192]. Currently, we are still awaiting results which may identify to which extent we can apply those escalation studies based on dose painting or if will need to include the entire cross tumor volume [Citation191,Citation193]. Most interestingly are we again back to placing our main focus on hypoxia, both the indication through biological imaging or through identification of predictive parameters [Citation189,Citation190,Citation194–196] and especially through exploration of therapeutic intervention [Citation185]. This takes e.g. place with the large international multicenter clinical trial (EORTC 1219/DAHANCA 29), which not only evaluates the importance of the hypoxic modifying sensitizer Nimorazole [Citation185,Citation189,Citation197,Citation198] but also investigates whether such potential benefit can be predicted by a hypoxic 15-gene probe [Citation188,Citation189]. The outcome of this study, together with similar other trials investigating Nimorazole [Citation197,Citation198], is likely to yield ultimate information on the role of hypoxic modification in head and neck cancer – and thereby answer a more than 100-years old question in clinical radiobiology [Citation199].

An overview of dose-volume-and fractionation effects in lung cancer was given by D. De Ruysscher, Maastricht. By SBRT, extreme high biological doses are delivered over a short time to small volumes. From a radiobiological point of view, it is not the most appropriate approach, but numerous studies have shown that SBRT of peripheral NSCLC is efficient and safe [Citation200–202]. Studies with very long follow-up confirm these findings [Citation203]. SBRT is now the preferred radical therapy of patients with severe chronic obstructive lung disease (who are in high risk for severe complication and death with surgery). With the current knowledge, there is no indication that SBRT of small volumes of lung result in deterioration of lung function [Citation204], but recent studies indicate that large doses to the heart may have detrimental effects and increase the patients risk for suffering non-cancer death [Citation205]. Advisory Committee on Radiation Oncology Practice (ACROP) guidelines providing advice on patient selection, equipment used and methodology of SBRT planning and delivery, quality assurance and patient follow-up have recently been published [Citation206].

SBRT of central tumors (within 2 cm from the bronchial tree) is related to a risk of fatal bleeding. Three of 71 patients in RTOG0813 had a grade 5 toxicity but in an analysis of ultra-central cases where PTV-overlap with the main bronchi and trachea this risk increased dramatically [Citation207,Citation208]. There is no current consensus on how to treat central tumors, but there is a general belief that they should be treated with softer fractionation if not conventional RT [Citation209].

Several randomized studies have aimed to test SBRT against surgical resection but were closed because of poor recruitment. A pooled analysis of two of the studies revealed a superior survival in the patients treated with SBRT compared to surgery, but the pooled analysis is not sufficiently powered to give a firm conclusion [Citation210]. A Scandinavian study compared SBRT with conventional fractionated RT [Citation211,Citation212]. It revealed no difference in survival and control of disease, but there was less toxicity in the SBRT treated patients. In stage III NSCLC, RT with total doses of 60–66 Gy concurrent with chemotherapy is considered the standard of care [Citation213]. Disease control and survival were expected to improve with dose escalation, but RTOG 0617 revealed inferior survival with the higher doses which has never been understood [Citation214]. Deaths because of excess toxicity are an obvious possibility and the toxicity should be further explored [Citation215–219]. A Danish study randomized patients with stage III NSCLC to 60 versus 66 Gy, both arms with concurrent oral vinorelbine. In this study, there was no difference between the study arms [Citation220]. Boost to FDG PET/CT-positive subvolumes of advanced stage NSCLC is currently being tested [Citation221].

Modern therapy of rectal cancer is truly multidisciplinary, involving surgeons, medical oncologists, radiologists, pathologists together with the radiation oncologist. Vincenzo Valentini gave a status of the radiation oncologist’s view on how radiation therapy should be implemented in the multimodality management of rectal cancer as it is described in the European Registration of Cancer Care consensus document [Citation222].

The Swedish Rectal Cancer Trial was the first to show that preoperative RT (pre-op RT) improved the outcome in patients with rectal cancer [Citation223]. A benefit in terms of improved local control, cancer-specific survival and overall survival appeared in all Dukes stages. Since then a number of randomized studies have confirmed that pre-op RT improves local control, but the Swedish study using short course 5 Gy ×5 without chemotherapy and with immediate surgery is still the only study to demonstrate a survival benefit [Citation224–228]. Studies using long-term chemoradiation with delayed surgery showed improved local control and 8–30% complete pathological responses without demonstrable viable cancer in the resected specimen [Citation229,Citation230]. A recent randomized Swedish three-arm study found no difference in local control between short course (5 Gy ×5) with and without delay before surgery or long course pre-op RT [Citation231–233]. The relatively high percentage of patients with the complete pathologic response has led to the hypothesis that surgery may be modified or omitted in some patients obtaining complete clinical remission after long course pre-op radio-chemotherapy. This is currently being tested in clinical trials. It is a general belief that pre-op RT is not needed for low-risk cancers if state-of-the-art total mesorectal excision is used and long course radio-chemotherapy is preferred in the more high-risk patients because of the high chance of local response compared to the short course regimen. The short course is often reserved for patients who are not amendable for pre-op chemotherapy. Pre-op RT comes to a prize with increased loss of sphincter control, but it seems less pronounced than after postoperative RT [Citation225,Citation234]. Due to limited evidence, the recommendations for the selection of patients for pre-op RT – for short or long course of pre-op RT and for adjuvant chemotherapy – are weak and there are large national differences in the practice of pre-op RT [Citation235].

In conclusion, the BiGART2017 meeting demonstrated a positive and strong development within the field of radiation oncology toward integrating clinical radiobiology, advanced imaging, state-of-the-art RT, and the most recent advances in ion beam therapy. This is a necessary development in order to secure that cancer patients will continue to be offered the most optimal therapy with photons or ion beam, in an integrated and evidence-based framework.

Disclosure statement

No potential conflict of interest was reported by the authors.

Additional information

Funding

This work was supported by a grant from Acta Oncological Foundation.

References

  • Grau C, Høyer M, Lindegaard J, et al. The emerging evidence for Stereotactic Body Radiotherapy. Acta Oncol. 2006;45:771–774.
  • Grau C, Muren LP, Høyer M, et al. Image-guided adaptive radiotherapy – integration of biology and technology to improve clinical outcome. Acta Oncol. 2008;47:1182–1185.
  • Muren LP, Rossi C, Hug E, et al. Establishing and expanding the indications for proton and particle therapy. Acta Oncol. 2013;52:459–462.
  • Grau C, Olsen DR, Overgaard J, et al. Biology-guided adaptive radiation therapy – presence or future? Acta Oncol. 2010;49:884–887.
  • Grau C, Høyer M, Alber M, et al. Biology-guided adaptive radiotherapy (BiGART) – more than a vision? Acta Oncol. 2013;52:1243–1247.
  • Grau C, Overgaard J, Høyer M, et al. Biology-guided adaptive radiotherapy (BiGART) is progressing towards clinical reality. Acta Oncol. 2015;54:1245–1250.
  • Caldas-Magalhaes J, Kooij N, Ligtenberg H, et al. The accuracy of target delineation in laryngeal and hypopharyngeal cancer. Acta Oncol. 2015;54:1181–1187.
  • Edmund JM, Andreasen D, Mahmood F, et al. Cone beam computed tomography guided treatment delivery and planning verification for magnetic resonance imaging only radiotherapy of the brain. Acta Oncol. 2015;54:1496–1500.
  • Grønborg C, Vestergaard A, Høyer M, et al. Intra-fractional bladder motion and margins in adaptive radiotherapy for urinary bladder cancer. Acta Oncol. 2015;54:1461–1466.
  • Mahmood F, Johannesen HH, Geertsen P, et al. The effect of region of interest strategies on apparent diffusion coefficient assessment in patients treated with palliative radiation therapy to brain metastases. Acta Oncol. 2015;54:1529–1534.
  • Martin S, Johnson C, Brophy M, et al. Impact of target volume segmentation accuracy and variability on treatment planning for 4D-CT-based non-small cell lung cancer radiotherapy. Acta Oncol. 2015;54:322–332.
  • Polders DL, Steggerda M, van Herk M, et al. Establishing implantation uncertainties for focal brachytherapy with I-125 seeds for the treatment of localized prostate cancer. Acta Oncol. 2015;54:839–846.
  • Casares-Magaz O, Thor M, Liao D, et al. An image-based method to quantify biomechanical properties of the rectum in radiotherapy of prostate cancer. Acta Oncol. 2015;54:1335–1342.
  • Feng Y, Welsh D, McDonald K, et al. Identifying the dominant prostate cancer focal lesion using image analysis and planning of a simultaneous integrated stereotactic boost. Acta Oncol. 2015;54:1543–1550.
  • Hoskin PJ. Hypoxia dose painting in prostate and cervix cancer. Acta Oncol. 2015;54:1259–1262.
  • Haack S, Tanderup K, Kallehauge JF, et al. Diffusion-weighted magnetic resonance imaging during radiotherapy of locally advanced cervical cancer-treatment response assessment using different segmentation methods. Acta Oncol. 2015;54:1535–1542.
  • Intven M, Monninkhof EM, Reerink O, et al. Combined T2w volumetry, DW-MRI and DCE-MRI for response assessment after neo-adjuvant chemoradiation in locally advanced rectal cancer. Acta Oncol. 2015;54:1729–1736.
  • Iversen AB, Ringgaard S, Laustsen C, et al. Hyperpolarized magnetic resonance spectroscopy for assessing tumor hypoxia. Acta Oncol. 2015;54:1393–1398.
  • Lakosi F, de Cuypere M, Viet Nguyen P, et al. Clinical efficacy and toxicity of radio-chemotherapy and magnetic resonance imaging-guided brachytherapy for locally advanced cervical cancer patients: a mono-institutional experience. Acta Oncol. 2015;54:1558–1566.
  • Møller S, Lundemann M, Law I, et al. Early changes in perfusion of glioblastoma during radio- and chemotherapy evaluated by T1-dynamic contrast enhanced magnetic resonance imaging. Acta Oncol. 2015;54:1521–1528.
  • Mönnich D, Welz S, Thorwarth D, et al. Robustness of quantitative hypoxia PET image analysis for predicting local tumor control. Acta Oncol. 2015;54:1364–1369.
  • Nkhali L, Thureau S, Edet-Sanson A, et al. FDG-PET/CT during concomitant chemo radiotherapy for esophageal cancer: reducing target volumes to deliver higher radiotherapy doses. Acta Oncol. 2015;54:909–915.
  • Noel CE, Parikh PJ, Spencer CR, et al. Comparison of onboard low-field magnetic resonance imaging versus onboard computed tomography for anatomy visualization in radiotherapy. Acta Oncol. 2015;54:1474–1482.
  • Pitman KE, Rusten E, Kristian A, et al. Variability of dynamic 18F-FDG-PET data in breast cancer xenografts. Acta Oncol. 2015;54:1399–1407.
  • Ramlov A, Kroon PS, Jürgenliemk-Schulz IM, et al. Impact of radiation dose and standardized uptake value of (18)FDG PET on nodal control in locally advanced cervical cancer. Acta Oncol. 2015;54:1567–1573.
  • Rasmussen GB, Vogelius IR, Rasmussen JH, et al. Immunohistochemical biomarkers and FDG uptake on PET/CT in head and neck squamous cell carcinoma. Acta Oncol. 2015;54:1408–1415.
  • Rasmussen JH, Vogelius IR, Aznar MC, et al. Spatio-temporal stability of pre-treatment 18F-Fludeoxyglucose uptake in head and neck squamous cell carcinomas sufficient for dose painting. Acta Oncol. 2015;54:1416–1422.
  • Sand HM, Brunø AH, Andersen LJ, et al. Correlation between pretreatment FDG-PET biological target volume and location of T-site failure after definitive radiation therapy for head and neck cancers. Acta Oncol. 2015;54:1682–1685.
  • van Dijk LK, Boerman OC, Kaanders JH, et al. Epidermal growth factor receptor imaging in human head and neck cancer xenografts. Acta Oncol. 2015;54:1263–1267.
  • Wack LJ, Mönnich D, van Elmpt W, et al. Comparison of [18F]-FMISO, [18F]-FAZA and [18F]-HX4 for PET imaging of hypoxia – a simulation study. Acta Oncol. 2015;54:1370–1377.
  • Wilbers J, Meijer FJ, Kappelle AC, et al. Magnetic resonance imaging of the carotid artery in long-term head and neck cancer survivors treated with radiotherapy. Acta Oncol. 2015;54:1175–1180.
  • Zegers CM, van Elmpt W, Hoebers FJ, et al. Imaging of tumour hypoxia and metabolism in patients with head and neck squamous cell carcinoma. Acta Oncol. 2015;54:1378–1384.
  • Zschaeck S, Haase R, Abolmaali N, et al. Spatial distribution of FMISO in head and neck squamous cell carcinomas during radio-chemotherapy and its correlation to pattern of failure. Acta Oncol. 2015;54:1355–1363.
  • Berkovic P, Paelinck L, Lievens Y, et al. Adaptive radiotherapy for locally advanced non-small cell lung cancer, can we predict when and for whom? Acta Oncol. 2015;54:1438–1444.
  • Joye I, Verstraete J, Bertoncini C, et al. Implementation of volumetric modulated arc therapy for rectal cancer: pitfalls and challenges. Acta Oncol. 2015;54:1677–1681.
  • Korhonen J, Kapanen M, Sonke JJ, et al. Feasibility of MRI-based reference images for image-guided radiotherapy of the pelvis with either cone-beam computed tomography or planar localization images. Acta Oncol. 2015;54:889–895.
  • Kurz C, Dedes G, Resch A, et al. Comparing cone-beam CT intensity correction methods for dose recalculation in adaptive intensity-modulated photon and proton therapy for head and neck cancer. Acta Oncol. 2015;54:1651–1657.
  • Majercakova K, Pötter R, Kirisits C, et al. Evaluation of planning aims and dose prescription in image-guided adaptive brachytherapy and radiochemotherapy for cervical cancer: Vienna clinical experience in 225 patients from 1998 to 2008. Acta Oncol. 2015;54:1551–1557.
  • Najim M, Perera L, Bendall L, et al. Volumetric and dosimetric changes to salivary glands during radiotherapy for head and neck cancer. Acta Oncol. 2015;54:1691–1693.
  • Nielsen MS, Østergaard LR, Carl J. A new method to validate thoracic CT-CT deformable image registration using auto-segmented 3D anatomical landmarks. Acta Oncol. 2015;54:1515–1520.
  • Nyeng TB, Nordsmark M, Hoffmann L. Dosimetric evaluation of anatomical changes during treatment to identify criteria for adaptive radiotherapy in oesophageal cancer patients. Acta Oncol. 2015;54:1467–1473.
  • Persoon LC, Podesta M, Hoffmann L, et al. Is integrated transit planar portal dosimetry able to detect geometric changes in lung cancer patients treated with volumetric modulated arc therapy? Acta Oncol. 2015;54:1501–1507.
  • Petersen PM, Aznar MC, Berthelsen AK, et al. Prospective phase II trial of image-guided radiotherapy in Hodgkin lymphoma: benefit of deep inspiration breath-hold. Acta Oncol. 2015;54:60–66.
  • Sibolt P, Ottosson W, Sjöström D, et al. Adaptation requirements due to anatomical changes in free-breathing and deep-inspiration breath-hold for standard and dose-escalated radiotherapy of lung cancer patients. Acta Oncol. 2015;54:1453–1460.
  • Fujii O, Demizu Y, Hashimoto N, et al. Particle therapy for clinically diagnosed stage I lung cancer: comparison with pathologically proven non-small cell lung cancer. Acta Oncol. 2015;54:315–321.
  • Góra J, Kuess P, Stock M, et al. ART for head and neck patients: on the difference between VMAT and IMPT. Acta Oncol. 2015;54:1166–1174.
  • Jakobi A, Stützer K, Bandurska-Luque A, et al. NTCP reduction for advanced head and neck cancer patients using proton therapy for complete or sequential boost treatment versus photon therapy. Acta Oncol. 2015;54:1658–1664.
  • Kole TP, Nichols RC, Lei S, et al. A dosimetric comparison of ultra-hypofractionated passively scattered proton radiotherapy and stereotactic body radiotherapy (SBRT) in the definitive treatment of localized prostate cancer. Acta Oncol. 2015;54:825–831.
  • Lassen-Ramshad Y, Petersen JB, Tietze A, et al. Pseudoprogression after proton radiotherapy for pediatric low grade glioma. Acta Oncol. 2015;54:1701–1702.
  • Makita C, Nakamura T, Takada A, et al. High-dose proton beam therapy for stage I non-small cell lung cancer: clinical outcomes and prognostic factors. Acta Oncol. 2015;54:307–314.
  • Belli ML, Fiorino C, Zerbetto F, et al. Early volume variation of positive lymph nodes assessed by in-room mega voltage CT images predicts risk of loco-regional relapses in head and neck cancer patients treated with intensity-modulated radiotherapy. Acta Oncol. 2015;54:1490–1495.
  • Haas RL, Gelderblom H, Sleijfer S, et al. A phase I study on the combination of neoadjuvant radiotherapy plus pazopanib in patients with locally advanced soft tissue sarcoma of the extremities. Acta Oncol. 2015;54:1195–1201.
  • Jakobsen A, Andersen F, Fischer A, et al. Neoadjuvant chemotherapy in locally advanced colon cancer. A phase II trial. Acta Oncol. 2015;54:1747–1753.
  • Lassen P, Eriksen JG, Krogdahl A, et al. The influence of HPV-associated p16-expression on accelerated fractionated radiotherapy in head and neck cancer: evaluation of the randomised DAHANCA 6&7 trial. Radiother Oncol. 2011;100:49–55.
  • Lilja-Fischer JK, Jensen K, Eskildsen HW, et al. Response evaluation of the neck in oropharyngeal cancer: value of magnetic resonance imaging and influence of p16 in selecting patients for post-radiotherapy neck dissection. Acta Oncol. 2015;54:1599–1606.
  • Pereira AA, Rego JF, Munhoz RR, et al. The impact of complete chemotherapy stop on the overall survival of patients with advanced colorectal cancer in first-line setting: a meta-analysis of randomized trials. Acta Oncol. 2015;54:1737–1746.
  • Rønjom MF, Brink C, Bentzen SM, et al. External validation of a normal tissue complication probability model for radiation-induced hypothyroidism in an independent cohort. Acta Oncol. 2015;54:1301–1309.
  • Stokkevåg CH, Engeseth GM, Hysing LB, et al. Risk of radiation-induced secondary rectal and bladder cancer following radiotherapy of prostate cancer. Acta Oncol. 2015;54:1317–1325.
  • Thor M, Olsson CE, Oh JH, et al. Relationships between dose to the gastro-intestinal tract and patient-reported symptom domains after radiotherapy for localized prostate cancer. Acta Oncol. 2015;54:1326–1334.
  • Tuomikoski L, Kapanen M, Collan J, et al. Toward a more patient-specific model of post-radiotherapy saliva secretion for head and neck cancer patients. Acta Oncol. 2015;54:1310–1316.
  • Van den Bergh L, Joniau S, Haustermans K, et al. Reliability of sentinel node procedure for lymph node staging in prostate cancer patients at high risk for lymph node involvement. Acta Oncol. 2015;54:896–902.
  • Winther M, Alsner J, Tramm T, et al. Evaluation of miR-21 and miR-375 as prognostic biomarkers in esophageal cancer. Acta Oncol. 2015;54:1582–1591.
  • Winther-Larsen A, Hoffmann L, Moeller DS, et al. Evaluation of factors associated with loco-regional failure and survival in limited disease small cell lung cancer patients treated with chemoradiotherapy. Acta Oncol. 2015;54:1574–1581.
  • Hansen CR, Johansen J, Kristensen CA, et al. Quality assurance of radiation therapy for head and neck cancer patients treated in DAHANCA 10 randomized trial. Acta Oncol. 2015;54:1669–1673.
  • Hermans BC, Persoon LC, Podesta M, et al. Weekly kilovoltage cone-beam computed tomography for detection of dose discrepancies during (chemo)radiotherapy for head and neck cancer. Acta Oncol. 2015;54:1483–1489.
  • Bøjen A, Vestergaard A, Hoffmann L, et al. A learning programme qualifying radiation therapists to manage daily online adaptive radiotherapy. Acta Oncol. 2015;54:1697–1701.
  • Borras JM, Lievens Y, Grau C. The need for radiotherapy in Europe in 2020: not only data but also a cancer plan. Acta Oncol. 2015;54:1268–1274.
  • Lambin P, Zindler J, Vanneste B, et al. Modern clinical research: how rapid learning health care and cohort multiple randomised clinical trials complement traditional evidence based medicine. Acta Oncol. 2015;54:1289–1300.
  • Lievens Y, Borras JM, Grau C. Cost calculation: a necessary step towards widespread adoption of advanced radiotherapy technology. Acta Oncol. 2015;54:1275–1281.
  • Massaccesi M, Corti M, Azario L, et al. Can automation in radiotherapy reduce costs? Acta Oncol. 2015;54:1282–1288.
  • Molinari AJ, Thorp SI, Portu AM, et al. Assessing advantages of sequential boron neutron capture therapy (BNCT) in an oral cancer model with normalized blood vessels. Acta Oncol. 2015;54:99–106.
  • Pignol JP, Janus C. The evaluation of innovation in radiation oncology – what can we do and what should we do? Acta Oncol. 2015;54:1251–1253.
  • Overgaard J. Radiotherapy. Gazing at the crystal ball of European radiotherapy. Nat Rev Clin Oncol. 2015;12:5–6.
  • Weber DC, Abrunhosa-Branquinho A, Bolsia A, et al. Profile of European proton and carbon ion therapy centers assessed by the EORTC facility questionnaire. Radiother Oncol. 2017;124:185–189.
  • Verma V, Simone CB, Wahl AO, et al. Proton radiotherapy for gynecologic neoplasms. Acta Oncol. 2016;55:1257–1265.
  • Seco J, Spadea MF. Imaging in particle therapy: state of the art and future perspective. Acta Oncol. 2015;54:1254–1258.
  • Malinen E, Søvik Å. Dose or ‘LET’ painting – what is optimal in particle therapy of hypoxic tumors? Acta Oncol. 2015;54:1614–1622.
  • Rieber JG, Kessel KA, Witt O, et al. Treatment tolerance of particle therapy in pediatric patients. Acta Oncol. 2015;54:1049–1055.
  • Baumann M, Krause M, Overgaard J, et al. Radiation oncology in the era of precision medicine. Nat Rev Cancer. 2016;16:234–249.
  • Paganetti H. Relative biological effectiveness (RBE) values for proton beam therapy. Variations as a function of biological endpoint, dose, and linear energy transfer. Phys Med Biol. 2014;59:R419–R472.
  • Mohan R, Peeler CR, Guan F, et al. Radiobiological issues in proton therapy. Acta Oncol. 2017;56:1367–1373.
  • Jones B. Clinical radiobiology of proton therapy: modeling of RBE. Acta Oncol. 2017;56:1374–1378.
  • Paganetti H. Relating the proton relative biological effectiveness to tumor control and normal tissue complication probabilities assuming inter-patient variability in α/β. Acta Oncol. 2017;56:1379–1386.
  • Peeler CR, Mirkovic D, Titt U, et al. Clinical evidence of variable proton biological effectiveness in pediatric patients treated for ependymoma. Radiother Oncol. 2016;121:395–401.
  • Saager M, Glowa C, Peschke P, et al. The relative biological effectiveness of carbon ion irradiations of the rat spinal cord increases linearly with LET up to 99 keV/μm. Acta Oncol. 2016;55:1512–1515.
  • Sørensen BS, Horsman MR, Alsner J, et al. Relative biological effectiveness of carbon ions for tumor control, acute skin damage and late radiation-induced fibrosis in a mouse model. Acta Oncol. 2015;54:1623–1630.
  • Sørensen BS, Bassler N, Nielsen S, et al. Relative biological effectiveness (RBE) and distal edge effects of proton radiation on early damage in vivo. Acta Oncol. 2017;56:1387–1391.
  • Nielsen S, Bassler N, Grzanka L, et al. Differential gene expression in primary fibroblasts induced by proton and cobalt-60 beam irradiation. Acta Oncol. 2017;56:1406–1412.
  • Andreassen CN, Rosenstein BS, Kerns SL, et al. Individual patient data meta-analysis shows a significant association between the ATM rs1801516 SNP and toxicity after radiotherapy in 5456 breast and prostate cancer patients. Radiother Oncol. 2016;121:431–439.
  • De Ruysscher D, Defraene G, Ramaekers BL, et al. Optimal design and patient selection for interventional trials using radiogenomic biomarkers: a REQUITE and Radiogenomics consortium statement. Radiother Oncol. 2016;121:440–446.
  • Alsner J, Andreassen CN, Overgaard J. Genetic markers for prediction of normal tissue toxicity after radiotherapy. Semin Radiat Oncol. 2008;18:126–135.
  • Grau C. The model-based approach to clinical studies in particle radiotherapy – a new concept in evidence based radiation oncology? Radiother Oncol. 2013;107:265–266.
  • Langendijk JA, Lambin P, De Ruysscher D, et al. Selection of patients for radiotherapy with protons aiming at reduction of side effects: the model-based approach. Radiother Oncol. 2013;107:267–273.
  • Blanchard P, Wong AJ, Gunn GB, et al. Toward a model-based patient selection strategy for proton therapy: external validation of photon-derived normal tissue complication probability models in a head and neck proton therapy cohort. Radiother Oncol. 2016;121:381–386.
  • Bijman RG, Breedveld S, Arts T, et al. Impact of model and dose uncertainty on model-based selection of oropharyngeal cancer patients for proton therapy. Acta Oncol. 2017;56:1444–1450.
  • Karsch L, Beyreyther E, Enghardt W, et al. Towards ion beam therapy based on laser plasma accelerators. Acta Oncol. 2017;56:1359–1366.
  • Henry T, Bassler N, Ureba A, et al. Development of an interlaced-crossfiring geometry for proton grid therapy. Acta Oncol. 2017;56:1437–1443.
  • Mako F, Tajima T. Collective ion acceleration by a reflexing electron beam: model and scaling. Phys Fluids. 1984;27:1815–1820.
  • Snavely RA, Key MH, Hatchet SP, et al. Intense high-energy proton beams from Petawatt-laser irradiation of solids. Phys Rev Lett. 2000;85:2945–2948.
  • Schwoerer H, Pfotenhauer S, Jäckel O, et al. Laser-plasma acceleration of quasi-monoenergetic protons from microstructured targets. Nature. 2006;439:445–448.
  • Schippers JM, Lomax AJ. Emerging technologies in proton therapy. Acta Oncol. 2011;50:838–850.
  • Köhler A. Une nouvelle méthode permettant de faire agir, dans la profondeur des tissus, de hautes doses de rayons Roentgen et un moyen nouveau de protection contre les radiodermites. Ann Electrobiol Radiol. 1909;10:661–664.
  • Mohiuddin M, Fujita M, Regine WF, et al. High-dose spatially-fractionated radiation (GRID): a new paradigm in the management of advanced cancers. Int J Radiat Oncol Biol Phys. 1999;45:721–727.
  • Jin JY, Zhao B, Kaminski JM, et al. A MLC-based inversely optimized 3D spatially fractionated grid radiotherapy technique. Radiother Oncol. 2015;117:483–486.
  • Narayanasamy G, Zhang X, Meigooni A, et al. Therapeutic benefits in grid irradiation on tomotherapy for bulky, radiation-resistant tumors. Acta Oncol. 2017;56:1043–1047.
  • Peng V, Suchowerska N, Rogers L, et al. Grid therapy using high definition multileaf collimators: realizing benefits of the bystander effect. Acta Oncol. 2017;56:1048–1059.
  • Bouchet A, Lemasson B, Christen T, et al. Synchrotron microbeam radiation therapy induces hypoxia in intracerebral gliosarcoma but not in the normal brain. Radiother Oncol. 2013;108:143–148.
  • Prezado Y, Fois GR. Proton-minibeam radiation therapy: a proof of concept. Med Phys. 2013;40:031712.
  • Hansen DC, Seco J, Sørensen TS, et al. A simulation study on proton computed tomography (CT) stopping power accuracy using dual energy CT scans as benchmark. Acta Oncol. 2015;54:1638–1642.
  • Hopfgartner J, Stock M, Knäusl B, et al. Robustness of IMPT treatment plans with respect to inter-fractional set-up uncertainties: impact of various beam arrangements for cranial targets. Acta Oncol. 2013;52:570–579.
  • Andersen AG, Casares-Magaz O, Muren LP, et al. A method for evaluation of proton plan robustness towards inter-fractional motion applied to pelvic lymph node irradiation. Acta Oncol. 2015;54:1643–1650.
  • Gorgisyan J, Perrin R, Lomax AJ, et al. Impact of beam angle choice on pencil beam scanning breath-hold proton therapy for lung lesions. Acta Oncol. 2017;56:853–859.
  • Gravgaard Andersen A, Casares-Magaz O, Petersen J, et al. Beam angle evaluation to improve inter-fraction motion robustness in pelvic lymph node irradiation with proton therapy. Acta Oncol. 2017;56:846–852.
  • Zeng C, Plastaras JP, James P, et al. Proton pencil beam scanning for mediastinal lymphoma: treatment planning and robustness assessment. Acta Oncol. 2016;55:1132–1138.
  • Kang M, Huang S, Solberg TD, et al. A study of the beam-specific interplay effect in proton pencil beam scanning delivery in lung cancer. Acta Oncol. 2017;56:531–540.
  • Santiago A, Fritz P, Mühlnickel W, et al. Changes in the radiological depth correlate with dosimetric deterioration in particle therapy for stage I NSCLC patients under high frequency jet ventilation. Acta Oncol. 2015;54:1631–1637.
  • Jensen CA, Roa AMA, Lund J-Å, et al. Intrafractional baseline drift during free breathing breast cancer radiation therapy. Acta Oncol. 2017;56:867–873.
  • Poulsen PR, Worm ES, Hansen R, et al. Respiratory gating based on internal electromagnetic motion monitoring during stereotactic liver radiation therapy: first results. Acta Oncol. 2015;54:1445–1452.
  • Lutz CM, Poulsen PR, Fledelius W, et al. Setup error and motion during deep inspiration breath-hold breast radiotherapy measured with continuous portal imaging. Acta Oncol. 2016;55:193–200.
  • Lens E, van der Horst A, Versteijne E, et al. Considerable pancreatic tumor motion during breath-holding. Acta Oncol. 2016;55:1360–1368.
  • Skyttä T, Kapanen N, Laaksomaa M, et al. Improving the reproducibility of voluntary deep inspiration breath hold technique during adjuvant left-sided breast cancer radiotherapy. Acta Oncol. 2016;55:970–975.
  • Müller J, Neubert C, von Neubeck C, et al. Proton radiography for inline treatment planning verification of small animals. Acta Oncol. 2017;56:1399–1405.
  • Ulrich S, Wieser H-P, Cao W, et al. Impact of respiratory motion on variable relative biological effectiveness in 4D-dose distributions of proton therapy. Acta Oncol. 2017;56:1420–1427.
  • Ödén J, Toma-Dasu I, Eriksson K, et al. The influence of breathing motion and a variable relative biological effectiveness in proton therapy of left-sided breast cancer. Acta Oncol. 2017;56:1428–1436.
  • Abravan A, Knudtsen I, Eide H, et al. A new method to assess pulmonary changes using 18F-fluoro-2-deoxyglucose positron emission tomography for lung cancer patients following radiotherapy. Acta Oncol. 2017;56:1601–1607.
  • Rao SD, Saleh ZH, Setton J, et al. Dose-volume factors correlating with trismus following chemoradiation for head and neck cancer. Acta Oncol. 2016;55:99–104.
  • Abe T, Shirai K, Saitoh J-I, et al. Incidence, risk factors, and dose-volume relationship of radiation-induced rib fracture after carbon ion radiotherapy for lung cancer. Acta Oncol. 2016;55:163–166.
  • Khalil AA, Hoffmann L, Møller DS, et al. New dose constraint reduces radiation-induced fatal pneumonitis in locally advanced non-small cell lung cancer patients treated with intensity-modulated radiotherapy. Acta Oncol. 2015;54:1343–1349.
  • Kirchheiner K, Nout RA, Lindegaard JC, et al. Dose-effect relationship and risk factors for vaginal stenosis after definitive radio(chemo)therapy with image-guided brachytherapy for locally advanced cervical cancer in the EMBRACE study. Radiother Oncol. 2016;118:160–166.
  • Schack LMH, Petersen SE, Nielsen S, et al. Validation of genetic predictors of late radiation-induced morbidity in prostate cancer patients. Acta Oncol. 2017;56:1518–1525.
  • Andreassen CN. Searching for genetic determinants of normal tissue radiosensitivity – are we on the right track? Radiother Oncol. 2010;97:1–8.
  • Barnett GC, Elliott RM, Alsner J, et al. Individual patient data meta-analysis shows no association between the SNP rs1800469 in TGFB and late radiotherapy toxicity. Radiother Oncol. 2012;105:289–295.
  • Oh JH, Kerns S, Ostrer H, et al. Computational methods using genome-wide association studies to predict radiotherapy complications and to identify correlative molecular processes. Sci Rep. 2017;7:43381.
  • Ramaekers BLT, Joore MA, Grutters JPC, et al. The impact of late treatment-toxicity on generic health-related quality of life in head and neck cancer patients after radiotherapy. Oral Oncol. 2011;47:768–774.
  • Bentzen SM, Vaeth M, Pedersen DE, et al. Why actuarial estimates should be used in reporting late normal-tissue effects of cancer treatment … now! Int J Radiat Oncol Biol Phys. 1995;32:1531–1534.
  • Pedersen D, Bentzen SM, Overgaard J. Early and late radiotherapeutic morbidity in 442 consecutive patients with locally advanced carcinoma of the uterine cervix. Int J Radiat Oncol Biol Phys. 1994;29:941–952.
  • Bentzen SM, Balslev I, Pedersen M, et al. Time to loco-regional recurrence after resection of Dukes’ B and C colorectal cancer with or without adjuvant postoperative radiotherapy. A multivariate regression analysis. Br J Cancer. 1992;65:102–107.
  • Bogowicz M, Riesterer O, Stark LS, et al. Comparison of PET and CT radiomics for prediction of local tumor recurrence in head and neck squamous cell carcinoma. Acta Oncol. 2017;56:1535–1540.
  • Leijenaar RTH, Carvalho S, Hoebers FJ, et al. External validation of a prognostic CT-based radiomic signature in oropharyngeal squamous cell carcinoma. Acta Oncol. 2015;54:1423–1429.
  • Huynh E, Coroller TP, Narayan V, et al. CT-based radiomic analysis of stereotactic body radiation therapy patients with lung cancer. Radiother Oncol. 2016;120:258–266.
  • van Timmeren JE, Leijenaar RTH, van Elmpt W, et al. Survival prediction of non-small cell lung cancer patients using radiomics analyses of cone-beam CT images. Radiother Oncol. 2017;123:363–369.
  • van Timmeren JE, Leijenaar RTH, van Elmpt W, et al. Feature selection methodology for longitudinal cone-beam CT radiomics. Acta Oncol. 2017;56:1541–1547.
  • Booth JT, Caillet V, Hardcastle N, et al. The first patient treatment of electromagnetic-guided real time adaptive radiotherapy using MLC tracking for lung SABR. Radiother Oncol. 2016;121:19–25.
  • Brouwer CL, Steenbakkers RJ, van der Schaaf A, et al. Selection of head and neck cancer patients for adaptive radiotherapy to decrease xerostomia. Radiother Oncol. 2016;120:36–40.
  • Brown E, Owen R, Harden F, et al. Predicting the need for adaptive radiotherapy in head and neck cancer. Radiother Oncol. 2015;116:57–63.
  • Fokdal L, Sturdza A, Mazeron R, et al. Image guided adaptive brachytherapy with combined intracavitary and interstitial technique improves the therapeutic ratio in locally advanced cervical cancer: analysis from the retroEMBRACE study. Radiother Oncol. 2016;120:434–440.
  • Hamming-Vrieze O, van Kranen SR, Heemsbergen WD, et al. Analysis of GTV reduction during radiotherapy for oropharyngeal cancer: implications for adaptive radiotherapy. Radiother Oncol. 2017;122:224–228.
  • Kirisits C, Federico M, Nkiwane K, et al. Quality assurance in MR image guided adaptive brachytherapy for cervical cancer: final results of the EMBRACE study dummy run. Radiother Oncol. 2015;117:548–554.
  • Lutkenhaus LJ, de Jong R, Geijsen ED, et al. Potential dosimetric benefit of an adaptive plan selection strategy for short-course radiotherapy in rectal cancer patients. Radiother Oncol. 2016;119:525–530.
  • Mazeron R, Fokdal LU, Kirchheiner K, et al. Dose-volume effect relationships for late rectal morbidity in patients treated with chemoradiation and MRI-guided adaptive brachytherapy for locally advanced cervical cancer: results from the prospective multicenter EMBRACE study. Radiother Oncol. 2016;120:412–419.
  • McPartlin AJ, Li XA, Kershaw LE, et al. MRI-guided prostate adaptive radiotherapy – a systematic review. Radiother Oncol. 2016;119:371–380.
  • Mohamed S, Lindegaard JC, de Leeuw AA, et al. Vaginal dose de-escalation in image guided adaptive brachytherapy for locally advanced cervical cancer. Radiother Oncol. 2016;120:480–485.
  • Møller DS, Holt MI, Alber M, et al. Adaptive radiotherapy for advanced lung cancer ensures target coverage and decreases lung dose. Radiother Oncol. 2016;121:32–38.
  • Tanderup K, Lindegaard JC, Kirisits C, et al. Image Guided Adaptive Brachytherapy in cervix cancer: a new paradigm changing clinical practice and outcome. Radiother Oncol. 2016;120:365–369.
  • Tuomikoski L, Valli A, Tenhunen M, et al. A comparison between two clinically applied plan library strategies in adaptive radiotherapy of bladder cancer. Radiother Oncol. 2015;117:448–452.
  • Vestergaard A, Hafeez S, Muren LP, et al. The potential of MRI-guided online adaptive re-optimisation in radiotherapy of urinary bladder cancer. Radiother Oncol. 2016;118:154–159.
  • Zhang P, Simon A, Rigaud B, et al. Optimal adaptive IMRT strategy to spare the parotid glands in oropharyngeal cancer. Radiother Oncol. 2016;120:41–47.
  • Berkovic P, Paelinck L, Gulyban A, et al. Adaptive radiotherapy for locally advanced non-small cell lung cancer: dosimetric gain and treatment outcome prediction. Acta Oncol. 2017;56:1660–1663.
  • Mahmood F, Johannesen HH, Geertsen P, et al. Ultra-early apparent diffusion coefficient change indicates irradiation and predicts radiotherapy outcome in brain metastases. Acta Oncol. 2017;56:1655–1657.
  • Jensen MB, Ejlertsen B, Mouridsen HT, et al. Improvements in breast cancer survival between 1995 and 2012 in Denmark: the importance of earlier diagnosis and adjuvant treatment. Acta Oncol. 2016;55(Suppl 2):24–35.
  • Lyngholm CD, Laurberg T, Alsner J, et al. Failure pattern and survival after breast conserving therapy. Long-term results of the Danish Breast Cancer Group (DBCG) 89 TM cohort. Acta Oncol. 2016;55:983–992.
  • Bodilsen A, Offersen BV, Christiansen P, et al. Pattern of relapse after breast conserving therapy, a study of 1519 early breast cancer patients treated in the Central Region of Denmark 2000–2009. Acta Oncol. 2016;55:964–969.
  • Poortmans PM, Arenas M, Livi L. Over-irradiation. Breast. 2017;31:295–302.
  • Taylor C, Correa C, Duane FK, et al. Estimating the risks of breast cancer radiotherapy: evidence from modern radiation doses to the lungs and heart and from previous randomized trials. J Clin Oncol. 2017;35:1641–1649.
  • Laurberg T, Alsner J, Tramm T, et al. Impact of age, intrinsic subtype and local treatment on long-term local-regional recurrence and breast cancer mortality among low-risk breast cancer patients. Acta Oncol. 2017;56:59–67.
  • Thorsen LB, Offersen BV, Danø H, et al. DBCG-IMN: a population-based cohort study on the effect of internal mammary node irradiation in early node-positive breast cancer. J Clin Oncol. 2016;34:314–320.
  • Santos AM, Marcu LG, Wong CM, et al. Risk estimation of second primary cancers after breast radiotherapy. Acta Oncol. 2016;55:1331–1337.
  • Moiseenko V, Einck J, Murphy J, et al. Clinical evaluation of QUANTEC guidelines to predict the risk of cardiac mortality in breast cancer patients. Acta Oncol. 2016;55:1506–1510.
  • Højris I, Andersen J, Overgaard M, et al. Late treatment-related morbidity in breast cancer patients randomized to postmastectomy radiotherapy and systemic treatment versus systemic treatment alone. Acta Oncol. 2000;39:355–372.
  • Tramm T, Kyndi M, Myhre S, et al. Relationship between the prognostic and predictive value of the intrinsic subtypes and a validated gene profile predictive of loco-regional control and benefit from post-mastectomy radiotherapy in patients with high-risk breast cancer. Acta Oncol. 2014;53:1337–1346.
  • Lyngholm CD, Overgaard J, Christiansen PM, et al. Validation of a gene expression profile predictive of the risk of radiation-induced fibrosis in women treated with breast conserving therapy. Acta Oncol. 2015;54:1665–1668.
  • Schack LH, Alsner J, Overgaard J, et al. Radiation-induced morbidity evaluated by high-frequency ultrasound. Acta Oncol. 2016;55:1498–1500.
  • Lin LL, Vennarini S, Dimofte A, et al. Proton beam versus photon beam dose to the heart and left anterior descending artery for left-sided breast cancer. Acta Oncol. 2015;54:1032–1039.
  • Knoop A, Andreasen PA, Andersen JA, et al. Prognostic significance of urokinase-type plasminogen activator and plasminogen activator inhibitor-1 in primary breast cancer. Br J Cancer. 1998;77:932–940.
  • Nielsen HM, Friis RB, Linnet S, et al. Loco-regional morbidity after breast conservation and axillary lymph node dissection for early breast cancer with or without regional nodes radiotherapy, perspectives in modern breast cancer treatment: the Skagen Trial 1 is active. Acta Oncol. 2017;56:713–718.
  • Overgaard J. Hypoxic modification of radiotherapy in squamous cell carcinoma of the head and neck – a systematic review and meta-analysis. Radiother Oncol. 2011;100:22–32.
  • Lacas B, Bourhis J, Overgaard J, et al. Role of radiotherapy fractionation in head and neck cancers (MARCH): an updated meta-analysis. Lancet Oncol. 2017;18:1221–1237.
  • Schernberg A, Blanchard P, Chargari C, et al. Neutrophils, a candidate biomarker and target for radiation therapy? Acta Oncol. 2017;56:1526–1534.
  • Rasmussen GB, Håkansson KE, Vogelius IR, et al. Immunohistochemical and molecular imaging biomarker signature for the prediction of failure site after chemoradiation for head and neck squamous cell carcinoma. Acta Oncol. 2017;56:1566–1574.
  • Lassen P. The role of Human papillomavirus in head and neck cancer and the impact on radiotherapy outcome. Radiother Oncol. 2010;95:371–380.
  • Sørensen BS, Busk M, Olthof N, et al. Radiosensitivity and effect of hypoxia in HPV positive head and neck cancer cells. Radiother Oncol. 2013;108:500–505.
  • Bøje CR, Dalton SO, Grønborg TK, et al. The impact of comorbidity on outcome in 12 623 Danish head and neck cancer patients: a population based study from the DAHANCA database. Acta Oncol. 2013;52:285–293.
  • Olsen MH, Bøje CR, Kjaer TK, et al. Socioeconomic position and stage at diagnosis of head and neck cancer – a nationwide study from DAHANCA. Acta Oncol. 2015;54:759–766.
  • Pagh A, Grau C, Overgaard J. Failure pattern and salvage treatment after radical treatment of head and neck cancer. Acta Oncol. 2016;55:625–632.
  • Bentzen J, Toustrup K, Eriksen JG, et al. Locally advanced head and neck cancer treated with accelerated radiotherapy, the hypoxic modifier nimorazole and weekly cisplatin. Results from the DAHANCA 18 phase II study. Acta Oncol. 2015;54:1001–1007.
  • Karlsson T, Bergström L, Ward E, et al. A prospective longitudinal study of voice characteristics and health-related quality of life outcomes following laryngeal cancer treatment with radiotherapy. Acta Oncol. 2016;55:693–699.
  • Møller PK, Tolstrup JS, Olsen MH, et al. Predictors of continuous tobacco smoking in a clinical cohort study of Danish laryngeal cancer patients smoking before treated with radiotherapy. Acta Oncol. 2015;54:685–692.
  • Linge A, Lohaus F, Löck S, et al. HPV status, cancer stem cell marker expression, hypoxia gene signatures and tumour volume identify good prognosis subgroups in patients with HNSCC after primary radiochemotherapy: a multicentre retrospective study of the German Cancer Consortium Radiation Oncology Group (DKTK-ROG). Radiother Oncol. 2016;121:364–373.
  • Toustrup K, Sørensen BS, Metwally MA, et al. Validation of a 15-gene hypoxia classifier in head and neck cancer for prospective use in clinical trials. Acta Oncol. 2016;55:1091–1098.
  • Horsman MR, Mortensen LS, Petersen JB, et al. Imaging hypoxia to improve radiotherapy outcome. Nat Rev Clin Oncol. 2012;9:674–687.
  • Mönnich D, Thorwarth D, Leibfarth S, et al. Overlap of highly FDG-avid and FMISO hypoxic tumour subvolumes in patients with head and neck cancer. Acta Oncol. 2017;56:1581–1586.
  • Zukauskaite R, Hansen CR, Brink C, et al. Analysis of CT-verified loco-regional recurrences after definitive IMRT for HNSCC using site of origin estimation methods. Acta Oncol. 2017;56:1558–1565.
  • Differding S, Sterpin E, Janssens G, et al. Methodology for adaptive and robust FDG-PET escalated dose painting by numbers in head and neck tumors. Acta Oncol. 2016;55:217–225.
  • Bittner MI, Wiedenmann N, Bucher S, et al. Analysis of relation between hypoxia PET imaging and tissue-based biomarkers during head and neck radiochemotherapy. Acta Oncol. 2016;55:1299–1304.
  • Wittenborn TR, Horsman MR. Targeting tumour hypoxia to improve outcome of stereotactic radiotherapy. Acta Oncol. 2015;54:1385–1392.
  • Tran LB, Bol A, Labar D, et al. Predictive value of (18)F-FAZA PET imaging for guiding the association of radiotherapy with nimorazole: a preclinical study. Radiother Oncol. 2015;114:189–194.
  • Hassan Metwally MA, Ali R, Kuddu M, et al. IAEA-HypoX. A randomized multicenter study of the hypoxic radiosensitizer nimorazole concomitant with accelerated radiotherapy in head and neck squamous cell carcinoma. Radiother Oncol. 2015;116:15–20.
  • Hassan Metwally MA, Ali R, Kuddu M, et al. Radiotherapy quality assurance of the IAEA-HypoX trial of the accelerated radiotherapy in the treatment of head and neck squamous cell carcinoma with or without the hypoxic radiosensitizer nimorazole. Acta Oncol. 2015;54:1673–1677.
  • Overgaard J. Hypoxic radiosensitization: adored and ignored. J Clin Oncol. 2007;25:4066–4074.
  • Grills IS, Hope AJ, Guckenberger M, et al. A collaborative analysis of stereotactic lung radiotherapy outcomes for early-stage non-small-cell lung cancer using daily online cone-beam computed tomography image-guided radiotherapy. J Thorac Oncol. 2012;7:1382–1393.
  • Baumann P, Nyman J, Høyer M, et al. Outcome in a prospective phase II trial of medically inoperable stage I non-small-cell lung cancer patients treated with stereotactic body radiotherapy. J Clin Oncol. 2009;27:3290–3296.
  • Jeppesen SS, Schytte T, Jensen HR, et al. Stereotactic body radiation therapy versus conventional radiation therapy in patients with early stage non-small cell lung cancer: an updated retrospective study on local failure and survival rates. Acta Oncol. 2013;52:1552–1558.
  • Lindberg K, Nyman J, Riesenfeld Kallskog V, et al. Long-term results of a prospective phase II trial of medically inoperable stage I NSCLC treated with SBRT – the Nordic experience. Acta Oncol. 2015;54:1096–1104.
  • Paludan M, Traberg Hansen A, Petersen J, et al. Aggravation of dyspnea in stage I non-small cell lung cancer patients following stereotactic body radiotherapy: is there a dose-volume dependency? Acta Oncol. 2006;45:818–822.
  • Stam B, Peulen H, Guckenberger M, et al. Dose to heart substructures is associated with non-cancer death after SBRT in stage I-II NSCLC patients. Radiother Oncol. 2017;123:370–375.
  • Guckenberger M, Andratschke N, Dieckmann K, et al. ESTRO ACROP consensus guideline on implementation and practice of stereotactic body radiotherapy for peripherally located early stage non-small cell lung cancer. Radiother Oncol. 2017;124:11–17.
  • Tekatli H, Haasbeek N, Dahele M, et al. Outcomes of hypofractionated high-dose radiotherapy in poor-risk patients with ‘ultracentral’ non-small cell lung cancer. J Thorac Oncol. 2016;11:1081–1089.
  • Dahele M, van Sörnsen de Koste JR, Verbakel WF, et al. An analysis of planned versus delivered airway doses during stereotactic lung radiotherapy for central tumors. Acta Oncol. 2016;55:934–937.
  • Lindblom E, Dasu A, Toma-Dasu I. Optimal fractionation in radiotherapy for non-small cell lung cancer – a modelling approach. Acta Oncol. 2015;54:1592–1598.
  • Moghanaki D, Karas T. Surgery versus SABR for NSCLC. Lancet Oncol. 2013;14:e490–e491.
  • Nyman J, Hallqvist A, Lund JÅ, et al. SPACE – A randomized study of SBRT vs conventional fractionated radiotherapy in medically inoperable stage I NSCLC. Radiother Oncol. 2016;121:1–8.
  • Jeppesen SS, Hansen NG, Schytte T, et al. Comparison of survival of chronic obstructive pulmonary disease patients with or without a localized non-small cell lung cancer. Lung Cancer. 2016;100:90–95.
  • Aupérin A, Le Péchoux C, Rolland E, et al. Meta-analysis of concomitant versus sequential radiochemotherapy in locally advanced non-small-cell lung cancer. J Clin Oncol. 2010;28:2181–2190.
  • Bradley JD, Paulus R, Komaki R, 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.
  • Halvorsen TO, Sundstrøm S, Flotten O, et al. Comorbidity and outcomes of concurrent chemo- and radiotherapy in limited disease small cell lung cancer. Acta Oncol. 2016;55:1349–1354.
  • Hoffmans D, Dahele M, Senan S, et al. Can the probability of radiation esophagitis be reduced without compromising lung tumor control: a radiobiological modeling study. Acta Oncol. 2016;55:926–930.
  • Tvilum M, Khalil AA, Møller DS, et al. Clinical outcome of image-guided adaptive radiotherapy in the treatment of lung cancer patients. Acta Oncol. 2015;54:1430–1437.
  • Farr KP, Møller DS, Khalil AA, et al. Loss of lung function after chemo-radiotherapy for NSCLC measured by perfusion SPECT/CT: correlation with radiation dose and clinical morbidity. Acta Oncol. 2015;54:1350–1354.
  • Hansen O, Schytte T, Nielsen M, et al. Age dependent prognosis in concurrent chemo-radiation of locally advanced NSCLC. Acta Oncol. 2015;54:333–339.
  • Hansen O, Knap MM, Khalil A, et al. A randomized phase II trial of concurrent chemoradiation with two doses of radiotherapy, 60Gy and 66Gy, concomitant with a fixed dose of oral vinorelbine in locally advanced NSCLC. Radiother Oncol. 2017;123:276–281.
  • Møller DS, Nielsen TB, Brink C, et al. Heterogeneous FDG-guided dose-escalation for locally advanced NSCLC (the NARLAL2 trial): design and early dosimetric results of a randomized, multi-centre phase-III study. Radiother Oncol. 2017;124:311–317.
  • Valentini V, Glimelius B, Haustermans K, et al. EURECCA consensus conference highlights about rectal cancer clinical management: the radiation oncologist’s expert review. Radiother Oncol. 2014;110:195–198.
  • Swedish Rectal Cancer Trial, Cedermark B, Dahlberg M, et al. Improved survival with preoperative radiotherapy in resectable rectal cancer. N Engl J Med. 1997;336:980–987.
  • Peeters ST, Dooms C, Van Baardwijk A, et al. Selective mediastinal node irradiation in non-small cell lung cancer in the IMRT/VMAT era: how to use E(B)US-NA information in addition to PET-CT for delineation? Radiother Oncol. 2016;120:273–278.
  • Sauer R, Liersch T, Merkel S, et al. Preoperative versus postoperative chemoradiotherapy for locally advanced rectal cancer: results of the German CAO/ARO/AIO-94 randomized phase III trial after a median follow-up of 11 years. J Clin Oncol. 2012;30:1926–1933.
  • Valentini V, Lambin P, Myerson RJ. Is it time for tailored treatment of rectal cancer? From prescribing by consensus to prescribing by numbers. Radiother Oncol. 2012;102:1–3.
  • Sebag-Montefiore D, Stephens RJ, Steele R, et al. Preoperative radiotherapy versus selective postoperative chemoradiotherapy in patients with rectal cancer (MRC CR07 and NCIC-CTG C016): a multicentre, randomised trial. Lancet. 2009;373:811–820.
  • Valentini V, van Stiphout RG, Lammering G, et al. Selection of appropriate end-points (pCR vs 2yDFS) for tailoring treatments with prediction models in locally advanced rectal cancer. Radiother Oncol. 2015;114:302–309.
  • Valentini V, Barba MC, Gambacorta MA. The role of multimodality treatment in M0 rectal cancer: evidence and research. Eur Rev Med Pharmacol Sci. 2010;14:334–341.
  • Hall MD, Schultheiss TE, Smith DD, et al. Effect of increasing radiation dose on pathologic complete response in rectal cancer patients treated with neoadjuvant chemoradiation therapy. Acta Oncol. 2016;55:1392–1399.
  • Erlandsson J, Holm T, Pettersson D, et al. Optimal fractionation of preoperative radiotherapy and timing to surgery for rectal cancer (stockholm III): a multicentre, randomised, non-blinded, phase 3, non-inferiority trial. Lancet Oncol. 2017;18:336–346.
  • Glimelius B. What is most relevant in preoperative rectal cancer chemoradiotherapy – the chemotherapy, the radiation dose or the timing to surgery? Acta Oncol. 2016;55:1381–1385.
  • Bujko K, Pietrzak L, Partycki M, et al. The feasibility of short-course radiotherapy in a watch-and-wait policy for rectal cancer. Acta Oncol. 2017;56:1152–1154.
  • Lundby L, Jensen VJ, Overgaard J, et al. Long-term colorectal function after postoperative radiotherapy for colorectal cancer. Lancet. 1997;350:564.
  • Glimelius B, Myklebust TA, Lundqvist K, et al. Two countries – two treatment strategies for rectal cancer. Radiother Oncol. 2016;121:357–363.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.