Back to Journals » Clinical Interventions in Aging » Volume 18

Geriatric Radiation Oncology: What We Know and What Can We Do Better?

Authors Roeder F, Jensen AD , Lindel K , Mattke M, Wolf F, Gerum S

Received 21 November 2022

Accepted for publication 22 April 2023

Published 4 May 2023 Volume 2023:18 Pages 689—711

DOI https://doi.org/10.2147/CIA.S365495

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 2

Editor who approved publication: Prof. Dr. Nandu Goswami



Falk Roeder,1 Alexandra D Jensen,2 Katja Lindel,3 Matthias Mattke,1 Frank Wolf,1 Sabine Gerum1

1Department of Radiation Therapy and Radiation Oncology, Paracelsus Medical University Hospital, Salzburg, Austria; 2Department of Radiation Oncology, University Hospital Marburg-Giessen, Giessen, Germany; 3Department of Radiation Oncology, Städtisches Klinikum, Karlsruhe, Germany

Correspondence: Falk Roeder, Department of Radiation Therapy and Radiation Oncology, Paracelsus Medical University Hospital, Müllner Hauptstrasse 48, Salzburg, 5020, Austria, Tel +43 57255 55569, Email [email protected]

Abstract: Elderly patients represent a growing subgroup of cancer patients for whom the role of radiation therapy is poorly defined. Older patients are still clearly underrepresented in clinical trials, resulting in very limited high-level evidence. Moreover, elderly patients are less likely to receive radiation therapy in similar clinical scenarios compared to younger patients. However, there is no clear evidence for a generally reduced radiation tolerance with increasing age. Modern radiation techniques have clearly reduced acute and late side effects, thus extending the boundaries of the possible regarding treatment intensity in elderly or frail patients. Hypofractionated regimens have further decreased the socioeconomic burden of radiation treatments by reducing the overall treatment time. The current review aims at summarizing the existing data for the use of radiation therapy or chemoradiation in elderly patients focusing on the main cancer types. It provides an overview of treatment tolerability and outcomes with current standard radiation therapy regimens, including possible predictive factors in the elderly population. Strategies for patient selection for standard or tailored radiation therapy approaches based on age, performance score or comorbidity, including the use of prediction tests or geriatric assessments, are discussed. Current and future possibilities for improvements of routine care and creation of high-level evidence in elderly patients receiving radiation therapy are highlighted.

Keywords: radiation therapy, elderly, geriatric assessment

Introduction

Due to demographic changes, radiation oncologists are facing an increasing number of elderly patients in their daily practice at least in developed countries.1 There is no clear preclinical, radiobiological or clinical evidence for a generally reduced radiation tolerance with increasing age.2 Nevertheless, an emerging body of evidence shows that older patients are still less likely to receive radiation therapy (RT) in similar clinical scenarios compared to younger patients, especially with regard to adjuvant treatment settings.3

A variety of factors may play a role in the clinical decision-making process: Elderly patients are clearly underrepresented or even excluded from most randomized trials creating the evidence for certain treatment decisions.4 Thus, for many physicians, it seems at least questionable if results commonly used for patient guidance can be simply transferred to the elderly population. Elderly patients are also more likely to have either distinct serious comorbidities or at least an accumulation of several minor limitations of organ functions. This may lead to a (anticipated) decreased treatment tolerance.5 Moreover, socioeconomic factors such as limited mobility or inadequate resources for general care and/or management of common side effects at home may further impair their (anticipated) ability for outpatient RT treatments.3 This seems especially true if long travel distances to the next RT facility are present. Different views on treatment aims focusing on the preservation on quality of life (Qol) and personal independence rather than pure survival may further prompt elderly patients more likely to refuse (anticipated) intense treatment approaches or long-lasting inpatient therapies.6

From a scientific point of view, addressing the question of an optimal RT approach in elderly patients is even more difficult. First of all, there is no generally agreed definition of the term “elderly”, which seems to be even disease- and/or treatment specific. While most physicians would agree to designate a 70-year-old person with glioblastoma as “elderly”, this would probably not been the case for prostate cancer. Moreover, there is no clear demarcation to patients with comorbidities and/or limited performance status. Many studies reporting outcomes of “elderly” patients include also “frail” patients without reporting subgroup analyses.3 Severe comorbidities of organs within the radiation volumes obviously can limit RT tolerance, but this does not represent a specific difference compared to younger patients. While limitations in organ functions outside the treated area usually do not compromise the ability to tolerate RT, this might be true for combination approaches.

Radiation techniques as well as supportive care have constantly evolved over time, resulting in less severe toxicities and/or improved capabilities to reduce side effects.7 The widespread adoption of intensity-modulated or stereotactic RT constantly reduced doses to adjacent organs at risk. Combined with the use of daily image-guidance, those advances resulted in improved tolerability of RT. Therefore, reports with outdated radiation techniques often describing increased toxicity in elderly patients should not be used anymore to draw conclusions on treatment tolerability.3 In contrast, non-randomized analyses reporting equal toxicity inherit always some risk of selection bias.3 Randomized comparisons specifically addressing elderly patients are desirable, however they likely face difficulties regarding the definitions of inclusion criteria: Because elderly patients show a greater variety in performance status and comorbidities, wide inclusion criteria would result in inhomogeneous cohorts. In contrast, strict inclusion criteria and/or stratification rules would compromise recruitment.

However, evolvement of radiation techniques offers also a chance to improve the adherence to RT in elderly and/or frail patients. Hypofractionated regimes using less fractions with higher single doses resulting in shorter overall treatment time have been widely adopted. This will likely reduce the impact of socioeconomic factors on treatment decisions on both sides (physicians and patients).8 Nevertheless, more research is warranted, specifically addressing the value and tolerability of RT and/or combination approaches in elderly patients. This should include investigations about the optimal study design for the evaluation of more personalized treatment approaches.

While clear evidence and consequently specific recommendations or guidelines are still rare, our review aims at summarizing the current evidence for RT in elderly patients to support radiation oncologists and other treating physicians in their decision-making process. We hereby focused on the most relevant disease types presented in daily practice of radiation oncology.

Central Nervous System Tumors

Approximately 310.000 new cases of central nervous system (CNS) tumours are diagnosed globally and 67.114 new cases in Europe each year corresponding to a crude incidence rate of 4.0/100.000 and 9.0/100.000.9 Almost half of them are diagnosed in patients aged 65 and older.9 Around 50–70% of those tumours are diagnosed as glioblastoma10 with incidence rates peaking at ages 60–80 years.11 Despite this fact, elderly patients are an underrepresented age group in clinical trials.

Outcome in patients diagnosed with glioblastoma is bleak: 5-year overall survival (OS) still ranges between 3% (patients >65 years) to 27% (patients between 20 and 39 years).10 Combined chemoradiation with temozolomide following resection has been established as treatment standard in 2005.12 Stupp et al12 could show impressive OS improvement by combining simultaneous radiochemotherapy with temozolomide plus adjuvant temozolomide. They observed an increase median survival from 12.1 months (radiotherapy only) to 14.6 months (chemoradiation).12 In their subgroup analysis of patients ≥60 years, chemoradiation still improved OS (11.8 months vs 10.9 months), but the observed effect was not as pronounced as in younger patients. The benefit also seemed to decrease with increasing RPA class corresponding to decreasing overall performance status.13 Patients with MGMT promotor methylation were shown to benefit from combined treatment significantly more than patients without methylation.14 However, age itself was not a significant prognostic factor in this post-hoc analysis.14

Treatment outcomes can be further improved by tumour-treating fields. Their addition after completion of a regular Stupp regimen resulted in improved OS without detrimental impact on Qol.15,16 Intensification of chemotherapy by the addition of CCNU to temozolomide in patients with MGMT promotor methylation in the CeTeG trial also increased median OS (48.1 vs 31.8 months) in patients ≤65 years.17 However, fewer elderly patients seem to exhibit favourable prognostic markers such as MGMT promotor methylation.18 Iwamoto et al analyzed patterns of care in 4137 glioblastoma patients aged >65 years and could show that age and comorbidities significantly influenced treatment choice (resection, chemotherapy, radiotherapy) in this cohort.19 Defining adequate treatment paths in elderly patients is therefore an important issue.

The considerable time burden of a standard course of radiotherapy may play an important role specifically in the elderly. Nevertheless, the French group showed that patients aged 70-85 years benefited significantly from 50.4Gy in 28 fractions vs best supportive care (BSC) in terms of OS and symptom control.20 In order to shorten overall treatment time, Roa et al21 established hypofractionated radiotherapy with 15×2.67Gy in elderly patients. Their regimen was equally effective compared to standard radiotherapy with 30x2Gy in a cohort with a median age of 71–72 years. No difference in either OS or performance score was noted. However, median survival was only 5.1 months (60Gy) and 5.6 months (40.05Gy).21

The EORTC-trial therefore investigated the use of combining hypofractionated radiotherapy with temozolomide in a trial randomizing 562 patients with a median age of 73 years. Hypofractionated chemoradiation improved OS from 7.6 months (RT alone) to 9.3 months (chemoradiation). Patients with MGMT-promotor methylation again benefited more from the addition of chemotherapy (13.5 months vs 7.7 months) than patients with unmethylated MGMT-promotor (10.0 months vs 7.9 months). Qol was not affected by treatment arm.22

With the aim to further reduce in-hospital treatment time, Wick et al23 investigated temozolomide only as an alternative to standard radiotherapy in patients with a median age of 71/72 years within the NOA-08 trial. Overall and event-free survival were higher in the radiotherapy group. In their subgroup analysis, MGMT-promotor methylated patients receiving temozolomide showed superior overall and event-free survival than MGMTpromotor methylated patients receiving radiotherapy. Hence, temozolomide only may be an option for this subset of patients.23

The NORDIC-trial was designed to clarify this question further: Malmstrom et al24 randomized patients aged 60–83 years to standard radiotherapy (60Gy), 10×3.4Gy or temozolomide. Protocol completion in the 60Gy and temozolomide groups were only 72% and 34% vs 95% in the hypofractionated group. OS in the temozolomide group was 8.3 months vs 7.5 months (hypofractionated) vs 6.0 months (standard RT).

As described above, patient performance score, comorbidities and age often influence treatment choice in clinical routine. Mirimanoff et al25 could show the influence of those aspects through analysis of the Stupp trial according to RPA classes: patients with lower RPA classes showed significantly improved outcomes. Gerstein et al26 and Combs et al27 retrospectively showed significant differences in outcome based on overall performance state or RPA class in elderly glioblastoma patients. Unfortunately, comorbidities are not reported in either of those trials specifically including elderly patients. Median age as well as minimum age also varies substantially between the trials. While two trials report Karnofsky performance and mini-mental state exam scores,22,23 Malmstrom et al24 does not include this information. Neither of those trials include any form of specific geriatric or frailty assessment, hence interpretation of those trials has some limits regarding those aspects.

As opposed to many other malignancies though, treatment of glioblastoma in elderly patients can be guided by prospective evidence albeit more detailed information is still somewhat scarce.

Elderly patients well enough to undergo treatment should at least receive hypofractionated radiotherapy. Elderly patients with MGMT-promotor methylation and good performance status may receive either combined chemoradiation or temozolomide alone, whereas patients with poor performance status may receive hypofractionated radiotherapy, temozolomide or BSC.28

Head and Neck Cancer

Squamous cell carcinoma represents the most common malignancy in the head and neck (HNSCC). Approximately 745.000 new cases were diagnosed in 2020 worldwide corresponding to a crude incidence rate of 9.6/100.000.29

Combined chemoradiation has been well established as a potentially curative treatment approach resulting in improved organ preservation, locoregional control and OS of around 60-70% in locally advanced HNSCC.30–37 A recent SEER database analysis could demonstrate that modern regimens have improved survival for HNSCC patients over the past decades for all age groups except for patients aged ≥75 years.38

Current cancer statistics in the UK show peak incidence rates of HNSCC in males at 70–74 years and in females at 85–89 years. Around 33% (male) to 36% (female) of all new HNSCC patients are ≥70 years,39 so a significant number of patients appear not to share the benefit from recent advances. Elderly patients (≥70–75 years) are often excluded from participation in clinical trials which represents a relevant knowledge gap in the treatment of patients in this age group.

We will therefore address the following issues in the treatment of elderly head and neck patients based on available – albeit mostly retrospective – data:

  1. Do elderly HNSCC patients routinely receive standard treatment?
  2. Is standard treatment feasible for elderly patients?
  3. Which factors adversely affect prognosis in elderly patients?
  4. Are there any objective criteria to guide treatment selection in elderly patients?

Do Elderly HNSCC Patients Routinely Receive Standard Treatment and is It Feasible for Elderly Patients?

A recent pattern-of-care survey in Germany, Austria and Switzerland reported that the majority of respondents apparently treat elderly patients within standard regimens.40 Several groups also report outcomes of elderly patients undergoing chemoradiation. Maggiore et al41 share their experience in 89 patients ≥70 years treated with a 5-FU/hydroxyurea-based regimen between 1997 and 2009. At a median follow-up of approx. 40 months, OS was 32%. Most patients (86.5%) completed their treatment; toxicity, however, was considerable: grade 3–4 neutropenia occurred in 34%, 44% of patients experienced aspiration, 62% required feeding tubes. The treatment was still deemed feasible in selected patients.41 Treatment toxicity in 246 elderly patients (65 years) mainly treated by chemoradiation was found to be high also in the Freiburg analyses: incidence of at least one grade 3/4 toxicity was 56%. While treatment adherence was high (87%), OS only reached 57% at 2 years.42 Brown et al43 reported the outcome of patients ≥80 years treated with intensity-modulated techniques from 2003 to 2015. Only 7% of those patients received chemoradiation. Twenty-six percent required in-patient treatment during radiotherapy. However, overall grade 3/4 late toxicity at a median follow-up of 25 months was only 2%. The authors concluded that treatment tolerance was high even in this age group.43 Considering low overall patient numbers and also low rates of patients treated with concomitant chemoradiation, it is doubtful whether elderly patients routinely receive standard treatment. As reported, standard treatment may be feasible for some patients, though selection is still largely subjective. Potentially, an evaluation of larger databases and registries regarding treatment specifics – which to our knowledge is currently lacking – may help to answer these questions.

Which Factors Adversely Affect Prognosis and are There Any Objective Criteria to Guide Treatment Selection in Elderly Patients?

As mentioned, chemoradiation is associated with considerable treatment-related toxicity. In cumulative analysis of RTOG studies, approximately 43% of patients developed late toxicities grade 3+.44 Patient age was one of the most important predictors for development of severe side effects.44 Age was also found to predict for higher rates of treatment modifications, interruptions or discontinuation in two retrospective cohorts of 272 and 40 elderly patients receiving chemoradiation.45,46 Moreover, age adversely affected outcomes in elderly patients receiving chemoradiation according to a SEER analysis.47 Comorbidity has also been shown to adversely impact survival48,49 and toxicity,50,51 while prevalence of comorbidity in HNSCC patients increases with age.51,52 Paleri et al53 analyzed 180 patients with larynx carcinomas: 64% of patients had comorbidities, 26% more than one comorbid condition. OS in patients without comorbidities was significantly higher than in patients with one or more comorbidities. Patients with independent neurological disorders as part of their comorbidities showed the highest mortality rate of 70% in this analysis.53 No reliable statement can be made regarding the frequency of treatment modifications in elderly patients (see above). However, Derks et al53 could show in their analyses that both age and comorbidities independently influenced treatment choice in HNSCC patients. Frailty is a relatively new term summarizing age-related physiological decline and functioning leading to increased vulnerability. De Vries and collaborators54 analyzed 160 patients undergoing radiotherapy for head and neck cancer from 2014 to 2016. Type of treatment and neither frailty or geriatric assessment were predictive of treatment toxicity in their cohort.54 In contrast, Morse et al45 found that patients showing sarcopenia as a surrogate for frailty were more prone to developing treatment toxicities during chemoradiation. This finding is supported by Chou et al55 and Karavolia et al,56 who showed associations between increased vulnerability and OS and toxicity. The most concordant parameter to predict for adverse outcome in the elderly seems to be overall performance state. Despite several attempts to identify other predictors, performance state was found to be an independent predictor for OS in several analyses.52,57–59 However, Nicolay et al59 recently proposed a novel prognostic score. Based on their initial cohort of 284 patients, they found performance state, comorbidity index, CRP level and tumour and nodal stage as independent prognostic factors. Their nomogram based on CRP, Charlson comorbidity index and Karnofsky performance state could predict for outcomes in their validation cohort (217 elderly patients) reasonably well and may be used to select treatment in the elderly population.59 Further prospective data are, however, pending.

In summary, data regarding standard treatment in elderly HNSCC patients are scarce. In addition, consensus regarding absolute age-groups termed “elderly” seems to be lacking. Retrospective analyses frequently report on comparatively low patient numbers in view of the considerable incidence of HNSCC in the elderly, hence one may safely assume that treatment is frequently modified and adjusted. Several factors, including age, comorbidities, frailty and performance score impact treatment as well as treatment outcome in HNSCC. Prospective data to guide objective scores and hence treatment recommendations are still lacking.

Lung Cancer

RT is an important component of lung cancer therapy alone or in combination with systemic treatments. With a median age of about 70 years at diagnosis, lung cancer is clearly a disease of the elderly, often associated with comorbidities and limited performance status. Many patients cannot receive standard treatment and were excluded from randomized trials, leading to limited evidence in the treatment of this patient group.60–62 Therefore, the decision-making process should aim at maximizing benefits and minimizing harms depending on patients' individual values and preferences.63 Lung cancer can be generally divided into small-cell (SCLC) and non-small-cell lung cancer (NSCLC). In SCLC, systemic therapy is the dominant option for the vast majority of patients, while the role of RT is restricted to early stages or palliative treatments. Within these situations, the used RT regimens (and their general limitations) for SCLC are similar to those for locally advanced or metastatic NSCLC. Therefore, it will not be specifically addressed in the following:

NSCLC Stage I/II

Standard of care in patients with Stage I/II is resection, alternatively stereotactic ablative body radiotherapy (SABR) is well established. A pooled analysis of the prospective randomized STARS and ROSEL trials showed even improved survival with SABR compared to surgery for resectable patients, although not focusing on elderly patients.64 In 2021, the updated propensity score matched results of the STARS trial confirmed that SABR is a comparable curative treatment with minimal toxicity, noninvasive character and possibility for an outpatient setting with a short treatment period (generally 1 week), which is often important for frail or elderly patients.65 Brooks et al66 retrospectively analysed 772 patients in Stage I/II NSCLC (n = 442 < 75 years, n = 330 >75 years) treated with SABR and found no difference in major endpoints (time to progression, lung-cancer-specific survival, 2yr-OS and toxicity) according to age. OS in the group >75 years deteriorated after 5 years, due to natural shorter life expectancy. Toxicity did also not differ, no grade 4/5 toxicity was observed in the elderly group.66 A large retrospective database confirmed these results.67 In conclusion, SABR should be offered preferably to elderly and frail patients.

NSCLC Stage IIIA/B (Resectable)

In potentially resectable Stage IIIA, multimodal treatment with neoadjuvant chemotherapy/chemoradiation followed by surgery is recommended. However, this seems feasible only in highly selected elderly patients without relevant comorbidities and high performance score.61 Although beneficial in other endpoints, prospective data showed no difference in OS comparing chemoradiation alone vs chemoradiation followed by surgery68,69 in well selected Stage IIIA/B tumors. The number of patients ≥70 years was not reported in the study of Eberhardt et al68 and very small (16%) in the study of Albain et al.69 No relevant prospective trials are available comparing trimodal therapy vs definitive chemoradiation in the elderly group. Both treatments are generally valid therapeutic options, while chemoradiation alone is less invasive and toxic and therefore likely the preferable option in frail or elderly patients. Furthermore, no prospective data are available addressing the role of surgery specifically in the elderly. Retrospective reports also included only very small numbers of patients ≥75 years (17%) evaluated for surgery70 due to the strict pulmonary/cardiac functional requirements for resections.61,71

One small prospective trial specifically assessed the role of RT in patients ≥75 years and reported a median survival of 19 months with very limited toxicity.72 Pignon et al found no significant differences regarding OS and toxicity after curative intent RT according to age.73 Although comorbidities and poor functional status may influence the tolerability of radiotherapy, modern radiation techniques have clearly increased the therapeutic index. Especially if conventional chemoradiation seems not tolerable, hypofractionated RT should be evaluated.74

NSCLC Stage IIIB/C (Unresectable)

Concurrent chemoradiation was found to be superior to RT alone already during the 1990s,75 but came along with increased toxicity. Consequently, subsequent trials included only small numbers of elderly patients. First, Atagi et al investigated the role of chemoradiation in patients ≥70 years.76–78 OS benefit for chemoradiation was confirmed, although the applied regimen (carboplatin only) was less intensive than usually used doublets. Similar results were shown in a meta-analysis for combined treatments despite increased toxicity.79 Esophageal and pulmonary toxicities are usually the clinically most relevant side effects with regard to treatment compliance and Qol and are clearly increased within combination approaches.80,81 Therefore, sequential chemoradiation or (hypofractionated) RT are still reasonable treatment options in elderly patients deemed unfit for concurrent chemoradiation, showing limited toxicity and promising results.74,81,82 A clinical trial comparing concurrent and sequential chemoradiation in patients ≥75 years regarding quality-adjusted survival is ongoing. Patients are stratified into frail, vulnerable and fit groups based on geriatric assessment.83

Immunotherapy has gained attraction also in non-metastatic NSCLC patients since the PACIFIC trial showed an OS benefit for adding consolidation therapy with Durvalumab after chemoradiation compared to chemoradiation alone.84,85 Immunotherapy generally inherits a different safety profile, which is not affected by age86,87 at least if used as a sole treatment. Therefore, checkpoint inhibitors seem worth to be evaluated either as a substitute for concomitant chemotherapy during chemoradiation or as an adjunct after less intensive chemoradiation specifically in elderly patients. The first approach is currently evaluated in the randomized Phase II TRADE-hypo trial (NCT04351256). It includes patients >70 years, ECOG 1/2 with locally advanced, unresectable NSCLC, who are unfit for chemotherapy because of age and/or frailty and compares concomitant durvalumab with either conventionally (60Gy in 30 fractions) or hypofractionated (55Gy in 20 fractions) RT.62 Moreover, a Japanese prospective trial evaluating conventionally fractionated RT with low-dose carboplatin and Durvalumab followed by Durvalumab consolidation in frail/elderly stage III NSCLC patients is ongoing.88 Efficacy and toxicity results, especially regarding pneumonitis rates with simultaneous application, are awaited.

NSCLC Stage IV (Metastatic)

If RT is used for palliation or prevention of symptoms (pain, bleeding, obstruction), hypofractionated regimens are clearly preferable. In case of oligometastatic disease, the individual situation has to be evaluated. Palma et al showed89 in his prospective trial that SABR in oligometastatic patients may improve OS with minimal toxicity compared to chemotherapy alone. Furthermore, SABR may help to avoid or delay the need for systemic therapies, which can improve Qol especially in frail/elderly patients. Adding RT to Pembrolizumab is also a promising option with improved outcome in metastatic lung cancer.90

Breast Cancer

Breast cancer (BC) remains the most common cancer diagnosis in women. Of those diagnosed with BC, about 30% are ≥70 years.91 In early stage BC, breast conserving surgery (BCS) is the first and one of the main pillars in oncological treatment. Even though RT is seen as a standard treatment after BCS,92 the treatment of elderly BC patients differs from the treatment of younger ones due to geriatric frailty or comorbidities – especially in low-risk constellations. Nevertheless, the range of general conditions in elderly patients is wide – from highly morbid patients in their early 70s to athletic patients in their late 80s – leaving the establishment of a standardized procedure hardly achievable. Especially in radiation oncology, where the side effects of a breast treatment decreased continually over the last decades, critical selection of patients is crucial, yet not always easy.

If patients are not eligible for postoperative RT, mastectomy was historically considered as the alternative surgical treatment.93 This dogma, that all patients who cannot receive postoperative radiotherapy have to undergo mastectomy, falters in low-risk constellations (pT1, pN0, ER/PR+, Her2-). Several analyses showed that in those situations, RT can most likely be dispensed – even after BCS. Hughes et al reported on the results of the CALGB 9343 trial, which showed an advantage of tamoxifen and postoperative RT compared to tamoxifen alone after 10 years (local control 98% vs 90%, sig.). Nevertheless, this did not translate in a significant difference in time to mastectomy, time to distant metastasis, cancer-specific survival or OS.94 A more recent analysis by Stueber et al came to similar conclusions with a chance of relapse within 5 years of <3% in patients aged 70 years and older with low-risk features.95 Furthermore, the authors recommend to spare elderly low-risk patients from mastectomy with the intention of avoiding postoperative RT. In elderly high-risk patients, RT following BCS is recommended. The recent NCCN-guidelines also feature the recommendation to consider omitting breast irradiation in patients aged 70 years and older with ER-positive, cN0, pT1 tumors who receive adjuvant endocrine therapy.96 The same is true for the national German guideline.92

If the omission of radiotherapy is no option, several hypofractionated options are eligible. The most elegant option is an intraoperative single-dose radiotherapy in which the “adjuvant” treatment is already completed intraoperatively. A major disadvantage may be that the full pathological report is not available at the time of the radiotherapy and that this option is not available in all centers. Nevertheless, the randomized TARGIT-IORT trial showed a comparable long-term control to the classic RT.97 Accelerated breast irradiation over the course of one week was also described as non-inferior to the standard treatment course of three weeks, though long-term follow-up concerning cosmesis is still pending.98 Partial breast irradiation after BCS is also often discussed as an additional RT technique. However, one meta-analysis reported on significantly higher rates of in breast recurrences.99 In cases with higher risk constellations (T3/T4, N+, ER/PR-, Her2+, TNBC) radiotherapy according to guidelines is clearly recommended with a significant effect on recurrence-free survival being proven and confirmed by a recent analysis.95

The variety of treatment options (especially in low-risk constellations) is wide and needs to be discussed in detail with the patient, desirably before surgery. The patient's clinical condition and the patient’s preferences must be taken into account to reach an informed consent on the best possible treatment for the individual case.

Gastrointestinal Cancer

Curative intent RT for gastrointestinal cancers is usually not used as a sole modality but often within a bimodal (combined with simultaneous chemotherapy) or even trimodal (followed by surgery) approach. This complicates the evaluation of its role in elderly patients. A relevant part of toxicities and treatment compliance will be related to the concurrently applied chemotherapy rather than to RT alone. Detailed discussion of all combination approaches within gastrointestinal cancer would exceed the scope of this review. Therefore, we focused on three major gastrointestinal entities with (chemo)radiation as a major part of curative intent therapy strategies.

Esophageal Cancer

Neoadjuvant or definitive chemoradiation represents the current standard of care in locally advanced esophageal cancer (EC) based on resectability, while RT alone has to be considered a palliative treatment.100 Although the median age at diagnosis for EC is 68–70 years and roughly 30–40% of patients will be ≥75 years,101,102 elderly patients are still underrepresented in randomized trials. This seems to be partly related to the inclusion of surgery into the treatment approach. For example, the median age within the CROSS trial (setting the standard of neoadjuvant chemoradiation followed by surgery in resectable locally advanced EC) was only 60 years.103 However, modern trials focusing on definitive chemoradiation analysed cohorts comparable to the general age distribution. In the randomized SCOPE-1 and ARTDECO trials,104,105 median age was 67 and 71 years, with the latter including patients up to 90 years. Nevertheless, two SEER analyses focusing on patients ≥65 years showed an underutilization of treatment in older patients per se:106,107 34% of patients ≥65 years did not receive any treatment, which was significantly correlated with decreased OS. Interestingly, increasing age was associated with the non-receipt of surgery or chemotherapy but not RT.

Trimodal Therapy

Several studies showed that trimodal therapy (chemoradiation followed by surgery) results in improved OS also in selected elderly patients compared to surgery alone.108–110 Guttman et al109 analysed 1364 patients >70 years and found lower R+ resection and increased OS rates with the addition of neoadjuvant chemoradiation. Postoperative morbidity and mortality were similar. However, most studies analyzing age-dependent outcomes after trimodal therapy or resection alone showed an increase in postoperative cardiopulmonary toxicity108,109,111 in elderly patients. This finding translated into consecutively increased postoperative mortality at least in some of the trials.108,109 Cardiopulmonary toxicity after trimodal therapy increased roughly linear with age in a pooled US analysis (+61% per decade).108 Similarly, postop. mortality after resection increased with age (65–69 yrs: 9%, 70–79 yrs: 13%, >80 yrs: 20%) in a large population-based study.112 However, this might be rather related to comorbidity than age, as a significant association between an increased Charlson score (CCI >2) with postoperative mortality was described in the latter analysis.112

Definitive Chemoradiation

Data on definitive chemoradiation focusing on elderly patients are limited and mainly retrospective. Some reports found decreased survival rates and increased major toxicities.113,114 However, most data suggest that chemoradiation is equally effective compared to younger patients without a major increase in adverse events.115–117 Clinical complete remissions (cCR) are achieved in 50–65% of the patients with median OS times of 12–26 months and 2-year OS-rates of 30–40%.118 Moreover, in the prospective SCOPE-1 trial, age (< vs ≥ 65 years) had no statistical impact on PFS or OS according to multivariate analyses.104 Only one prospective trial specifically addressed chemoradiation feasibility in (selected) elderly patients: Servagi-Vernat et al119 included 22 patients (mean 79 years) if they had a CCI ≤4, a baseline weight loss <15% and ECOG ≤2. They were treated with 50Gy and concurrent cisplatin. The treatment compliance was 100%, 64% achieved cCR at 6 weeks and 1-year OS was 62% with no acute grade 4 toxicities. The authors concluded that chemoradiation is well tolerated using these inclusion criteria, however some data suggest an increase in pulmonary complications in patients ≥80 years.120 Tolerability of chemoradiation may depend also on several treatment factors. Carboplatin/Paclitaxel has shown similar OS and DFS with lower toxicity rates compared to the long-time standard of Cisplatin/5-FU also in the definitive setting.121 The introduction of modern radiation techniques (namely image-guided intensity-modulated RT) has resulted in similar or improved outcomes with clearly reduced side effects in patients of all age groups.122,123 The issue of the necessity of elective nodal irradiation is not finally solved in the absence of randomized trials. Indeed, increasing evidence suggests the use of smaller treatment volumes confined to the areas of gross disease because of increased tolerability.123,124 A recent randomized trial further suggests that dose escalation beyond 50Gy to the primary tumor does not result in significantly improved outcome, but may result in increased toxicity.105 Therefore, it seems reasonable to restrict the total dose in elderly/frail patients to 50Gy. Nutritional status (based on nutritional risk index) or weight loss at baseline have been identified as major prognostic factors for DFS and OS in large retrospective series.125,126 There is no clear evidence supporting the prognostic value of further weight loss or deterioration of nutritional status nor its therapeutic correction during chemoradiation. Nevertheless, care should be taken to ensure adequate nutritional support especially in elderly patients.

In summary, elderly patients in good shape (especially with no or limited cardiopulmonary comorbidity) might be selected for trimodal therapy. For most patients, definitive chemoradiation (with selective reevaluation for surgery) should be preferred because of better treatment compliance and less treatment-related mortality. Chemoradiation should be performed preferably with limited treatment volumes and total doses not exceeding 50Gy. Modern imaging for treatment planning and modern RT techniques should be used. Care should be taken especially for adequate nutritional support. Future trials specifically designed for elderly cohorts are warranted. They should include the evaluation of geriatric assessment tools for the prediction of chemoradiation outcomes as for example in the ongoing OSAGE trial.127

Rectal Cancer

Neoadjuvant (short course) RT or chemoradiation therapy represents the current standard of care in locally advanced rectal cancer (LARC) to improve locoregional control and/or resectability.128 Recently, neoadjuvant (chemo)radiation has been incorporated into so-called total neoadjuvant therapy (TNT) concepts combined with induction or consolidation chemotherapy prior to surgery.128 Several randomized trials have shown increased overall response, pathologic complete remissions (pCR) and distant control rates compared to the standard approach.129,130 In parallel, so-called NOM approaches omitting surgery in cases of cCR after neoadjuvant chemoradiation or TNT have gained attraction, especially if sphincter-sparing surgery seems not possible.131 Both concepts have not been specifically studied in elderly patients. As TNT concepts usually include doublet or triplet consolidation or induction chemotherapy regimens, they seem hardly suitable for the majority of elderly patients. Nevertheless, alternative (non-surgical) treatment concepts might offer new options in elderly patients hardly suitable for extended surgery.

Median age at diagnosis of rectal cancer is roughly 70 years with a peak incidence at 80–85 years, which also represents the peak prevalence age for comorbidities.132,133 In comparison, median age in major landmark trials was roughly 10 years less.134,135 Moreover, several population-based or retrospective studies showed an underutilization of surgery, neoadjuvant radiotherapy and palliative systemic treatment with less adherence to guidelines compared to younger patients (<70–75 years).136–141 In contrast, elderly patients received more often palliative and/or hypofractionated RT.140,141 While treatment outcome in terms of OS has clearly improved in the last decades in younger patients (5y-OS increased from 60% to 70%), this was not the case in patients aged >75 years (5-year OS remained stable at around 40%).142

As the vast majority of LARC patients is treated within multimodal concepts, suitability for subsequent treatments has to be included into the evaluation of radiation approaches. Most available data indicate no distinct differences in treatment compliance or tolerance within neoadjuvant (chemo)radiation concepts comparing younger and older patients (cutoff typically 70–75 years).143–147 Some trials showed higher rates of acute G3+ toxicity with limited clinical consequences. In contrast, a large Dutch study found a clear and steady increase in 1- and 6-month mortality after surgery with increasing age starting at 75 years.133,142 While postoperative complication rates per se showed no significant difference, the onset of a complication resulted in clearly worse outcome in elderly patients.133,142 For example, anastomotic leakage occurred at a rate of roughly 10% but resulted in a mortality rate of 8% in younger vs 57% in older patients.133 Interestingly, the same studies did not find a significant association of preoperative RT with postoperative complication rates.133,142 However, they showed an improved outcome with the addition of RT to surgery in elderly, which is also supported by a SEER analysis.148 The authors concluded that RT had little or no impact on postoperative complication rates or mortality, while the surgical trauma itself remains most important.133 In contrast, comorbidity seems to be clearly linked with postoperative complications and 30-day-mortality. A Dutch study restricted to patients >75 years showed only a moderate difference in postoperative complication rates and none in 30- day-mortality with or without neoadjuvant short-course RT in the entire cohort. However, they described a roughly 5-fold increase in complication rates and a more than 10-fold increase in 30- day-mortality, if severe comorbidity like COPD, diabetes or cerebrovascular disease was present.149 Therefore, the most important question to answer prior to RT is suitability for major pelvic surgery. If a patient is deemed suitable, indication for neoadjuvant (chemo)radiation can usually follow standard recommendations. Several retrospective studies showed high compliance rates and similar results in patients aged >70 and deemed fit for surgery compared to younger ones.143,146 There is no clear evidence for a distinctly different outcome comparing neoadjuvant short-course RT with long-course chemoradiation specifically in elderly patients, thus the indication may follow the institutional standards and general recommendations. However, short-course RT might be preferred with regard to patient’s convenience.

In elderly patients deemed less suitable for major pelvic surgery, several algorithms have been proposed.132,150 Paradoxically, some include treatment intensification of chemoradiation because of a higher likeliness of response. Moderate treatment intensification by localized dose escalation either via external beam RT or addition of brachytherapy can usually be achieved without a major increase in side effects.151 A consequently pronounced response then might enable local excision (LE) or even omission of surgery without compromising the overall results. Several trials suggested similar local control rates at least in node-negative patients with chemoradiation followed by LE in responding patients compared to more extended surgery,152–154 although not specifically for elderly patients. Short-course RT seems less suitable for this approach because several reports suggest increased complication rates with LE after short-course RT compared to chemoradiation.153–155 In frail (medically inoperable) patients, chemoradiation alone132,150 or EBRT with or without brachytherapy are reasonable options.150,156–158 If RT alone is used, hypofractionation should be strongly considered, because it achieved similar results to conventional fractionation.159 Short-course RT is an effective regimen for palliation with >80% complete or partial symptom relief at four weeks and reasonable rates of colostomy-free and overall survival.160 The addition of brachytherapy may further enhance the results as shown by a recent Phase I trial: increasing weekly doses of brachytherapy were added to a moderately hypofractionated EBRT concept and resulted in good response rates (cCR 61%) and acceptable OS.161 However, within the MTD cohort of this trial, rectal grade 3+ toxicity was roughly 30% prompting the authors to recommend some form of optimization.

Several structured reviews have proposed similar treatment algorithms for elderly patients with rectal cancer.132,150 Based on the available evidence, elderly patients in good shape (suitable for major pelvic surgery) should be preferably treated according to standard recommendations. Patients with intermediate features (some comorbidity, less suitable for major surgery) might be treated with chemoradiation (with or without dose escalation) and LE or omission or surgery in case of partial or complete response. In patients with severe comorbidity (unlikely to undergo surgery at all), hypofractionated radiotherapy achieves good palliation and seems feasible in most cases. Addition of brachytherapy can improve results but has to be weighed against increased complications risks.

Anal Cancer

In contrast to many other gastrointestinal malignancies, most patients with anal cancer are managed without surgery. The cornerstone of treatment is definitive chemoradiation with curative intent. Chemoradiation results in significantly improved outcomes compared to RT alone based on randomized trials.162–164 The median age at diagnosis is 60–65 years and roughly onethird of the patients are aged ≥70 years.165,166 Standard treatment usually includes a doublet chemotherapy regimen (Mitomycin C or Cisplatin combined with 5-FU or capecitabine) simultaneously applied to RT. This may result in considerable rates of acute gastrointestinal, hematological and skin toxicities. Therefore, its suitability to elderly patients is often questioned by clinicians. Data specifically addressing elderly patients are rare, mainly retrospective and therefore susceptible for selection biases. Moreover, the vast majority of published series used outdated staging modalities and radiation techniques (2D- or 3D-conformal RT). In contrast, modern techniques like intensity-modulated RT (IMRT) have shown clearly reduced rates of side effects in younger populations.167 Nevertheless, most published data suggest that age per se is not limiting the capability to tolerate standard therapy. In a large population-based analysis of roughly 12,000 patients treated with curative intent, age was not an independent factor for receiving chemoradiation in multivariate analysis.168 However, patients with two or more comorbidities were more likely to receive RT alone.168 Several authors analyzed elderly cohorts with cutoffs of 70–80 years treated by chemoradiation or RT alone.169–172 They showed high treatment compliance for RT but 25–50% of the patients needed dose reductions of chemotherapy, especially if comorbidity was present.169,170 Addition of chemotherapy resulted in significantly increased toxicity but also in improved outcome in most reports, including colostomy-free survival.169–171 Some investigators analyzed patients treated with either RT or chemoradiation according to age.173,174 They observed less CHT use in elderly patients, which showed also worse performance scores. If only patients with chemoradiation were compared, overall toxicity was not clearly increased in elderly patients, but they tended to have less skin but more hematological side effects. Outcome parameters were reported only for the overall cohorts. While cCR, colostomy and LC rates were similar, MFS, DFS and OS were worse in older patients, probably related to less chemotherapy use and increased mortality by other causes.173,174 Recent studies analyzed larger cohorts receiving chemoradiation by age groups.165,175 They consistently found reduced treatment compliance (mainly to chemotherapy) with increasing age but no clear difference in overall toxicity. However, none of the measured outcome parameters was associated with age, while comorbidity was associated with more toxicity, more dose reductions and worse LC.175 Thus, it seems reasonable to offer standard regimens also to elderly patients at least if performance score is good and no severe comorbidity is present.

In patients with comorbidities or limited performance status, several adjustments are possible with still curative intent. Chemoradiation can be performed using only one sensitizing agent (either 5-FU, Mitomycin or Cisplatin). Although chemoradiation with simultaneous doublet therapy (MMC/5-FU) was superior to 5-FU mono regarding LC, MFS and DFS in RTOG 87–04, no significant survival difference was reported.176 However, patients with cardiopulmonary comorbidities might be less suitable for 5-FU or capecitabine therapy. In those patients, MMC or Cisplatin mono may represent a reasonable alternative, although not supported by sufficient data. In patients deemed unsuitable for chemotherapy at all, conventionally fractionated RT alone may still result in good outcome with less toxicity. In the RT only arms of both randomized trials showing superiority of chemoradiation, 5-year OS was still >50%. RT alone can be tolerated by most patients of advanced age even in the presence of comorbidities,169–171,173 especially if modern radiation techniques are used. RTOG 0529167 evaluated IMRT for anal cancer in a single-arm phase II design and demonstrated significant reductions in acute toxicity compared to historic data from RTOG 98–11.177 Moreover, they showed that moderate de-escalation of dose did not result in decreased outcome. Therefore, the use of modern RT techniques should be mandatory in elderly patients. In frail patients, further adjustments can be made regarding target volume and dose. Charnley et al178 reported the results of limited treatment (30Gy in 10 fractions to the primary tumor excluding elective nodal irradiation combined with low-dose 5-FU) in frail patients and found 100% treatment compliance with low toxicity and still tolerable outcomes.

In summary, elderly patients in good shape should be preferably treated according to standard recommendations. In patients with comorbidities or limited performance status, stepwise adjustments can be made, ranging from chemoradiation with a single agent to standard RT alone. While toxicity will be clearly reduced with every step, long-term survival is still likely in the majority of patients. In patients with severe comorbidity, hypofractionated radiotherapy with limited volumes still achieves good palliation. The use of modern RT techniques like IMRT is strongly recommended to reduce acute and late side effects.

Gynecological Cancer

RT is an integral part of the treatment in many gynecological cancers, however often used as an adjunct to surgery. We therefore focused on cervical cancer as the main entity using nonsurgical radiation-containing approaches. Recently, the American Cancer Society reported that more than 15% of cervical cancer cases were found in women aged over 65 years.179 In large clinical studies, only a few elderly patients were included. Venkatesulu et al conducted a systematic review about patterns of care of cervical cancer in elderly and found only 24 out of 17,338 publications addressing the outcome in elderly cohorts.180 In these publications, 11,279 out of 14,479 patients aged ≥60 years (78%) received EBRT with or without concurrent chemotherapy and/or brachytherapy. With regard to the latter, low dose rate (LDR) was the most common modality, followed by high dose rate (HDR). However, in some studies with scheduled brachytherapy, up to 30% of the patients did not receive it due technical reasons (48.7%), comorbidities (69.4%) or patient refusal (38.3%). Five-year OS was generally inferior (27–69%) for elderly patients compared to younger populations (58–75%). Suboptimal radiation dose resulted in clearly reduced 5-year OS (11%) compared to patients treated with chemoradiation followed by brachytherapy (74%). However, it remains unclear if elderly patients with cervical cancer have a worse prognosis per se, due to comorbidity or limited performance scores or due to limited treatment application or tolerability. Limited historical data showed inconsistent results regarding generally worse181,182 or similar183,184 survival outcomes in elderly compared to younger patients. The performance status has been reported as a significant prognostic factor for OS and PFS.185–187 However, a retrospective registry-based study showed that patients ≥60 years were less likely to receive standard therapy compared to younger ones, mainly because treatment was not recommended.188 In a Japanese study, some elderly patients experienced severe toxicity, although radiotherapy was generally effective for them.189 These conditions might further contribute to the adverse effect on the prognosis of elderly patients.190 Interestingly, Hou et al191 reported comparable treatment outcomes with regard to CSS and loco-regional control in elderly patients with cervical cancer despite receiving less comprehensive treatment (including less concurrent chemoradiation, less brachytherapy, lower total RT dose and limited EBRT volumes). The authors concluded that, for patients ≥70 years a conservative treatment strategy with RT alone could be appropriate, especially in those with a favorable stage or histopathology. Similarly, Shimamoto et al192 found equivalent DFS but worse OS in patients aged ≥65 years compared to younger ones.

Modern EBRT techniques with a reduction of treatment-related side effects might be helpful for elderly patients to improve outcomes and RT completion rates. Several studies have confirmed clearly reduced side effects with IMRT techniques compared to conventional 2D- or 3D-conformal treatments.180,193 Brachytherapy was also often refused in elderly patients due to fear of toxicities or other reasons in elderly patients,180,183,191 but seems applicable with acceptable toxicity using modern image-guided approaches. In a prospective cohort study, Seppenwoolde et al194 analyzed morbidity and dose–volume effects in definitive radiochemotherapy for locally advanced cervical cancer. This cohort was treated with modern treatment techniques according to the EMBRACE protocol (94% received IMRT techniques). Most common gastrointestinal toxicities were low-grade diarrhea, stool urgency, rectal incontinence, rectal bleeding and weight loss. Most common genitourinary toxicities were dysuria and bladder incontinence. Only stool urgency, rectal or bladder incontinence and weight loss showed significantly increasing rates over the entire dose range. Correlations of toxicities with certain dose-volume factors (V40Gy) were observed, linking dysuria to bladder dose and stool urgency and rectal incontinence to bowel and rectum doses. Based on these data, the following treatment planning objectives were recommended to minimize stool urgency, rectal and urinary incontinence: bowel V40Gy≤250cm3, rectum V40Gy≤80% and bladder V40Gy≤80–90%, respectively.194 Using modern EBRT techniques considering those assumptions during treatment planning will likely increase treatment tolerability and ensure treatment completion in a larger proportion of elderly patients in the future.

In summary, age influences treatment strategies for cervical cancer world-wide, although based on scarce evidence. Only very few studies addressed treatment outcome in elderly patients with cervical cancer. However, most elderly patients may be treated with curative intent using modern radiation techniques with adequate supportive care. Recent studies have provided more insights into dose constraints for organs at risk to minimize acute and late toxicity by sophisticated treatment planning. The use of concurrent chemotherapy should be carefully evaluated based on patients´ performance status and comorbidities balancing possible benefit and harms. Brachytherapy should be an integral part of the treatment regimens in elderly patients as it can be safely applied by modern image-guided approaches with acceptable toxicities.

Prostate Cancer

Prostate cancer is a disease of the elderly and age remains to be the most important risk factor for its development. Due to its high prevalence and since the population will continue to grow older, prostate cancer and its management is a major health and socioeconomic factor.

The population-wide PSA screening combined with the good prognosis of prostate cancer carries the inherent risk of overtreatment, which is aggravated in elderly patients with a limited life span due to comorbidities or frailty. This has to be taken into account in screening strategies and treatment decisions. EAU, S3 as well as international NCCN guidelines have abandoned general population-based PSA screening in favor of offering early detection programs to select patients after discussion of pros and cons. For men ≥70 years, EAU guidelines for example give a grade D recommendation (rather than C for men aged 55–69) to inform about the possibility of PSA screening.195 A definition of ‘select patients’ is not provided, but data from the ERSPC and PIVOT trial suggest that men with a life expectancy of ≤15 years are unlikely to benefit from screening.196,197 However, after decades of routine PSA screening of men aged ≥45, the current recommendations do not seem to be fully adopted in clinical practice yet.

The same is true for the primary treatment of elderly patients with very low risk and low risk prostate cancer for whom active surveillance is the preferred approach when the life expectancy is below 20 and 10 years, respectively. The ProtecT trial has shown that active surveillance had similar very high 10-year-OS compared to surgery or primary RT.198 For patients with a life expectancy below 5 years, observation (not active surveillance!) is suggested in NCCN guidelines.199

The dependency on life expectancy underlines the importance of geriatric assessment of prostate cancer patients as a basis of treatment decisions. The task force of the International Society of Geriatric Oncology has issued recommendations for the management of prostate cancer in elderly patients.200 They emphasize that patients should be managed according to their individual health status rather than age. Therefore, initial evaluation of health status is mandatory. A two-step process is suggested using the validated G8 screening tool which identifies patients who should undergo full comprehensive geriatric assessment (CGA).

For elderly patients having aggressive (Gleason grade 8–10) or locally advanced disease, immediate treatment is usually warranted. Local complications such as bleeding, urinary retention and problems arising from distant metastases may occur within a short timeframe. In these patients, EBRT is the treatment of choice since it is less invasive and can be better tailored to the individual situation in terms of total dose and treatment time. For such patients, ultrahypofractionated radiation schedules, using 4–7 high-dose fractions daily or every other day, may be the preferred treatment. Such regimens reduce strain on patients in terms of required hospital visits (and transportation) and have been shown to be non-inferior to moderate or normofractionated schedules in randomized Phase III trials201,202 and metaanalyses.203 Androgen deprivation therapy (ADT) should be added according to current guidelines. However, cardiovascular risk and long-term complications as a result of reduced bone density need to be considered in the elderly. Notably, Qol (hot flashes, depression, fatigue) is typically less affected by long-term ADT in elderly patients than in younger patients featuring higher testosterone levels.

In the oligometastatic setting, data have shown OS benefits for both, RT of the primary tumor204–206 as well as metastases directed therapy89 for patients with low-volume disease. These treatments should not be held back thoughtlessly in elderly patients. They inherit a very low risk of severe toxicity, especially when delivered via SBRT. Thus, elderly patients will likely benefit in a similar range than younger ones. If life expectancy is severely impaired by comorbidities such as secondary cancers, palliative androgen deprivation monotherapy may be considered.

Discussion

Generally, RT is an important tool in the treatment of cancer patients. It has been estimated that more than 50% of all cancer patients benefit from some form of RT.207 With the latest improvements in radiation technique, cure rates have further increased, paralleled by a reduction in side effects. Thus, RT is a particularly attractive local treatment in elderly patients because of its non-invasive nature with limited systemic toxicities.207 Recently, hypofractionated regimens using higher single doses but fewer fractions have been established in many cancer entities. They achieved similar rates of tumor control and toxicity compared to conventionally fractionated approaches.8 This seems specifically relevant for older and/or frail patients with lack of social, financial or practical support for transportation and may further improve patient’s acceptance and tolerance of RT.

However, many curative intent radiation therapies still involve simultaneous applications of chemotherapy (Chemoradiation), which impedes strong hypofractionation approaches. Nevertheless, many older patients can tolerate conventionally fractionated chemoradiation regimens without increased side effects as shown by our summarized data. This is especially true, if older patients present in good performance score without significant comorbidities. Therefore, age alone should not be a decisive factor for treatment assignment. However, some of the mentioned studies also showed decreased treatment tolerance or the need for treatment adaptions, especially within combination approaches in relevant parts of elderly cohorts. Therefore, tools for an adequate selection of elderly patients for standard regimens as well as for adapted regimens or even omission of RT are urgently needed.

Several authors have used different assessment tools to predict treatment tolerance or side effects in elderly cancer patients with regard to surgery, chemotherapy and RT.208–210 The most sophisticated tool is a comprehensive geriatric assessment (CGA). This includes a multidisciplinary diagnostic process that evaluates medical, psychological, social and functional capacity.211 However, CGA is time consuming (it takes roughly two hours) and therefore unlikely to be applied in a relevant proportion of elderly patients in daily routine practice prior to RT.211 Therefore, screening tools have been developed either as surrogate markers to guide treatment decisions directly or at least to reduce the proportion of elderly patients amended to a full CGA. For example, Hurria et al209 developed an 11-item scale that correctly predicted the risk of severe toxicities in the majority of patients treated with chemotherapy in an external validation cohort. However, most of these screening tools still lack adequate discriminative power for the prediction of tolerability according to a recent review.208 Moreover, most of the studies did not involve patients with combined chemoradiation approaches. In studies specifically addressing elderly patients with planned RT, the use of either screening tools or CGA was even less promising. Szumacher et al211 performed a systematic review and found no significant association with RT-related toxicity in the majority of the included studies. One possible reason lies within the different spectrum of acute side effects caused by RT. In contrast to chemotherapy, most severe side effects are locoregionally limited, site-specific and depend on irradiated volume, total dose and fractionation. For example, lung cancer treatment by a small-volume SABR in 3 fractions with total doses equivalent to ≥80Gy in conventional fractionation will hardly result in any severe acute toxicity. Therefore, it can be used even in very old, frail patients with severe pulmonal comorbidity. In contrast, large field chemoradiation for lung cancer (even with much lower equivalent doses) will likely result in unacceptable side effects in similar patients. Moreover, RT inherits the risk of causing late toxicities possibly limiting long-term Qol. Again, those are site-specific, depend on volume, dose and fractionation and additionally on preexisting mainly organ-specific comorbidities. While an older or frail patient with limited pulmonary reserve will be at higher risk for all major surgeries, this is not true for all high-dose radiation treatments. Moreover, the timely onset of late radiation toxicities is highly variable and their detection needs long and careful follow-up strategies. Taken together, it is unlikely that common CGA or screening tests will be able to accurately predict tolerability or toxicity caused by RT, because they do not include site-specific and treatment-specific variables. Therefore, one key priority of future radiation research must be the development of modified evaluation tools taken the specific conditions of RT into account. Ideally, they would use at least some information already ascertained during routine RT planning.207 Prediction tools using such information could be more conveniently incorporated into daily routine compared to long lasting assessments. For example, radiomics and deep-learning algorithms might be used to gain more site-specific information already inherited in routinely performed treatment planning CTs. The latter methods also allow the incorporation of multiple information into new models.207 This may include site-specific clinical information, molecular data, imaging and (RT)-treatment-related factors resulting in more complex, nevertheless easily usable models.

Aside from patient selection for standard treatments, underrepresentation of elderly and/or comorbid patients in clinical trials is a major issue. Some progress has been made since major study groups recommended to skip an upper age limit within the eligibility criteria of randomized trials.3 This has already resulted in an increased rate of protocols allowing the inclusion of older patients.3 However, real inclusion of those patients is far from being adequate.4,212 Several patient-related barriers for study participation of elderly patients have been suggested: poor performance status, number of comorbidities, need for emergent therapy, lack of social support, reluctance to travel or difficulties due to travel distance (especially regarding fractionated RT), unwillingness to participate in trials per se, unability to manage the intensified follow-up procedures.213 While some of these factors may truly prevent patients from entering trials or from being eligible, there is no reliable evidence of a distinctly different attitude of elderly patients towards study participation.214 In fact, physicians recommendations and bias may play a more important role.214 Restrictive inclusion criteria regarding organ function, comorbidity and performance status215 and/or complex trial designs with multiple endpoints and/or additional preclinical research213 may further reduce trial participation of elderly patients. Moreover, most trials rarely address endpoints of particular interest for older adults such as preservation of physical or cognitive function.6,213

While there is clearly a need to generate more high-level evidence regarding the optimal treatment for elderly patients, the way to do so is challenging and debatable. One option would be to establish general rules for trials to ensure enrolling of a particular percentage of older patients.207 However, this may lengthen the trial period or increase the number of prematurely stopped trials if accrual in the elderly age group is slower than in younger patients. Even if achieved, subgroup analyses according to age will often lack statistical power for definitive conclusions. Another option is the design of trials specifically addressing elderly patients.213 This approach would allow adaptions of the study design: inclusion of geriatric assessments and recording of parameters related to frailty, but less complex trial procedures and more adequate endpoints to increase the willingness to participate. Several studies suggest that age-specific trials will result in the inclusion of much older patients, many of whom otherwise will not have entered a clinical trial.216,217 Moreover, tailored treatment regimens based on combinations of age, performance status, comorbidity and results of geriatric assessments could be evaluated. However, caution is mandatory in exploring tailored treatment approaches: age-specific trials should aim at defining an optimal treatment rather than simply testing less aggressive interventions.216 Otherwise, undertreatment may result in suboptimal outcomes. Consensus recommendations for the design of therapeutic trials in older and frail patients have been already published.213

In summary, RT plays an important role in the treatment of elderly patients because of its noninvasive nature. In several situations, RT can replace surgical interventions with lower risks for severe complications but similar outcome. Most treatments using RT as the sole modality are well tolerated by elderly patients. If RT is embedded into multimodal treatment approaches (for example combined with simultaneous chemotherapy or as neoadjuvant/adjuvant treatment), tolerability of the overall treatment is mainly dependent on the systemic or surgical therapy. Nevertheless, the (limited) available data suggest that standard treatments including RT are feasible and well tolerated in a large proportion of elderly patient with similar outcomes to younger ones. Therefore, age alone should not be a decisive factor with regard to treatment selection. Geriatric assessments or screening might be used for patient selection. However, the value of the available tests is limited with regard to predictability of specific RT side effects. Care should be taken to use modern radiation techniques, which clearly have reduced toxicity rates. Hypofractionated regimens might be preferred due to a reduction in socioeconomic burden for the patients. Future research should focus on the development of specific assessment tools for RT and improved representation of elderly patients in randomized trials. Moreover, specific trials addressing the optimal treatment strategy for elderly patients including endpoints of particular interest for elderly should be developed and conducted.

Abbreviations

RT, Radiation therapy; OS, overall survival; Qol, quality of life; RPA, recursive partitioning analysis; MGMT, O-6-methylguanine-DNA methyltransferase; CCNU, lomustine; Gy, Gray; EORTC, European Organisation for Research and Treatment of Cancer; BSC, best supportive care; HNSCC, head and neck squamous cell cancer; SEER, surveillance, epidemiology, and end results; 5-FU, 5-fluorouracil; RTOG, Radiation Therapy Oncology Group; CRP, C-reactive protein; SCLC, small cell lung cancer; NSCLC, non-small cell lung cancer; SABR, stereotactic ablative radiation therapy; ECOG, Eastern Cooperative Group performance score; BC, breast cancer; BCS, breast conserving surgery; ER, estrogen receptor; PR, progesterone receptor; Her2, human epidermal growth factor receptor 2; CALGB, Cancer and leukemia group B; NCCN, National Comprehensive Cancer network; N+, node-positive; EC, esophageal cancer; R+, margin-positive resection US, United States; yrs, years; CCI, Charlson comorbidity score; cCR, clinical complete remission PFS, progression-free survival; DFS, disease-free survival; LARC, locally advanced rectal cancer TNT, total neoadjuvant therapy pCR, pathologic complete remission NOM, non-operative management; COPD, chronic obstructive pulmonal disease; LE, local excision; EBRT, external beam radiation therapy; MTD, maximum tolerated dose; IMRT, intensity-modulated radiation therapy; 2D, two-dimensional; 3D, three-dimensional; LC, local control; MFS, metastases-free survival; MMC, Mitomycin C; LDR, low dose rate; HDR, high dose rate; CSS, cancer-specific survival; PSA, prostate-specific antigen; EAU, European Association of Urology; CGA, comprehensive geriatric assessment; ADT, androgen deprivation therapy; CT, computed tomography.

Disclosure

Falk Roeder received travel grants and lecture honoraria from Intraop Medical and PharmaMar. All other authors reported no competing interests.

References

1. Exterman M, Brain E, Canin B, et al. Priorities for the global advancement of care for older adults with cancer: an update of the international society of geriatric oncology priorities initiative. Lancet Oncol. 2021;22:e29–e36. doi:10.1016/S1470-2045(20)30473-3

2. Gomez-Millan J. Radiation therapy in the elderly: more side effects and complications? Crit Rev Oncol Hematol. 2009;71:70–78. doi:10.1016/j.critrevonc.2008.11.004

3. Horiot JC. Radiation therapy and the geriatric oncology patient. J Clin Oncol. 2007;25:1930–1935. doi:10.1200/JCO.2006.10.5312

4. Ludmir E, Mainwaring W, Lin TA, et al. Factors associated with age disparities among cancer clinical trial participants. JAMA Oncol. 2019;5:1769–1773. doi:10.1001/jamaoncol.2019.2055

5. VanderWalde N, Hurria A, Jasgsi R. Improving consistency and quality of care for older adults with cancer: the challenges of developing consensus guidelines for radiation therapy. Int J Radiat Oncol Biol Phys. 2017;98:721–724. doi:10.1016/j.ijrobp.2016.11.042

6. Dhakal P, Wichman CS, Pozehl B, et al. Preferences of adults with cancer for systemic cancer treatment: do preferences differ based on age? Future Oncol. 2022;18:311–321. doi:10.2217/fon-2021-0260

7. Dawson LA, Jaffray DA. Advances in image-guided radiation therapy. J Clin Oncol. 2007;25:938–946. doi:10.1200/JCO.2006.09.9515

8. Aitken K, Mukherjee S. When less is more: the rising tide of hypofractionation. Clin Oncol. 2022;34:277–279. doi:10.1016/j.clon.2022.03.002

9. WHO International Agency for research on cancer - Cancer today. Available from: https://gco.iarc.fr/today/online-analysis-table?v=2020&mode=population&mode_population=continents&population=900&populations=900&key=asr&sex=0&cancer=31&type=0&statistic=5&prevalence=0&population_group=0&ages_group%5B%5D=0&ages_group%5B%5D=17&group_cancer=1&include_nmsc=0&include_nmsc_other=1. Accessed November 01, 2022.

10. Miller KD, Ostrom QT, Kruchko C, et al. Brain and other central nervous system tumor statistics, 2021. CA Cancer J Clin. 2021;71(5):381–406. PMID: 34427324. doi:10.3322/caac.21693

11. Tumorregister München-Glioblastoma incidence and mortality. Available from: https://www.tumorregister-muenchen.de/en/facts/base/bC71G_E-ICD-10-C71-Glioblastomaincidence-and-mortality.pdf. Accessed November 01, 2022.

12. Stupp R, Mason WP, van den Bent MJ, et al; European Organisation for Research and Treatment of Cancer Brain Tumor and Radiotherapy Groups; National Cancer Institute of Canada Clinical Trials Group. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352:987–996. PMID: 15758009. doi:10.1056/NEJMoa043330

13. Stupp R, Hegi ME, Mason WP, et al; European Organisation for Research and Treatment of Cancer Brain Tumour and Radiation Oncology Groups; National Cancer Institute of Canada Clinical Trials Group. Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol. 2009;10(5):459–466. PMID: 19269895. doi:10.1016/S1470-2045(09)70025-7

14. Hegi ME, Diserens AC, Gorlia T, et al. MGMT gene silencing and benefit from temozolomide in glioblastoma. N Engl J Med. 2005;352(10):997–1003. PMID: 15758010. doi:10.1056/NEJMoa043331

15. Stupp R, Taillibert S, Kanner A, et al. Effect of tumor-treating fields plus maintenance temozolomide vs maintenance temozolomide alone on survival in patients with glioblastoma: a randomized clinical trial. JAMA. 2017;318(23):2306–2316. Erratum in: JAMA. 2018 319(17):1824.PMID: 29260225; PMCID: PMC5820703. doi:10.1001/jama.2017.18718

16. Taphoorn MJB, Dirven L, Kanner AA, et al. Influence of treatment with tumor-treating fields on health-related quality of life of patients with newly diagnosed glioblastoma: a secondary analysis of a randomized clinical trial. JAMA Oncol. 2018;4(4):495–504. PMID: 29392280; PMCID: PMC5885193. doi:10.1001/jamaoncol.2017.5082

17. Herrlinger U, Tzaridis T, Mack F, et al; Neurooncology Working Group of the German Cancer Society. Lomustine-temozolomide combination therapy versus standard temozolomide therapy in patients with newly diagnosed glioblastoma with methylated MGMT promoter (CeTeG/NOA-09): a randomised, open-label, Phase 3 trial. Lancet. 2019;393(10172):678–688. PMID: 30782343. doi:10.1016/S0140-6736(18)31791-4

18. Wiestler B, Claus R, Hartlieb SA, et al; Neurooncology Working Group (NOA) of the German Cancer Society. Malignant astrocytomas of elderly patients lack favorable molecular markers: an analysis of the NOA-08 study collective. Neuro Oncol. 2013;15(8):1017–1026. PMID: 23595628; PMCID: PMC3714152. doi:10.1093/neuonc/not043

19. Iwamoto FM, Reiner AS, Panageas KS, Elkin EB, Abrey LE. Patterns of care in elderly glioblastoma patients. Ann Neurol. 2008;64(6):628–634. PMID: 19107984. doi:10.1002/ana.21521

20. Keime-Guibert F, Chinot O, Taillandier L, et al.; Association of French-Speaking Neuro-Oncologists. Radiotherapy for glioblastoma in the elderly. N Engl J Med. 2007;356(15):1527–1535. PMID: 17429084. doi:10.1056/NEJMoa065901

21. Roa W, Brasher PM, Bauman G, et al. Abbreviated course of radiation therapy in older patients with glioblastoma multiforme: a prospective randomized clinical trial. J Clin Oncol. 2004;22(9):1583–1588. PMID: 15051755. doi:10.1200/JCO.2004.06.082

22. Perry JR, Laperriere N, O’Callaghan CJ, et al.; Trial Investigators. Short-course radiation plus temozolomide in elderly patients with glioblastoma. N Engl J Med. 2017;376(11):1027–1037. PMID: 28296618. doi:10.1056/NEJMoa1611977

23. Wick W, Platten M, Meisner C, et al; NOA-08 Study Group of Neuro-oncology Working Group (NOA) of German Cancer Society. Temozolomide chemotherapy alone versus radiotherapy alone for malignant astrocytoma in the elderly: the NOA-08 randomised, phase 3 trial. Lancet Oncol. 2012;13(7):70715. PMID: 22578793. doi:10.1186/1471-2407-9-68

24. Malmström A, Grønberg BH, Marosi C, et al.; Nordic Clinical Brain Tumour Study Group (NCBTSG). Temozolomide versus standard 6-week radiotherapy versus hypofractionated radiotherapy in patients older than 60 years with glioblastoma: the Nordic randomised, phase 3 trial. Lancet Oncol. 2012;13(9):1108–1117. PMID: 22877848. doi:10.1016/S1470-2045(12)70265-6

25. Mirimanoff RO, Gorlia T, Mason W, et al. Radiotherapy and temozolomide for newly diagnosed glioblastoma: recursive partitioning analysis of the EORTC 26981/22981-NCIC CE3 phase III randomized trial. J Clin Oncol. 2006;24(16):2563–2569. PMID: 16735709. doi:10.1200/JCO.2005.04.5963

26. Gerstein J, Franz K, Steinbach JP, et al. Postoperative radiotherapy and concomitant temozolomide for elderly patients with glioblastoma. Radiother Oncol. 2010;97(3):382–386. PMID: 20850883. doi:10.1016/j.radonc.2010.06.014

27. Combs SE, Wagner J, Bischof M, et al. Postoperative treatment of primary glioblastoma multiforme with radiation and concomitant temozolomide in elderly patients. Int J Radiat Oncol Biol Phys. 2008;70(4):1537–1546. PMID: 17967509. doi:10.1016/j.ijrobp.2007.07.2368

28. National Comprehensive Cancer Network (NCCN). Clinical Practice Guidelines in Oncology (NCCN Guidelines)- central Nervous System Cancers- Version 2; 2022. Available from: https://www.nccn.org/professionals/physician_gls/pdf/cns.pdf. Accessed November 1, 2022.

29. WHO International Agency for Research on Cancer - Cancer today. Available from: https://gco.iarc.fr/today/online-analysis-table?v=2020&mode=cancer&mode_population=continents&population=900&populations=900&key=asr&sex=0&cancer=39&type=0&statistic=5&prevalence=0&population_group=0&ages_group%5B%5D=0&ages_group%5B%5D=17&group_cancer=1&include_nmsc=0&include_nmsc_other=1. Accessed October 25, 2022.

30. Forastiere AA, Goepfert H, Maor M, et al. Concurrent chemotherapy and radiotherapy for organ preservation in advanced laryngeal cancer. N Engl J Med. 2003;349(22):2091–2098. doi:10.1056/NEJMoa031317

31. Bernier J, Domenge C, Ozsahin M, et al. Postoperative irradiation with or without concomitant chemotherapy for locally advanced head and neck cancer. N Engl J Med. 2004;350(19):1945–1952. doi:10.1056/NEJMoa032641

32. Cooper JS, Pajak TF, Forastiere AA, et al. Postoperative concurrent radiotherapy and chemotherapy for high-risk squamous-cell carcinoma of the head and neck. N Engl J Med. 2004;350(19):1937–1944. doi:10.1056/NEJMoa032646

33. Adelstein DJ, Saxton JP, Rybicki LA, et al. Multiagent concurrent chemoradiotherapy for locoregionally advanced squamous cell head and neck cancer: mature results from a single institution. J Clin Oncol. 2006;24(7):1064–1071. doi:10.1200/JCO.2005.01.5867

34. Bonner JA, Harari PM, Giralt J, et al. Radiotherapy plus cetuximab for squamous-cell carcinoma of the head and neck. N Engl J Med. 2006;354(6):567–578. doi:10.1056/NEJMoa053422

35. Bhide SA, Ahmed M, Barbachano Y, Newbold K, Harrington KJ, Nutting CM. Sequential induction chemotherapy followed by radical chemo-radiation in the treatment of locoregionally advanced head-and-neck cancer. Br J Cancer. 2008;99(1):57–62. doi:10.1038/sj.bjc.6604444

36. Blanchard P, Baujat B, Holostenco V, et al. Meta-analysis of chemotherapy in head and neck cancer (MACH-NC): a comprehensive analysis by tumour site. Radiother Oncol. 2011;100(1):33–40. doi:10.1016/j.radonc.2011.05.036

37. Pignon JP, le Maitre A, Maillard E, Bourhis J. Meta-analysis of chemotherapy in head and neck cancer (MACH-NC): an update on 93 randomised trials and 17,346 patients. Radiother Oncol. 2009;92(1):4–14. doi:10.1016/j.radonc.2009.04.014

38. Pulte D, Brenner H. Changes in survival in head and neck cancers in the late 20th and early 21st century: a period analysis. Oncologist. 2010;15:994–1001. doi:10.1634/theoncologist.2009-0289

39. Cancer Research UK - Cancer statistics - Head and neck cancers. Available from: https://www.cancerresearchuk.org/health-professional/cancer-statistics/statistics-by-cancertype/head-and-neck-cancers/incidence#heading-One. Accessed October 25 2022.

40. Haehl E, Rühle A, Spohn S, et al. Patterns-ofCare analysis for radiotherapy of elderly head-and-neck cancer patients: a trinational survey in Germany, Austria and Switzerland. Front Oncol. 2022;11:723716. PMID: 35047384; PMCID: PMC8761738. doi:10.3389/fonc.2021.723716

41. Maggiore RJ, Curran EK, Witt ME, Haraf DJ, Vokes EE, Cohen EE. Survival and selected outcomes of older adults with locally advanced head/neck cancer treated with chemoradiation therapy. J Geriatr Oncol. 2013;4(4):327–333. PMID: 24472475; PMCID: PMC4413907. doi:10.1016/j.jgo.2013.05.003

42. Haehl E, Rühle A, David H, et al. Radiotherapy for geriatric head-and-neck cancer patients: what is the value of standard treatment in the elderly? Radiat Oncol. 2020;15(1):31. PMID: 32019576; PMCID: PMC7001207. doi:10.1186/s13014-020-1481-z

43. Brown ML, Glanzmann C, Huber G, Bredell M, Rordorf T, Studer G. IMRT/VMAT for malignancies in the head-and-neck region: outcome in patients aged 80. Strahlenther Onkol. 2016;192(8):52636. PMID: 27306747; PMCID: PMC4956718. doi:10.1007/s00066-016-0986-8

44. Machtay M, Moughan J, Trotti A, et al. Factors associated with severe late toxicity after concurrent chemoradiation for locally advanced head and neck cancer: an RTOG analysis. J Clin Oncol. 2008;26(21):3582–3589. PMID: 18559875; PMCID: PMC4911537. doi:10.1200/JCO.2007.14.8841

45. Morse RT, Ganju RG, Gan GN, et al. Sarcopenia and treatment toxicity in older adults undergoing chemoradiation for head and neck cancer: identifying factors to predict frailty. Cancers. 2022;14(9):2094. PMID: 35565223; PMCID: PMC9103923. doi:10.3390/cancers14092094.

46. Daly ME, Lau DH, Farwell DG, Luu Q, Donald PJ, Chen AM. Feasibility and toxicity of concurrent chemoradiation for elderly patients with head and neck cancer. Am J Otolaryngol. 2013;34(6):631–635. PMID: 23954137. doi:10.1016/j.amjoto.2013.07.010

47. Lu Y, Hua J, Yan F, et al. Combined radiotherapy and chemotherapy versus radiotherapy alone in elderly patients with nasopharyngeal carcinoma: a SEER population-based study. Medicine. 2021;100(29):e26629. PMID: 34398019; PMCID: PMC8294920. doi:10.1097/MD.0000000000026629.

48. Paleri V, Wight RG, Silver CE, et al. Comorbidity in head and neck cancer: a critical appraisal and recommendations for practice. Oral Oncol. 2010;46(10):712–719. PMID: 20850371. doi:10.1016/j.oraloncology.2010.07.008

49. Hall SF, Groome PA, Rothwell D. The impact of comorbidity on the survival of patients with squamous cell carcinoma of the head and neck. Head Neck. 2000;22(4):317–322. doi:10.1002/1097-0347(200007)22:4<317::AID-HED1>3.0.CO;2-0

50. van Deudekom FJ, Schimberg AS, Kallenberg MH, Slingerland M, van der Velden L-A, Mooijaart SP. Functional and cognitive impairment, social environment, frailty and adverse health outcomes in older patients with head and neck cancer, a systematic review. Oral Oncol. 2017;64:27–36. doi:10.1016/j.oraloncology.2016.11.013

51. Eytan DF, Blackford AL, Eisele DW, Fakhry C. Prevalence of comorbidities among older head and neck cancer survivors in the United States. Otolaryngol Head Neck Surg. 2019;160:85–92. doi:10.1177/0194599818796163

52. Sanabria A, Carvalho AL, Vartanian JG, Magrin J, Ikeda MK, Kowalski LP. Comorbidity is a prognostic factor in elderly patients with head and neck cancer. Ann Surg Oncol. 2007;14(4):1449–1457. doi:10.1245/s10434-006-9296-1

53. Derks W, de Leeuw JR, Hordijk GJ, Winnubst JA. Reasons for non-standard treatment in elderly patients with advanced head and neck cancer. Eur Arch Otorhinolaryngol. 2005;262:21–26. doi:10.1007/s00405-004-0744-x

54. de Vries J, Poelman A, Sidorenkov G, et al. The association of frailty and outcomes of geriatric assessment with acute radiation-induced toxicity in patients with head and neck cancer. Oral Oncol. 2022;130:105933. PMID: 35665634. doi:10.1016/j.oraloncology.2022.105933

55. Chou WC, Chang PH, Chen PT, et al. Clinical significance of vulnerability assessment in patients with primary head and neck cancer undergoing definitive concurrent chemoradiation therapy. Int J Radiat Oncol Biol Phys. 2020;108(3):602–611. PMID: 31987971. doi:10.1016/j.ijrobp.2020.01.004

56. Karavolia E, van Rijn-dekker MI, Van den Bosch L, et al. Impact of sarcopenia on acute radiation-induced toxicity in head and neck cancer patients. Radiother Oncol. 2022;170:122–128. PMID: 35304862. doi:10.1016/j.radonc.2022.03.009

57. Rühle A, Haehl E, David H, et al. The value of laboratory parameters for anemia, renal function, systemic inflammation and nutritional status as predictors for outcome in elderly patients with head-and neck cancers. Cancers. 2020;12(6):1698. PMID: 32604773; PMCID: PMC7352755. doi:10.3390/cancers12061698

58. Sprave T, Rühle A, Stoian R, et al. Radiotherapy for nonagenarians: the value of biological versus chronological age. Radiat Oncol. 2020;15(1):113. PMID: 32430009; PMCID: PMC7236131. doi:10.1186/s13014-020-01563-x

59. Rühle A, Stromberger C, Haehl E, et al. Development and validation of a novel prognostic score for elderly head-and-neck cancer patients undergoing radiotherapy or chemoradiation. Radiother Oncol. 2021;154(276–282):276–282. PMID: 33245947. doi:10.1016/j.radonc.2020.11.023

60. Miller ED, Fisher JL, Haglund KE, et al. Identifying patterns of care for elderly patients with nonsurgically treated stage III non-small cell lung cancer: an analysis of the national cancer database. Radiat Oncol. 2018;13(1):196. doi:10.1186/s13014-018-1142-7

61. Bonanno L, Attili I, Pavan A, et al. Treatment strategies for locally advanced non-small cell lung cancer in elderly patients: translating scientific evidence into clinical practice. Crit Rev Oncol Hematol. 2021;163:103378. doi:10.1016/j.critrevonc.2021.103378

62. Bozogmehr F, Chung I, Christopoulos P, et al. Thoracic radiotherapy plus durvalumab in elderly and/or frail NSCLC stage III patients unfit for chemotherapy – employing optimized (hypofractionated) radiotherapy to foster durvalumab efficacy: study protocol of the TRADE-hypo trial. BMC Cancer. 2020;20:806. doi:10.1186/s12885-020-07264-8

63. Dumontier C, Loh KP, Soto-perez-de-celis E, et al. Decision making in older adults with cancer. J Clin Oncol. 2021;39(19):2164–2174. doi:10.1200/JCO.21.00165

64. Chang JY, Senan S, Paul MA, et al. Stereotactic ablative radiotherapy versus lobectomy for operable stage I non-small-cell lung cancer: a pooled analysis of two randomised trials. Lancet Oncol. 2015;16:630–637. doi:10.1016/S1470-2045(15)70168-3

65. Chang JY, Mehran RJ, Feng L, et al. Stereotactic ablative radiotherapy for operable stage I nonsmall-cell lung cancer (revised STARS): long-term results of a single-arm, prospective trial with prespecified comparison to surgery. Lancet Oncol. 2021;22(10):1448–1457. doi:10.1016/S14702045(21)00401-0

66. Brooks ED, Sun B, Zhao L, et al. Stereotactic ablative radiation therapy is highly safe and effective for elderly patients with early-stage non-small cell lung cancer. Int J Radiat Oncol Biol Phys. 2017;98(4):900–907. doi:10.1016/j.ijrobp.2016.12.022

67. Giuliani M, Hope A, Guckenberger M, et al. Stereotactic body radiation therapy in octo- and nonagenarians for the treatment of early-stage lung cancer. Int J Radiat Oncol Biol Phys. 2017;98(4):893–899. doi:10.1016/j.ijrobp.2017.01.019

68. Eberhardt WE, Pöttgen C, Gauler TC, et al. Phase III study of surgery versus definitive concurrent chemoradiotherapy boost in patients with resectable stage IIIA(N2) and selected IIIB non-small-cell lung cancer after induction chemotherapy and concurrent chemoradiotherapy (ESPATUE). J Clin Oncol. 2015;33(35):4194–4201. doi:10.1200/JCO.2015.62.6812

69. Albain KS, Swann RS, Rusch V, et al. Radiotherapy plus chemotherapy with or without surgical resection for stage III non-small-cell lung cancer: a phase III randomised controlled trial. Lancet. 2009;374(9687):379–386. doi:10.1016/S0140-6736(09)60737-6

70. Sawada S, Komori E, Nogami N, et al. Advanced age is not correlated with either short-term or long-term postoperative results in lung cancer patients in good clinical condition. Chest. 2005;128(3):1557–1563. doi:10.1378/chest.128.3.1557

71. Die Loucou J, Pagès P-B, Falcoz P-E, et al. Validation and update of the thoracic surgery scoring system (Thoracoscore) risk model. Eur J Cardiothorac Surg. 2020;58(2):350–356. doi:10.1093/ejcts/ezaa056

72. Pergolizzi S, Santacaterina A, De Renzis C, et al. Older people with non small cell lung cancer in clinical stage IIIA and co-morbid conditions. Is curative irradiation feasible? Final results of a prospective study. Lung Cancer. 2002;37(2):201–206. doi:10.1016/s0169-5002(02)00038-7

73. Pignon T, Gregor A, Schaake Koning C, et al. Age has no impact on acute and late toxicity of curative thoracic radiotherapy. Radiother Oncol. 1998;46(3):239–248. doi:10.1016/s0167-8140(97)00188-6

74. Lyengar P, Zhang-Velten E, Court L, et al. Accelerated hypofractionated image-guided vs conventional radiotherapy for patients with stage II/III non-small cell lung cancer and poor performance status: a randomized clinical trial. JAMA Oncol. 2021;7(10):1497–1505. doi:10.1001/jamaoncol.2021.3186

75. Dillman RO, Seagren SL, Propert KJ, et al. A randomized trial of induction chemotherapy plus high-dose radiation versus radiation alone in stage III non-small-cell lung cancer. N Engl J Med. 1990;323(14):940–945. doi:10.1056/NEJM199010043231403

76. Atagi S, Kawahara M, Tamura T, et al. Standard thoracic radiotherapy with or without concurrent daily low-dose carboplatin in elderly patients with locally advanced non-small cell lung cancer: a phase III trial of the Japan Clinical Oncology Group (JCOG9812). Jpn J Clin Oncol. 2005;35(4):195–201. doi:10.1093/jjco/hyi060

77. Atagi S, Kawahara M, Yokoyama A, et al. Thoracic radiotherapy with or without daily low-dose carboplatin in elderly patients with non-small-cell lung cancer: a randomised, controlled, phase 3 trial by the Japan Clinical Oncology Group (JCOG0301). Lancet Oncol. 2012;13(7):671–678. doi:10.1016/S1470-2045(12)70139-0

78. Atagi S, Mizusawa J, Ishikura S, et al. Chemoradiotherapy in elderly patients with non-small-cell lung cancer: long-term follow-up of a randomized trial (JCOG0301). Clin Lung Cancer. 2018;19(5):e619–e627. doi:10.1016/j.cllc.2018.04.018

79. Dawe DE, Christiansen D, Swaminath A, et al. Chemoradiotherapy versus radiotherapy alone in elderly patients with stage III non-small cell lung cancer: a systematic review and meta-analysis. Lung Cancer. 2016;99:180–185. doi:10.1016/j.lungcan.2016.07.016

80. Auperin 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(13):2181–2190. doi:10.1200/JCO.2009.26.2543

81. Dilling TJ, Extermann M, Kim J, et al. Phase 2 Study of concurrent cetuximab plus definitive thoracic radiation therapy followed by consolidation docetaxel plus cetuximab in poor prognosis or elderly patients with locally advanced non-small cell lung cancer. Int J Radiat Oncol Biol Phys. 2014;90:828–833. PMID: 25216856 Clinical Trial. doi:10.1016/j.ijrobp.2014.07.023

82. Franceschini D, De Rose F, Cozzi L, et al. Radical hypo-fractionated radiotherapy with volumetric modulated arc therapy in lung cancer A retrospective study of elderly patients with stage II disease. Strahlenther Onkol. 2017;193:385–391. doi:10.1007/s00066-017-1103-3

83. Driessen EJM, Janssen-Heijnen MLG, Maas HA, et al. Study protocol of the NVALT25-ELDAPT trial: selecting the optimal treatment for older patients with stage III non-small-cell lung cancer. Clin Lung Cancer. 2018;19(6):e849–e852. doi:10.1016/j.cllc.2018.07.003

84. Gray JE, Villegas A, Daniel D, et al. Three-year overall survival with durvalumab after chemoradiotherapy in stage III NSCLC-update from PACIFIC. J Thorac Oncol. 2020;15(2):288–293. doi:10.1016/j.jtho.2019.10.002

85. Antonia SJ, Villegas A, Daniel D, et al. Overall survival with durvalumab after chemoradiotherapy in stage III NSCLC. N Engl J Med. 2018;379:2342–2350. PMID: 30280658 Clinical Trial. doi:10.1056/NEJMoa1809697

86. Grossi F, Crinò L, Logroscino A, et al. Use of nivolumab in elderly patients with advanced squamous non-small-cell lung cancer: results from the Italian cohort of an expanded access programme. Eur J Cancer. 2018;100:126–134. doi:10.1016/j.ejca.2018.05.015

87. Galli G, De Toma A, Pagani F, et al. Efficacy and safety of immunotherapy in elderly patients with non-small cell lung cancer. Lung Cancer. 2019;137:38–42. doi:10.1016/j.lungcan.2019.08.030

88. Kaira K, Mouri A, Kato S, et al. A phase II study of daily carboplatin plus irradiation followed by durvalumab for stage III non-small cell lung cancer patients with PS 2 up to 74 years old and patients with PS 0 or 1 from 75 years: NEJ039A (trial in progress). BMC Cancer. 2020;20:961. doi:10.1186/s12885-020-07406-y

89. Palma DA, Olson R, Harrow S, et al. Stereotactic ablative radiotherapy versus standard of care palliative treatment in patients with oligometastatic cancers (SABR-COMET): a randomised, phase 2, open-label trial. Lancet. 2019;393:2051–2058. doi:10.1016/S0140-6736(18)32487-5

90. Theelen W, Chen D, Verma V, et al. Pembrolizumab with or without radiotherapy for metastatic non-small-cell lung cancer: a pooled analysis of two randomised trials. Lancet Respir Med. 2021;9:467–475. doi:10.1016/S2213-2600(20)30391-X

91. American Cancer Society. 2019–2020, breast cancer facts & figures. Available from: https://www.cancer.org/content/dam/cancer-org/research/cancer-facts-and-statistics/breastcancer-facts-and-figures/breast-cancer-facts-and-figures-2019-2020.pdf. Accessed October 06, 2022.

92. S3-Leitlinie Mammakarzinom; 2021. Available from: https://www.leitlinienprogramm-onkologie.de/fileadmin/user_upload/Downloads/Leitlinien/Mammakarzinom_4_0/Version_4.4/LL_Mammakarzinom_Langversion_4.4.pdf. Accessed October 06, 2022.

93. Gu J, Groot G, Boden C, et al. Review of factors influencing women’s choice of mastectomy versus breast conserving therapy in early stage breast cancer: a systematic review. Clin Breast Cancer. 2018;18:e539–e554. doi:10.1016/j.clbc.2017.12.013

94. Hughes K, Schnaper L, Bellon J, et al. Lumpectomy plus tamoxifen with or without irradiation in women age 70 years or older with early breast cancer: long-term follow-up of CALGB 9343. J Clin Oncol. 2013;31:19. doi:10.1200/JCO.2012.45.2615

95. Stueber T, Diessner J, Bartmann C, et al. Effect of adjuvant radiotherapy in elderly patients with breast cancer. PLoS One. 2020;15(5):e0229518. doi:10.1371/journal.pone.0229518

96. NCCN. NCCN guideline; 2022. Available from: https://www.nccn.org/professionals/physician_gls/pdf/breast.pdf. Accessed October 06, 2022.

97. Vaidya J, Bulsara M, Baum M, et al. Long term survival and local control outcomes from single dose targeted intraoperative radiotherapy during lumpectomy (TARGIT-IORT) for early breast cancer: TARGIT-A randomised clinical trial. BMJ. 2020:m2836. doi:10.1136/bmj.m2836

98. Murray Brunt A, Haviland J, Wheatley D, et al. Hypofractionated breast radiotherapy for 1 week versus 3 weeks (FAST-Forward): 5-year efficacy and late normal tissue effects results from a multicentre, non-inferiority, randomised, phase 3 trial. Lancet. 2020;395(10237):1613–1626. doi:10.1016/S0140-6736(20)30932-6

99. Marta G, Macedo C, Carvalho HA, Hanna S, da Silva J, Riera R. Accelerated partial irradiation for breast cancer: systematic review and meta-analysis of 8653 women in eight randomized trials. Radiother Oncol. 2015;114(1):42–49. doi:10.1016/j.radonc.2014.11.014

100. Herskovic A, Martz K, Al-Sarraf M, et al. Combined chemotherapy and radiotherapy compared with radiotherapy alone in patients with cancer of the esophagus. N Eng J Med. 1992;326(24):1593–1598. doi:10.1056/NEJM199206113262403

101. Bracken-Clark D, Farroq AR, Horgan AM. Management of locally advanced and metastatic esophageal cancer in the older population. Curr Oncol Rep. 2018;20(12):99. doi:10.1007/s11912-018-0745-3

102. Won E. Issues in the management of esophageal cancer and geriatric patients. Chin Clin Oncol. 2017;6(5):51. doi:10.21037/cco.2017.10.05

103. van Hagen P, Hulshof MC, van Lanschot JJ, et al. Preoperative chemoradiotherapy for esophageal or junctional cancer. N Engl J Med. 2012;366(22):2074–2084. doi:10.1056/NEJMoa1112088

104. Crosby T, Hurt CN, Falk S, et al. Chemoradiotherapy with or without cetuximab in patients with oesophageal cancer (SCOPE-1): a multicenter, phase 2/3 randomised trial. Lancet Oncol. 2013;14:627–637. doi:10.1016/j.eururo.2018.09.008

105. Hulshof MC, Geijsen ED, Rozema T, et al. Randomized study on dose escalation in definitive chemoradiation for patients with locally advanced esophageal cancer (ARTDECO study). J Clin Oncol. 2021;39:2816–2824. doi:10.1200/JCO.20.03697

106. Molena D, Stem M, Blackford AL, Lidor AO. Esophageal cancer treatment is underutilized among elderly patients in the United States. J Gastrointest Surg. 2017;21:126–136. doi:10.1007/s11605-016-3229-5

107. Steyerberg EW, Neville B, Weeks JC, Earle CC. Referral patterns, treatment choices, and outcomes in locoregional esophageal cancer: a population-based analysis of elderly patients. J Clin Oncol. 2007;25:2389–2396. doi:10.1200/JCO.2006.09.7931

108. Lester SC, Lin SH, Chuong M, et al. A multi-institutional analysis of trimodality therapy for esophageal cancer in elderly patients. Int J Radiat Oncol Biol Phys. 2017;98:820–828. doi:10.1016/j.ijrobp.2017.02.021

109. Guttman DM, Mitra N, Metz JM, et al. Neoadjuvant chemoradiation is associated with improved overall survival in older patients with esophageal cancer. J Geriatr Oncol. 2018;9:40–46. doi:10.1016/j.jgo.2017.08.010

110. Smith GL, Smith BD, Buchholz TA, et al. Patterns of care and locoregional treatment outcomes in older esophageal cancer patients: the SEER-medicare cohort. Int J Radiat Oncol Biol Phys. 2009;74:482–489. doi:10.1016/j.ijrobp.2008.08.046

111. Fogh SE, Yu A, Kubicek GJ, et al. Do elderly patients experience increased perioperative or postoperative morbidity or mortality when given neoadjuvant chemoradiation before esophagectomy? Int J Radiat Oncol Biol Phys. 2011;80:1372–1376. doi:10.1016/j.ijrobp.2010.04.055

112. Finlaysson E, Fan Z, Birkmeyer JD. Outcomes in octogenarians undergoing high-risk cancer operation: a national study. J Am Coll Surg. 2007;205:729–734. doi:10.1016/j.jamcollsurg.2007.06.307

113. Mak RH, Mamon HJ, Ryan DP, et al. Toxicity and outcome after chemoradiation for esophageal cancer in patients age 75 or older. Dis Esophagus. 2010;23:316–323. doi:10.1111/j.1442-2050.2009.01014.x

114. Takeuchi S, Ohtsu A, Doi T, et al. A retrospective study of definitive chemoradiotherapy for elderly patients with esophageal cancer. Am J Clin Oncol. 2007;30:607–611. doi:10.1097/COC.0b013e3180ca7c84

115. Tougeron D, Di Fiore F, Thureau S, et al. Safety and outcome of definitive chemoradiotherapy in elderly patients with oesophageal cancer. Br J Cancer. 2008;99(10):1586–1592. doi:10.1038/sj.bjc.6604749

116. Anderson SE, Minsky BD, Bains M, et al. Combined modality chemoradiation in elderly oesophageal cancer patients. Br J Cancer. 2007;96(12):1823–1827. doi:10.1038/sj.bjc.6603821

117. Tougeron D, Hamidou H, Scotte M, et al. Esophageal cancer in the elderly: an analysis of the factors associated with treatment decisions and outcomes. BMC Cancer. 2010;10(1):510. doi:10.1186/1471-2407-10-510

118. Won E, Ilson DH. Management of localized esophageal cancer in the older patient. Oncologist. 2014;19(4):367–374. doi:10.1634/theoncologist.2013-0178

119. Servagi-Vernat S, Crehange G, Roullet B, et al. Phase II study of a platinum-based adapted chemotherapy regime combined with radiotherapy in patients 75 years and older with esophageal cancer. Drugs Aging. 2015;32:487–493. doi:10.1007/s40266-015-0275-8

120. Xu C, Xi M, Moreno A, et al. Definitive chemoradiation therapy for esophageal cancer in the elderly: clinical outcomes for patients exceeding 80 years old. Int J Radiat Oncol Biol Phys. 2017;98:811–819. doi:10.1016/j.ijrobp.2017.02.097

121. Honing J, Smit JK, Mujis CT, et al. A comparison of carboplatin and paclitaxel with cisplatinum and 5-fluorouracil in definitive chemoradiation in esophageal cancer patients. Ann Oncol. 2014;25:638–643. doi:10.1093/annonc/mdt589

122. Roeder F, Nicolay NH, Ngyen T, et al. Intensity modulated radiotherapy (IMRT) with concurrent chemotherapy as definitive treatment of locally advanced esophageal cancer. Radiat Oncol. 2014;9:191. doi:10.1186/1748-717X-9-191

123. Walter F, Böckle D, Schmidt-Hegemann NS, et al. Clinical outcome of elderly patients (≥70 years) with esophageal cancer undergoing definitive or neoadjuvant radio(chemo)therapy: a retrospective single center analysis. Radiat Oncol. 2018;13:93. doi:10.1186/s13014-018-1044-8

124. Wang X, Miao C, Chen Z. Can involved-field irradiation replace elective nodal irradiation in chemoradiotherapy for esophageal cancer? A systematic review and meta-analysis. Onco Targets Ther. 2017;10:2087–2095. doi:10.2147/OTT.S130285

125. Clavier JB, Antoni D, Atlani D, et al. Baseline nutritional status is prognostic factor after definitive radiochemotherapy for esophageal cancer. Dis Esophagus. 2014;2:560–567. doi:10.1111/j.1442-2050.2012.01441.x

126. Di Fiori F, Lecleire S, Pop D, et al. Baseline nutritional status is predictive of response to treatment and survival in patients treated by definitive chemoradiotherapy for a locally advanced esophageal cancer. Am J Gastroenterol. 2007;102:2557–2563. doi:10.1111/j.1572-0241.2007.01437.x

127. Servagi-Vernat S, Crehange G, Bonnetain F, et al. Chemoradiation in elderly esophageal cancer patients: rationale and design of a phase I/II multicenter study (OSAGE). BMC Cancer. 2017;17:483. doi:10.1186/s12885-017-3465-4

128. Roeder F, Meldolesi E, Gerum S, et al. Recent advances in (chemo-)radiation therapy for rectal cancer: a comprehensive review. Radiat Oncol. 2020;15(1):262. doi:10.1186/s13014-020-01695-0

129. Bahadoer RR, Dijkstra EA, van Etten B, et al. Short-course radiotherapy followed by chemotherapy before total mesorectal excision (TME) versus preoperative chemoradiotherapy, TME, and optional adjuvant chemotherapy in locally advanced rectal cancer (RAPIDO): a randomized, open-label, phase 3 trial. Lancet Oncol. 2021;22:29–42. doi:10.1200/JCO.2005.03.7465

130. Conroy T, Bosset JF, Etienne PL, et al. Neoadjuvant chemotherapy with FOLFIRINOX and preoperative chemoradiotherapy for patients with locally advanced rectal cancer (UNICANCERPRODIGE 23): a multicentre, randomized, open-label phase 3 trial. Lancet Oncol. 2021;22:702–715. doi:10.1016/S1470-2045(21)00079-6

131. Garcia-Aguilar J, Patil S, Gollub MJ, et al. Organ preservation in patients treated with rectal adenocarcinoma treated with total neoadjuvant therapy. J Clin Oncol. 2022;40:2546–2556. doi:10.1200/JCO.22.00032

132. De Felice F, Crocetti D, Maiuri V, et al. Locally advanced rectal cancer: treatment approach in elderly patients. Curr Treat Options Oncol. 2020;21:1. doi:10.1007/s11864-019-0692-8

133. Rutten HJ, den Dulk M, Lemmens VE, et al. Controversies of total mesorectal excision for rectal cancer in elderly patients. Lancet Oncol. 2008;9(5):494–501. doi:10.1016/S1470-2045(08)70129-3

134. Kapiteijn E, Marijnen CA, Nagtegaal ID, et al. Preoperative radiotherapy combined with total mesorectal excision for resectable rectal cancer. N Engl J Med. 2001;345:638–646. doi:10.1056/NEJMoa010580

135. Sauer R, Becker H, Hohenberger W, et al. Preoperative versus postoperative chemoradiotherapy for rectal cancer. N Engl J Med. 2004;351(17):1731–1740. doi:10.1056/NEJMoa040694

136. Banghu A, Kiran RP, Audisio R, Tekkis P. Survival outcome of operated and non-operated elderly patients with rectal cancer: a surveillance, epidemiology and end results analysis. Eur J Surg Oncol. 2014;40:1510–1516. doi:10.1016/j.ejso.2014.02.239

137. Chang GJ, Skibber JM, Feig BW, et al. Are we undertreating rectal cancer in the elderly? An epidemiologic study. Ann Surg. 2007;246(2):215–221. doi:10.1097/SLA.0b013e318070838f

138. Jung B, Pahlman L, Johansson R, Nilsson E. Rectal cancer treatment and outcome in the elderly: an audit based on the Swedish rectal cancer registry 1995–2004. BMC Cancer. 2009;9:68. doi:10.1186/1471-2407-9-68

139. Mourad AP, Shella de Robles M, Putnis S, Winn RD. Current treatment approaches and outcomes in the management of rectal cancer above the age of 80. Curr Oncol. 2021;38:1388–1401. doi:10.3390/curroncol28020132

140. Schiphorst AH, Verweij NM, Pronk A, Hamaker ME. Age-related guideline adherence and outcome in low rectal cancer. Dis Colon Rectum. 2014;57:967–975. doi:10.1097/DCR.0000000000000145

141. Guillerme F, Clavier JB, Nehme-Schuster H, et al. Age impacts the pattern of care for elderly patients with rectal cancer. Int J Colorectal Dis. 2014;29:157–163. doi:10.1007/s00384-013-1778-6

142. Rutten H, den Dulk M, Lemmens V, et al. Survival of elderly rectal cancer patients not improved: analysis of populations based data on the impact of TME surgery. Eur J Cancer. 2007;43:2295–2300. doi:10.1016/j.ejca.2007.07.009

143. Choi Y, Kim JH, Kim JW, et al. Preoperative chemoradiotherapy for elderly patients with locally advanced rectal cancer – a real world outcome study. Jpn J Clin Oncol. 2016;46:1108–1117. doi:10.1093/jjco/hyw126

144. Jiang DM, Raissouni S, Mercer J, et al. Clinical outcomes of elderly patients receiving neoadjuvant chemoradiation for locally advanced rectal cancer. Ann Oncol. 2015;26:2102–2106. doi:10.1093/annonc/mdv331

145. Guimas V, Boustani J, Schipman B, et al. Preoperative chemoradiotherapy for rectal cancer in patients aged 75 years or older.: acute toxicity, compliance with treatment, and early results. Drugs Aging. 2016;33:419–425. doi:10.1007/s40266-016-0367-0

146. Pasetto LM, Friso ML, Pucciarelli S, et al. Rectal cancer neoadjuvant treatment in elderly patients. Anticancer Res. 2006;2006:3913–3923.

147. Hathout L, Maloney-Patel N, Malhotra U, et al. Management of locally advanced rectal cancer in the elderly: a critical review and algorithm. J Gastrointest Oncol. 2018;9:363–376. doi:10.21037/jgo.2017.10.10

148. Wan JF, Zhu J, Li GC, et al. Implications for determining the optimal treatment for locally advanced rectal cancer in elderly patients aged 75 years and older. Oncotarget. 2015;6:30337–30383. doi:10.18632/oncotarget.4599

149. Maas HA, Lemmens VE, Nijhuis PH, et al. Benefits and drawbacks of short-course preoperative radiotherapy in rectal cancer patients aged 75 years and older. Eur J Surg Oncol. 2013;39(10):1087–1093. doi:10.1016/j.ejso.2013.07.094

150. Wang SJ, Hathout L, Malhotra U, et al. Decision-making strategy for rectal cancer management using radiation therapy for elderly or comorbid patients. Int J Radiat Oncol Biol Phys. 2017;100:92644.

151. Appelt AL, Ploen J, Harling H, et al. High-dose chemoradiotherapy and watchful waiting for distal rectal cancer: a prospective observational study. Lancet Oncol. 2015;16(8):919–927. doi:10.1016/S1470-2045(15)00120-5

152. Lezoche E, Baldarelli M, Lezoche G, et al. Randomized clinical trial of endoluminal locoregional resection versus laparoscopic total mesorectal excision for T2 rectal cancer after neoadjuvant therapy. Br J Surg. 2012;99(9):1211–1218. doi:10.1002/bjs.8821

153. Garcia-Aguilar J, Renfro LA, Chow OS, et al. Organ preservation for clinical T2N0 distal rectal cancer using neoadjuvant chemoradiotherapy and local excision (ACOSOG Z6041): results of an open-label, single-arm, multi-institutional, phase 2 trial. Lancet Oncol. 2015;16(15):1537–1546. doi:10.1016/S1470-2045(15)00215-6

154. Verselveld M, de Graaf EJ, Verhoef C, et al. Chemoradiation therapy for rectal cancer in the distal rectum followed by organ-sparing transanal endoscopic resection (CARTS study). Br J Surg. 2015;102:853–860. doi:10.1002/bjs.9809

155. Arezzo A, Arolfo S, Allaix ME, et al. Results of neoadjuvant short-course radiation therapy followed by transanal endoscopic microsurgery for T1-2 N0 extraperitoneal rectal cancer. Int J Radiat Oncol Biol Phys. 2015;92:299–306. doi:10.1016/j.ijrobp.2015.01.024

156. Aumock A, Birnbaum EH, Fleshman JW, et al. Treatment of rectal adenocarcinoma with endocavitary and external beam radiotherapy: results for 199 patients with localized tumors. Int J radiat Oncol Biol Phys. 2011;51:363–370. doi:10.1016/S0360-3016(01)01677-7

157. Gerard JP, Romestaing P, Chapet O. Radiotherapy alone in the curative treatment of rectal carcinoma. Lancet Oncol. 2003;4:158–166. doi:10.1016/S1470-2045(03)01020-9

158. Gerard JP, Frin AC, Doyen J, et al. Organ preservation in rectal adenocarcinoma (T1) T2-T3 Nx M0. Historical overview of the Lyon Sud – nice experience using contact x-ray brachytherapy and external beam radiotherapy for 120 patients. Acta Oncol. 2015;54:545–551. doi:10.3109/0284186X.2014.975840

159. 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. doi:10.1016/S1470-2045(17)30086-4

160. Picardi V, Deodato F, Guido A, et al. Palliative short-course radiation therapy in rectal cancer: a phase 2 study. Int J Radiat Oncol Biol Phys. 2016;95:1184–1190. doi:10.1016/j.ijrobp.2016.03.010

161. Rijkmans EC, Cats A, Nout RA, et al. Endorectal brachytherapy boost after external beam radiation therapy in elderly or medically inoperable patients with rectal cancer: primary outcomes of the Phase 1 HERBERT study. Int J Radiat Oncol Biol Phys. 2017;98:908–917. doi:10.1016/j.ijrobp.2017.01.033

162. UKCCCR Anal Cancer Trial Working Party. Epidermoid anal cancer: results from the UKCCCR randomized trial of radiotherapy alone versus radiotherapy, 5-fluorouracil, and mitomycin. Lancet. 1996;348(9034):1049–1054. doi:10.1016/S0140-6736(96)03409-5

163. Northover J, Glynne-Jones R, Sebag-Montefiore D, et al. Chemoradiation for the treatment of epidermoid anal cancer: 13-year follow-up of the first randomized UKCCR anal cancer trial (ACT I). Br J Cancer. 2010;102:1123–1128. doi:10.1038/sj.bjc.6605605

164. Bartelink H, Roelofson F, Eschwege F, et al. Concomitant radiotherapy and chemotherapy is superior to radiotherapy alone in the treatment of locally advanced anal cancer: results of a phase III randomized trial of the European organization for research and treatment of cancer radiotherapy and gastrointestinal cooperative groups. J Clin Oncol. 1997;15:2040–2049.

165. Foo M, Link E, Leong T, et al. Impact of advancing age on treatment and outcomes of anal cancer. Acta Oncol. 2014;53:909–916. doi:10.3109/0284186X.2013.876513

166. Johnson G, Madeleine MM, Newcomer LM, et al. Anal cancer incidence and survival: the surveillance, epidemiology, and end results experience, 1973–2000. Cancer. 2004;101(2):218. doi:10.1002/cncr.20364

167. Kachnic LA, Winter K, Myerson RJ, et al. RTOG 0529: a phase 2 evaluation of dose-painted intensity modulated radiation therapy in combination with 5-fluorouracil and mitomycin-c for the reduction of acute morbidity in carcinoma of the anal canal. Int J Radiat Oncol Biol Phys. 2013;86(1):2733. doi:10.1016/j.ijrobp.2012.09.023

168. Geltzeiler CB, Tsikitis VL, Kim JS, et al. Variation in the use of chemoradiotherapy for stage II and III anal cancer: analysis of the national cancer data base. Ann Surg Oncol. 2016;23(12):3934–3940. doi:10.1245/s10434-016-5431-9

169. Fallai C, Cerotta A, Valvo F, et al. Anal carcinoma of the elderly treated with radiotherapy alone or with concomitant radio-chemotherapy. Crit Rev Oncol Hematol. 2007;61(3):261–268. doi:10.1016/j.critrevonc.2006.09.003

170. Lestrade L, Bari B, Montbarbon X, et al. Radiochemotherapy and brachytherapy could be the standard treatment for anal canal cancer in elderly patients? A retrospective single-centre analysis. Med Oncol. 2013;30(1):402. doi:10.1007/s12032-012-0402-x

171. Allal AS, Obradovic M, Laurencet F, et al. Treatment of anal carcinoma in the elderly: feasibility and outcome of radical radiotherapy with or without concomitant chemotherapy. Cancer. 1999;85(1):26–31. doi:10.1002/(SICI)1097-0142(19990101)85:1<26::AID-CNCR4>3.0.CO;2-0

172. Valentini V, Morganti AG, Luzi S, et al. Is chemoradiation feasible in elderly patients? A study of 17 patients with anorectal carcinoma. Cancer. 1997;80(8):1387–1392. doi:10.1002/(SICI)1097-0142(19971015)80:8<1387::AID-CNCR4>3.0.CO;2-C

173. Daly JE, Sebjornson S, Leh S, et al. Multimodal therapy is feasible in elderly anal cancer patients. Acta Oncol. 2017;56:81–87. doi:10.1080/0284186X.2016.1244356

174. Claren A, Doyen J, Falk AT, et al. Results of age-dependent anal canal cancer treatment: a single centre retrospective study. Dig Liver Dis. 2014;46(5):460–464. doi:10.1016/j.dld.2014.01.004

175. Saarilahti K, Arponen P, Vaalavirta L, et al. Chemoradiotherapy of anal cancer is feasible in elderly patients: treatment results of mitomycin-5-Fu combined with radiotherapy at Helsinki University central hospital 1992–2003. Acta Oncol. 2006;45(6):736–742. doi:10.1080/02841860600849075

176. Flam M, John M, Pajak TF, et al. Role of mitomycin in combination with fluorouracil and radiotherapy and of salvage chemoradiation in the definitive nonsurgical treatment of epidermoid carcinoma of the anal canal: result of a phase III randomized intergroup study. J Clin Oncol. 1996;14:2527–2539. doi:10.1200/JCO.1996.14.9.2527

177. Ajani JA, Winter KA, Gunderson LL, et al. Fluorouracil, mitomycin, and radiotherapy vs fluorouracil, cisplatin, and radiotherapy for carcinoma of the anal canal: a randomized controlled trial. JAMA. 2008;299:1914–1921. doi:10.1001/jama.299.16.1914

178. Charnley N, Choudhury A, Chesser P, et al. Effective treatment of anal cancer in the elderly with low-dose chemoradiotherapy. Br J Cancer. 2005;92:1221–1225. doi:10.1038/sj.bjc.6602486

179. Street W. Cancer facts & figures 2018. American Cancer Society. Available from: https://onlinelibrary.wiley.com/page/journal/15325415/homepage/forauthors.html. AccessedJanuary 17, 2019.

180. Venkatesulu BP, Mallick S, Rath GK. Patterns of care of cervical cancer in the elderly: a qualitative literature review. J Geriatr Oncol. 2017;8:108–116. doi:10.1016/j.jgo.2016.12.004

181. Darlin L, Borgfeldt C, Widen E, Kannisto P. Elderly women above screening age diagnosed with cervical cancer have a worse prognosis. Anticancer Res. 2014;34:5147–5151.

182. Wright JD, Gibb RK, Geevarghese S, et al. Cervical carcinoma in the elderly: an analysis of patterns of care and outcome. Cancer. 2005;103:85–91. doi:10.1002/cncr.20751

183. Sharma C, Deutsch I, Horowitz DP, et al. Patterns of care and treatment outcomes for elderly women with cervical cancer: cervical cancer in the elderly. Cancer. 2012;118:3618–3626. doi:10.1002/cncr.26589

184. Gao Y, Ma J-L, Gao F, Song L-P. The evaluation of older patients with cervical cancer. Clin Interv Aging. 2013;8:783–788. doi:10.2147/CIA.S45613

185. Boussios S, Seraj E, Zarkavelis G, et al. Management of patients with recurrent/advanced cervical cancer beyond first line platinum regimens: where do we stand? A literature review. Crit Rev Oncol Hematol. 2016;108:164–174. doi:10.1016/j.critrevonc.2016.11.006

186. Monk BJ, Sill MW, McMeekin DS, et al. Phase III trial of four cisplatin-containing doublet combinations in stage IVB, recurrent, or persistent cervical carcinoma: a gynecologic oncology group study. J Clin Oncol. 2009;27:4649–4655. doi:10.1200/JCO.2009.21.8909

187. Goodheart M, Jacobson G, Smith B, Zhou L. Chemoradiation for invasive cervical cancer in elderly patients: outcomes and morbidity. Int J Gynecol Cancer. 2008;18:95–103. doi:10.1111/j.1525-1438.2007.00967.x

188. Eggemann H, Ignatov T, Geyken CH, Seitz S, Ignatov A. Management of elderly women with cervical cancer. J Cancer Res Clin Oncol. 2018;144:961–967. doi:10.1007/s00432-018-2617-5

189. Yoshida K, Sasaki R, Nishimura H, et al. Radiotherapy for Japanese elderly patients with cervical cancer: preliminary survival outcomes and evaluation of treatment-related toxicity. Arch Gynecol Obstet. 2011;284:1007–1014. doi:10.1007/s00404-010-1777-6

190. Mitchell PA, Waggoner S, Rotmensch J, Mundt AJ. Cervical cancer in the elderly treated with radiation therapy. Gynecol Oncol. 1998;71:291–298. doi:10.1006/gyno.1998.5180

191. Hou P, Hsieh C, Wei M, Hsiao S, Shueng P. Differences in treatment outcomes and prognosis between elderly and younger patients receiving definitive radiotherapy for cervical cancer. Int J Environ Res Public Health. 2020;17(12):4510. PMID: 32585933. doi:10.3390/ijerph17124510

192. Shimamoto K, Saito T, Kitade S, et al. A study of treatments and outcomes in elderly women with cervical cancer. Eur J Obstet. 2018;228:174–179. doi:10.1016/j.ejogrb.2018.06.032

193. Kidd EA, Siegel BA. Clinical outcomes of definitive intensity-modulated radiation therapy with fluorodeoxyglucose-positron emission tomography simulation in patients with locally advanced cervical cancer. Int J Radiat Oncol Biol Phys. 2010;77:1085–1091. doi:10.1016/j.ijrobp.2009.06.041

194. Seppenwoolde Y, Majercakova K, Buschmann M, Elke Dörr AE, Sturdza MP, Schmid RP. Dietmar Georg early morbidity and dose–volume effects in definitive radiochemotherapy for locally advanced cervical cancer: a prospective cohort study covering modern treatment techniques. Strahlenther Onkol. 2021;197(6):505–519. doi:10.1007/s00066-021-01781-6

195. EAU Guidelines. Edn. presented at the EAU annual congress Amsterdam. ISBN 978-9492671-16-5; 2022. Available from: https://uroweb.org/guidelines/prostate-cancer. Accessed April 24, 2023.

196. Schröder FH, Hugosson J, Roobol MJ, et al. Screening and prostate cancer mortality: results of the European Randomised Study of Screening for Prostate Cancer (ERSPC) at 13 years of follow-up. Lancet. 2014;384(9959):2027–2035. PMID: 25108889; PubMed Central PMCID: PMCPMC4427906. doi:10.1016/s0140-6736(14)60525-0

197. Wilt TJ, Jones KM, Barry MJ, et al. Follow-up of prostatectomy versus observation for early prostate cancer. N Engl J Med. 2017;377(2):132–142. PubMed PMID: 28700844. doi:10.1056/NEJMoa1615869

198. Hamdy FC, Donovan JL, Lane JA, et al. 10-year outcomes after monitoring, surgery, or radiotherapy for localized prostate cancer. N Engl J Med. 2016;375(15):1415–1424. doi:10.1056/NEJMoa1606220

199. NCCN Clinical Practice. Guidelines in oncology (NCCN Guidelines®) prostate cancer, version 4.2022 - May 10, 2022; 2022. Available from: https://www.nccn.org/professionals/physician_gls/pdf/prostate.pdf. Accessed April 24, 2023.

200. Droz JP, Albrand G, Gillessen S, et al. Management of prostate cancer in elderly patients: recommendations of a task force of the international society of geriatric oncology. Eur Urol. 2017;72(4):521–531. PubMed PMID: 28089304. doi:10.1016/j.eururo.2016.12.025

201. Widmark A, Gunnlaugsson A, Beckman L, et al. Ultra-hypofractionated versus conventionally fractionated radiotherapy for prostate cancer: 5-year outcomes of the HYPO-RT-PC randomised, non-inferiority, phase 3 trial. Lancet. 2019;394(10196):385–395. PubMed PMID: 31227373. doi:10.1016/s0140-6736(19)31131-6

202. Brand DH, Tree AC, Ostler P, et al. Intensity-modulated fractionated radiotherapy versus stereotactic body radiotherapy for prostate cancer (PACE-B): acute toxicity findings from an international, randomised, open-label, phase 3, non-inferiority trial. Lancet Oncol. 2019;20(11):1531–1543. PubMed PMID: 31540791; PubMed Central PMCID: PMCPMC6838670. doi:10.1016/s1470-2045(19)30569-8

203. Jackson WC, Silva J, Hartman HE, et al. Stereotactic body radiation therapy for localized prostate cancer: a systematic review and meta-analysis of over 6000 patients treated on prospective studies. Int J Radiat Oncol Biol Phys. 2019;104(4):778–789. PubMed PMID: 30959121; PubMed Central PMCID: PMCPMC6770993. doi:10.1016/j.ijrobp.2019.03.051

204. Parker CC, James ND, Brawley CD, et al. Radiotherapy to the primary tumour for newly diagnosed, metastatic prostate cancer (STAMPEDE): a randomised controlled phase 3 trial. Lancet. 2018;392(10162):2353–2366. PubMed PMID: 30355464; PubMed Central PMCID: PMCPMC6269599. doi:10.1016/s0140-6736(18)32486-3

205. Ost P, Reynders D, Decaestecker K, et al. Surveillance or metastasis-directed therapy for oligometastatic prostate cancer recurrence: a prospective, randomized, multicenter phase II trial. J Clin Oncol. 2018;36(5):446–453. PubMed PMID: 29240541. doi:10.1200/jco.2017.75.4853

206. Boeve LMS, Hulshof M, Vis AN, et al. Effect on survival of androgen deprivation therapy alone compared to androgen deprivation therapy combined with concurrent radiation therapy to the prostate in patients with primary bone metastatic prostate cancer in a prospective randomised clinical trial: data from the HORRAD trial. Eur Urol. 2019;75(3):410–418. PubMed PMID: 30266309. doi:10.1016/j.eururo.2018.09.008

207. Amini A, Morris L, Ludmir EB, et al. Radiation therapy in older adults with cancer: a critical modality in geriatric oncology. J Clin Oncol. 2022;40:1806–1811. doi:10.1200/JCO.21.02656

208. Hamaker M, JOnker JM, de Rooij S, et al. Frailty screening methods for predicting outcome of a comprehensive geriatric assessment in elderly patients with cancer: a systematic review. Lancet Oncol. 2012;13:e437–e444. doi:10.1016/S1470-2045(12)70259-0

209. Hurria A, Mohile S, Gajra A, et al. Validation of a prediction tool for chemotherapy toxicity in older adults with cancer. J Clin Oncol. 2016;34:2366–2371. doi:10.1200/JCO.2015.65.4327

210. Shinde A, Vazquez J, Novak J, et al. The role of comprehensive geriatric assessment in radiation oncology. J Geriatr Oncol. 2020;11:194–196. doi:10.1016/j.jgo.2019.08.012

211. Szumacher E, Sattar S, Neve M, et al. Use of comprehensive geriatric assessment and geriatric screening for older adults in the radiation oncology setting: a systematic review. Clin Oncol. 2018;30:578–588. doi:10.1016/j.clon.2018.04.008

212. Scher K, Hurria A. Under-representation of older adults in cancer registration trials: known problem, little progress. J Clin Oncol. 2012;30:2036–2038. doi:10.1200/JCO.2012.41.6727

213. Hurria A, Dale W, Mooney M, et al. Designing therapeutic clinical trials for older and frail adults with cancer: U13 conference recommendations. J Clin Oncol. 2014;32:2587–2594. doi:10.1200/JCO.2013.55.0418

214. Townsley CA, Chan KK, Pond GR, et al. Understanding the attitudes of the elderly towards enrolment into cancer clinical trials. BMC Cancer. 2006;6:34. doi:10.1186/1471-2407-6-34

215. Townsley CA, DSElby R, Siu LL. Systematic review of barriers tot he recruitment of older patients with cancer onto clinical trials. J Clin Oncol. 2005;23(13):3112–3124. doi:10.1200/JCO.2005.00.141

216. Dao D, Zemla T, Jatoi A, et al. Older-patient-specific cancer trials: a pooled analysis of 2277 patients (A151715). Oncologist. 2019;24:e284–e291.

217. Jatoi A, Hillman S, Stella P, et al. Should elderly non-small-cell lung cancer patients be offered older-age-specific trials? Results of a pooled analysis from the North central cancer treatment group. J Clin Oncol. 2005;23:9113–9119.

Creative Commons License © 2023 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, v3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.