Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Exploring prehabilitation interventions for patients with gynaecological cancer undergoing radiotherapy: A scoping review

  • Elizabeth McGladrigan ,

    Roles Conceptualization, Formal analysis, Investigation, Methodology, Visualization, Writing – original draft, Writing – review & editing

    b.mcgladrigan@lancaster.ac.uk

    Affiliation Lancaster Medical School, Faculty of Health and Medicine, Lancaster University, Lancaster, Lancashire, United Kingdom

  • Elizabeth Wrench,

    Roles Investigation, Validation, Writing – review & editing

    Affiliation Lancaster Medical School, Faculty of Health and Medicine, Lancaster University, Lancaster, Lancashire, United Kingdom

  • Ewan Dean,

    Roles Investigation, Validation, Writing – review & editing

    Affiliation Lancaster Medical School, Faculty of Health and Medicine, Lancaster University, Lancaster, Lancashire, United Kingdom

  • Aneurin O’Neil,

    Roles Investigation, Validation, Writing – review & editing

    Affiliation UK Centre for Ecology & Hydrology, Lancaster Environment Centre, Lancaster, Lancashire, United Kingdom

  • Lisa Ashmore ,

    Contributed equally to this work with: Lisa Ashmore, Christopher Gaffney

    Roles Conceptualization, Methodology, Supervision, Writing – review & editing

    Affiliation Lancaster Medical School, Faculty of Health and Medicine, Lancaster University, Lancaster, Lancashire, United Kingdom

  • Christopher Gaffney

    Contributed equally to this work with: Lisa Ashmore, Christopher Gaffney

    Roles Conceptualization, Methodology, Supervision, Writing – review & editing

    Affiliation Lancaster Medical School, Faculty of Health and Medicine, Lancaster University, Lancaster, Lancashire, United Kingdom

Abstract

Purpose

Radiotherapy imposes a significant physiological and psychological burden on gynaecological cancer patients. Prehabilitation is being increasingly used to prepare individuals for cancer treatment and improve their well-being and resilience. Whilst prehabilitation has demonstrated benefit for individuals undergoing cancer surgery, the structure, role and implementation of prehabilitation prior to radiotherapy are poorly defined and relatively unexplored. This scoping review aims to provide a comprehensive overview of the current literature regarding prehabilitation interventions for individuals with gynaecological cancer undergoing radiotherapy.

Methods

This review was conducted following the gold-standard Joanna Briggs Institute guidelines for scoping reviews. Literature searches were completed in October 2024 across: the Allied and Complementary Medicine Database; British Nursing Index; Cumulative Index to Nursing and Allied Health Literature; Cochrane library (Controlled trials and systematic reviews); Embase; Medical Literature Analysis and Retrieval System Online; and the Psychological Information Database. Grey literature searches were conducted via Google Scholar, Overton.io, and Trip Pro Medical Database.

Results

Ninety records met the inclusion criteria, pertaining to 56 studies. Cervical cancer was the most represented gynaecological cancer type across studies. A small number of multimodal prehabilitation studies were identified (n = 4). Studies evaluating unimodal interventions were more common, with nutritional interventions (n = 24) being the most frequent, followed by psychological (n = 22) and physical exercise (n = 6) interventions. There was considerable variation across studies in respect to intervention initiation, duration, delivery and outcome measures.

Conclusions

The physiological and psychological impacts of cancer diagnosis and treatment are closely entwined. Further development of multimodal prehabilitation to cohesively address these is an important area for future research. Studies evaluating exercise interventions are relatively unexplored in this patient population and the potential barriers to engagement must be considered. Future research should focus on complete and transparent reporting of interventions, with input from those with lived experience, and adopting a standardised set of outcome measures reported across all trials.

Introduction

Radiotherapy is a mainstay in the treatment of gynaecological malignancies and may be given as a primary treatment, in combination with chemotherapy, and in a neo-adjuvant or adjuvant setting, e.g., for patients receiving surgery [1]. Highly conformal external-beam radiotherapy and brachytherapy techniques have led to reduced morbidity and mortality [2]. However, these treatments still impose a significant physiological and psychosocial burden on patients [1,3].

Radiation-induced cellular death and injury, key to its anti-neoplastic mechanisms, cause adverse effects such as urinary tract and gastrointestinal injury which may present as radiation cystitis, diarrhoea, frequency, urgency, nausea, or bloating [1,2]. Whilst the acute toxicities generally resolve within 12 weeks of completing radiotherapy, many patients experience chronic side-effects that persist months or years later [2,4]. Most radiotherapy side effects are localised to the treatment area, yet fatigue is a commonly reported symptom that may occur acutely or persist long-term [5]. Cancer-related fatigue is characterised by physical, emotional, and/or cognitive tiredness, exhaustion, or weakness related to cancer and its treatment which can significantly alter daily living [5]. Disease and treatment-related burden can also profoundly impact a person’s sexual well-being. Brachytherapy, a technique requiring the insertion of an applicator into the vagina and uterus for prolonged periods, has been highlighted as a source of trauma and distress for gynaecological cancer survivors resulting from factors such as pain, vulnerability, anxiety, and loss of autonomy [6,7]. Unsurprisingly, the acute and chronic side-effects experienced by gynaecological cancer survivors have a profound impact on quality of life, affecting important aspects such as intimacy, social activities, and employment [3,8].

The period between diagnosis and treatment provides an opportunity for early engagement with prehabilitation activities that are designed to optimise patients’ physiological and psychological well-being and resilience [9]. Whilst unimodal exercise regimens have been used by some prior to surgery, prehabilitation is ideally delivered via a multimodal regimen comprising targeted exercise, nutrition and psychological interventions and support [9,10].

Prehabilitation benefits patients undergoing cancer surgery, including reduced complication severity and length of hospital stay [1113] and prehabilitation interventions for gynaecological cancer patients prior to surgery have been explored in a recent scoping [14] and systematic review [15]. However, the evidence base for prehabilitation is highly variable and there is a comparative dearth of research for prehabilitation prior to non-surgical cancer treatments such as radiotherapy [16]. Additionally, the term prehabilitation is not used ubiquitously in the literature and has only become more frequently used in recent years [9] making it difficult to determine the true extent of the evidence base relating to prehabilitation in this population. Unlike surgery, radiotherapy treatment may be given over a period of weeks providing additional time for patients to participate in health optimising activities. As such, the timing and duration of prehabilitation is less clearly defined for individuals scheduled to undergo radiotherapy treatment. Furthermore, chemotherapy, surgery, radiotherapy, and the various combinations of these treatments all pose unique challenges that necessitate tailored support, meaning prehabilitation developed for surgical cohorts may not be the best approach for those treated with radiotherapy [17]. Therefore, it is of interest to explore prehabilitation in the context of individuals with gynaecological malignancies receiving radiotherapy. In particular, establishing: (1) what prehabilitation interventions are being delivered, (2) their underpinning rationale, (3) the point of delivery and duration of these interventions, and (4) what outcome measures are being reported within the literature. The objective of this scoping review is to provide a comprehensive overview of the current literature regarding prehabilitation interventions for gynaecological cancer patients undergoing radiotherapy.

Methods

Scoping reviews are a form of evidence synthesis ideal for identifying and mapping the breadth of evidence available for a given topic, clarifying key concepts and highlighting gaps in the literature [18]. They are particularly useful for examining emerging evidence where it is less clear if more precise questions, such as those regarding efficacy, can be asked and suitably addressed in a systematic review [19]. Unlike systematic reviews, the exploratory nature of a scoping review allows for broader research questions and is not intended to assess effectiveness or validity of studies [19].

The methodology for this scoping review was conducted following Joanna Briggs Institute (JBI) guidelines for scoping reviews [20] and reported in accordance with the Preferred Reporting Items for Systematic reviews and Meta-Analyses extension for Scoping Reviews (PRISMA-ScR) checklist [21] (S1 Checklist). The study protocol outlining the objectives, inclusion criteria and methods was registered a priori with the Open Science Framework (https://osf.io/jgrv3). This study was a scoping review of the literature and did not require ethical approval.

Preliminary searches of the Cochrane Database of Systematic Reviews, Open Science Framework, JBI Evidence Synthesis and Google were completed in December 2023 and no current or in-progress reviews on this topic for the specified patient population were identified. A subsequent search was conducted in January 2024, following the identification of a recently published scoping review addressing prehabilitation for radiotherapy. However, this review included studies evaluating prehabilitation in any adult patient undergoing radiotherapy, not limited by tumour site, and there were no studies specifically addressing gynaecological cancer. The research questions and inclusion criteria of this proposed review are sufficiently different to those addressed by Flores et al [22] to warrant a further scoping review.

Inclusion/exclusion criteria

As advised for scoping reviews, the inclusion criteria was developed using the Population, Concept and Context (PCC) framework [20].

We included prospective or retrospective studies that reported on or evaluated (1) prehabilitation for (2) adult (≥18 years old), female patients with a gynaecological malignancy prior to or during radiotherapy, with or without chemotherapy, (3) in any setting where care is provided or an intervention can be delivered to this population. We defined prehabilitation as an intervention prior to or during radiotherapy where unimodal addressed either physical, psychological or nutritional well-being and multimodal delivery was the combination of at least two different categories, e.g., physical and psychological. Preliminary searches indicated there were only a small number of studies evaluating multimodal interventions, as such the authors agreed that inclusion of unimodal prehabilitation interventions was necessary to suitably address the research questions and explore the breadth of prehabilitation literature in this population. Despite prehabilitation sitting within the broader context of health improvement, this review did not include studies focused primarily on smoking cessation, alcohol reduction or medication management, as they as are not encompassed by the primary prehabilitation interventions – tailored physical exercise, nutritional support and psychological support.

Grey literature, conference proceedings, clinical trial protocols and records; and peer-reviewed qualitative, quantitative or mixed-methods publications, were all considered for inclusion. This breadth aligns with a scoping methodology and was considered important to fully gauge the variety of studies evaluating interventions for this population and the various aspects of applying these clinically. Social media posts, animal studies, blogs, and podcasts were excluded. Any relevant systematic or scoping reviews were excluded and hand-searched for primary studies that met the above criteria. Sources were excluded if they were not available in English language due to limited resources available for translation. The authors agreed that studies evaluating prehabilitation interventions in a broader population (e.g., a mixed population of individuals receiving radiotherapy, chemotherapy and/or surgery) would be considered for inclusion if there was sufficient separation in the results for the population of interest, in addition to ongoing clinical trials, as they could provide valuable insight into the review questions.

Search strategy

An initial search was conducted in Medical Literature Analysis and Retrieval System Online (MEDLINE) and Cumulative Index to Nursing and Allied Health Literature (CINAHL) with an adapted version of the terms used by Saggu et al [14]. The full search strategy was then developed using an analysis of text words contained within the titles and abstracts of retrieved papers and any index terms used to describe the articles. This search strategy was adapted and applied to each database including: Allied and Complementary Medicine Database (AMED) (Ovid and ProQuest Dialog); British Nursing Index (BNI) (ProQuest); CINAHL (EBSCO); Cochrane library (Controlled trials and systematic reviews); Embase (Ovid); MEDLINE (EBSCO); Psychological Information Database (PsycINFO) (EBSCO). Each search strategy was then peer reviewed by a research librarian and modified according to feedback. A search for grey literature was also conducted via Google Scholar, Overton.io, and Trip Pro Medical Database. Final searches were completed in October 2024. The full search strategies are available in the supplementary materials (S1 Table and S2 Table). Included sources and identified reviews were hand-searched for additional eligible articles.

Study selection

All identified records were uploaded to EndNote (Clarivate Analytics, PA, USA) and duplicates removed. The remaining abstracts were then uploaded to Rayyan (Qatar Computing Research Institute, Doha, Qatar) for title and abstract screening by the primary reviewer (EM) and the secondary screening was divided equally between two independent reviewers (ED and EW). The full-text screening for any potentially relevant sources were then screened in duplicate by the reviewers (EM, EW, ED or AON). Any conflicts were resolved by discussion between the relevant reviewers. A small number of sources (n = 8) were unable to be uploaded to Rayyan, the title and abstracts of each source were reviewed against the inclusion criteria and were found not to be relevant to the review.

Data charting

Relevant data were charted using a standardised form developed for this study by a reviewer (EM). This data extraction form was developed in accordance with JBI guidance [20] and piloted across different sources by two reviewers (EM and EW). Where multiple sources for the same studies were identified, the records were collated for data extraction to minimise risk of double counting [23]. A random sample (n = 8) were then verified by independent reviewers (ED or AON). Data items were extracted as published, including: title, author, year of publication, aims/purpose, population, study design, intervention type, comparator; and outcome measures, along with items from the Template for Intervention Description and Replication (TIDieR) checklist [24]. In the case of ongoing trials, all data items were extracted from available clinical trial records or published study protocols including planned intervention components and outcome measures. Authors were contacted for missing or additional information with a subsequent follow-up email after a minimum of two weeks, as necessary. Critical appraisal of the quality of included studies was not performed as this is not generally recommended for scoping reviews [19,21]. Data are presented in tabular and diagrammatic formats accompanied by a descriptive summary in a manner that aligns with the review objectives. Figures were prepared in GraphPad Prism 10 (GraphPad Software 2365 Northside Dr. Suite 560 San Diego, CA 92108) and the GNU Image Manipulation Programme (GIMP 2.10.38, gimp.org), unless otherwise stated.

Results

Study selection and descriptive characteristics of the studies

The systematic search identified 8,921 records (Fig 1). Following deduplication in EndNote, 6,835 titles and abstracts were screened against the inclusion criteria. A total of 234 records were then further assessed for eligibility, 90 were included pertaining to 56 studies.

The characteristics and details of the included studies are summarised in Tables 16. The 56 studies were categorised as multimodal (n = 4) [2533], physical/exercise (n = 6) [3442], psychological (n = 22) [43–79], and nutritional (n = 24) [80–114]. The studies were conducted in Australia (n = 4), Brazil (n = 2), Canada (n = 2), Chile (n = 1), China (n = 5), England (n = 1), France (n = 2), Finland (n = 1), Germany (n = 2), Italy (n = 1), India (n = 8), Japan (n = 1), Republic of Korea (n = 1), Mexico (n = 4), Myanmar (n = 1), Norway (n = 2), Spain (n = 5), Sweden (n = 2), Thailand (n = 2), Turkey (n = 1), and the USA (n = 8). Several ongoing clinical trials were included (n = 10) [28,36,39,43,51,72,73,91,102,108] and the earliest included record was published in 1958 [104]. Sources included peer-reviewed journal articles (n = 50), conference abstracts (n = 13), clinical trial records (n = 20), peer-reviewed study protocols (n = 4), a pre-print author manuscript (n = 1) and dissertations (n = 2). Cervical cancer was the most highly represented gynaecological cancer type across study populations, however, studies also included endometrial, uterine, vaginal, vulval, ovarian and fallopian tube malignancies (Fig 2).

thumbnail
Table 2. Characteristics of included ongoing clinical trials.

https://doi.org/10.1371/journal.pone.0319518.t002

thumbnail
Table 3. Overview of the studies evaluating multimodal prehabilitation interventions using a modified TIDieR checklist.

https://doi.org/10.1371/journal.pone.0319518.t003

thumbnail
Table 4. Overview of the studies evaluating unimodal physical exercise interventions using a modified TIDieR checklist.

https://doi.org/10.1371/journal.pone.0319518.t004

thumbnail
Table 5. Overview of the studies evaluating unimodal psychological interventions using a modified TIDieR checklist.

https://doi.org/10.1371/journal.pone.0319518.t005

thumbnail
Table 6. Overview of the studies evaluating unimodal nutritional interventions using a modified TIDieR checklist.

https://doi.org/10.1371/journal.pone.0319518.t006

thumbnail
Fig 2. Number of studies including different gynaecological cancers by type.

https://doi.org/10.1371/journal.pone.0319518.g002

What interventions are being reported in the literature?

Multimodal.

Studies evaluating multimodal prehabilitation utilised multidisciplinary teams to address the complex needs of their target population including: dieticians; physiotherapists; radiation oncologists; and prehabilitation exercise specialists (Table 3). Some of the studies also utilised early referrals or input from mental health services or social work teams within their prehabilitation interventions [25,28,32]. Kaliamurthi et al. [27] provided cervical cancer patients with a programme consisting of counselling, nutritional support and physiotherapy. Only one study did not specifically mention including a physical exercise element, this was targeted to patients undergoing brachytherapy so the intervention focused on carbohydrate loading, pain control and psychosocial support [32]. RadBone [28] is an ongoing clinical trial that utilises the resources of an established prehabilitation service to provide tailored exercise, nutritional and psychological support in combination with a targeted musculoskeletal health package.

Unimodal – Physical/Exercise.

Of the six studies reporting on a unimodal physical exercise intervention, only two implemented a physical activity programme that resemble those more commonly used in prehabilitation prior to surgery. Of these, one was a case study providing a completely tailored exercise intervention for an individual during treatment [37] and the other is an ongoing clinical trial involving a variety of patient groups undergoing radiotherapy using an activity tracker-based programme [36]. Two single-arm (pre-post) interventional studies focused on teaching pelvic floor muscle exercises (PFMEs) and encouraging participants to perform these exercises at home facilitated by educational materials and/or pre-recorded instructions [35,38]. The final category of physical interventions was targeted towards post-surgical patients prior to adjuvant treatment. These studies focus on prophylactic complex physiotherapy including manual lymphatic drainage, skin care, use of compression stockings, and functional exercises that could be performed at home [34,39].

Unimodal – Psychological.

The majority of studies addressing psychological wellbeing (n = 17) [43,51,54,56,5865,6769,71,72] incorporated relaxation and stress reduction techniques such as controlled breathing exercises, aromatherapy, progressive muscle relaxation, guided imagery, videos, music, meditation, mindfulness, and stress management coaching. Three of which also evaluated the effect of the complementary therapies: healing touch [54], reiki [43], and reflexology with aromatherapy [62]. In the PeNTAGOn trial, and an earlier pilot study [69], Schofield et al. [68] used both specialist nurses and matched peers to deliver a tailored psychoeducational intervention that involved elements of relaxation combined with timely delivery of information and support, to address patients’ concerns and psychosexual needs. Kpoghomou et al. [70] and Varre et al. [53] also provided nurse-led counselling to support and manage patients’ psychological and psychosexual wellbeing, whereas, Jeffries et al. [52] favoured a small group approach with taught elements and peer discussion facilitated by healthcare professionals. In their ongoing trial, Oluloro et al. [73] are evaluating the implementation of virtual support groups hosted by a trained peer-support and specialist co-facilitators to promote group conversation and discussion of structured topics including mental well-being, finances and family dynamics. The support group intervention is also being compared against enhanced care including more targeted written information and a 1:1 support intervention with a peer trained by the Endometrial Cancer American Network for African-Americans [73].

Unimodal – nutritional.

Twelve (50%) of the studies evaluating nutritional interventions were randomised, placebo-controlled trials. Several of these studies focused on the introduction of probiotics or synbiotics (prebiotics and probiotics) into the participants’ diets. The strain and combination of bacteria used, varied between studies including: Lactobacillus casei DN-114 001 with standard starters Streptococcus thermophilus and Lactobacillus delbrueckii subsp. Bulgaricus [88]; Lactobacillus acidophilus LA-5 and Bifidobacterium animalis subsp. lactis BB-12 [93]; Lactiplantibacillus plantarum HEAL9 and 299 [81]; Lactobacillus acidophilus with Bifidobacterium bifidum [82]; Lactobacillus acidophilus (NCDO 1748) [97]; and Lactobacillus acidophilus NCFM, Bifidobacterium lactis Bi-07 with blue agave inulin [85]. Garcia-Peris et al. [86] instead opted for a mixture of prebiotics, inulin and fructo-oligosaccharide, to stimulate proliferation of Lactobacillus spp and Bifidobacterium spp populations.

Two studies provided patients with Omega-3 supplements, with Chitapanarux et al. [83] additionally including arginine and glutamine into their supplement regimen. Whereas, Muecke et al. [96] and Yang et al. [106] provided selenium supplements to cervical cancer patients. Some focused on modifying factors through dietary changes, such as no or low-residue (little to no fibre) [104]; anti-inflammatory [112]; low fat and lactose [110]; or low fermentable oligosaccharides, disaccharides, monosaccharides and polyols (FODMAP) [100] diets. Other studies included glutamine [105], hydrolysed rice bran [90], oral resistant starch [99], immunonutrient [108] or elemental [84] supplements. One ongoing trial is evaluating the effect of time-restricted eating compared to nutritional counselling during radiotherapy [91,107].

What are the rationales underpinning these interventions?.

Any identified theories and/or rationales for each study are outlined in Tables 3-6. More broadly, multimodal interventions were introduced to improve outcomes and prevent or reduce treatment-related morbidity. Edbrooke et al. [25] identified that gynaecological cancer patients require increased emotional and social support due to high-levels of distress. They highlighted that their target patient population did not meet nutritional and exercise recommendations and had expressed the need for external motivation to drive behavioural changes [25]. Therefore, they used a multidisciplinary team to provide psychological, nutritional and exercise support underpinned by HealthChange® methodologies with features such as pedometers and text prompts to provide additional motivation [25].

Lymphoedema is a chronic process, experienced by many gynaecological cancer survivors, that is associated with physical, psychosocial and financial burden [34,39,115]. Daggez et al. [34] and Zou et al. [39] both utilise a technique that is currently used for the conservative management of lower extremity lymphoedema but introduced it in a prophylactic setting. The exercise program used by Daggez et al. [34] was chosen specifically to induce muscle function and promote lymphatic drainage. Adjuvant radiotherapy is a risk factor for lymphoedema therefore the studies included, or intend to include, post-surgical patients scheduled for radiotherapy. Similarly, as radiotherapy is also considered a risk factor for pelvic floor dysfunction, Jagdish et al. [38] and Sacomori et al. [35] adopted pelvic floor strengthening techniques more commonly used in a rehabilitation setting but initiated them prior to radiotherapy as a preventative measure. Hauth et al. [36] acknowledged that whilst a personal supervised physical activity programme is desirable for prehabilitation, it is not always feasible due to cost, geographical limitations and resources. Instead, they designed a physical activity programme that can be carried out at home, is less resource intensive, and more easily implemented into a busy radiotherapy workflow [36]. The ongoing Onkofit II trial [36] uses a wearable activity tracker to monitor patient activity and provide a motivational element to the intervention. Interestingly, when developing a physical activity programme, Tórtola-Navarro et al. [37] altered their exercise prescription relative to the timing of treatment, reducing the intensity of physical activity in the days following chemotherapy to account for the anticipated increase in side-effects experienced by the patient.

Psychological interventions aimed to reduce stress and help patients cope with the complex psychological burden associated with treatment and disease. Relaxation techniques were favoured by researchers due to their safety, accessibility, low-cost and effectiveness amongst similar populations in the literature [60,63,65,67,71]. De Oliveira Santana et al. [67] used virtual reality technology to provide a more immersive guided-imagery experience that can be applied in a clinical environment to overcome physical barriers of accessing nature. Of the studies focused specifically on patients receiving brachytherapy, interventions promoting relaxation and pain reduction were particularly prevalent due to the high levels of discomfort and distress experienced by patients. Chi et al. [56] introduced combined audiovisual stimulation to modify pain perception and maximise the anxiolytic effects of both music and guided imagery. The music was chosen with consideration of the literature suggesting a slow, constant rhythm with predictable dynamics and harmonic consonance at a tempo, similar to the resting human heart rate, was optimal for relaxation [56]. In a similar study, Lim [71] prioritised music based on individual preference and projected nature videos onto the ceiling to ensure the participant could engage with the intervention whilst lying on the therapy bed. Several studies particularly emphasised the relevance of mind-body connection as a core element of the intervention to promote relaxation [51,59,62,64,67,72]. Tagliaferri et al. [58] took a multidisciplinary approach to assess the needs of brachytherapy patients from different perspectives and developed a number of interventions and recommendations to address the needs and concerns expressed by patients. As an example, they allowed patients to select music and relaxing videos to create a relaxing environment; altered language used to reduce stressful trigger words for patients; provided an information booklet in a timely manner prior to the treatment to facilitate shared decision making; and increased staff presence in the interventional room to decrease feelings of isolation [50,58].

The prevalence of physical and psychosexual concerns following radiotherapy and the complexities of compliance with rehabilitative strategies, such as vaginal dilation, prompted researchers to counteract this with timely intervention strategies. Bergin et al. [69] and Schofield et al. [68] took a psychoeducational approach to improve coping, and reduce the psychological impact of treatment. The nurse-led consultations were included to provide tailored information at relevant timepoints throughout treatment to improve patient recall and involved elements such as radiotherapy department orientation to enhance patients’ sense of preparedness [68,69]. Peer support was a key component of the intervention to help patients make sense of their journey and provide social support [68,69]. Similarly, Jeffries et al. [52] developed a psychoeducational intervention, underpinned by a behaviour change model, involving groups of peers to normalise patient experience and overcome attitudinal barriers to care strategies. Kpoghomou et al. [70] introduced a programme in response to the lack of support patients are currently given regarding their sexual concerns, aiming to reduce side effects through supportive and educational measures.

Many of the nutritional interventions aimed to prevent or reduce the adverse gastrointestinal effects experienced by gynaecological patients, such as radiation enteritis, due to the impact on quality of life and treatment tolerability. Radiotherapy can disrupt the gut microbiota and cause intestinal inflammation and injury. A number of studies cited the positive effects of probiotics in patients with other inflammatory bowel conditions and theorised that probiotics may correct dysbiosis, improve or maintain intestinal barrier function, and/or offer beneficial immunomodulatory effects [81,82,88,93,97]. Whilst probiotic strains varied between studies, generally they were chosen due to safety and/or benefit in previous studies. Dietary changes and recommendations were often implemented to reduce or modify factors that may influence gastrointestinal toxicities. Bye et al. [110] theorised a low-fat, low-lactose diet would reduce diarrhoea during treatment by minimising faecal bile salt excretion and fluid accumulation within the intestinal lumen. However, they also highlighted that inadequate nutrition could impact healing so included compensatory measures and tailored advice to achieve sufficient energy intake and minimise weight loss. More recently, Soto-Lugo and colleagues [100] evaluated a low FODMAP diet to explore if gastrointestinal toxicity could be managed more effectively by modifying multiple factors rather than a single dietary element. Both Aredes et al. [103] and Medina-Jiménez et al. [95] aimed to reduce malnutrition and fat-free mass loss due to their association with treatment toxicity and poor outcomes. Aredes et al. [103] provided patients with nutritional risk omega-3 and nutritional formula supplements whereas Medina-Jiménez et al. [95] opted for a tailored nutritional intervention with individual counselling due to the reported benefits of this approach in other radiotherapy patient populations compared to standard recommendations.

What are the timings and duration of these interventions?.

Interventions were broadly divided between those beginning prior to radiotherapy (external-beam or brachytherapy) (n = 27) and those initiated at the point of or shortly after beginning radiotherapy (n = 29) (Fig 3). Point of intervention and duration was variable and difficult to determine for some studies due to ambiguous reporting. Most spanned the radiotherapy treatment period with some continuing for longer. The timings of individual studies are outlined in Tables 3-6, the duration of the interventions and number of sessions varied depending on factors such as treatment length (e.g., brachytherapy alone compared to chemoradiotherapy) and use of familiarisation periods.

thumbnail
Fig 3. Point of initiation for unimodal and multimodal interventions.

https://doi.org/10.1371/journal.pone.0319518.g003

What outcome measures are being reported within the literature?.

Outcome measures varied both across and within study categories. Quality of life was the most consistent outcome measured across studies. This was assessed by a variety of tools, used alone or in combination, including the European Organisation For Research And Treatment Of Cancer (EORTC) Core Quality of Life Questionnaire QLQ-C30 (n = 11) [27,31,37,39,53,86,88,94,100,103,108], EORTC QLQ-CX24 (cervical cancer module) (n = 3) [27,31,100], EORTC QLQ-EN24 (endometrial cancer module) (n = 1) [100], EORTC QLQ-36 (n = 1) [111], EuroQol (EQ)-5D-3L (n = 1) [103], EQ-5D-5L (n = 1) [28], Cuestionario de Calidad de Vida QL-CA-AFex (CCV) (n = 1) [60], Functional Assessment of Cancer Therapy (FACT) -General subscale (n = 5) [25,36,54,57,68], and FACT-Cervix (n = 1) [63].

Both ENABLE [25] and the ongoing RadBone trial [28] assess feasibility considering recruitment rate, attrition, and study eligibility. The ENABLE study utilised the Patient Generated-Subjective Global Assessment (PG-SGA), bioelectrical impedance, and accelerometery to assess nutritional and physical activity outcomes. Additionally, they included the Physical Activity Questionnaire-Short form and Physical Activity Assessment Inventory to measure patient-reported physical activity and self-efficacy levels respectively. Whilst the PG-SGA is used as part of the tailored nutritional intervention in Prehab4Cancer, the secondary outcome measures in RadBone centre around longitudinal changes including incidence of radiotherapy related insufficiency fractures measured by Magnetic Resonance Imaging (MRI); bone mineral density measured by Dual-Energy X-ray Absorptiometry (DEXA); biochemical markers of bone turnover; and fracture risk using the FRAX® assessment tool [28]. Alongside the EQ-5D-5L, Grigoriadou et al. [28,33] are using the Patient-Reported Outcome Version of the Common Terminology Criteria for Adverse Events (PRO- CTCAE) and the Short Form Musculoskeletal Function Assessment to assess quality of life. Both Kaliamurthi et al. [27] and Andring et al. measured treatment related morbidity taking into consideration factors such as patient reported toxicity, hospital admissions and/or length of stay.

The distinct subgroups within the physical/exercise studies resulted in some shared outcomes. The studies evaluating prophylactic physiotherapy [34,39] both utilise patient-reported symptoms from the Gynecologic Cancer Lymphedema Questionnaire to identify risk and incidence of lower extremity lymphoedema. Pelvic floor strength was a primary outcome for the pelvic floor exercise studies, with Sacomori et al. [35] opting to measure pelvic floor strength using vaginal bidigital evaluation grading with the modified Oxford scale by a physical therapist and Jagdish et al. [38] using a perineometer. Both studies also used the International Consultation on Incontinence Questionnaire-Urinary Incontinence Short Form to assess urinary incontinence. The CTCAE was chosen by Tórtola-Navarro et al. [37] and Hauth et al. [36] to assess patient reported toxicity and adverse events. In the Tórtola-Navarro et al. [37] case study, the main outcome measure was feasibility of the programme determined by patient compliance but they were also interested in functional and exercise capacity changes using the Karnofsky index and both the Six Minute Walk Test and Five Times Sit to Stand Test.

In studies evaluating psychological interventions, common outcome measures included levels of psychological distress, anxiety and/or depression using tools such as: the Distress Thermometer (n = 1) [58]; Patient Reported Outcome Measurement Information System (PROMIS) Emotional-Distress Anxiety Short Form 4a (n = 1) [73]; the Hospital Anxiety and Depression Scale (n = 8) [43,46,53,5860,68,72]; State-Trait Anxiety Inventory (n = 3) [56,62,67]; Six-item Spielberger State Anxiety Scale (n = 1) [43]; the Self-rating Anxiety Scale (n = 2) [63,65]; an 11-point anxiety numeric rating scale (n = 1) [71]; the Self-rating Depression scale (n = 1) [63]; the Beck Depression inventory (n = 1) [65]; and the Center for Epidemiological Studies Depression Scale (n = 1) [54]. Other outcomes included level of fatigue, assessed with the Chalder (n = 1) [61] Piper (n = 1) [63], or visual analogue (n = 1) [72] fatigue scales and pain levels using a numeric (n = 3) [61,62,71] or visual (n = 3) [43,56,72] rating scale. In addition to secondary outcomes of interest such as fatigue and pain levels, Texier and Meignant [72] are utilising a 10-item to perceived stress score to evaluate the impact of an educational physiotherapy yoga on perceived stress in cervical cancer patients undergoing brachytherapy. In the ongoing SISTER study, the primary outcome of interest is treatment completion rate measured by relative dose (expected vs actual dose received at six month follow-up) [73]. As a secondary measure, Oluloro et al. [73] are also evaluating the impact of the virtual support interventions on patient-reported social isolation using the Social Provisions Scale-24 and the PROMIS Short Form Social Isolation 4a.

For nutritional studies, incidence and severity of gastrointestinal toxicities were generally measured by versions of the CTCAE (n = 11) [82,83,86,88,91,93,96,99,100,103,112] and the Radiation Therapy Oncology Group (RTOG) criteria (n = 4) [84,95,99,105]. Additionally, some studies included the Bristol stool chart to monitor change in consistency or incidence of watery stools (n = 5) [80,81,85,86,88] and monitored the administration of intestinal regulators and/or anti-diarrhoeal agents during interventions (n = 7) [81,82,84,88,90,93,97]. A subset of studies (n = 4) [85,86,105,112] monitored faecal calprotectin levels as an indicator of intestinal inflammation whereas others monitored inflammation using markers such as c-reactive protein (n = 2) [80,105]. Body mass loss (n = 2) [100,110], body mass change (n = 4) [82,84,97,112] and change in body composition (n = 2) [95,103] were also frequently used to evaluate the impact of nutritional interventions. Like the ENABLE study, Aredes et al. [103] used the PG-SGA to assess nutritional status. However, they also utilised computed tomography (CT) images to evaluate changes body composition including skeletal muscle quantity and quality. Similarly, in an ongoing trial evaluating an immunonutrition intervention for individuals with nutritional risk, prevalence of malnutrition and sarcopenia are being assessed using a modified Global Leadership Initiative on Malnutrition criteria and the Skeletal Muscle Index respectively [108]. Other outcome measures for this study focus on survival including dose-limiting toxicity-free survival, two-year overall survival, and two-year progression free survival [108].

Discussion

This scoping review identified 56 studies, spanning 66 years, evaluating unimodal or multimodal prehabilitation interventions for patients with gynaecological malignancies treated with radiotherapy. It is not entirely unexpected that cervical cancer was the most highly represented gynaecological cancer type across studies considering it is the 8th most common cancer globally [116] and is more often treated with techniques such as brachytherapy than other tumour sites. Our findings indicate there are fewer studies focusing on multimodal prehabilitation programmes. Although, it is encouraging that ongoing trials like RadBone [28] are able to utilise established prehabilitation services and integrate them into a package tailored to radiotherapy patients. Despite many studies evaluating unimodal interventions, there is a clear imbalance between the categories, with physical exercise interventions being comparatively underrepresented.

The heterogeneity of studies in respect to intervention components, initiation, duration, delivery, outcome measures and the tools used to assess these, complicates future work in the form of a systematic review or meta-analysis to build on our findings. As the scope of this review is very broad, including a wide range of records from various countries spanning six decades, some variation was anticipated. Similar issues were identified by Saggu et al. [14] in their scoping review of prehabilitation interventions for gynaecological cancer patients receiving surgery. This is further compounded by inconsistent and incomplete reporting of interventions. Study heterogeneity and ambiguous reporting are frequently cited limitations of systematic reviews and meta-analyses in similar populations [1,15,117120]. Promisingly, tools such as the TIDieR checklist are being used in more recent studies which has improved the completeness of reporting and replicability of interventions [25]. This is particularly important when considering the rationale of a complex intervention. Despite many authors expressing the need for an intervention to address the burden patients experience, few provided clear theory or rationale underpinning design and delivery. Prehabilitation is understudied in this population therefore comprehensive and transparent reporting is essential to facilitate future intervention development and inform changes in practice [121].

The total number of outcomes measured across studies and tools used to assess these was extensive, and incomplete reporting further impeded data charting. Despite the variation in outcome measures, there were a small number of validated instruments favoured by researchers. The EORTC QLQ-C30 is designed to measure health-related quality of life specifically in individuals with cancer [122]. The frequent use of this instrument is not surprising due to its length of circulation and availability in multiple languages. However, only a small proportion of studies used it alongside the recommended disease or symptom-specific modules. Similarly, the CTCAE provides an internationally recognised standard criteria for the reporting of adverse events in cancer therapy and clinical trials. Although more recently it has been acknowledged that additional use of PRO-CTCAE can provide greater insight into patient’s perception of toxicities which is beneficial to establishing tolerability of an intervention [123]. Several patient-reported outcome measures were also used across studies which highlights growing recognition of the importance of patient experience when developing and evaluating interventions [124]. The agreement of a core outcome set (COS) that are consistently measured by widely-accepted, validated tools will facilitate greater comparison between studies to inform future decisions regarding prehabilitation and this has also been recently acknowledged in surgical populations [125]. A project to agree standardised outcome measures for prehabilitation in cancer is currently being undertaken as part of the Core Outcome Measures in Effectiveness Trials (COMET) initiative [126]. However, it is important that consideration is given to how outcomes of interest and measures of efficacy may vary between different treatment populations [127].

Nutritional interventions in this population currently aim to address malnutrition and/or minimise GI toxicities. Despite advances in radiotherapy resulting in reduced morbidity and mortality, pelvic radiotherapy patients still experience acute and chronic GI toxicities which can impact quality of life and treatment tolerability [1]. Interruptions in treatment due toxicities and increase in overall treatment time impacts radiotherapy efficacy [1,128]. Therefore, nutritional support for gynaecological patients receiving radiotherapy is an important element of prehabilitation. Minimising the impact of radiation-induced dysbiosis and intestinal injury through the introduction of pre- and probiotics was a recurring concept in the literature, yet it is acknowledged more evidence is required to gauge the effectiveness of these interventions [1]. Additionally, malnutrition may occur due to changes in gastrointestinal absorption and digestive functioning. Nutritional interventions such as dietary changes are a proactive approach to addressing GI toxicity and improving nutritional status. However, the observed variation across studies highlights that there is not yet a clear approach for these interventions. Globally, differences in radiotherapy techniques, resources and cultural differences, including dietary preferences, may account for some of the observed variation in nutritional interventions.

The physical exercise interventions discussed in this review differ from those frequently used in prehabilitation for cancer surgery. Both the studies evaluating prophylactic physiotherapy for lymphoedema [34,39] and pelvic floor strengthening [35,38] exercises, highlight how prehabilitation can be adapted to address the unique challenges of a target population. There are several factors which may explain the comparative lack of studies evaluating physical exercise interventions. Prior to radiotherapy, patients have a CT scan to develop a highly conformal plan for treatment delivery. As such, subsequent changes in body mass are avoided as they can alter the planned dose to the target volumes and organs at risk [129]. Acute side effects such as vaginal mucositis, nausea; faecal and urinary urgency; and fatigue can present during radiotherapy treatment [1,2] which may influence patients’ desire and ability to exercise. Moreover, travelling to a radiotherapy department multiple times a week for several weeks restricts patients time to participate in additional activities, frequently cited as one of the major barriers to exercise [130]. In their case study, Tórtola-Navarro et al. [37] were able to deliver a highly tailored physical activity programme with consideration for factors such as delivery of chemotherapy which impacts patients’ general wellbeing and energy levels. In the pilot randomised controlled trial ENABLE [25], healthcare professionals worked with participants to develop a home-based physical activity programme with patient-centred nutritional and activity goals, yet some patients still expressed challenges engaging with the programme whilst navigating the side-effects of treatment.

It is well-established that gynaecological cancer and its subsequent treatment has a profound impact on psychological well-being [3,6,8]. The risk of traumatisation during brachytherapy further strengthens the need for psychological support in prehabilitation for these patients [6,7]. Studies have shown that gynaecological cancer survivors have unmet informational needs exacerbating existing psychological distress including increased anxiety and feelings of embarrassment [131134]. Tailored information, provided in a timely manner, including involvement from both healthcare professionals and peers with lived experience can contribute to engagement, patient satisfaction and feelings of preparedness [135]. It is promising that a several studies identified this as an issue and incorporated elements of patient education, in diverse formats, to address this. The psychological interventions in this review ranged from brief interventions for relaxation during brachytherapy to more complex psychoeducational interventions addressing psychosexual concerns and providing social support. The multifactorial nature of psychological distress in gynaecological cancer patients necessitates a multifaceted approach, providing patients with relevant support at an appropriate time to minimise the impact of treatment-related burden.

Prehabilitation interventions are generally considered to be those delivered between diagnosis and the beginning of acute treatment [136]. This review included studies that introduce interventions both before and during the delivery of radiotherapy, with some spanning the pre-, intra- and post-treatment periods. Prehabilitation is an evolving concept and consideration must be given to each population’s unique needs [127]. As acknowledged by Flores et al. [22], the timing of intervention delivery is influenced by the nature of radiotherapy including its role within multimodal cancer care and delayed development of acute side effects. Therefore, including interventions implemented during radiotherapy is key to understanding prehabilitation in the context of radiotherapy, particularly if multiphasic prehabilitation becomes more prevalent in the future. In the multiphasic framework, prehabilitation is viewed as a health optimising strategy initiated at multiple time points after diagnosis [127]. Prehabilitation interventions are tailored to current and anticipated experiences throughout multimodal cancer care to minimise adverse effects and reduce treatment delay [127]. Prehabilitation for a gynaecological cancer patient prior to surgery may include aerobic exercise, resistance training, psychological support and pre-operative carbohydrate loading [137]. This patient may then go on to receive prehabilitation tailored for adjuvant radiotherapy such as prophylactic complex physiotherapy, nutritional interventions to reduce GI toxicity, pelvic floor strengthening, and psychosexual support including radiotherapy-specific education. If the patient is then scheduled for brachytherapy, they will receive further targeted interventions. As prehabilitation for non-surgical therapies continues to develop, we must examine the boundaries between prehabilitation and rehabilitation. If the end of prehabilitation continues to be defined by the beginning of treatment, there may be a gap for interventions during the treatment period that align with the principles of prehabilitation and go beyond standard supportive care.

The strengths of this scoping review lie in its comprehensive and inclusive approach to exploring a relatively understudied area. By including studies evaluating unimodal interventions, this review provides a holistic summary of the evidence base and offers invaluable insight into the components, timing and rationale of prehabilitation interventions in this population. Additionally, the broad inclusion criteria ensures that this review captures a wide range of evidence, including emerging research and recent innovations. This inclusive methodology strengthens the relevance and applicability of these findings to inform future research.

There are only a handful of studies evaluating multimodal prehabilitation in this population and these are limited to smaller studies and feasibility trials with limited published results. Whilst such studies provide valuable insight, larger randomised controlled trials are required in the future to strengthen the evidence base for prehabilitation in individuals with gynaecological malignancies undergoing radiotherapy. Future studies should prioritise complete and transparent reporting of interventions and adoption of a COS informed by multiple-stakeholders to be reported as a minimum across all cancer prehabilitation trials. This will reduce ambiguity and observed heterogeneity in the literature and facilitate data synthesis to establish efficacy and inform changes in practice and policy. Physical exercise interventions remain relatively unexplored and mixed-methods approaches, as was used in ENABLE [29], can provide valuable insight as to how we can adapt interventions to overcome perceived barriers to exercise in prehabilitation for patients undergoing (chemo)radiotherapy. Involving individuals with lived experience in the intervention development process, such as in the HAPPY [56], SISTER [71] and PeNTAGOn [66] trials, is an important element for ongoing research to ensure patients’ needs are being addressed in a way that is acceptable to them.

Limitations

Despite an extensive literature search, it is possible that some studies may have been missed particularly as the term “prehabilitation” is not consistently used across the literature and only English language studies could be included due to limited resources. However, a significant amount of time was spent developing the search strategies and inclusion criteria, including extensive search terms, broad publication date ranges and information sources, to maximise the inclusion of relevant studies. It is outside the scope of this review to comment on the effectiveness of interventions and, as a risk of bias for studies was not performed, the results of this review are limited in terms of informing clinical guidance or policy. Nonetheless, this review captures the current landscape of prehabilitation in this population and highlights the gaps in current research.

Conclusion

This review highlights the diverse research relating to prehabilitation for gynaecological cancer patients undergoing radiotherapy. Interventions to reduce functional impairment and address adverse patient experiences are not necessarily a new concept but there is growing consideration for the complexities of managing treatment and disease-related burden, with increased involvement of those with lived experiences during study development. Studies evaluating unimodal interventions are more prevalent and there remains gaps in knowledge and literature that need to be addressed. Physical exercise interventions are still relatively unexplored in this patient population and consideration must be given to the barriers to physical activity experienced by this patient group. The physical and psychological impacts of cancer diagnosis and treatment are closely entwined; therefore, further development of multimodal prehabilitation interventions to cohesively address these is an important area for future research. Larger, randomised controlled trials will be useful for establishing efficacy of prehabilitation for gynaecological cancer patients undergoing radiotherapy although researchers must recognise that a nuanced approach is required. Complete and transparent reporting of interventions, along with greater consistency in outcome measures, will allow for a more cohesive approach to prehabilitation for this patient population and facilitate change in both practice and policy.

Supporting information

S1 Table. Search strategies for all databases (February 2024).

https://doi.org/10.1371/journal.pone.0319518.s002

(DOCX)

S2 Table. Search strategies for all databases (October 2024).

https://doi.org/10.1371/journal.pone.0319518.s003

(DOCX)

Acknowledgments

The authors would like to thank Eva Thackery and the research librarians at Lancashire Teaching Hospitals for their assistance informing the search strategy and peer review.

References

  1. 1. Lawrie TA, Green JT, Beresford M, Wedlake L, Burden S, Davidson SE, et al. Interventions to reduce acute and late adverse gastrointestinal effects of pelvic radiotherapy for primary pelvic cancers. Cochrane Database Syst Rev. 2018;1(1):CD012529. pmid:29360138.
  2. 2. Viswanathan AN, Lee LJ, Eswara JR, Horowitz NS, Konstantinopoulos PA, Mirabeau-Beale KL, et al. Complications of pelvic radiation in patients treated for gynecologic malignancies. Cancer. 2014;120(24):3870–83. pmid:25056522.
  3. 3. Biran A, Dobson C, Rees C, Brooks-Pearson R, Cunliffe A, Durrant L, et al. From pelvic radiation to social isolation: a qualitative study of survivors’ experiences of chronic bowel symptoms after pelvic radiotherapy. J Cancer Surviv. 202410.1007/s11764-023-01527–6. pmid:38182936.
  4. 4. Lind H, Waldenström A-C, Dunberger G, al-Abany M, Alevronta E, Johansson K-A, et al. Late symptoms in long-term gynaecological cancer survivors after radiation therapy: a population-based cohort study. Br J Cancer. 2011;105(6):737–45. pmid:21847122.
  5. 5. Smet S, Spampinato S, Pötter R, Jürgenliemk-Schulz IM, Nout RA, Chargari C, et al. Risk Factors for Late Persistent Fatigue After Chemoradiotherapy in Patients With Locally Advanced Cervical Cancer: An Analysis From the EMBRACE-I Study. Int J Radiat Oncol Biol Phys. 2022;112(5):1177–89. pmid:34838868.
  6. 6. Humphrey P, Dures E, Hoskin P, Cramp F. Patient Experiences of Brachytherapy for Locally Advanced Cervical Cancer: Hearing the Patient Voice Through Qualitative Interviews. Int J Radiat Oncol Biol Phys. 2024;119(3):902–11. pmid:38154511.
  7. 7. Kirchheiner K, Czajka-Pepl A, Ponocny-Seliger E, Scharbert G, Wetzel L, Nout RA, et al. Posttraumatic stress disorder after high-dose-rate brachytherapy for cervical cancer with 2 fractions in 1 application under spinal/epidural anesthesia: incidence and risk factors. Int J Radiat Oncol Biol Phys. 2014;89(2):260–7. pmid:24721589.
  8. 8. Stewart H, Ashmore L, Kragh-Furbo M, Singleton V, Hutton D. Ghosts in the machinery: Living with and beyond radiotherapy treatment for gynaecological cancer. Health (London). 2024;28(1):90–107. pmid:35900050
  9. 9. Lukez A, Baima J. The Role and Scope of Prehabilitation in Cancer Care. Semin Oncol Nurs. 2020;36(1):150976. pmid:31987643
  10. 10. Lambert J, Subar D, Gaffney C. Prehabilitation for Gastrointestinal Cancer Surgery. In: Faintuch J, Faintuch S, editors. Recent Strategies in High Risk Surgery. Cham: Springer Nature Switzerland; 2024. p. 69–80.
  11. 11. de Klerk M, van Dalen DH, Nahar-van Venrooij LMW, Meijerink WJHJ, Verdaasdonk EGG. A multimodal prehabilitation program in high-risk patients undergoing elective resection for colorectal cancer: A retrospective cohort study. Eur J Surg Oncol. 2021;47(11):2849–56. pmid:34103244
  12. 12. Molenaar CJL, Minnella EM, Coca-Martinez M, Ten Cate DWG, Regis M, Awasthi R, et al. Effect of Multimodal Prehabilitation on Reducing Postoperative Complications and Enhancing Functional Capacity Following Colorectal Cancer Surgery: The PREHAB Randomized Clinical Trial. JAMA Surg. 2023;158(6):572–81. pmid:36988937
  13. 13. Lambert JE, Hayes LD, Keegan TJ, Subar DA, Gaffney CJ. The Impact of Prehabilitation on Patient Outcomes in Hepatobiliary, Colorectal, and Upper Gastrointestinal Cancer Surgery: A PRISMA-Accordant Meta-analysis. Ann Surg. 2021;274(1):70–7. pmid:33201129.
  14. 14. Saggu RK, Barlow P, Butler J, Ghaem-Maghami S, Hughes C, Lagergren P, et al. Considerations for multimodal prehabilitation in women with gynaecological cancers: a scoping review using realist principles. BMC Womens Health. 2022;22(1):300. pmid:35854346.
  15. 15. Dhanis J, Keidan N, Blake D, Rundle S, Strijker D, van Ham M, et al. Prehabilitation to Improve Outcomes of Patients with Gynaecological Cancer: A New Window of Opportunity?. Cancers (Basel). 2022;14(14):3448. pmid:35884512.
  16. 16. Macmillan Cancer Support. Principles and guidance for prehabilitation within the management and support of people with cancer [Internet]. 2020 [cited 2024 Nov 6]. Available from: https://www.macmillan.org.uk/healthcare-professionals/news-and-resources/guides/principles-and-guidance-for-prehabilitation
  17. 17. Galica J, Saunders S, Romkey-Sinasac C, Silva A, Ethier J-L, Giroux J, et al. The needs of gynecological cancer survivors at the end of primary treatment: A scoping review and proposed model to guide clinical discussions. Patient Educ Couns. 2022;105(7):1761–82. pmid:34865888.
  18. 18. Munn Z, Pollock D, Khalil H, Alexander L, Mclnerney P, Godfrey CM, et al. What are scoping reviews? Providing a formal definition of scoping reviews as a type of evidence synthesis. JBI Evid Synth. 2022;20(4):950–2. pmid:35249995.
  19. 19. Munn Z, Peters MDJ, Stern C, Tufanaru C, McArthur A, Aromataris E. Systematic review or scoping review? Guidance for authors when choosing between a systematic or scoping review approach. BMC Med Res Methodol. 2018;18(1):143. pmid:30453902.
  20. 20. Peters M, Godfrey C, McInerey P, Munn Z, Tricco A, Khalil H. Scoping reviews. JBI Manual for Evidence Synthesis. 2020:406–51.
  21. 21. Tricco A, Lillie E, Zarin W, O’Brien K, Colquhoun H, Levac D, et al. PRISMA extension for scoping reviews (PRISMA-ScR): checklist and explanation. Annals of Internal Medicine. 2018;169(7):467–73.
  22. 22. Flores LE, Westmark D, Katz NB, Hunter TL, Silver EM, Bryan KM, et al. Prehabilitation in radiation therapy: a scoping review. Support Care Cancer. 2024;32(1):83. pmid:38177946.
  23. 23. Pollock D, Peters MDJ, Khalil H, McInerney P, Alexander L, Tricco AC, et al. Recommendations for the extraction, analysis, and presentation of results in scoping reviews. JBI Evid Synth. 2023;21(3):520–32. pmid:36081365.
  24. 24. Hoffmann TC, Glasziou PP, Boutron I, Milne R, Perera R, Moher D, et al. Better reporting of interventions: template for intervention description and replication (TIDieR) checklist and guide. BMJ. 2014;348g1687. pmid:24609605
  25. 25. Edbrooke L, Khaw P, Freimund A, Carpenter D, McNally O, Joubert L, et al. ENhAncing Lifestyle Behaviors in EndometriaL CancEr (ENABLE): A Pilot Randomized Controlled Trial. Integr Cancer Ther. 2022;2115347354211069885. pmid:35045735.
  26. 26. Andring LM, Kelsey C, Weng J, Manzar GS, Bailard N, Fellman B, et al. Baseline Characteristics of Patients Undergoing Brachytherapy for Gynecologic Cancer (GYN-BT) and the Role for an Enhanced Recovery Pathway (ERP). Int J Radiat Oncol Biol Phys. 2022;114(3):e449–50.
  27. 27. Kaliamurthi P, Karunya J, Sathyamurthy A, John NO, Ram TS, Babu A, et al. Prehabilitation in Locally Advanced Cervical Cancer Patients Receiving Radiotherapy. Int J Radiat Oncol Biol Phys. 2022;114(3):e258.
  28. 28. Chatzimavridou Grigoriadou V, Barraclough LH, Baricevic-Jones I, Bristow RG, Eden M, Haslett K, et al. RadBone: bone toxicity following pelvic radiotherapy - a prospective randomised controlled feasibility study evaluating a musculoskeletal health package in women with gynaecological cancers undergoing pelvic radiotherapy. BMJ Open. 2022;12(6):e056600. pmid:35701060.
  29. 29. Edbrooke L, Khaw P, Freimund A, Carpenter D, McNally O, Joubert L. ENABLE: ENhAncing lifestyle Behaviours in endometriaL cancEr: A pilot randomised controlled trial. Asia Pac J Clin Oncol. 2020;16(S8):108–207.
  30. 30. Edbrooke L. Australia New Zealand Clinical Trials Registry [Internet]. Sydney (NSW): NHMRC Clinical Trials Centre, University of Sydney (Australia); 2005 – Identifier ACTRN12619000631101. ENABLE: ENhAncing lifestyle Behaviours in endometriaL cancEr; 2019 Apr 29 [cited 2024 Jun 2]. Available from: https://uat.anzctr.org.au/Trial/Registration/TrialReview.aspx?id=377200
  31. 31. Andring LM, Corrigan K, Manzar G, Bailard N, Fellman B, Domingo M, et al. GPP06  Presentation Time: 8:50 AM. Brachytherapy. 2022;21(6):S19.
  32. 32. Andring L, Corrigan K, Bailard N, Rooney M, Domingo M, Varkey J, et al. PD-0414 ERAS pathway for gynecologic cancer patients receiving brachytherapy: A prospective clinical trial. Radiother Oncol. 2023;182S314–5.
  33. 33. The Christie NHS Foundation Trust (Sponsor). ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine(US); 2000 – Identifier NCT04555317. Bone Toxicity Following Pelvic Radiotherapy (RadBone); 2020 Sept 18 [cited 2024 Jun 2]. Available from: https://clinicaltrials.gov/study/NCT04555317?term=NCT04555317&rank=1&tab=table
  34. 34. Daggez M, Koyuncu EG, Kocabaş R, Yener C. Prophylactic complex physiotherapy in gynecologic cancer survivors: patient-reported outcomes based on a lymphedema questionnaire. Int J Gynecol Cancer. 2023;33(12):1928–33. pmid:37844965.
  35. 35. Sacomori C, Araya-Castro P, Diaz-Guerrero P, Ferrada I, Martínez-Varas A, Zomkowski K. Pre-rehabilitation of the pelvic floor before radiation therapy for cervical cancer: a pilot study. Int Urogynecol J. 2020;31(11):2411–8.
  36. 36. Hauth F, Gehler B, Nieß AM, Fischer K, Toepell A, Heinrich V, et al. An Activity Tracker-Guided Physical Activity Program for Patients Undergoing Radiotherapy: Protocol for a Prospective Phase III Trial (OnkoFit I and II Trials). JMIR Res Protoc. 2021;10(9):e28524. pmid:34550079
  37. 37. Tórtola-Navarro A, Martínez-García J, Cano-Martínez A, Serradilla A. Exercise during treatment for advanced cervical cancer. BMJ Support Palliat Care. 2024;13(e3):e960–3. pmid:36792345.
  38. 38. Jagdish P, Bhosale S, Gurram L, Chopra S, Daptardar A. Strengthening of pelvic floor muscles for incontinence in cervical cancer. Int J Gynecol Cancer. 2022;32A24.
  39. 39. Zou D. ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine(US); 2000 -Identifier NCT05793749. Early Intervention to Prevent Lower Limb Lymphedema of Gynecological Malignancy; 2023 Mar 31 [cited 2024 Jun 3. ]. Available from: https://clinicaltrials.gov/study/NCT05793749?term=NCT05793749&rank=1&tab=table
  40. 40. Araya-Castro P, Sacomori C, Diaz-Guerrero P, Gayán P, Román D, Sperandio F. Vaginal dilator and pelvic floor exercises for vaginal stenosis, sexual health and quality of life among cervical cancer patients treated with radiation: Clinical report. J Sex Marital Ther. 2020;46(6):513–27.
  41. 41. Gani C. ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine(US); 2000 – Identifier NCT04517019. Randomised Trial Evaluating the Benefit of a Fitness Tracker Based Workout During Radiotherapy (OnkoFit II); 2020 Aug 18 [cited 2024 Jun 1. ]. Available from: https://clinicaltrials.gov/study/NCT04517019?term=NCT04517019&rank=1&tab=table
  42. 42. Jagdish P, Daptadar A. A Study to Assess the Effect of Pelvic Floor Muscle Strengthening Exercises on Urinary Incontinence in Patients With Cervical Cancer Undergoing Radiation Therapy at a Tertiary Cancer Center [Preprint V2]. Qeios. 2024 [cited 2024 Jun 2]. Available from: https://www.qeios.com/read/87EV5F.2
  43. 43. Burt L. ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine(US). 2000 – Identifier - NCT05979610. Using Reiki Therapy to Improve Symptoms Associated With Brachytherapy in Patients With Gynecological Malignancies (Reiki-Brachy); 2023 Jul 7 [cited 2024 Jun 3. ]. Available from: https://clinicaltrials.gov/study/NCT05979610?term=NCT05979610&rank=1&tab=table
  44. 44. Jeffries SA. Sexuality and Gynaecological Cancer: A Randomised Control Trial to Evaluate the Effectiveness of a Group Psychoeducational Intervention in Improving the Sexual Health of Women with Gynaecological Cancer Treated with Radiotherapy. [Calgary]: University of Calgary; 2002.
  45. 45. de Oliveira Santana E. Registro Brasileiro de Ensaios Clinicos [Internet]. Rio de Janeiro (RJ): Instituto de Informação Científica e Tecnológica em Saúde (Brazil); 2010 - Identifier (RBR-7ssvytb). Relaxation with Guided Image for patients in treatment of radiotherapy combined to chemotherapy randomized clinical trial; 2021 Sep 8 [cited 2024 Jun 3. ]. Available from: https://ensaiosclinicos.gov.br/rg/RBR-7ssvytb
  46. 46. Schofield P. Australia New Zealand Clinical Trials Registry [Internet]. Sydney (NSW): NHMRC Clinical Trials Centre, University of Sydney (Australia); 2005 – Identifier ACTRN12609000312246. A nurse led psychosocial intervention with peer support to reduce psychosocial needs in women with gynaecological cancer; 2009 May 15 [cited 2024 Jun 4. ]. Available from: https://anzctr.org.au/Trial/Registration/TrialReview.aspx?ACTRN=12609000312246
  47. 47. Gough K, Pascoe MC, Bergin R, Drosdowsky A, Schofield P. Differential adherence to peer and nurse components of a supportive care package-The appeal of peer support may be related to women’s health and psychological status. Patient Educ Couns. 2022;105(3):762–8. pmid:34244032.
  48. 48. Schofield P. Australia New Zealand Clinical Trials Registry [Internet]. Sydney (NSW): NHMRC Clinical Trials Centre, University of Sydney (Australia); 2005 – Identifier ACTRN12611000744954. PeNTAGOn: Peer & Nurse support Trial to Assist women in Gynaecological Oncology; 2011 Jul 15 [cited 2024 Jun 4. ]. Available from: https://anzctr.org.au/Trial/Registration/TrialReview.aspx?ACTRN=ACTRN12611000744954
  49. 49. Schofield P, Juraskova I, Bergin R, Gough K, Mileshkin L, Krishnasamy M, et al. A nurse- and peer-led support program to assist women in gynaecological oncology receiving curative radiotherapy, the PeNTAGOn study (peer and nurse support trial to assist women in gynaecological oncology): study protocol for a randomised controlled trial. Trials. 2013;1439. pmid:23399476.
  50. 50. Lancellotta V, De Sanctis V, Cornacchione P, Barbera F, Fusco V, Vidali C, et al. HAPPY - Humanity Assurance Protocol in interventional radiotheraPY (brachytherapy) - an AIRO Interventional Radiotherapy Study Group project. J Contemp Brachytherapy. 2019;11(6):510–5. pmid:31969908.
  51. 51. Ramondetta LM. ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine(US). 2000 – Identifier NCT04622670. Yoga Therapy During Chemotherapy and Radiation Treatment for the Improvement of Physical and Emotional Well-Being in Patients With Stage IB2-IIIB Cervical Cancer; 2020 Nov 10 [cited 2024 Jun 4. ]. Available from: https://clinicaltrials.gov/study/NCT04622670
  52. 52. Jeffries SA, Robinson JW, Craighead PS, Keats MR. An effective group psychoeducational intervention for improving compliance with vaginal dilation: a randomized controlled trial. Int J Radiat Oncol Biol Phys. 2006;65(2):404–11. pmid:16542794.
  53. 53. Varre P, Jacobsen AM, Flovik AM, Skovlund E, Fosså SD. Regular counselling by an oncology nurse increases coping with side effects during outpatients radiotherapy of gynecological malignancies. Eur J Cancer. 1999;35S10.
  54. 54. Lutgendorf SK, Mullen-Houser E, Russell D, Degeest K, Jacobson G, Hart L, et al. Preservation of immune function in cervical cancer patients during chemoradiation using a novel integrative approach. Brain Behav Immun. 2010;24(8):1231–40. pmid:20600809.
  55. 55. Hart LK, Freel MI, Haylock PJ, Lutgendorf SK. The use of healing touch in integrative oncology. Clin J Oncol Nurs. 2011;15(5):519–25. pmid:21951738.
  56. 56. Chi G, Young A, McFarlane J, Watson M, Coleman R, Eifel P, et al. Effects of music relaxation video on pain and anxiety for women with gynaecological cancer receiving intracavitary brachytherapy: a randomised controlled trial. J Res Nurs. 2015;20(2):129–44.
  57. 57. Dey T, Mukerjee A, Rai B, Arora M, Kumar D, Srinivasa GY, et al. Early integration of palliative care in cervical cancer: Experiences from a pilot study. J Family Med Prim Care. 2023;12(2):366–70. pmid:37090997.
  58. 58. Tagliaferri L, Mancini S, Lancellotta V, Dinapoli L, Capocchiano N, Cornacchione P, et al. The Impact of HAPPY (Humanity Assurance Protocol in Interventional Radiotherapy) on the Psychological Well-being of Gynecological Cancer Patients. Curr Radiopharm. 2024;17(2):168–73. pmid:37807408.
  59. 59. Nuzhath FJ, Patil NJ, Sheela SR, Manjunath GN. A Randomized Controlled Trial on Pranayama and Yoga Nidra for Anxiety and Depression in Patients With Cervical Cancer Undergoing Standard of Care. Cureus. 2024;16(3):e55871. pmid:38595893.
  60. 60. León-Pizarro C, Gich I, Barthe E, Rovirosa A, Farrús B, Casas F, et al. A randomized trial of the effect of training in relaxation and guided imagery techniques in improving psychological and quality-of-life indices for gynecologic and breast brachytherapy patients. Psychooncology. 2007;16(11):971–9.
  61. 61. Thakur A, Agnihotri M, Kaur S, Rai B. Effectiveness of support group approach on the level of fatigue and pain in patients with cervical cancer: A quasi-experimental study. Nurs Midwifery Res J. 2021;17(2):83–91.
  62. 62. Blackburn L, Hill C, Lindsey A, Sinnott L, Thompson K, Quick A. Effect of foot reflexology and aromatherapy on anxiety and pain during brachytherapy for cervical cancer. Oncol Nurs Forum. 2021;48(3):265–76.
  63. 63. An F, Dan X, Yun A, Zhou L. Effects of mindfulness-based stress reduction on cervical cancer patients undergoing concurrent radiochemotherapy. Int J Clin Exp Med. 2020;13(7):5076–83.
  64. 64. D’cunha R, Pappachan B, D’souza O, Tonse R, Elroy Saldnha E, Baliga M. Effectiveness of Yoga Nidra in Mitigating Stress in Women Undergoing Curative Radiotherapy for Cervical Cancer. Middle East J Cancer. 2021;12(1):117–27.
  65. 65. Kaur M, Agnihotri M, Das K, Rai B, Ghai S. Effectiveness of an interventional package on the level of anxiety, depression, and fatigue among patients with cervical cancer. Asia Pac J Oncol Nurs. 2018;5(2):195–200.
  66. 66. Chi G. Music Relaxation Video and Pain Control: A Randomised Controlled Trial for Women Receiving Intracavitary Brachytherapy for Gynaecological Cancer. Denton, Texas: Texas Woman’s University; 2009.
  67. 67. Santana E de O, Silva LDS, Silva LAA da, Lemos JL de A, Marcondes L, Guimarães PRB, et al. Effect of guided imagery relaxation on anxiety in cervical cancer: randomized clinical trial. Rev Bras Enferm. 2023;76(5):e20210874. pmid:37820123.
  68. 68. Schofield P, Gough K, Pascoe M, Bergin R, White K, Mileshkin L, et al. A nurse- and peer-led psycho-educational intervention to support women with gynaecological cancers receiving curative radiotherapy: The PeNTAGOn randomised controlled trial - ANZGOG 1102. Gynecol Oncol. 2020;159(3):785–93. pmid:32962898.
  69. 69. Bergin RJ, Grogan SM, Bernshaw D, Juraskova I, Penberthy S, Mileshkin LR, et al. Developing an Evidence-Based, Nurse-Led Psychoeducational Intervention With Peer Support in Gynecologic Oncology. Cancer Nurs. 2016;39(2):E19-30. pmid:25881806.
  70. 70. Kpoghomou M, Geneau M, Menard J, Stiti M, Almont T, Ghose B, et al. Assessment of an onco-sexology support and follow-up program in cervical or vaginal cancer patients undergoing brachytherapy. Support Care Cancer. 2021;29(8):4311–8.
  71. 71. Lim KH. Effects of Integrated Music-Video Therapy on Pain and Anxiety During High-Dose-Rate Brachytherapy. Pain Manag Nurs. 2024;25(3):e223–9. pmid:38423804
  72. 72. Texier E, Meignant L. ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (US). 2000 – identifier NCT06263283. Impact of an Educational Physiotherapy-Yoga Intervention on Perceived Stress in Women Treated With Brachytherapy for Cervical Cancer (KYOCOL); 2024 Feb 2 [cited 2024 Nov 7. ]. Available from: https://clinicaltrials.gov/study/NCT06263283?term=NCT06263283&rank=1
  73. 73. Oluloro A, Comstock B, Monsell SE, Gross M, Wolff EM, Sage L, et al. Study Protocol for the Social Interventions for Support During Treatment for Endometrial Cancer and Recurrence (SISTER) study: a community engaged national randomized trial. J Comp Eff Res. 2024;13(3):e230159. pmid:38348827.
  74. 74. Chi GC, Young A, McFarlane J, Watson M, Coleman RL, Eifel P, et al. Music Relaxation Video and Pain Control: A Randomized Controlled Trial for Women Receiving Intracavitary Brachytherapy for Gynecological Cancer. Int J Radiat Oncol Biol Phys. 2011;81(2):S189.
  75. 75. Rai B. Clinical Trials Registry India [Internet]. New Delhi: The Indian Council of Medical Research (ICMR) (India). 2007 – identifier CTRI/2017/05/008704. Impact of early institution of Palliative care on Quality of Life of patients with locally advanced cancer of the uterine cervix- A prospective randomized study. - EIPAQ-CX. Clinical Trials Registry India. 2017 May 31 [cited 2024 Jun 4. ]. Available from: https://ctri.nic.in/Clinicaltrials/advancesearchmain.php
  76. 76. Lancellotta V, Dinapoli L, Mancini S, Salvati A, Cornacchione P, Colloca GF, et al. PO-1434 HAPPY protocol impact on psychological well-being in patients with gynecological cancer. Radiother Oncol. 2023;182:S1160–1.
  77. 77. Patil N. Clinical Trials Registry India [Internet]. New Delhi: The Indian Council of Medical Research (ICMR) (India). 2007 – identifier CTRI/2022/02/040423. Yoga for Cancer of the Cervix. 2022 Feb 18 [cited 2024 Jun 4. ]. Available from: https://ctri.nic.in/Clinicaltrials/advancesearchmain.php
  78. 78. Thakur A. Clinical Trials Registry India [Internet]. New Delhi: The Indian Council of Medical Research (ICMR) (India). 2007 – identifier CTRI/2019/07/020057. A study to assess effectiveness of social support group among cervical cancer patients and their caregivers. 2019 Jul 5 [cited 2024 Jun 4. ]. Available from: https://ctri.nic.in/Clinicaltrials/advancesearchmain.php
  79. 79. Kaur M. Clinical Trials Registry India [Internet]. New Delhi: The Indian Council of Medical Research (ICMR) (India). 2007 – identifier CTRI/2017/06/008732. Intervention on anxiety, depression, fatigue, treatment related knowledge and practices in cervical cancer patients. 2017 Jun 1 [cited 2024 Jun 4. ]. Available from: https://ctri.nic.in/Clinicaltrials/advancesearchmain.php
  80. 80. Ahlin R, Bergmark K, Bull C, Devarakonda S, Landberg R, Sigvardsson I, et al. A Preparatory Study for a Randomized Controlled Trial of Dietary Fiber Intake During Adult Pelvic Radiotherapy. Front Nutr. 2021;8756485. pmid:34950688.
  81. 81. Ahrén IL, Bjurberg M, Steineck G, Bergmark K, Jeppsson B. Decreasing the Adverse Effects in Pelvic Radiation Therapy: A Randomized Controlled Trial Evaluating the Use of Probiotics. Adv Radiat Oncol. 2022;8(1):101089. pmid:36483069.
  82. 82. Chitapanarux I, Chitapanarux T, Traisathit P, Kudumpee S, Tharavichitkul E, Lorvidhaya V. Randomized controlled trial of live lactobacillus acidophilus plus bifidobacterium bifidum in prophylaxis of diarrhea during radiotherapy in cervical cancer patients. Radiat Oncol. 2010;5:31. pmid:20444243.
  83. 83. Chitapanarux I, Traisathit P, Chitapanarux T, Jiratrachu R, Chottaweesak P, Chakrabandhu S, et al. Arginine, glutamine, and fish oil supplementation in cancer patients treated with concurrent chemoradiotherapy: A randomized control study. Curr Probl Cancer. 2020;44(1):100482. pmid:31146957
  84. 84. Craighead PS, Young S. Phase II study assessing the feasibility of using elemental supplements to reduce acute enteritis in patients receiving radical pelvic radiotherapy. Am J Clin Oncol. 1998;21(6):573–8. pmid:9856658.
  85. 85. De Loera Rodriguez L, Ortiz G, Rivero Moragrega P, Velázquez Brizuela I, Santoscoy Gutiérrez J, Rincón Sánchez A. Effect of symbiotic supplementation on fecal calprotectin levels and lactic acid bacteria, Bifidobacteria, Escherichia coli and Salmonella DNA in patients with cervical cancer. Nutr Hosp. 2018;35(6):1394–400.
  86. 86. Garcia-Peris P, Velasco C, Hernandez M, Lozano M, Paron L, de la Cuerda C, et al. Effect of inulin and fructo-oligosaccharide on the prevention of acute radiation enteritis in patients with gynecological cancer and impact on quality-of-life: a randomized, double-blind, placebo-controlled trial. Eur J Clin Nutr. 2016;70(2):170–4.
  87. 87. García-Peris P, Velasco C, Lozano MA, Moreno Y, Paron L, de la Cuerda C, et al. Effect of a mixture of inulin and fructo-oligosaccharide on Lactobacillus and Bifidobacterium intestinal microbiota of patients receiving radiotherapy: a randomised, double-blind, placebo-controlled trial. Nutr Hosp. 2012;27(6):1908–15. pmid:23588438.
  88. 88. Giralt J, Regadera J, Verges R, Romero J, de la Fuente I, Biete A. Effects of probiotic Lactobacillus casei DN-114 001 in prevention of radiation-induced diarrhea: Results from multicenter, randomized, placebo-controlled nutritional trial. Int J Radiat Oncol. 2008;71(4):1213–9.
  89. 89. Huang C. Chinese Clinical Trial Register [Internet]. Chengdu (Sichuan): Ministry of Health (China). 2007 – Identifier ChiCTR2100043379. The effect of selenium supplementation on the efficacy of concurrent radiotherapy for cervical cancer: a randomized, double-blind, placebo-controlled phase II clinical trial. 2021 Feb 12 [cited 2024 Jun 5. ]. Available from: https://www.chictr.org.cn/showprojEN.html?proj=121064
  90. 90. Itoh Y, Mizuno M, Ikeda M, Nakahara R, Kubota S, Ito J, et al. A randomized, double-blind pilot trial of hydrolyzed rice bran versus placebo for radioprotective effect on acute gastroenteritis secondary to chemoradiotherapy in patients with cervical cancer. Evid Based Complement Alternat Med. 2015;20151–6.
  91. 91. Li Y. ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (US). 2000 – identifier NCT05722288. Time-Restricted Eating Versus Nutritional Counseling for the Reduction of Radiation or Chemoradiation Tx Side Effects in Patients With Prostate, Cervical, or Rectal Cancers. ClinicalTrials.gov. 2023 Feb 10 [cited 2024 Jun 5. ]. Available from: https://clinicaltrials.gov/study/NCT05722288?term=NCT05722288&rank=1
  92. 92. Aredes MA, da Camara AO, de Paula NS, Chaves GV. MON-P088: Effects of Supplementation with Omega-3 on Body Composition and Functional Capacity in Patients with Cervical Cancer in Chemoradiotherapy: Preliminary Results. Clin Nutr. 2017;36S212.
  93. 93. Linn YH, Thu KK, Win NHH. Effect of Probiotics for the Prevention of Acute Radiation-Induced Diarrhoea Among Cervical Cancer Patients: a Randomized Double-Blind Placebo-Controlled Study. Probiotics Antimicrob Proteins. 2019;11(2):638–47. pmid:29550911.
  94. 94. Luvián-Morales J, Delgadillo-González M, Cetina-Pérez L, Castro-Eguiluz D. The effect of an anti-inflammatory diet on cervical cancer patients. Clin Nutr ESPEN. 2023;58504.
  95. 95. Medina-Jiménez AK, Monroy-Torres R. Repurposing Individualized Nutritional Intervention as a Therapeutic Component to Prevent the Adverse Effects of Radiotherapy in Patients With Cervical Cancer. Front Oncol. 2020;10595351. pmid:33364195.
  96. 96. Muecke R, Schomburg L, Glatzel M, Berndt-Skorka R, Baaske D, Reichl B, et al. Multicenter, phase 3 trial comparing selenium supplementation with observation in gynecologic radiation oncology. Int J Radiat Oncol Biol Phys. 2010;78(3):828–35. pmid:20133068.
  97. 97. Salminen E, Elomaa I, Minkkinen J, Vapaatalo H, Salminen S. Preservation of intestinal integrity during radiotherapy using live Lactobacillus acidophilus cultures. Clin Radiol. 1988;39(4):435–7. pmid:3141101.
  98. 98. Sasidharan B, Viswanathan P, Prasanna S, Ramadass B, Pugazhendhi S, Ramakrishna B. PV-0124: Does daily intake of resistant starch reduce the acute bowel symptoms in pelvic radiotherapy? RCT. Radiother Oncol. 2016;119(4):S56-7.
  99. 99. Sasidharan BK, Ramadass B, Viswanathan PN, Samuel P, Gowri M, Pugazhendhi S, et al. A phase 2 randomized controlled trial of oral resistant starch supplements in the prevention of acute radiation proctitis in patients treated for cervical cancer. J Cancer Res Ther. 2019;15(6):1383–91. pmid:31898677.
  100. 100. Soto-Lugo JH, Souto-Del-Bosque MA, Vázquez Martínez CA. Effectiveness of a nutritional intervention in the reduction of gastrointestinal toxicity during teletherapy in women with gynaecological tumours. Revista Médica del Hospital General de México. 2018;81(1):7–14.
  101. 101. Soto-Lugo J, Souto-Del Bosque M, Vázquez-Martínez C. Effectiveness of nutritional intervention in reduction of gastrointestinal toxicity during external beam radiotherapy in women with gynecological tumors. Gac Mex Oncol. 2017;16(2):84–90.
  102. 102. Steineck G. ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (US). 2000 – identifier NCT04534075. Dietary Fiber During Radiotherapy - a Placebo-controlled Randomized Trial (FIDURA). ClinicalTrials.gov. 2020 Sep 1 [cited 2024 Jun 5. ]. Available from: https://clinicaltrials.gov/study/NCT04534075?term=NCT04534075&rank=1
  103. 103. Aredes MA, da Camara AO, de Paula NS, Fraga KYD, do Carmo M das GT, Chaves GV. Efficacy of ω-3 supplementation on nutritional status, skeletal muscle, and chemoradiotherapy toxicity in cervical cancer patients: A randomized, triple-blind, clinical trial conducted in a middle-income country. Nutrition. 2019;67–68:110528. pmid:31445316.
  104. 104. TURNER CN. Dietary regime during radiation therapy for carcinoma of the uterus. Med J Aust. 1958;45(7):227–8. pmid:13577147.
  105. 105. Vidal‐Casariego A, Calleja‐Fernández A, de Urbina‐González J, Cano‐Rodríguez I, Cordido F, Ballesteros‐Pomar M. Efficacy of glutamine in the prevention of acute radiation enteritis. J Parenter Enteral Nutr. 2014;38(2):205–13.
  106. 106. Yang M, Pei B, Hu Q, Li X, Fang X, Huang X, et al. Effects of selenium supplementation on concurrent chemoradiotherapy in patients with cervical cancer: A randomized, double-blind, placebo-parallel controlled phase II clinical trial. Front Nutr. 2023;101094081. pmid:36819673.
  107. 107. Eustace NJ, Huang Z, Abuali T, Mercier B, Feng Q, Li C, et al. A Randomized, Phase II Clinical Trial of Time-Restricted Eating vs. Dietary Counseling to Improve Effect of Radiotherapy (TIDIER). Int J Radiat Oncol Biol Phys. 2024;120(2):e440.
  108. 108. Shuang-zheng J. ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (US). 2000 – identifier NCT06349148. The Effects of Immunonutrition Therapy on Locally Advanced Cervical Cancer Patients. ClinicalTrials.gov. 2024 April 5 [cited 2024 Nov 7. ]. Available from: https://clinicaltrials.gov/study/NCT06349148?term=NCT06349148&rank=1
  109. 109. Bye A, Kaasa S, Ose T, Sundfør K, Tropé C. The influence of low fat, low lactose diet on diarrhoea during pelvic radiotherapy. Clin Nutr. 1992;11(3):147–53. pmid:16839990.
  110. 110. Bye A, Ose T, Kaasa S. The effect of a low fat, low lactose diet on nutritional status during pelvic radiotherapy. Clin Nutr. 1993;12(2):89–95. pmid:16843293.
  111. 111. Bye A, Ose T, Kaasa S. Quality of life during pelvic radiotherapy. Acta Obstet Gynecol Scand. 1995;74(2):147–52. pmid:7900512.
  112. 112. Cetina L. ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (US). 2000 – identifier NCT03994055. Effect of an Anti-inflammatory Diet on Patients With Cervical Cancer. ClinicalTrials.gov. 2019 Jun 21 [cited 2024 Jun 5. ]. Available from: https://clinicaltrials.gov/study/NCT03994055?term=NCT03994055&rank=1
  113. 113. Chaves G. ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (US). 2000 – identifier NCT02779868. Omega-3 Supplementation in Cervix Cancer Patients Undergoing Chemoradiotherapy. ClinicalTrials.gov. 2016 May 23 [cited 2024 Jun 5. ]. Available from: https://clinicaltrials.gov/study/NCT02779868?term=NCT02779868&rank=1
  114. 114. Chen Y. Changes of immune response and side effects before and after nutritional intervention in cervical cancer patients with concurrent chemoradiotherapy. Journal of Hainan Medical University. 2018;24(13):70–3.
  115. 115. Dessources K, Aviki E, Leitao MM Jr. Lower extremity lymphedema in patients with gynecologic malignancies. Int J Gynecol Cancer. 2020;30(2):252–60. pmid:31915136.
  116. 116. Ferlay J, Ervik M, Lam F, Laversanne M, Colombet M, Mery L, et al. Global Cancer Observatory: Cancer Today – Cervix Uteri. Lyon, France: International Agency for Research on Cancer. 2024 [cited 29 Jun 2024. ]. Available from: https://gco.iarc.who.int/today
  117. 117. Lin K-Y, Frawley HC, Denehy L, Feil D, Granger CL. Exercise interventions for patients with gynaecological cancer: a systematic review and meta-analysis. Physiotherapy. 2016;102(4):309–19. pmid:27553642.
  118. 118. Arthur E, Wills C, Menon U. A systematic review of interventions for sexual well-being in women with gynecologic, anal, or rectal cancer. Oncol Nurs Forum. 2018;45(4):469–82.
  119. 119. Croisier E, Brown T, Bauer J. The Efficacy of Dietary Fiber in Managing Gastrointestinal Toxicity Symptoms in Patients with Gynecologic Cancers undergoing Pelvic Radiotherapy: A Systematic Review. J Acad Nutr Diet. 2021;121(2):261-277.e2. pmid:33127328.
  120. 120. Allenby TH, Crenshaw ML, Mathis K, Champ CE, Simone NL, Schmitz KH, et al. A systematic review of home-based dietary interventions during radiation therapy for cancer. Tech Innov Patient Support Radiat Oncol. 2020;16:10–6. pmid:32995577.
  121. 121. Duncan E, O’Cathain A, Rousseau N, Croot L, Sworn K, Turner KM, et al. Guidance for reporting intervention development studies in health research (GUIDED): an evidence-based consensus study. BMJ Open. 2020;10(4):e033516. pmid:32273313.
  122. 122. Cocks K, Wells JR, Johnson C, Schmidt H, Koller M, Oerlemans S, et al. Content validity of the EORTC quality of life questionnaire QLQ-C30 for use in cancer. Eur J Cancer. 2023;178128–38. pmid:36436330.
  123. 123. Minasian LM, O’Mara A, Mitchell SA. Clinician and Patient Reporting of Symptomatic Adverse Events in Cancer Clinical Trials: Using CTCAE and PRO-CTCAE® to Provide Two Distinct and Complementary Perspectives. Patient Relat Outcome Meas. 2022;13:249–58. pmid:36524232.
  124. 124. Hutton D, Ashmore L, Furbo MK, Stewart H, Singleton V. Patient voices: An essential piece in Radiotherapy’s data jigsaw. Radiography (Lond). 2023;29 Suppl 1S8–10. pmid:36806324.
  125. 125. Fleurent-Grégoire C, Burgess N, Denehy L, Edbrooke L, Engel D, Testa GD, et al. Outcomes reported in randomised trials of surgical prehabilitation: a scoping review. Br J Anaesth. 2024;133(1):42–57. pmid:38570300.
  126. 126. Parkington T, Myers A, Humphreys L, Moore J, Jhanji S, Copeland R. COMET database [Internet]; 2011. Development of a core outcome set for cancer prehabilitation interventions. 2023 Oct [cited 2024 Jul 2]. Available from: https://www.comet-initiative.org/Studies/Details/3098
  127. 127. Santa Mina D, van Rooijen SJ, Minnella EM, Alibhai SMH, Brahmbhatt P, Dalton SO, et al. Multiphasic Prehabilitation Across the Cancer Continuum: A Narrative Review and Conceptual Framework. Front Oncol. 2021;10598425. pmid:33505914.
  128. 128. Tanderup K, Fokdal LU, Sturdza A, Haie-Meder C, Mazeron R, van Limbergen E, et al. Effect of tumor dose, volume and overall treatment time on local control after radiochemotherapy including MRI guided brachytherapy of locally advanced cervical cancer. Radiother Oncol. 2016;120(3):441–6. pmid:27350396.
  129. 129. Pair ML, Du W, Rojas HD, Kanke JE, McGuire SE, Lee AK, et al. Dosimetric effects of weight loss or gain during volumetric modulated arc therapy and intensity-modulated radiation therapy for prostate cancer. Med Dosim. 2013;38(3):251–4. pmid:23540491.
  130. 130. Hoare E, Stavreski B, Jennings GL, Kingwell BA. Exploring Motivation and Barriers to Physical Activity among Active and Inactive Australian Adults. Sports (Basel). 2017;5(3):47. pmid:29910407.
  131. 131. Morgan O, Schnur J, Caban-Martinez AJ, Duenas-Lopez M, Huang M, Portelance L, et al. A qualitative analysis of female patient perspectives on physician communication regarding sexual dysfunction associated with pelvic radiotherapy. J Sex Med. 2023;20(6):813–20. pmid:37037772.
  132. 132. Long D, Friedrich-Nel HS, Joubert G. Patients’ informational needs while undergoing brachytherapy for cervical cancer. Int J Qual Health Care. 2016;28(2):200–8. pmid:26803540.
  133. 133. Jennings A, O’Connor L, Durand H, Finnerty M. Women’s psychosexual experiences following radical radiotherapy for gynaecological cancer: A qualitative exploration. J Psychosoc Oncol. 2023;41(3):355–71. pmid:36073853.
  134. 134. Powlesland C, MacGregor F, Swainston K. Women’s experiences of information, education and support when undergoing pelvic radiotherapy for gynaecological cancer: An exploratory qualitative study. Radiography (Lond). 2023;29(1):70–5. pmid:36327517.
  135. 135. Corish S, Fulton BA, Galbraith L, Coltart K, Duffton A. Impact of patient information format on the experience of cancer patients treated with radiotherapy. Tech Innov Patient Support Radiat Oncol. 2024;30100252. pmid:38779037.
  136. 136. Fleurent-Grégoire C, Burgess N, McIsaac DI, Chevalier S, Fiore JF Jr, Carli F, et al. Towards a common definition of surgical prehabilitation: a scoping review of randomised trials. Br J Anaesth. 2024;133(2):305–15. pmid:38677949.
  137. 137. Miralpeix E, Mancebo G, Gayete S, Corcoy M, Solé-Sedeño J-M. Role and impact of multimodal prehabilitation for gynecologic oncology patients in an Enhanced Recovery After Surgery (ERAS) program. Int J Gynecol Cancer. 2019;29(8):1235–43. pmid:31473663.